1
|
Li Q, Lin J, Luo S, Schmitz‐Abe K, Agrawal R, Meng M, Moghadaszadeh B, Beggs AH, Liu X, Perrella MA, Agrawal PB. Integrated multi-omics approach reveals the role of striated muscle preferentially expressed protein kinase in skeletal muscle including its relationship with myospryn complex. J Cachexia Sarcopenia Muscle 2024; 15:1003-1015. [PMID: 38725372 PMCID: PMC11154751 DOI: 10.1002/jcsm.13470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Autosomal-recessive mutations in SPEG (striated muscle preferentially expressed protein kinase) have been linked to centronuclear myopathy with or without dilated cardiomyopathy (CNM5). Loss of SPEG is associated with defective triad formation, abnormal excitation-contraction coupling, calcium mishandling and disruption of the focal adhesion complex in skeletal muscles. To elucidate the underlying molecular pathways, we have utilized multi-omics tools and analysis to obtain a comprehensive view of the complex biological processes and molecular functions. METHODS Skeletal muscles from 2-month-old SPEG-deficient (Speg-CKO) and wild-type (WT) mice were used for RNA sequencing (n = 4 per genotype) to profile transcriptomics and mass spectrometry (n = 4 for WT; n = 3 for Speg-CKO mice) to profile proteomics and phosphoproteomics. In addition, interactomics was performed using the SPEG antibody on pooled muscle lysates (quadriceps, gastrocnemius and triceps) from WT and Speg-CKO mice. Based on the multi-omics results, we performed quantitative real-time PCR, co-immunoprecipitation and immunoblot to verify the findings. RESULTS We identified that SPEG interacts with myospryn complex proteins CMYA5, FSD2 and RyR1, which are critical for triad formation, and that SPEG deficiency results in myospryn complex abnormalities (protein levels decreased to 22 ± 3% for CMYA5 [P < 0.05] and 18 ± 3% for FSD2 [P < 0.01]). Furthermore, SPEG phosphorylates RyR1 at S2902 (phosphorylation level decreased to 55 ± 15% at S2902 in Speg-CKO mice; P < 0.05), and its loss affects JPH2 phosphorylation at multiple sites (increased phosphorylation at T161 [1.90 ± 0.24-fold], S162 [1.61 ± 0.37-fold] and S165 [1.66 ± 0.13-fold]; decreased phosphorylation at S228 and S231 [39 ± 6%], S234 [50 ± 12%], S593 [48 ± 3%] and S613 [66 ± 10%]; P < 0.05 for S162 and P < 0.01 for other sites). On analysing the transcriptome, the most dysregulated pathways affected by SPEG deficiency included extracellular matrix-receptor interaction (P < 1e-15) and peroxisome proliferator-activated receptor signalling (P < 9e-14). CONCLUSIONS We have elucidated the critical role of SPEG in the triad as it works closely with myospryn complex proteins (CMYA5, FSD2 and RyR1), it regulates phosphorylation levels of various residues in JPH2 and S2902 in RyR1, and its deficiency is associated with dysregulation of several pathways. The study identifies unique SPEG-interacting proteins and their phosphorylation functions and emphasizes the importance of using a multi-omics approach to comprehensively evaluate the molecular function of proteins involved in various genetic disorders.
Collapse
Affiliation(s)
- Qifei Li
- Division of Neonatology, Department of PediatricsUniversity of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health SystemMiamiFLUSA
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Jasmine Lin
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Shiyu Luo
- Division of Neonatology, Department of PediatricsUniversity of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health SystemMiamiFLUSA
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Klaus Schmitz‐Abe
- Division of Neonatology, Department of PediatricsUniversity of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health SystemMiamiFLUSA
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Rohan Agrawal
- Division of Neonatology, Department of PediatricsUniversity of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health SystemMiamiFLUSA
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Melissa Meng
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Behzad Moghadaszadeh
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Alan H. Beggs
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMAUSA
- Department of Pediatric Newborn MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Mark A. Perrella
- Division of Pulmonary and Critical Care MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMAUSA
- Department of Pediatric Newborn MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Pankaj B. Agrawal
- Division of Neonatology, Department of PediatricsUniversity of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health SystemMiamiFLUSA
- Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
- The Manton Center for Orphan Disease ResearchBoston Children's Hospital, Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
2
|
Luo P, Yang J, Jian L, Dong J, Yin S, Luo C, Zhou S. Knockdown of PGBD5 inhibits the malignant progression of glioma through upregulation of the PPAR pathway. Int J Oncol 2024; 64:55. [PMID: 38577941 PMCID: PMC11015917 DOI: 10.3892/ijo.2024.5643] [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: 11/01/2023] [Accepted: 02/05/2024] [Indexed: 04/06/2024] Open
Abstract
Glioma is the most common type of primary intracranial malignant tumor, and because of its high invasiveness and recurrence, its prognosis remains poor. The present study investigated the biological function of piggyBac transportable element derived 5 (PGBD5) in glioma. Glioma and para-cancerous tissues were obtained from five patients. Reverse transcription-quantitative PCR and western blotting were used to detect the expression levels of PGBD5. Transwell assay and flow cytometry were used to evaluate cell migration, invasion, apoptosis and cell cycle distribution. In addition, a nude mouse tumor transplantation model was established to study the downstream pathways of PGBD5 and the molecular mechanism was analyzed using transcriptome sequencing. The mRNA and protein expression levels of PGBD5 were increased in glioma tissues and cells. Notably, knockdown of PGBD5 in vitro could inhibit the migration and invasion of glioma cells. In addition, the knockdown of PGBD5 expression promoted apoptosis and caused cell cycle arrest in the G2/M phase, thus inhibiting cell proliferation. Furthermore, in vivo experiments revealed that knockdown of PGBD5 expression could inhibit Ki67 expression and slow tumor growth. Changes in PGBD5 expression were also shown to be closely related to the peroxisome proliferator-activated receptor (PPAR) signaling pathway. In conclusion, interference with PGBD5 could inhibit the malignant progression of glioma through the PPAR pathway, suggesting that PGBD5 may be a potential molecular target of glioma.
Collapse
Affiliation(s)
- Pengren Luo
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Yunnan 650500, P.R. China
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan 650500, P.R. China
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Jinhong Yang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Lipeng Jian
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Yunnan 650500, P.R. China
| | - Jigen Dong
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Yunnan 650500, P.R. China
| | - Shi Yin
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Yunnan 650500, P.R. China
| | - Chao Luo
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Yunnan 650500, P.R. China
| | - Shuai Zhou
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Yunnan 650500, P.R. China
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Yunnan 650500, P.R. China
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
3
|
Firouzjaei AA, Mahmoudi A, Almahmeed W, Teng Y, Kesharwani P, Sahebkar A. Identification and analysis of the molecular targets of statins in colorectal cancer. Pathol Res Pract 2024; 256:155258. [PMID: 38522123 DOI: 10.1016/j.prp.2024.155258] [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: 01/16/2024] [Revised: 02/05/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world. According to several types of research, statins may impact the development and treatment of CRC. This work aimed to use bioinformatics to discover the relationship between statin targets and differentially expressed genes (DEGs) in CRC patients and determine the possible molecular effect of statins on CRC suppression. We used CRC datasets from the GEO database to select CRC-related DEGs. DGIdb and STITCH databases were used to identify gene targets of subtypes of statin. Further, we identified the statin target of CRC DEGs hub genes by using a Venn diagram of CRC DEGs and statin targets. Funrich and enrichr databases were carried out for the KEGG pathway and gene ontology (GO) enrichment analysis, respectively. GSE74604 and GSE10950 were used to identify CRC DEGs. After analyzing datasets,1370 genes were identified as CRC DEGs, and 345 targets were found for statins. We found that 35 genes are CRC DEGs statin targets. We found that statin targets in CRC were enriched in the receptor and metallopeptidase activity for molecular function, cytoplasm and plasma membrane for cellular component, signal transduction, and cell communication for biological process genes were substantially enriched based on FunRich enrichment. Analysis of the KEGG pathways revealed that the overexpressed DEGs were enriched in the IL-17, PPAR, and Toll-like receptor signaling pathways. Finally, CCNB1, DNMT1, AURKB, RAC1, PPARGC1A, CDKN1A, CAV1, IL1B, and HSPD1 were identified as hub CRC DEGs statin targets. The genetic and molecular aspects of our findings reveal that statins might have a therapeutic effect on CRC.
Collapse
Affiliation(s)
- Ali Ahmadizad Firouzjaei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Mahmoudi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Amirhossein Sahebkar
- Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Chávez-Ortega MP, Almanza-Pérez JC, Sánchez-Muñoz F, Hong E, Velázquez-Reyes E, Romero-Nava R, Villafaña-Rauda S, Pérez-Ontiveros A, Blancas-Flores G, Huang F. Effect of Supplementation with Omega-3 Polyunsaturated Fatty Acids on Metabolic Modulators in Skeletal Muscle of Rats with an Obesogenic High-Fat Diet. Pharmaceuticals (Basel) 2024; 17:222. [PMID: 38399437 PMCID: PMC10892617 DOI: 10.3390/ph17020222] [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: 10/26/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 02/25/2024] Open
Abstract
Previous studies provided evidence of the benefits of omega-3 polyunsaturated fatty acids (ω-3 PUFA) on the cardiovascular system and inflammation. However, its possible effect on skeletal muscle is unknown. This study aimed to evaluate whether ω-3 PUFA reverses the dysregulation of metabolic modulators in the skeletal muscle of rats on a high-fat obesogenic diet. For this purpose, an animal model was developed using male Wistar rats with a high-fat diet (HFD) and subsequently supplemented with ω-3 PUFA. Insulin resistance was assessed, and gene and protein expression of metabolism modulators in skeletal muscle was also calculated using PCR-RT and Western blot. Our results confirmed that in HFD rats, zoometric parameters and insulin resistance were increased compared to SD rats. Furthermore, we demonstrate reduced gene and protein expression of peroxisome proliferator-activated receptors (PPARs) and insulin signaling molecules. After ω-3 PUFA supplementation, we observed that glucose (24.34%), triglycerides (35.78%), and HOMA-IR (40.10%) were reduced, and QUICKI (12.16%) increased compared to HFD rats. Furthermore, in skeletal muscle, we detected increased gene and protein expression of PPAR-α, PPAR-γ, insulin receptor (INSR), insulin receptor substrate 1 (ISR-1), phosphatidylinositol-3-kinase (PI3K), and glucose transporter 4 (GLUT-4). These findings suggest that ω-3 PUFAs decrease insulin resistance of obese skeletal muscle.
Collapse
Affiliation(s)
- Mara Patricia Chávez-Ortega
- Posgrado en Biología Experimental, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 02200, Mexico;
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico;
| | - Julio Cesar Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 02200, Mexico; (J.C.A.-P.); (E.V.-R.)
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico;
| | - Enrique Hong
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico;
| | - Elihu Velázquez-Reyes
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 02200, Mexico; (J.C.A.-P.); (E.V.-R.)
| | - Rodrigo Romero-Nava
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (R.R.-N.); (S.V.-R.)
| | - Santiago Villafaña-Rauda
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (R.R.-N.); (S.V.-R.)
| | - Alfredo Pérez-Ontiveros
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico;
| | - Gerardo Blancas-Flores
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 02200, Mexico; (J.C.A.-P.); (E.V.-R.)
| | - Fengyang Huang
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico;
| |
Collapse
|
5
|
Pi A, Villivalam SD, Kang S. The Molecular Mechanisms of Fuel Utilization during Exercise. BIOLOGY 2023; 12:1450. [PMID: 37998049 PMCID: PMC10669127 DOI: 10.3390/biology12111450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
Exercise is widely recognized for its positive impact on human health and well-being. The process of utilizing substrates in skeletal muscle during exercise is intricate and governed by complex mechanisms. Carbohydrates and lipids serve as the primary fuel sources for skeletal muscle during exercise. It is now understood that fuel selection during exercise is not solely determined by physical activity itself but is also influenced by the overall metabolic state of the body. The balance between lipid and carbohydrate utilization significantly affects exercise capacity, including endurance, fatigue, and overall performance. Therefore, comprehensively understanding the regulation of substrate utilization during exercise is of utmost importance. The aim of this review is to provide an extensive overview of the current knowledge regarding the pathways involved in the regulation of substrate utilization during exercise. By synthesizing existing research, we can gain a holistic perspective on the intricate relationship between exercise, metabolism, and fuel selection. This advanced understanding has the potential to drive advancements in the field of exercise science and contribute to the development of personalized exercise strategies for individuals looking to optimize their performance and overall health.
Collapse
Affiliation(s)
| | | | - Sona Kang
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
6
|
Shi Y, Wang M, Wu L, Li X, Liao Z. COVID-19 associated liver injury: An updated review on the mechanisms and management of risk groups. LIVER RESEARCH 2023. [DOI: 10.1016/j.livres.2023.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
7
|
Dioh W, Narkar V, Singh A, Malik F, Ferrucci L, Tourette C, Mariani J, van Maanen R, Fielding RA. Novel Potential Targets for Function-Promoting Therapies: Orphan Nuclear Receptors, Anti-inflammatory Drugs, Troponin Activators, Mas Receptor Agonists, and Urolithin A. J Gerontol A Biol Sci Med Sci 2023; 78:44-52. [PMID: 37325960 PMCID: PMC10272986 DOI: 10.1093/gerona/glad072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Indexed: 06/17/2023] Open
Abstract
In recent years, several new classes of therapies have been investigated with their potential for restoring or improving physical functioning in older adults. These have included Mas receptor agonists, regulators of mitophagy, skeletal muscle troponin activators, anti-inflammatory compounds, and targets of orphan nuclear receptors. The present article summarizes recent developments of the function-promoting effects of these exciting new compounds and shares relevant preclinical and clinical data related to their safety and efficacy. The development of novel compounds in this area is expanding and likely will need the advent of a new treatment paradigm for age-associated mobility loss and disability.
Collapse
Affiliation(s)
| | - Vihang Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA
| | | | - Fady Malik
- Cytokinetics, Inc., San Francisco, California, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, U.S. Department of Health and Human Services, Baltimore, Maryland, USA
| | | | - Jean Mariani
- Biophytis, UMPC—BC9, Paris, France
- Sorbonne Université, CNRS—Institute de Biologie Paris Seine (UMR B2A), Paris, France
| | | | - Roger A Fielding
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
8
|
El-Bassouny DR, Mansour AA, Ellakkany AS, Ayuob NN, AbdElfattah AA. Can coenzyme Q10 alleviate the toxic effect of fenofibrate on skeletal muscle? Histochem Cell Biol 2023:10.1007/s00418-023-02205-5. [PMID: 37270716 PMCID: PMC10386954 DOI: 10.1007/s00418-023-02205-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/05/2023]
Abstract
Fenofibrate (FEN) is an antilipidemic drug that increases the activity of the lipoprotein lipase enzyme, thus enhancing lipolysis; however, it may cause myopathy and rhabdomyolysis in humans. Coenzyme Q10 (CoQ10) is an endogenously synthesized compound that is found in most living cells and plays an important role in cellular metabolism. It acts as the electron carrier in the mitochondrial respiratory chain. This study aimed to elucidate FEN-induced skeletal muscle changes in rats and to evaluate CoQ10 efficacy in preventing or alleviating these changes. Forty adult male rats were divided equally into four groups: the negative control group that received saline, the positive control group that received CoQ10, the FEN-treated group that received FEN, and the FEN + CoQ10 group that received both FEN followed by CoQ10 daily for 4 weeks. Animals were sacrificed and blood samples were collected to assess creatine kinase (CK). Soleus muscle samples were taken and processed for light and electron microscopic studies. This study showed that FEN increased CK levels and induced inflammatory cellular infiltration and disorganization of muscular architecture with lost striations. FEN increased the percentage of degenerated collagen fibers and immune expression of caspase-3. Ultrastructurally, FEN caused degeneration of myofibrils with distorted cell organelles. Treatment with CoQ10 could markedly ameliorate these FEN-induced structural changes and mostly regain the normal architecture of muscle fibers due to its antifibrotic and antiapoptotic effects. In conclusion, treatment with CoQ10 improved muscular structure by suppressing oxidative stress, attenuating inflammation, and inhibiting apoptosis.
Collapse
Affiliation(s)
- Dalia R El-Bassouny
- Medical Histology & Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Alyaa A Mansour
- Medical Histology & Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amany S Ellakkany
- Medical Histology & Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nasra N Ayuob
- Medical Histology Department, Faculty of Medicine, Damietta University, Damietta, Egypt
- Yousef Abdullatif Jameel Chair of Prophetic Medical Applications (YAJCPMA), Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amany A AbdElfattah
- Medical Histology & Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
- Department of Basic Medical Sciences, Faculty of Medicine, King Salman International University, South Sinai, El-Tor, Egypt.
| |
Collapse
|
9
|
Sun J, Yu L, Qu X, Huang T. The role of peroxisome proliferator-activated receptors in the tumor microenvironment, tumor cell metabolism, and anticancer therapy. Front Pharmacol 2023; 14:1184794. [PMID: 37251321 PMCID: PMC10213337 DOI: 10.3389/fphar.2023.1184794] [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: 03/12/2023] [Accepted: 05/05/2023] [Indexed: 05/31/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been extensively studied for over 3 decades and consist of three isotypes, including PPARα, γ, and β/δ, that were originally considered key metabolic regulators controlling energy homeostasis in the body. Cancer has become a leading cause of human mortality worldwide, and the role of peroxisome proliferator-activated receptors in cancer is increasingly being investigated, especially the deep molecular mechanisms and effective cancer therapies. Peroxisome proliferator-activated receptors are an important class of lipid sensors and are involved in the regulation of multiple metabolic pathways and cell fate. They can regulate cancer progression in different tissues by activating endogenous or synthetic compounds. This review emphasizes the significance and knowledge of peroxisome proliferator-activated receptors in the tumor microenvironment, tumor cell metabolism, and anti-cancer treatment by summarizing recent research on peroxisome proliferator-activated receptors. In general, peroxisome proliferator-activated receptors either promote or suppress cancer in different types of tumor microenvironments. The emergence of this difference depends on various factors, including peroxisome proliferator-activated receptor type, cancer type, and tumor stage. Simultaneously, the effect of anti-cancer therapy based on drug-targeted PPARs differs or even opposes among the three peroxisome proliferator-activated receptor homotypes and different cancer types. Therefore, the current status and challenges of the use of peroxisome proliferator-activated receptors agonists and antagonists in cancer treatment are further explored in this review.
Collapse
Affiliation(s)
- Jiaao Sun
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Liyan Yu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Xueling Qu
- Dalian Women and Children’s Medical Center(Group), Dalian, Liaoning, China
| | - Tao Huang
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
10
|
Delgadillo-Puga C, Sánchez-Castillo DR, Cariño-Cervantes YY, Torre-Villalvazo I, Tovar-Palacio C, Vásquez-Reyes S, Furuzawa-Carballeda J, Acevedo-Carabantes JA, Camacho-Corona MDR, Guzmán-Mar JL, Cisneros-Zevallos L, Tovar AR, Rebollar-Vega R, Hernández-Montes G, Ulloa-Aguirre A, Palacios-Gonzalez B, Noriega LG. Vachellia farnesiana Pods or a Polyphenolic Extract Derived from Them Exert Immunomodulatory, Metabolic, Renoprotective, and Prebiotic Effects in Mice Fed a High-Fat Diet. Int J Mol Sci 2023; 24:ijms24097984. [PMID: 37175691 PMCID: PMC10178983 DOI: 10.3390/ijms24097984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/09/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Obesity causes systemic inflammation, hepatic and renal damage, as well as gut microbiota dysbiosis. Alternative vegetable sources rich in polyphenols are known to prevent or delay the progression of metabolic abnormalities during obesity. Vachellia farnesiana (VF) is a potent source of polyphenols with antioxidant and anti-inflammatory activities with potential anti-obesity effects. We performed an in vivo preventive or an interventional experimental study in mice and in vitro experiments with different cell types. In the preventive study, male C57BL/6 mice were fed with a Control diet, a high-fat diet, or a high-fat diet containing either 0.1% methyl gallate, 10% powdered VFP, or 0.5%, 1%, or 2% of a polyphenolic extract (PE) derived from VFP (Vachellia farnesiana pods) for 14 weeks. In the intervention study, two groups of mice were fed for 14 weeks with a high-fat diet and then one switched to a high-fat diet with 10% powdered VFP for ten additional weeks. In the in vitro studies, we evaluated the effect of a VFPE (Vachellia farnesiana polyphenolic extract) on glucose-stimulated insulin secretion in INS-1E cells or of naringenin or methyl gallate on mitochondrial activity in primary hepatocytes and C2C12 myotubes. VFP or a VFPE increased whole-body energy expenditure and mitochondrial activity in skeletal muscle; prevented insulin resistance, hepatic steatosis, and kidney damage; exerted immunomodulatory effects; and reshaped fecal gut microbiota composition in mice fed a high-fat diet. VFPE decreased insulin secretion in INS-1E cells, and its isolated compounds naringenin and methyl gallate increased mitochondrial activity in primary hepatocytes and C2C12 myotubes. In conclusion VFP or a VFPE prevented systemic inflammation, insulin resistance, and hepatic and renal damage in mice fed a high-fat diet associated with increased energy expenditure, improved mitochondrial function, and reduction in insulin secretion.
Collapse
Affiliation(s)
- Claudia Delgadillo-Puga
- Departamento de Nutrición Animal Dr. Fernando Pérez-Gil Romo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | | | - Yonatan Y Cariño-Cervantes
- Departamento de Nutrición Animal Dr. Fernando Pérez-Gil Romo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Claudia Tovar-Palacio
- Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Sarai Vásquez-Reyes
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Janette Furuzawa-Carballeda
- Departamento de Cirugía Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Joshua Ayork Acevedo-Carabantes
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - María Del Rayo Camacho-Corona
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León (UANL), Av. Universidad s/n, Ciudad Universitaria, San Nicolás de Los Garza 66455, Mexico
| | - Jorge Luis Guzmán-Mar
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León (UANL), Av. Universidad s/n, Ciudad Universitaria, San Nicolás de Los Garza 66455, Mexico
| | - Luis Cisneros-Zevallos
- Department of Horticultural Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| | - Rosa Rebollar-Vega
- Red de Apoyo a la Investigación, Universidad Nacional de Autónoma de México, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Georgina Hernández-Montes
- Red de Apoyo a la Investigación, Universidad Nacional de Autónoma de México, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional de Autónoma de México, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Berenice Palacios-Gonzalez
- Unidad de Vinculación Científica Facultad de Medicina, Instituto Nacional de Medicina Genómica 14, (INMEGEN), Mexico City 16080, Mexico
| | - Lilia G Noriega
- Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City 14080, Mexico
| |
Collapse
|
11
|
Li Q, Lin J, Luo S, Schmitz-Abe K, Agrawal R, Meng M, Moghadaszadeh B, Beggs AH, Liu X, Perrella MA, Agrawal PB. Integrated multi-omics approach reveals the role of SPEG in skeletal muscle biology including its relationship with myospryn complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.538136. [PMID: 37162921 PMCID: PMC10168260 DOI: 10.1101/2023.04.24.538136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Autosomal-recessive mutations in SPEG (striated muscle preferentially expressed protein kinase) have been linked to centronuclear myopathy. Loss of SPEG is associated with defective triad formation, abnormal excitation-contraction coupling, and calcium mishandling in skeletal muscles. To elucidate the underlying molecular pathways, we have utilized multi-omics tools and analysis to obtain a comprehensive view of the complex biological processes. We identified that SPEG interacts with myospryn complex proteins (CMYA5, FSD2, RyR1), and SPEG deficiency results in myospryn complex abnormalities. In addition, transcriptional and protein profiles of SPEG-deficient muscle revealed defective mitochondrial function including aberrant accumulation of enlarged mitochondria on electron microscopy. Furthermore, SPEG regulates RyR1 phosphorylation at S2902, and its loss affects JPH2 phosphorylation at multiple sites. On analyzing the transcriptome, the most dysregulated pathways affected by SPEG deficiency included extracellular matrix-receptor interaction and peroxisome proliferator-activated receptors signaling, which may be due to defective triad and mitochondrial abnormalities. In summary, we have elucidated the critical role of SPEG in triad as it works closely with myospryn complex, phosphorylates JPH2 and RyR1, and demonstrated that its deficiency is associated with mitochondrial abnormalities. This study emphasizes the importance of using multi-omics techniques to comprehensively analyze the molecular anomalies of rare diseases. Synopsis We have previously linked mutations in SPEG (striated preferentially expressed protein) with a recessive form of centronuclear myopathy and/or dilated cardiomyopathy and have characterized a striated muscle-specific SPEG-deficient mouse model that recapitulates human disease with disruption of the triad structure and calcium homeostasis in skeletal muscles. In this study, we applied multi-omics approaches (interactomic, proteomic, phosphoproteomic, and transcriptomic analyses) in the skeletal muscles of SPEG-deficient mice to assess the underlying pathways associated with the pathological and molecular abnormalities. SPEG interacts with myospryn complex proteins (CMYA5, FSD2, RyR1), and its deficiency results in myospryn complex abnormalities.SPEG regulates RyR1 phosphorylation at S2902, and its loss affects JPH2 phosphorylation at multiple sites.SPEGα and SPEGβ have different interacting partners suggestive of differential function.Transcriptome analysis indicates dysregulated pathways of ECM-receptor interaction and peroxisome proliferator-activated receptor signaling.Mitochondrial defects on the transcriptome, proteome, and electron microscopy, may be a consequence of defective calcium signaling.
Collapse
|
12
|
Unsihuay D, Hu H, Qiu J, Latorre-Palomino A, Yang M, Yue F, Yin R, Kuang S, Laskin J. Multimodal high-resolution nano-DESI MSI and immunofluorescence imaging reveal molecular signatures of skeletal muscle fiber types. Chem Sci 2023; 14:4070-4082. [PMID: 37063787 PMCID: PMC10094364 DOI: 10.1039/d2sc06020e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
The skeletal muscle is a highly heterogeneous tissue comprised of different fiber types with varying contractile and metabolic properties. The complexity in the analysis of skeletal muscle fibers associated with their small size (30-50 μm) and mosaic-like distribution across the tissue tnecessitates the use of high-resolution imaging to differentiate between fiber types. Herein, we use a multimodal approach to characterize the chemical composition of skeletal fibers in a limb muscle, the gastrocnemius. Specifically, we combine high-resolution nanospray desorption electrospray ionization (nano-DESI) mass spectrometry imaging (MSI) with immunofluorescence (IF)-based fiber type identification. Computational image registration and segmentation approaches are used to integrate the information obtained with both techniques. Our results indicate that the transition between oxidative and glycolytic fibers is associated with shallow chemical gradients (<2.5 fold change in signals). Interestingly, we did not find any fiber type-specific molecule. We hypothesize that these findings might be linked to muscle plasticity thereby facilitating a switch in the metabolic properties of fibers in response to different conditions such as exercise and diet, among others. Despite the shallow chemical gradients, cardiolipins (CLs), acylcarnitines (CAR), monoglycerides (MGs), fatty acids, highly polyunsaturated phospholipids, and oxidized phospholipids, were identified as molecular signatures of oxidative metabolism. In contrast, histidine-related compounds were found as molecular signatures of glycolytic fibers. Additionally, the presence of highly polyunsaturated acyl chains in phospholipids was found in oxidative fibers whereas more saturated acyl chains in phospholipids were found in glycolytic fibers which suggests an effect of the membrane fluidity on the metabolic properties of skeletal myofibers.
Collapse
Affiliation(s)
- Daisy Unsihuay
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Philadelphia PA 19104 USA
| | - Hang Hu
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University West Lafayette IN 47907 USA
| | | | - Manxi Yang
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
| | - Feng Yue
- Department of Animal Sciences, Purdue University West Lafayette IN 47907 USA
| | - Ruichuan Yin
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University West Lafayette IN 47907 USA
| | - Julia Laskin
- Department of Chemistry, Purdue University West Lafayette IN 47907 USA
| |
Collapse
|
13
|
Nutraceuticals and the Network of Obesity Modulators. Nutrients 2022; 14:nu14235099. [PMID: 36501129 PMCID: PMC9739360 DOI: 10.3390/nu14235099] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Obesity is considered an increasingly widespread disease in the world population, regardless of age and gender. Genetic but also lifestyle-dependent causes have been identified. Nutrition and physical exercise play an important role, especially in non-genetic obesity. In a three-compartment model, the body is divided into fat mass, fat-free mass and water, and obesity can be considered a condition in which the percentage of total fat mass is in excess. People with a high BMI index or overweight use self-medications, such as food supplements or teas, with the aim to prevent or treat their problem. Unfortunately, there are several obesity modulators that act both on the pathways that promote adipogenesis and those that inhibit lipolysis. Moreover, these pathways involve different tissues and organs, so it is very difficult to identify anti-obesity substances. A network of factors and cells contributes to the accumulation of fat in completely different body districts. The identification of natural anti-obesity agents should consider this network, which we would like to call "obesosome". The nutrigenomic, nutrigenetic and epigenetic contribute to making the identification of active compounds very difficult. This narrative review aims to highlight nutraceuticals that, in vitro or in vivo, showed an anti-obesity activity or were found to be useful in the control of dysfunctions which are secondary to obesity. The results suggest that it is not possible to use a single compound to treat obesity, but that the studies have to be addressed towards the identification of mixtures of nutraceuticals.
Collapse
|
14
|
Ballav S, Biswas B, Sahu VK, Ranjan A, Basu S. PPAR-γ Partial Agonists in Disease-Fate Decision with Special Reference to Cancer. Cells 2022; 11:3215. [PMID: 36291082 PMCID: PMC9601205 DOI: 10.3390/cells11203215] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/03/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2023] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ) has emerged as one of the most extensively studied transcription factors since its discovery in 1990, highlighting its importance in the etiology and treatment of numerous diseases involving various types of cancer, type 2 diabetes mellitus, autoimmune, dermatological and cardiovascular disorders. Ligands are regarded as the key determinant for the tissue-specific activation of PPAR-γ. However, the mechanism governing this process is merely a contradictory debate which is yet to be systematically researched. Either these receptors get weakly activated by endogenous or natural ligands or leads to a direct over-activation process by synthetic ligands, serving as complete full agonists. Therefore, fine-tuning on the action of PPAR-γ and more subtle modulation can be a rewarding approach which might open new avenues for the treatment of several diseases. In the recent era, researchers have sought to develop safer partial PPAR-γ agonists in order to dodge the toxicity induced by full agonists, akin to a balanced activation. With a particular reference to cancer, this review concentrates on the therapeutic role of partial agonists, especially in cancer treatment. Additionally, a timely examination of their efficacy on various other disease-fate decisions has been also discussed.
Collapse
Affiliation(s)
- Sangeeta Ballav
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Bini Biswas
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Vishal Kumar Sahu
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Amit Ranjan
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Soumya Basu
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| |
Collapse
|
15
|
Molecular Mechanisms of Inflammation in Sarcopenia: Diagnosis and Therapeutic Update. Cells 2022; 11:cells11152359. [PMID: 35954203 PMCID: PMC9367570 DOI: 10.3390/cells11152359] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 01/10/2023] Open
Abstract
Sarcopenia is generally an age-related condition that directly impacts the quality of life. It is also related to chronic diseases such as metabolic dysfunction associated with diabetes and obesity. This means that everyone will be vulnerable to sarcopenia at some point in their life. Research to find the precise molecular mechanisms implicated in this condition can increase knowledge for the better prevention, diagnosis, and treatment of sarcopenia. Our work gathered the most recent research regarding inflammation in sarcopenia and new therapeutic agents proposed to target its consequences in pyroptosis and cellular senescence. Finally, we compared dual X-ray absorptiometry (DXA), magnetic resonance imaging (MRI), and ultrasound (US) as imaging techniques to diagnose and follow up on sarcopenia, indicating their respective advantages and disadvantages. Our goal is for the scientific evidence presented here to help guide future research to understand the molecular mechanisms involved in sarcopenia, new treatment strategies, and their translation into clinical practice.
Collapse
|
16
|
Tissue-Specific Variations in Transcription Factors Elucidate Complex Immune System Regulation. Genes (Basel) 2022; 13:genes13050929. [PMID: 35627314 PMCID: PMC9140347 DOI: 10.3390/genes13050929] [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: 03/22/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
Abstract
Gene expression plays a key role in health and disease. Estimating the genetic components underlying gene expression can thus help understand disease etiology. Polygenic models termed “transcriptome imputation” are used to estimate the genetic component of gene expression, but these models typically consider only the cis regions of the gene. However, these cis-based models miss large variability in expression for multiple genes. Transcription factors (TFs) that regulate gene expression are natural candidates for looking for additional sources of the missing variability. We developed a hypothesis-driven approach to identify second-tier regulation by variability in TFs. Our approach tested two models representing possible mechanisms by which variations in TFs can affect gene expression: variability in the expression of the TF and genetic variants within the TF that may affect the binding affinity of the TF to the TF-binding site. We tested our TF models in whole blood and skeletal muscle tissues and identified TF variability that can partially explain missing gene expression for 1035 genes, 76% of which explains more than the cis-based models. While the discovered regulation patterns were tissue-specific, they were both enriched for immune system functionality, elucidating complex regulation patterns. Our hypothesis-driven approach is useful for identifying tissue-specific genetic regulation patterns involving variations in TF expression or binding.
Collapse
|
17
|
Rocha GLD, Rupcic IF, Mizobuti DS, Hermes TDA, Covatti C, Silva HNMD, Araujo HN, Lourenço CCD, Silveira LDR, Pereira ECL, Minatel E. Cross-talk between TRPC-1, mTOR, PGC-1α and PPARδ in the dystrophic muscle cells treated with tempol. Free Radic Res 2022; 56:245-257. [PMID: 35549793 DOI: 10.1080/10715762.2022.2074842] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background Ca2+ dysregulation and oxidative damage appear to have a central role in Duchenne muscular dystrophy (DMD) progression. The current study provides muscle cell-specific insights into the effect of Tempol on the TRPC 1 channel; on the positive and negative regulators of muscle cell differentiation; on the antioxidant enzymatic system; on the activators of mitochondrial biogenesis; and on the inflammatory process in the dystrophic primary muscle cells in culture. METHODS Mdx myotubes were treated with Tempol (5 mM) for 24 h. Untreated mdx myotubes and C57BL/10 myotubes were used as controls. RESULTS The Trypan Blue, MTT and Live/Dead Cell assays showed that Tempol (5 mM) presented no cytotoxic effect on the dystrophic muscle cells. The Tempol treated-mdx muscle cells showed significantly lower levels in the fluorescence intensity of intracellular calcium; TRPC-1 channel; MyoD; H2O2 and O2•- production; 4-HNE levels; SOD2, CAT and GPx levels; and TNF levels. On the other hand, SOD, CAT and GR mRNA relative expression were significantly higher in Tempol treated-mdx muscle cells. In addition, higher levels of Myogenin, MHC-Slow, mTOR, PGC-1α and PPARδ were also observed in Tempol treated-mdx muscle cells. CONCLUSION Our findings demonstrated that Tempol decreased intracellular calcium and oxidative stress in primary dystrophic muscle cells, promoting a cross-talk between TRPC-1, mTOR, PGC-1α and PPARδ.
Collapse
Affiliation(s)
- Guilherme Luiz da Rocha
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Ian Feller Rupcic
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Daniela Sayuri Mizobuti
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Túlio de Almeida Hermes
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Caroline Covatti
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | | | - Hygor Nunes Araujo
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Caroline Caramano de Lourenço
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Leonardo Dos Reis Silveira
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Elaine Cristina Leite Pereira
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil.,Universidade de Brasília (UnB), Faculdade de Ceilândia, Brasília, Brazil
| | - Elaine Minatel
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
18
|
Ahmad SS, Ahmad K, Shaikh S, You HJ, Lee EY, Ali S, Lee EJ, Choi I. Molecular Mechanisms and Current Treatment Options for Cancer Cachexia. Cancers (Basel) 2022; 14:cancers14092107. [PMID: 35565236 PMCID: PMC9105812 DOI: 10.3390/cancers14092107] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The primary characteristics of cancer cachexia are weakness, weight loss, atrophy, fat reduction, and systemic inflammation. Cachexia is strongly associated with cancers involving the lungs, pancreas, esophagus, stomach, and liver, which account for half of all cancer deaths. TGF-β, MSTN, activin, IGF-1/PI3K/AKT, and JAK-STAT signaling pathways are known to underlie muscle atrophy and cachexia. Anamorelin (appetite stimulation), megestrol acetate, eicosapentaenoic acid, phytocannabinoids, targeting MSTN/activin, and molecules targeting proinflammatory cytokines, such as TNF-α and IL-6, are being tested as treatment options for cancer cachexia. Abstract Cancer cachexia is a condition marked by functional, metabolic, and immunological dysfunctions associated with skeletal muscle (SM) atrophy, adipose tissue loss, fat reduction, systemic inflammation, and anorexia. Generally, the condition is caused by a variety of mediators produced by cancer cells and cells in tumor microenvironments. Myostatin and activin signaling, IGF-1/PI3K/AKT signaling, and JAK-STAT signaling are known to play roles in cachexia, and thus, these pathways are considered potential therapeutic targets. This review discusses the current state of knowledge of the molecular mechanisms underlying cachexia and the available therapeutic options and was undertaken to increase understanding of the various factors/pathways/mediators involved and to identify potential treatment options.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
| | - Hye Jin You
- Tumor Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Gyeonggi-do, Korea; (H.J.Y.); (E.-Y.L.)
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, 323, Ilsan-ro, Ilsandong-gu, Goyaan 10408, Gyeonggi-do, Korea
| | - Eun-Young Lee
- Tumor Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Gyeonggi-do, Korea; (H.J.Y.); (E.-Y.L.)
| | - Shahid Ali
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
- Correspondence: (E.J.L.); (I.C.)
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
- Correspondence: (E.J.L.); (I.C.)
| |
Collapse
|
19
|
Wang T, Xiao Y, Hu Z, Gu J, Hua R, Hai Z, Chen X, Zhang JV, Yu Z, Wu T, Yeung WSB, Liu K, Guo C. MFN2 Deficiency Impairs Mitochondrial Functions and PPAR Pathway During Spermatogenesis and Meiosis in Mice. Front Cell Dev Biol 2022; 10:862506. [PMID: 35493072 PMCID: PMC9046932 DOI: 10.3389/fcell.2022.862506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondria are highly dynamic organelles and their activity is known to be regulated by changes in morphology via fusion and fission events. However, the role of mitochondrial dynamics on cellular differentiation remains largely unknown. Here, we explored the molecular mechanism of mitochondrial fusion during spermatogenesis by generating an Mfn2 (mitofusin 2) conditional knock-out (cKO) mouse model. We found that depletion of MFN2 in male germ cells led to disrupted spermatogenesis and meiosis during which the majority of Mfn2 cKO spermatocytes did not develop to the pachytene stage. We showed that in these Mfn2 cKO spermatocytes, oxidative phosphorylation in the mitochondria was affected. In addition, RNA-Seq analysis showed that there was a significantly altered transcriptome profile in the Mfn2 deficient pachytene (or pachytene-like) spermatocytes, with a total of 262 genes up-regulated and 728 genes down-regulated, compared with wild-type (control) mice. Pathway enrichment analysis indicated that the peroxisome proliferator-activated receptor (PPAR) pathway was altered, and subsequent more detailed analysis showed that the expression of PPAR α and PPAR γ was up-regulated and down-regulated, respectively, in the MFN2 deficient pachytene (or pachytene-like) spermatocytes. We also demonstrated that there were more lipid droplets in the Mfn2 cKO cells than in the control cells. In conclusion, our study demonstrates a novel finding that MFN2 deficiency negatively affects mitochondrial functions and alters PPAR pathway together with lipid metabolism during spermatogenesis and meiosis.
Collapse
Affiliation(s)
- Tianren Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- *Correspondence: Tianren Wang, ; Chenxi Guo,
| | - Yuan Xiao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhe Hu
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jingkai Gu
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Renwu Hua
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhuo Hai
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xueli Chen
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jian V. Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhiying Yu
- Department of Gynecology, Shenzhen Second People’s Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Ting Wu
- Department of Gynecology and Obstetrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kui Liu
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chenxi Guo
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Tianren Wang, ; Chenxi Guo,
| |
Collapse
|
20
|
Tavasoli M, Lahire S, Sokolenko S, Novorolsky R, Reid SA, Lefsay A, Otley MOC, Uaesoontrachoon K, Rowsell J, Srinivassane S, Praest M, MacKinnon A, Mammoliti MS, Maloney AA, Moraca M, Pedro Fernandez-Murray J, McKenna M, Sinal CJ, Nagaraju K, Robertson GS, Hoffman EP, McMaster CR. Mechanism of action and therapeutic route for a muscular dystrophy caused by a genetic defect in lipid metabolism. Nat Commun 2022; 13:1559. [PMID: 35322809 PMCID: PMC8943011 DOI: 10.1038/s41467-022-29270-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 02/23/2022] [Indexed: 12/01/2022] Open
Abstract
CHKB encodes one of two mammalian choline kinase enzymes that catalyze the first step in the synthesis of the membrane phospholipid phosphatidylcholine. In humans and mice, inactivation of the CHKB gene (Chkb in mice) causes a recessive rostral-to-caudal muscular dystrophy. Using Chkb knockout mice, we reveal that at no stage of the disease is phosphatidylcholine level significantly altered. We observe that in affected muscle a temporal change in lipid metabolism occurs with an initial inability to utilize fatty acids for energy via mitochondrial β-oxidation resulting in shunting of fatty acids into triacyglycerol as the disease progresses. There is a decrease in peroxisome proliferator-activated receptors and target gene expression specific to Chkb−/− affected muscle. Treatment of Chkb−/− myocytes with peroxisome proliferator-activated receptor agonists enables fatty acids to be used for β-oxidation and prevents triacyglyerol accumulation, while simultaneously increasing expression of the compensatory choline kinase alpha (Chka) isoform, preventing muscle cell injury. Mutations in the CHKB gene cause muscular dystrophy. Here, the authors show that in mouse models of the disease changes in lipid metabolism are associated with decreased PPAR signaling, and show PPAR agonists can rescue expression of injury markers in myocytes in vitro.
Collapse
Affiliation(s)
- Mahtab Tavasoli
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Sarah Lahire
- University of Reims Champagne-Ardenne, Reims, France
| | - Stanislav Sokolenko
- Department of Process Engineering & Applied Science, Dalhousie University, Halifax, NS, Canada
| | - Robyn Novorolsky
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Sarah Anne Reid
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Abir Lefsay
- Mass Spectrometry Core Facility, Dalhousie University, Halifax, NS, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | - Kanneboyina Nagaraju
- Agada Biosciences Inc., Halifax, NS, Canada.,School of Pharmacy and Pharmaceutical Sciences, Binghamton University, State University of New York (SUNY), Binghamton, NY, USA
| | - George S Robertson
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada.,Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Eric P Hoffman
- Agada Biosciences Inc., Halifax, NS, Canada.,School of Pharmacy and Pharmaceutical Sciences, Binghamton University, State University of New York (SUNY), Binghamton, NY, USA
| | | |
Collapse
|
21
|
Davoli R, Vegni J, Cesarani A, Dimauro C, Zappaterra M, Zambonelli P. Identification of differentially expressed genes in early-postmortem Semimembranosus muscle of Italian Large White heavy pigs divergent for glycolytic potential. Meat Sci 2022; 187:108754. [DOI: 10.1016/j.meatsci.2022.108754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/18/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
|
22
|
Valentine WJ, Mostafa SA, Tokuoka SM, Hamano F, Inagaki NF, Nordin JZ, Motohashi N, Kita Y, Aoki Y, Shimizu T, Shindou H. Lipidomic Analyses Reveal Specific Alterations of Phosphatidylcholine in Dystrophic Mdx Muscle. Front Physiol 2022; 12:698166. [PMID: 35095541 PMCID: PMC8791236 DOI: 10.3389/fphys.2021.698166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), lack of dystrophin increases the permeability of myofiber plasma membranes to ions and larger macromolecules, disrupting calcium signaling and leading to progressive muscle wasting. Although the biological origin and meaning are unclear, alterations of phosphatidylcholine (PC) are reported in affected skeletal muscles of patients with DMD that may include higher levels of fatty acid (FA) 18:1 chains and lower levels of FA 18:2 chains, possibly reflected in relatively high levels of PC 34:1 (with 16:0_18:1 chain sets) and low levels of PC 34:2 (with 16:0_18:2 chain sets). Similar PC alterations have been reported to occur in the mdx mouse model of DMD. However, altered ratios of PC 34:1 to PC 34:2 have been variably reported, and we also observed that PC 34:2 levels were nearly equally elevated as PC 34:1 in the affected mdx muscles. We hypothesized that experimental factors that often varied between studies; including muscle types sampled, mouse ages, and mouse diets; may strongly impact the PC alterations detected in dystrophic muscle of mdx mice, especially the PC 34:1 to PC 34:2 ratios. In order to test our hypothesis, we performed comprehensive lipidomic analyses of PC and phosphatidylethanolamine (PE) in several muscles (extensor digitorum longus, gastrocnemius, and soleus) and determined the mdx-specific alterations. The alterations in PC 34:1 and PC 34:2 were closely monitored from the neonate period to the adult, and also in mice raised on several diets that varied in their fats. PC 34:1 was naturally high in neonate’s muscle and decreased until age ∼3-weeks (disease onset age), and thereafter remained low in WT muscles but was higher in regenerated mdx muscles. Among the muscle types, soleus showed a distinctive phospholipid pattern with early and diminished mdx alterations. Diet was a major factor to impact PC 34:1/PC 34:2 ratios because mdx-specific alterations of PC 34:2 but not PC 34:1 were strictly dependent on diet. Our study identifies high PC 34:1 as a consistent biochemical feature of regenerated mdx-muscle and indicates nutritional approaches are also effective to modify the phospholipid compositions.
Collapse
Affiliation(s)
- William J. Valentine
- Department of Molecular Therapy, National Center for Neurology and Psychiatry (NCNP), National Institute of Neuroscience, Kodaira, Tokyo, Japan
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
- *Correspondence: William J. Valentine,
| | - Sherif A. Mostafa
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
- Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Suzumi M. Tokuoka
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Fumie Hamano
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
- Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Natsuko F. Inagaki
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
| | - Joel Z. Nordin
- Department of Molecular Therapy, National Center for Neurology and Psychiatry (NCNP), National Institute of Neuroscience, Kodaira, Tokyo, Japan
- Department of Laboratory Medicine, Centre for Biomolecular and Cellular Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Norio Motohashi
- Department of Molecular Therapy, National Center for Neurology and Psychiatry (NCNP), National Institute of Neuroscience, Kodaira, Tokyo, Japan
| | - Yoshihiro Kita
- Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Center for Neurology and Psychiatry (NCNP), National Institute of Neuroscience, Kodaira, Tokyo, Japan
- Yoshitsugu Aoki,
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
- Hideo Shindou,
| |
Collapse
|
23
|
Yang Q, Chan P. Skeletal Muscle Metabolic Alternation Develops Sarcopenia. Aging Dis 2022; 13:801-814. [PMID: 35656108 PMCID: PMC9116905 DOI: 10.14336/ad.2021.1107] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/28/2021] [Indexed: 11/23/2022] Open
Abstract
Sarcopenia is a new type of senile syndrome with progressive skeletal muscle mass loss with age, accompanied by decreased muscle strength and/or muscle function. Sarcopenia poses a serious threat to the health of the elderly and increases the burden of family and society. The underlying pathophysiological mechanisms of sarcopenia are still unclear. Recent studies have shown that changes of skeletal muscle metabolism are the risk factors for sarcopenia. Furthermore, the importance of the skeletal muscle metabolic microenvironment in regulating satellite cells (SCs) is gaining significant attention. Skeletal muscle metabolism has intrinsic relationship with the regulation of skeletal muscle mass and regeneration. This review is to discuss recent findings regarding skeletal muscle metabolic alternation and the development of sarcopenia, hoping to contribute better understanding and treatment of sarcopenia.
Collapse
Affiliation(s)
- Qiumei Yang
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Piu Chan
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China.
- Clinical Center for Parkinson’s Disease, Capital Medical University, Beijing Institute of Geriatrics, Beijing, China.
- Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson’s Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
- Correspondence should be addressed to: Dr. Piu Chan, Department of Neurobiology, Xuanwu Hospital of Capital Medical University, 45 Changchun Road, Beijing 100053, China. .
| |
Collapse
|
24
|
Liao Z, Yeo HL, Wong SW, Zhao Y. Cellular Senescence: Mechanisms and Therapeutic Potential. Biomedicines 2021; 9:1769. [PMID: 34944585 PMCID: PMC8698401 DOI: 10.3390/biomedicines9121769] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Cellular senescence is a complex and multistep biological process which cells can undergo in response to different stresses. Referring to a highly stable cell cycle arrest, cellular senescence can influence a multitude of biological processes-both physiologically and pathologically. While phenotypically diverse, characteristics of senescence include the expression of the senescence-associated secretory phenotype, cell cycle arrest factors, senescence-associated β-galactosidase, morphogenesis, and chromatin remodelling. Persistent senescence is associated with pathologies such as aging, while transient senescence is associated with beneficial programmes, such as limb patterning. With these implications, senescence-based translational studies, namely senotherapy and pro-senescence therapy, are well underway to find the cure to complicated diseases such as cancer and atherosclerosis. Being a subject of major interest only in the recent decades, much remains to be studied, such as regarding the identification of unique biomarkers of senescent cells. This review attempts to provide a comprehensive understanding of the diverse literature on senescence, and discuss the knowledge we have on senescence thus far.
Collapse
Affiliation(s)
- Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden
| | - Han Lin Yeo
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| | - Siaw Wen Wong
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore;
| | - Yan Zhao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| |
Collapse
|
25
|
Pohl A, Schünemann F, Bersiner K, Gehlert S. The Impact of Vegan and Vegetarian Diets on Physical Performance and Molecular Signaling in Skeletal Muscle. Nutrients 2021; 13:3884. [PMID: 34836139 PMCID: PMC8623732 DOI: 10.3390/nu13113884] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Muscular adaptations can be triggered by exercise and diet. As vegan and vegetarian diets differ in nutrient composition compared to an omnivorous diet, a change in dietary regimen might alter physiological responses to physical exercise and influence physical performance. Mitochondria abundance, muscle capillary density, hemoglobin concentration, endothelial function, functional heart morphology and availability of carbohydrates affect endurance performance and can be influenced by diet. Based on these factors, a vegan and vegetarian diet possesses potentially advantageous properties for endurance performance. Properties of the contractile elements, muscle protein synthesis, the neuromuscular system and phosphagen availability affect strength performance and can also be influenced by diet. However, a vegan and vegetarian diet possesses potentially disadvantageous properties for strength performance. Current research has failed to demonstrate consistent differences of performance between diets but a trend towards improved performance after vegetarian and vegan diets for both endurance and strength exercise has been shown. Importantly, diet alters molecular signaling via leucine, creatine, DHA and EPA that directly modulates skeletal muscle adaptation. By changing the gut microbiome, diet can modulate signaling through the production of SFCA.
Collapse
Affiliation(s)
- Alexander Pohl
- Department of Biosciences of Sport Science, Institute of Sport Science, University of Hildesheim, 31141 Hildesheim, Germany; (F.S.); (K.B.); (S.G.)
| | - Frederik Schünemann
- Department of Biosciences of Sport Science, Institute of Sport Science, University of Hildesheim, 31141 Hildesheim, Germany; (F.S.); (K.B.); (S.G.)
| | - Käthe Bersiner
- Department of Biosciences of Sport Science, Institute of Sport Science, University of Hildesheim, 31141 Hildesheim, Germany; (F.S.); (K.B.); (S.G.)
| | - Sebastian Gehlert
- Department of Biosciences of Sport Science, Institute of Sport Science, University of Hildesheim, 31141 Hildesheim, Germany; (F.S.); (K.B.); (S.G.)
- Department for Molecular and Cellular Sports Medicine, German Sports University Cologne, 50933 Cologne, Germany
| |
Collapse
|
26
|
Chi T, Wang M, Wang X, Yang K, Xie F, Liao Z, Wei P. PPAR-γ Modulators as Current and Potential Cancer Treatments. Front Oncol 2021; 11:737776. [PMID: 34631571 PMCID: PMC8495261 DOI: 10.3389/fonc.2021.737776] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Worldwide, cancer has become one of the leading causes of mortality. Peroxisome Proliferator-Activated Receptors (PPARs) is a family of critical sensors of lipids as well as regulators of diverse metabolic pathways. They are also equipped with the capability to promote eNOS activation, regulate immunity and inflammation response. Aside from the established properties, emerging discoveries are also made in PPAR's functions in the cancer field. All considerations are given, there exists great potential in PPAR modulators which may hold in the management of cancers. In particular, PPAR-γ, the most expressed subtype in adipose tissues with two isoforms of different tissue distribution, has been proven to be able to inhibit cell proliferation, induce cell cycle termination and apoptosis of multiple cancer cells, promote intercellular adhesion, and cripple the inflamed state of tumor microenvironment, both on transcriptional and protein level. However, despite the multi-functionalities, the safety of PPAR-γ modulators is still of clinical concern in terms of dosage, drug interactions, cancer types and stages, etc. This review aims to consolidate the functions of PPAR-γ, the current and potential applications of PPAR-γ modulators, and the challenges in applying PPAR-γ modulators to cancer treatment, in both laboratory and clinical settings. We sincerely hope to provide a comprehensive perspective on the prospect of PPAR-γ applicability in the field of cancer treatment.
Collapse
Affiliation(s)
- Tiange Chi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Mina Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Department of Acupuncture and Moxibustion, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xu Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ke Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Feiyu Xie
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Oncology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Peng Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
27
|
The Regulatory Roles of PPARs in Skeletal Muscle Fuel Metabolism and Inflammation: Impact of PPAR Agonism on Muscle in Chronic Disease, Contraction and Sepsis. Int J Mol Sci 2021; 22:ijms22189775. [PMID: 34575939 PMCID: PMC8465345 DOI: 10.3390/ijms22189775] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
The peroxisome proliferator-activated receptor (PPAR) family of transcription factors has been demonstrated to play critical roles in regulating fuel selection, energy expenditure and inflammation in skeletal muscle and other tissues. Activation of PPARs, through endogenous fatty acids and fatty acid metabolites or synthetic compounds, has been demonstrated to have lipid-lowering and anti-diabetic actions. This review will aim to provide a comprehensive overview of the functions of PPARs in energy homeostasis, with a focus on the impacts of PPAR agonism on muscle metabolism and function. The dysregulation of energy homeostasis in skeletal muscle is a frequent underlying characteristic of inflammation-related conditions such as sepsis. However, the potential benefits of PPAR agonism on skeletal muscle protein and fuel metabolism under these conditions remains under-investigated and is an area of research opportunity. Thus, the effects of PPARγ agonism on muscle inflammation and protein and carbohydrate metabolism will be highlighted, particularly with its potential relevance in sepsis-related metabolic dysfunction. The impact of PPARδ agonism on muscle mitochondrial function, substrate metabolism and contractile function will also be described.
Collapse
|
28
|
Ahn J, Son HJ, Seo HD, Ha TY, Ahn J, Lee H, Shin SH, Jung CH, Jang YJ. γ-Oryzanol Improves Exercise Endurance and Muscle Strength by Upregulating PPARδ and ERRγ Activity in Aged Mice. Mol Nutr Food Res 2021; 65:e2000652. [PMID: 33932312 DOI: 10.1002/mnfr.202000652] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/05/2021] [Indexed: 12/24/2022]
Abstract
SCOPE γ-Oryzanol, a well-known antioxidant, has been used by body builders and athletes to boost strength and increase muscle gain, without major side effects. However, the effect of γ-Oryzanol on sarcopenia and the underlying molecular mechanism is poorly understood. RESULTS Aged mice fed with the γ-Oryzanol diet do not show significant changes in muscle weight, but show increased running endurance as well as improved grip strength. The expression and activity of PPARδ and ERRγ are increased in skeletal muscle of γ-Oryzanol supplemented mice. γ-Oryzanol upregulates oxidative muscle fibers by MEF2 transcription factor, and PGC-1α and ERRα expressions. Fatty acid oxidation related genes and mitochondria biogenesis are upregulated by γ-Oryzanol. In addition, γ-Oryzanol inhibits TGF-β-Smad-NADPH oxidase 4 pathway and inflammatory cytokines such as TNF-α, IL-1β, IL-6, and p65 NF-κB subunit, which cause skeletal muscle weakness. Collectively, γ-Oryzanol attenuates muscle weakness pathway and increases oxidative capacity by increasing PPARδ and ERRγ activity, which contributes to enhance strength and improve oxidative capacity in muscles, consequently enhancing exercise capacity in aged mice. Particularly, γ-Oryzanol directly binds to PPARδ. CONCLUSIONS These are the first findings showing that γ-Oryzanol enhances skeletal muscle function in aged mice by regulating PPARδ and ERRγ activity without muscle gain.
Collapse
Affiliation(s)
- Jisong Ahn
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Department of Food Science and Technology, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - Hyo Jeong Son
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
| | - Hyo Deok Seo
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
| | - Tae Youl Ha
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jiyun Ahn
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Hyunjung Lee
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
| | - Seung Ho Shin
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828, Republic of Korea
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Chang Hwa Jung
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Young Jin Jang
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Major of Food Science & Technology, Seoul Women's University, Seoul, 01797, Republic of Korea
| |
Collapse
|
29
|
Camps J, Breuls N, Sifrim A, Giarratana N, Corvelyn M, Danti L, Grosemans H, Vanuytven S, Thiry I, Belicchi M, Meregalli M, Platko K, MacDonald ME, Austin RC, Gijsbers R, Cossu G, Torrente Y, Voet T, Sampaolesi M. Interstitial Cell Remodeling Promotes Aberrant Adipogenesis in Dystrophic Muscles. Cell Rep 2021; 31:107597. [PMID: 32375047 DOI: 10.1016/j.celrep.2020.107597] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 03/06/2020] [Accepted: 04/10/2020] [Indexed: 12/26/2022] Open
Abstract
Fibrosis and fat replacement in skeletal muscle are major complications that lead to a loss of mobility in chronic muscle disorders, such as muscular dystrophy. However, the in vivo properties of adipogenic stem and precursor cells remain unclear, mainly due to the high cell heterogeneity in skeletal muscles. Here, we use single-cell RNA sequencing to decomplexify interstitial cell populations in healthy and dystrophic skeletal muscles. We identify an interstitial CD142-positive cell population in mice and humans that is responsible for the inhibition of adipogenesis through GDF10 secretion. Furthermore, we show that the interstitial cell composition is completely altered in muscular dystrophy, with a near absence of CD142-positive cells. The identification of these adipo-regulatory cells in the skeletal muscle aids our understanding of the aberrant fat deposition in muscular dystrophy, paving the way for treatments that could counteract degeneration in patients with muscular dystrophy.
Collapse
Affiliation(s)
- Jordi Camps
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium; Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Natacha Breuls
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Alejandro Sifrim
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; Wellcome Genome Campus, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Nefele Giarratana
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Marlies Corvelyn
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Laura Danti
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Hanne Grosemans
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Sebastiaan Vanuytven
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Irina Thiry
- Laboratory for Molecular Virology and Gene Therapy, and Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
| | - Marzia Belicchi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122 Milan, Italy
| | - Mirella Meregalli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122 Milan, Italy
| | - Khrystyna Platko
- Department of Medicine, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada
| | - Melissa E MacDonald
- Department of Medicine, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada
| | - Richard C Austin
- Department of Medicine, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada
| | - Rik Gijsbers
- Laboratory for Molecular Virology and Gene Therapy, and Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Yvan Torrente
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122 Milan, Italy
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; Wellcome Genome Campus, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Maurilio Sampaolesi
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium; Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
30
|
da Silva HNM, Covatti C, da Rocha GL, Mizobuti DS, Mâncio RD, Hermes TDA, Kido LA, Cagnon VHA, Pereira ECL, Minatel E. Oxidative Stress, Inflammation, and Activators of Mitochondrial Biogenesis: Tempol Targets in the Diaphragm Muscle of Exercise Trained- mdx Mice. Front Physiol 2021; 12:649793. [PMID: 33981250 PMCID: PMC8107395 DOI: 10.3389/fphys.2021.649793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022] Open
Abstract
The mdx mouse phenotype aggravated by chronic exercise on a treadmill makes this murine model more reliable for the study of muscular dystrophy. Thus, to better assess the Tempol effect on dystrophic pathways, the analyses in this study were performed in the blood samples and diaphragm muscle from treadmill trained adult (7–11-weeks old) mdx animals. The mdx mice were divided into three groups: mdxSed, sedentary controls (n = 28); mdxEx, exercise-trained animals (n = 28); and mdxEx+T, exercise-trained animals with the Tempol treatment (n = 28). The results demonstrated that the Tempol treatment promoted muscle strength gain, prevented muscle damage, reduced the inflammatory process, oxidative stress, and angiogenesis regulator, and up regulated the activators of mitochondrial biogenesis. The main new findings of this study are that Tempol reduced the NF-κB and increased the PGC1-α and PPARδ levels in the exercise-trained-mdx mice, which are probably related to the ability of this antioxidant to scavenge excessive ROS. These results reinforce the use of Tempol as a potential therapeutic strategy in DMD.
Collapse
Affiliation(s)
| | - Caroline Covatti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Guilherme Luiz da Rocha
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Rafael Dias Mâncio
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Larissa Akemi Kido
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Valéria Helena Alves Cagnon
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Elaine Cristina Leite Pereira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil.,Faculty of Ceilândia, University of Brasília (UnB), Brasília, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
31
|
Hatami Marbini M, Amiri F, Sajadi Hezaveh Z. Dietary glycemic index, glycemic load, insulin index, insulin load and risk of diabetes-related cancers: A systematic review of cohort studies. Clin Nutr ESPEN 2021; 42:22-31. [PMID: 33745582 DOI: 10.1016/j.clnesp.2021.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/05/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND & AIMS It is believed that diets high in glycemic index (GI), glycemic load (GL), Insulin index (II), and Insulin load (IL) are associated with the increased risks of certain cancers through increasing serum glucose or insulin levels. METHODS We conducted this systematic review of cohort studies to evaluate the possible relation between GI, GL, II, and IL with diabetes-related cancers, including colorectal, bladder, breast, endometrium, liver, pancreas, and prostate cancers. Two separate investigators conducted a literature search through PubMed/Medline, Scopus, and Web of Science databases up to February 2020, plus reference lists of relevant articles. RESULTS Fifty-three cohort studies with a total of 100 098 cancer cases were included in this systematic review. Fifteen out of eighteen studies among breast cancer cases reported no significant association between GI/GL and cancer risk. These numbers were 4 out of 13 for colorectal cancer, 7 out of 9 for endometrial cancer, 2 out of 3 for liver cancer, 8 out of 10 for pancreatic cancer, and 3 out of 3 for prostate cancer. Only one cohort investigated this association in terms of bladder cancer and reported a significant association. Also, five studies reported this relation in terms of II/IL, and only one cohort among endometrial cancer patients observed a significant positive association between the risk of cancer and IL. CONCLUSION We concluded a weak association between dietary GI/GL and no association between II/IL with diabetes-related cancer risk. More cohort studies are required to be performed regarding II/IL and the risk of cancer.
Collapse
Affiliation(s)
- Motahare Hatami Marbini
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemehsadat Amiri
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| | - Zohreh Sajadi Hezaveh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
33
|
Moustafa EM, Rashed ER, Rashed RR. Pterostilbene Inhibits Dyslipidemia-Induced Activation of Progenitor Adipose Gene Under High-Fat Diet and Radiation Stressor. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211001267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Repeated exposure to ionizing radiation has been reported to increase the risk of chronic metabolic disorders such as systemic hyperlipidemia and intracellular lipid accumulation that might lead to diabetes-induced heart disease. The purpose of this study was to investigate the effect of pterostilbene on high-fat diet rats suffering from ionizing radiation-induced hyperlipidemia. High-fat diet rats showed an increase in body weight and body fat compared with rats fed with normal chow. Pterostilbene and Orlistat treatments resulted in lower body weight and body fat gain, insulin resistance, reduced lipid peroxidation with attenuated liver enzyme levels, and regulated lipogenesis-related genes in the HFD + IR rat group. Regulation of Peroxisome proliferator-activated receptor-γ (PPAR-γ) mRNA enhanced paraoxonase-1 (PON-1) and arylesterase (AE) activities and inhibited that of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA). It also increased the activities of plasma lecithin-cholesterol acyltransferase (LCAT) and lipoprotein lipase (LPL). Pterostilbene and Orlistat also corrected the alterations of serum leptin and adiponectin levels in lipidemic rats. Such findings provide evidence that Pterostilbene and Orlistat can act as normolipidemic agents that possess lipid-lowering effects and potential as a radioprotector.
Collapse
Affiliation(s)
- Enas M. Moustafa
- Department of Radiation Biology, National Center for Radiation Research & Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Nasr City, Egypt
| | - Engy R. Rashed
- Department of Drug Radiation Research, National Center for Radiation Research & Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Nasr City, Egypt
| | - Rasha R. Rashed
- Department of Drug Radiation Research, National Center for Radiation Research & Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Nasr City, Egypt
| |
Collapse
|
34
|
Alatshan A, Benkő S. Nuclear Receptors as Multiple Regulators of NLRP3 Inflammasome Function. Front Immunol 2021; 12:630569. [PMID: 33717162 PMCID: PMC7952630 DOI: 10.3389/fimmu.2021.630569] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Nuclear receptors are important bridges between lipid signaling molecules and transcription responses. Beside their role in several developmental and physiological processes, many of these receptors have been shown to regulate and determine the fate of immune cells, and the outcome of immune responses under physiological and pathological conditions. While NLRP3 inflammasome is assumed as key regulator for innate and adaptive immune responses, and has been associated with various pathological events, the precise impact of the nuclear receptors on the function of inflammasome is hardly investigated. A wide variety of factors and conditions have been identified as modulators of NLRP3 inflammasome activation, and at the same time, many of the nuclear receptors are known to regulate, and interact with these factors, including cellular metabolism and various signaling pathways. Nuclear receptors are in the focus of many researches, as these receptors are easy to manipulate by lipid soluble molecules. Importantly, nuclear receptors mediate regulatory mechanisms at multiple levels: not only at transcription level, but also in the cytosol via non-genomic effects. Their importance is also reflected by the numerous approved drugs that have been developed in the past decade to specifically target nuclear receptors subtypes. Researches aiming to delineate mechanisms that regulate NLRP3 inflammasome activation draw a wide range of attention due to their unquestionable importance in infectious and sterile inflammatory conditions. In this review, we provide an overview of current reports and knowledge about NLRP3 inflammasome regulation from the perspective of nuclear receptors, in order to bring new insight to the potentially therapeutic aspect in targeting NLRP3 inflammasome and NLRP3 inflammasome-associated diseases.
Collapse
Affiliation(s)
- Ahmad Alatshan
- Departments of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Benkő
- Departments of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
35
|
Skeletal Muscle Metabolomic Responses to Endurance and Resistance Training in Rats under Chronic Unpredictable Mild Stress. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041645. [PMID: 33572176 PMCID: PMC7914905 DOI: 10.3390/ijerph18041645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/31/2022]
Abstract
The objectives of this study were to compare the antidepressant effects between endurance and resistance exercise for optimizing interventions and examine the metabolomic changes in different types of skeletal muscles in response to the exercise, using a rat model of chronic unpredictable mild stress (CUMS)-induced depression. There were 32 male Sprague-Dawley rats randomly divided into a control group (C) and 3 experimental groups: CUMS control (D), endurance exercise (E), and resistance exercise (R). Group E underwent 30 min treadmill running, and group R performed 8 rounds of ladder climbing, 5 sessions per week for 4 weeks. Body weight, sucrose preference, and open field tests were performed pre and post the intervention period for changes in depressant symptoms, and the gastrocnemius and soleus muscles were sampled after the intervention for metabolomic analysis using the 1H-NMR technique. The results showed that both types of exercise effectively improved the depression-like symptoms, and the endurance exercise appeared to have a better effect. The levels of 10 metabolites from the gastrocnemius and 13 metabolites from the soleus of group D were found to be significantly different from that of group C, and both types of exercise had a callback effect on these metabolites, indicating that a number of metabolic pathways were involved in the depression and responded to the exercise interventions.
Collapse
|
36
|
Chi T, Lin J, Wang M, Zhao Y, Liao Z, Wei P. Non-Coding RNA as Biomarkers for Type 2 Diabetes Development and Clinical Management. Front Endocrinol (Lausanne) 2021; 12:630032. [PMID: 34603195 PMCID: PMC8484715 DOI: 10.3389/fendo.2021.630032] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Diabetes, a metabolic disease characterized by high blood glucose and other complications, has undefined causes and multiple risk factors, including inappropriate diet, unhealthy lifestyles, and genetic predisposition. The two most distinguished types of diabetes are type 1 and type 2 diabetes, resulting from the autoimmune impairment of insulin-generating pancreatic β cells and insulin insensitivity, respectively. Non-coding RNAs (ncRNAs), a cohort of RNAs with little transcriptional value, have been found to exert substantial importance in epigenetic and posttranscriptional modulation of gene expression such as messenger RNA (mRNA) silencing. This review mainly focuses on the pathology of type 2 diabetes (T2D) and ncRNAs as potential biomarkers in T2D development and clinical management. We consolidate the pathogenesis, diagnosis, and current treatments of T2D, and present the existing evidence on changes in multiple types of ncRNAs in response to various pathological changes and dysfunctions in different stages of T2D.
Collapse
Affiliation(s)
- Tiange Chi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaran Lin
- Department of Nephrology and Endocrinology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Mina Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| | - Yihan Zhao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Peng Wei, ; Zehuan Liao,
| | - Peng Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Peng Wei, ; Zehuan Liao,
| |
Collapse
|
37
|
Tan Y, Wang M, Yang K, Chi T, Liao Z, Wei P. PPAR-α Modulators as Current and Potential Cancer Treatments. Front Oncol 2021; 11:599995. [PMID: 33833983 PMCID: PMC8021859 DOI: 10.3389/fonc.2021.599995] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the leading causes of mortality worldwide. PPAR modulators may hold great potential for the management of cancer patients. Indeed, PPARs are critical sensors and regulators of lipid, and they are able to promote eNOS activation, regulate immunity and inflammation response, and affect proliferation and differentiation of cancer cells. Cancer, a name given to a group of diseases, is characterized by multiple distinctive biological behaviors, including angiogenesis, abnormal cell proliferation, aerobic glycolysis, inflammation, etc. In the last decade, emerging evidence has shown that PPAR-α, a nuclear hormone receptor, can modulate carcinogenesis via exerting effects on one or several characteristic pathological behaviors of cancer. Therefore, the multi-functional PPAR modulators have substantial promise in various types of cancer therapies. This review aims to consolidate the functions of PPAR-α, as well as discuss the current and potential applications of PPAR-α agonists and antagonists in tackling cancer.
Collapse
Affiliation(s)
- Yan Tan
- School of Traditional Chinese Medicine and School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mina Wang
- School of Traditional Chinese Medicine and School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Acupuncture Neuromodulation, Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ke Yang
- School of Traditional Chinese Medicine and School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Tiange Chi
- The First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Stockholm, Sweden
- Zehuan Liao
| | - Peng Wei
- School of Traditional Chinese Medicine and School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Peng Wei
| |
Collapse
|
38
|
Manickam R, Duszka K, Wahli W. PPARs and Microbiota in Skeletal Muscle Health and Wasting. Int J Mol Sci 2020; 21:ijms21218056. [PMID: 33137899 PMCID: PMC7662636 DOI: 10.3390/ijms21218056] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 10/24/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle is a major metabolic organ that uses mostly glucose and lipids for energy production and has the capacity to remodel itself in response to exercise and fasting. Skeletal muscle wasting occurs in many diseases and during aging. Muscle wasting is often accompanied by chronic low-grade inflammation associated to inter- and intra-muscular fat deposition. During aging, muscle wasting is advanced due to increased movement disorders, as a result of restricted physical exercise, frailty, and the pain associated with arthritis. Muscle atrophy is characterized by increased protein degradation, where the ubiquitin-proteasomal and autophagy-lysosomal pathways, atrogenes, and growth factor signaling all play an important role. Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family of transcription factors, which are activated by fatty acids and their derivatives. PPARs regulate genes that are involved in development, metabolism, inflammation, and many cellular processes in different organs. PPARs are also expressed in muscle and exert pleiotropic specialized responses upon activation by their ligands. There are three PPAR isotypes, viz., PPARα, -β/δ, and -γ. The expression of PPARα is high in tissues with effective fatty acid catabolism, including skeletal muscle. PPARβ/δ is expressed more ubiquitously and is the predominant isotype in skeletal muscle. It is involved in energy metabolism, mitochondrial biogenesis, and fiber-type switching. The expression of PPARγ is high in adipocytes, but it is also implicated in lipid deposition in muscle and other organs. Collectively, all three PPAR isotypes have a major impact on muscle homeostasis either directly or indirectly. Furthermore, reciprocal interactions have been found between PPARs and the gut microbiota along the gut–muscle axis in both health and disease. Herein, we review functions of PPARs in skeletal muscle and their interaction with the gut microbiota in the context of muscle wasting.
Collapse
Affiliation(s)
- Ravikumar Manickam
- Department of Pharmaceutical Sciences, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA;
| | - Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria;
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
- Toxalim, INRAE, Chemin de Tournefeuille 180, F-31027 Toulouse, France
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Correspondence:
| |
Collapse
|
39
|
Lai N, Fealy CE, Kummitha CM, Cabras S, Kirwan JP, Hoppel CL. Mitochondrial Utilization of Competing Fuels Is Altered in Insulin Resistant Skeletal Muscle of Non-obese Rats (Goto-Kakizaki). Front Physiol 2020; 11:677. [PMID: 32612543 PMCID: PMC7308651 DOI: 10.3389/fphys.2020.00677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/26/2020] [Indexed: 12/25/2022] Open
Abstract
Aim Insulin-resistant skeletal muscle is characterized by metabolic inflexibility with associated alterations in substrate selection, mediated by peroxisome-proliferator activated receptor δ (PPARδ). Although it is established that PPARδ contributes to the alteration of energy metabolism, it is not clear whether it plays a role in mitochondrial fuel competition. While nutrient overload may impair metabolic flexibility by fuel congestion within mitochondria, in absence of obesity defects at a mitochondrial level have not yet been excluded. We sought to determine whether reduced PPARδ content in insulin-resistant rat skeletal muscle of a non-obese rat model of T2DM (Goto-Kakizaki, GK) ameliorate the inhibitory effect of fatty acid (i.e., palmitoylcarnitine) on mitochondrial carbohydrate oxidization (i.e., pyruvate) in muscle fibers. Methods Bioenergetic function was characterized in oxidative soleus (S) and glycolytic white gastrocnemius (WG) muscles with measurement of respiration rates in permeabilized fibers in the presence of complex I, II, IV, and fatty acid substrates. Mitochondrial content was measured by citrate synthase (CS) and succinate dehydrogenase activity (SDH). Western blot was used to determine protein expression of PPARδ, PDK isoform 2 and 4. Results CS and SDH activity, key markers of mitochondrial content, were reduced by ∼10-30% in diabetic vs. control, and the effect was evident in both oxidative and glycolytic muscles. PPARδ (p < 0.01), PDK2 (p < 0.01), and PDK4 (p = 0.06) protein content was reduced in GK animals compared to Wistar rats (N = 6 per group). Ex vivo respiration rates in permeabilized muscle fibers determined in the presence of complex I, II, IV, and fatty acid substrates, suggested unaltered mitochondrial bioenergetic function in T2DM muscle. Respiration in the presence of pyruvate was higher compared to palmitoylcarnitine in both animal groups and fiber types. Moreover, respiration rates in the presence of both palmitoylcarnitine and pyruvate were reduced by 25 ± 6% (S), 37 ± 6% (WG) and 63 ± 6% (S), 57 ± 8% (WG) compared to pyruvate for both controls and GK, respectively. The inhibitory effect of palmitoylcarnitine on respiration was significantly greater in GK than controls (p < 10-3). Conclusion With competing fuels, the presence of fatty acids diminishes mitochondria ability to utilize carbohydrate derived substrates in insulin-resistant muscle despite reduced PPARδ content.
Collapse
Affiliation(s)
- Nicola Lai
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, United States.,Biomedical Engineering Institute, Old Dominion University, Norfolk, VA, United States.,Department of Mechanical, Chemical and Materials Engineering, University of Cagliari, Cagliari, Italy.,Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States.,Center for Mitochondrial Disease, Case Western Reserve University, Cleveland, OH, United States
| | - Ciarán E Fealy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Chinna M Kummitha
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Silvia Cabras
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - John P Kirwan
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States.,Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Charles L Hoppel
- Center for Mitochondrial Disease, Case Western Reserve University, Cleveland, OH, United States.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States.,Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
40
|
Wang M, Yang Y, Liao Z. Diabetes and cancer: Epidemiological and biological links. World J Diabetes 2020; 11:227-238. [PMID: 32547697 PMCID: PMC7284016 DOI: 10.4239/wjd.v11.i6.227] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/24/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
The incidence of diabetes and cancer has increased significantly in recent years. Furthermore, there are many common risk factors for both diabetes and cancer, such as obesity, sedentary lifestyle, smoking, and ageing. A large body of epidemiological evidence has indicated that diabetes is considered as an independent risk factor for increased rates of heterogeneous types of cancer occurrence and death. The incidence and mortality of various types of cancer, such as pancreas, liver, colorectal, breast, endometrial, and bladder cancers, have a modest growth in diabetics. However, diabetes may work as a protective factor for prostate cancer. Although the underlying biological mechanisms have not been totally understood, studies have validated that insulin/insulin-like growth factor (IGF) axis (including insulin resistance, hyperinsulinemia, and IGF), hyperglycemia, inflammatory cytokines, and sex hormones provide good circumstances for cancer cell proliferation and metastasis. Insulin/IGF axis activates several metabolic and mitogenic signaling pathways; hyperglycemia provides energy for cancer cell growth; inflammatory cytokines influence cancer cell apoptosis. Thus, these three factors affect all types of cancer, while sex hormones only play important roles in breast cancer, endometrial cancer, and prostate cancer. This minireview consolidates and discusses the epidemiological and biological links between diabetes and various types of cancer.
Collapse
Affiliation(s)
- Mina Wang
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
- The Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing 100010, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yingying Yang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna 17177, Sweden
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Solna 17177, Sweden
| |
Collapse
|
41
|
Kadayat TM, Shrestha A, Jeon YH, An H, Kim J, Cho SJ, Chin J. Targeting Peroxisome Proliferator-Activated Receptor Delta (PPARδ): A Medicinal Chemistry Perspective. J Med Chem 2020; 63:10109-10134. [DOI: 10.1021/acs.jmedchem.9b01882] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tara Man Kadayat
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Aarajana Shrestha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Republic of Korea
| | - Hongchan An
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Republic of Korea
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| |
Collapse
|
42
|
Wang M, Liu L, Zhang CS, Liao Z, Jing X, Fishers M, Zhao L, Xu X, Li B. Mechanism of Traditional Chinese Medicine in Treating Knee Osteoarthritis. J Pain Res 2020; 13:1421-1429. [PMID: 32606908 PMCID: PMC7304682 DOI: 10.2147/jpr.s247827] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/24/2020] [Indexed: 12/15/2022] Open
Abstract
Knee osteoarthritis (KOA) is a degenerative disease, making a unique contribution to chronic pain, edema, and limited mobility of knee joint. Traditional Chinese Medicine (TCM) is a common complementary therapy for KOA and has been found effective. The aim of this review is to consolidate the current knowledge about the mechanism of four interventions of TCM: acupuncture, moxibustion, herbs, and massage in treating KOA, and how they alleviate symptoms such as pain, swelling, and dysfunction. Furthermore, this review highlights that four therapies have different mechanisms but all of them can manage KOA through inhibiting inflammation, which indicates that alternative therapies should be considered as a viable complementary treatment for pain management in clinical practice.
Collapse
Affiliation(s)
- Mina Wang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, People's Republic of China.,Graduate School, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China
| | - Lu Liu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, People's Republic of China.,Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Claire Shuiqing Zhang
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore 637551.,Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Stockholm SE-17177, Sweden
| | - Xianghong Jing
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Marc Fishers
- Department of Neurology, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, USA
| | - Luopeng Zhao
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, People's Republic of China.,Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, People's Republic of China
| | - Xiaobai Xu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, People's Republic of China
| | - Bin Li
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, People's Republic of China
| |
Collapse
|
43
|
Phua WWT, Tan WR, Yip YS, Hew ID, Wee JWK, Cheng HS, Leow MKS, Wahli W, Tan NS. PPARβ/δ Agonism Upregulates Forkhead Box A2 to Reduce Inflammation in C2C12 Myoblasts and in Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21051747. [PMID: 32143325 PMCID: PMC7084392 DOI: 10.3390/ijms21051747] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 01/10/2023] Open
Abstract
Daily activities expose muscles to innumerable impacts, causing accumulated tissue damage and inflammation that impairs muscle recovery and function, yet the mechanism modulating the inflammatory response in muscles remains unclear. Our study suggests that Forkhead box A2 (FoxA2), a pioneer transcription factor, has a predominant role in the inflammatory response during skeletal muscle injury. FoxA2 expression in skeletal muscle is upregulated by fatty acids and peroxisome proliferator-activated receptors (PPARs) but is refractory to insulin and glucocorticoids. Using PPARβ/δ agonist GW501516 upregulates FoxA2, which in turn, attenuates the production of proinflammatory cytokines and reduces the infiltration of CD45+ immune cells in two mouse models of muscle inflammation, systemic LPS and intramuscular injection of carrageenan, which mimic localized exercise-induced inflammation. This reduced local inflammatory response limits tissue damage and restores muscle tetanic contraction. In line with these results, a deficiency in either PPARβ/δ or FoxA2 diminishes the action of the PPARβ/δ agonist GW501516 to suppress an aggravated inflammatory response. Our study suggests that FoxA2 in skeletal muscle helps maintain homeostasis, acting as a gatekeeper to maintain key inflammation parameters at the desired level upon injury. Therefore, it is conceivable that certain myositis disorders or other forms of painful musculoskeletal diseases may benefit from approaches that increase FoxA2 activity in skeletal muscle.
Collapse
Affiliation(s)
- Wendy Wen Ting Phua
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; (W.W.T.P.); (Y.S.Y.); (I.D.H.); (J.W.K.W.); (H.S.C.)
- NTU Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University Singapore, Singapore 637551, Singapore
| | - Wei Ren Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (W.R.T.); (M.K.S.L.); (W.W.)
| | - Yun Sheng Yip
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; (W.W.T.P.); (Y.S.Y.); (I.D.H.); (J.W.K.W.); (H.S.C.)
| | - Ivan Dongzheng Hew
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; (W.W.T.P.); (Y.S.Y.); (I.D.H.); (J.W.K.W.); (H.S.C.)
| | - Jonathan Wei Kiat Wee
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; (W.W.T.P.); (Y.S.Y.); (I.D.H.); (J.W.K.W.); (H.S.C.)
| | - Hong Sheng Cheng
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; (W.W.T.P.); (Y.S.Y.); (I.D.H.); (J.W.K.W.); (H.S.C.)
| | - Melvin Khee Shing Leow
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (W.R.T.); (M.K.S.L.); (W.W.)
- Department of Endocrinology, Division of Medicine, Endocrine and Diabetes Clinic, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (W.R.T.); (M.K.S.L.); (W.W.)
- INRA ToxAlim, UMR1331, Chemin de Tournefeuille, Toulouse Cedex 3, 31300 Toulouse, France
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| | - Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; (W.W.T.P.); (Y.S.Y.); (I.D.H.); (J.W.K.W.); (H.S.C.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (W.R.T.); (M.K.S.L.); (W.W.)
- Correspondence: ; Tel.: +65-6904-1295; Fax: +65-6339-2889
| |
Collapse
|
44
|
Gogulothu R, Nagar D, Gopalakrishnan S, Garlapati VR, Kallamadi PR, Ismail A. Disrupted expression of genes essential for skeletal muscle fibre integrity and energy metabolism in Vitamin D deficient rats. J Steroid Biochem Mol Biol 2020; 197:105525. [PMID: 31705962 DOI: 10.1016/j.jsbmb.2019.105525] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/16/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022]
Abstract
Vitamin D, a secosteroid that regulates mineral homeostasis via its actions in intestine, bone, kidneys and parathyroid glands, has many other target tissues, including skeletal muscle. In the present study, we used rats to examine if diet-induced vitamin D deficiency or insufficiency altered protein synthesis in muscle via the mTOR pathway, and impaired skeletal muscle quality by changing expression of genes needed for its function. Vitamin D deficiency resulted in reduced levels of phosphorylated mTOR, and suppressed mTOR-dependent phosphorylation of 4E-BP1 and p70-S6K, implying a decrease in activity of the protein synthesis machinery. These changes were coupled with up regulation of genes that are negative regulators of muscle growth (Fbxo32 & Trim63), leading to a net loss of skeletal muscle mass. Vitamin D deficiency or insufficiency also led to a decrease in expression of both myosin and actin-associated proteins (Myh1, Myh2, Myh7, Tnnc1& Tnnt1), which are essential for generation of the mechanical force needed for muscle contraction. We also detected a decrease in expression of glycolytic and oxidative enzyme genes (Hk2, Pfkm, Cs, Pdk4 & βHad) and transcriptional coactivator genes (Ppargc-1α & Ppargc-1β) which indicate a low oxidative capacity of skeletal muscle in the vitamin D deficient state. Furthermore, decreased citrate synthase activity corroborates a decrease in mitochondrial density and aerobic capacity of the muscle. In conclusion, our study demonstrates that chronic vitamin D deficiency or insufficiency reduced the size of skeletal muscle fibres, altered their composition, and decreased their oxidative potential. Most of the changes observed were reversible, either partially or completely, by restoring vitamin D to the diet of the deficient rats.
Collapse
Affiliation(s)
- Ramesh Gogulothu
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | - Devika Nagar
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | | | - Venkat R Garlapati
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | | | - Ayesha Ismail
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
45
|
Wang M, Tan Y, Shi Y, Wang X, Liao Z, Wei P. Diabetes and Sarcopenic Obesity: Pathogenesis, Diagnosis, and Treatments. Front Endocrinol (Lausanne) 2020; 11:568. [PMID: 32982969 PMCID: PMC7477770 DOI: 10.3389/fendo.2020.00568] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Sarcopenic obesity and diabetes are two increasing health problems worldwide, which both share many common risk factors, such as aging, and general obesity. The pathogenesis of sarcopenic obesity includes aging, physical inactivity, malnutrition, low-grade inflammation, insulin resistance, and hormonal changes. Nevertheless, there are two major reasons to cause diabetes: impaired insulin secretion and impaired insulin action. Furthermore, the individual diagnosis of obesity and sarcopenia should be combined to adequately define sarcopenic obesity. Also, the diagnosis of diabetes includes fasting plasma glucose test (FPG), 2-h oral glucose tolerance test (OGTT), glycated hemoglobin (A1C), and random plasma glucose coupled with symptoms. Healthy diet and physical activity are beneficial to both sarcopenic obesity and diabetes, but there are only recommended drugs for diabetes. This review consolidates and discusses the latest research in pathogenesis, diagnosis, and treatments of diabetes and sarcopenic obesity.
Collapse
Affiliation(s)
- Mina Wang
- School of Traditional Chinese Medicine, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Acupuncture Neuromodulation, Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yan Tan
- School of Traditional Chinese Medicine, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yifan Shi
- School of Traditional Chinese Medicine, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Wang
- School of Traditional Chinese Medicine, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Stockholm, Sweden
- Zehuan Liao
| | - Peng Wei
- School of Traditional Chinese Medicine, School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Peng Wei
| |
Collapse
|
46
|
Espinosa CD, Fry RS, Kocher ME, Stein HH. Effects of copper hydroxychloride on growth performance and abundance of genes involved in lipid metabolism of growing pigs. J Anim Sci 2020; 98:skz369. [PMID: 31901093 PMCID: PMC6978892 DOI: 10.1093/jas/skz369] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/02/2020] [Indexed: 12/16/2022] Open
Abstract
An experiment was conducted to test the hypothesis that copper (Cu) hydroxychloride improves growth performance by upregulating the mRNA transcription of genes involved in lipid metabolism of pigs fed a diet based on corn, soybean meal (SBM), and distillers dried grains with solubles (DDGS). Thirty-two pigs (15.05 ± 0.98 kg) were allotted to 2 dietary treatments with 2 pigs per pen for a total of 8 replicate pens per treatment. Pigs were fed a corn-SBM-DDGS control diet that included Cu to meet the requirement. A second diet was formulated by adding 150 mg Cu/kg from copper hydroxychloride to the control diet. On the last day of the experiment, one pig per pen was sacrificed, and samples from liver, skeletal muscle, and subcutaneous adipose tissue were collected to analyze relative mRNA abundance of genes involved in lipid metabolism. Results indicated that overall ADG and G:F were greater (P < 0.05) for pigs fed the diet containing copper hydroxychloride compared with pigs fed the control diet. Pigs fed the diet supplemented with copper hydroxychloride also had increased (P < 0.05) abundance of cluster of differentiation 36 in the liver and increased (P < 0.05) abundance of fatty acid-binding protein 4 and lipoprotein lipase in subcutaneous adipose tissue. Inclusion of copper hydroxychloride also tended to increase (P < 0.10) the abundance of fatty acid-binding protein 1, peroxisome proliferator-activated receptor α, and carnitine palmitoyltransferase 1B in the liver, skeletal muscle, and subcutaneous adipose tissue, respectively. This indicates that dietary Cu may affect signaling pathways associated with lipid metabolism by improving the uptake, transport, and utilization of fatty acids. In conclusion, supplementation of copper hydroxychloride to the control diet improved growth performance and upregulated the abundance of some genes involved in postabsorptive metabolism of lipids.
Collapse
Affiliation(s)
| | | | | | - Hans H Stein
- Department of Animal Sciences, University of Illinois, Urbana
| |
Collapse
|
47
|
Yang Y, Yang T, Liu S, Cao Z, Zhao Y, Su X, Liao Z, Teng X, Hua J. Concentrated ambient PM 2.5 exposure affects mice sperm quality and testosterone biosynthesis. PeerJ 2019; 7:e8109. [PMID: 31799077 PMCID: PMC6885350 DOI: 10.7717/peerj.8109] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/28/2019] [Indexed: 12/18/2022] Open
Abstract
Background Studies suggested that PM2.5 exposure could lead to adverse reproductive effects on male animals. However, the underlying mechanism is still not clear. Besides, animals in the majority of previous studies were exposed to PM2.5 through intratracheal instillation which should be improved. In addition, limited amount of research has been conducted in China where the PM2.5 concentration is higher and the PM2.5 components are different. The aim of this work is to explore the effects of concentrated ambient PM2.5 (CAP) on mice sperm quality and testosterone biosynthesis. Methods A total of 12 male C57BL/6 mice were exposed to filtered air (FA) or CAP for 125 days using the Shanghai Meteorological and Environmental Animal Exposure System. The mice sperm concentration, sperm motility, DNA fragmentation index, high DNA stainability and plasma testosterone were analyzed. Testicular histology and sperm morphology were observed through optical microscope. Testosterone biosynthesis related gene expressions were analyzed using real-time PCR, including cytochrome P450 CHOL side-chain cleavage enzyme (P450scc), steroidogenic acute regulatory protein (StAR), 3β-hydroxysteroid dehydrogenase (3β HSD), 17β-hydroxysteroid dehydrogenase, cytochrome P450 aromatase (P450arom), estrogen receptor (ER), androgen receptor (AR) and follicle stimulating hormone receptor (FSHR). Results Exposure to CAP resulted in disturbance of various stages of spermatogenesis and significant higher percentage of abnormal sperm (FA vs. CAP: 24.37% vs. 44.83%) in mice testis. CAP exposure significantly decreased sperm concentration (43.00 × 106 vs. 25.33 × 106) and motility (PR: 63.58% vs. 55.15%; PR + NP: 84.00% vs. 77.08%) in epididymis. Plasma testosterone concentration were significantly declined (0.28 ng/ml vs. 0.69 ng/ml) under CAP exposure. Notably, the levels of testosterone biosynthesis related genes, StAR, P450scc, P450arom, ER and FSHR were significantly decreased with CAP exposure. Conclusion Concentrated ambient PM2.5 exposure altered mice sperm concentration, motility and morphology, which might be mediated primarily by the decline in testosterone concentration and testosterone biosynthesis process.
Collapse
Affiliation(s)
- Yingying Yang
- Department of Women and Children's Health Care, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Tingting Yang
- Department of Social Medicine, School of Public Health, Fudan University, Shanghai, China
| | - Shengxin Liu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Zhijuan Cao
- Department of Women and Children's Health Care, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Zhao
- Department of Women and Children's Health Care, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiujuan Su
- Department of Women and Children's Health Care, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Xiaoming Teng
- Department of Assisted Reproductive Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Hua
- Department of Women and Children's Health Care, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Nutrigenomic effect of conjugated linoleic acid on growth and meat quality indices of growing rabbit. PLoS One 2019; 14:e0222404. [PMID: 31600212 PMCID: PMC6786800 DOI: 10.1371/journal.pone.0222404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/29/2019] [Indexed: 12/20/2022] Open
Abstract
Conjugated linoleic acid was detected in rabbit caecotrophs, due to the presence of microbial lipid activity in rabbit cecum. However, the effect of CLA as a functional food in growing rabbit is not well established. Therefore, this study was conducted to determine the effect of CLA on production, meat quality, and its nutrigenomic effect on edible parts of rabbit carcass including skeletal muscle, liver, and adipose tissue. Therefore, seventy five weaned V-Line male rabbits, 30 days old, were randomly allocated into three dietary treatments receiving either basal control diet, diet supplemented with 0.5% (CLAL), or 1% CLA (CLAH). Total experimental period (63 d) was segmented into 7 days adaptation and 56 days experimental period. Dietary supplementation of CLA did not alter growth performance, however, the fat percentage of longissimus lumborum muscle was decreased, with an increase in protein and polyunsaturated fatty acids (PUFA) percentage. Saturated fatty acids (SFA) and mono unsaturated fatty acids (MUFA) were not increased in CLA treated groups. There was tissue specific sensing of CLA, since subcutaneous adipose tissue gene expression of PPARA was downregulated, however, CPT1A tended to be upregulated in liver of CLAL group only (P = 0.09). In skeletal muscle, FASN and PPARG were upregulated in CLAH group only (P ≤0.01). Marked cytoplasmic vacuolation was noticed in liver of CLAH group without altering hepatocyte structure. Adipocyte size was decreased in CLA fed groups, in a dose dependent manner (P <0.01). Cell proliferation determined by PCNA was lower (P <0.01) in adipose tissue of CLA groups. Our data indicate that dietary supplementation of CLA (c9,t11-CLA and t10,c12- CLA) at a dose of 0.5% in growing rabbit diet produce rabbit meat rich in PUFA and lower fat % without altering growth performance and hepatocyte structure.
Collapse
|
49
|
Lake JA, Papah MB, Abasht B. Increased Expression of Lipid Metabolism Genes in Early Stages of Wooden Breast Links Myopathy of Broilers to Metabolic Syndrome in Humans. Genes (Basel) 2019; 10:E746. [PMID: 31557856 PMCID: PMC6826700 DOI: 10.3390/genes10100746] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022] Open
Abstract
Wooden breast is a muscle disorder affecting modern commercial broiler chickens that causes a palpably firm pectoralis major muscle and severe reduction in meat quality. Most studies have focused on advanced stages of wooden breast apparent at market age, resulting in limited insights into the etiology and early pathogenesis of the myopathy. Therefore, the objective of this study was to identify early molecular signals in the wooden breast transcriptional cascade by performing gene expression analysis on the pectoralis major muscle of two-week-old birds that may later exhibit the wooden breast phenotype by market age at 7 weeks. Biopsy samples of the left pectoralis major muscle were collected from 101 birds at 14 days of age. Birds were subsequently raised to 7 weeks of age to allow sample selection based on the wooden breast phenotype at market age. RNA-sequencing was performed on 5 unaffected and 8 affected female chicken samples, selected based on wooden breast scores (0 to 4) assigned at necropsy where affected birds had scores of 2 or 3 (mildly or moderately affected) while unaffected birds had scores of 0 (no apparent gross lesions). Differential expression analysis identified 60 genes found to be significant at an FDR-adjusted p-value of 0.05. Of these, 26 were previously demonstrated to exhibit altered expression or genetic polymorphisms related to glucose tolerance or diabetes mellitus in mammals. Additionally, 9 genes have functions directly related to lipid metabolism and 11 genes are associated with adiposity traits such as intramuscular fat and body mass index. This study suggests that wooden breast disease is first and foremost a metabolic disorder characterized primarily by ectopic lipid accumulation in the pectoralis major.
Collapse
Affiliation(s)
- Juniper A Lake
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19711, USA.
| | - Michael B Papah
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA.
| | - Behnam Abasht
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
50
|
Khan RMM, Chua ZJY, Tan JC, Yang Y, Liao Z, Zhao Y. From Pre-Diabetes to Diabetes: Diagnosis, Treatments and Translational Research. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E546. [PMID: 31470636 PMCID: PMC6780236 DOI: 10.3390/medicina55090546] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/16/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022]
Abstract
Diabetes, a silent killer, is one of the most widely prevalent conditions of the present time. According to the 2017 International Diabetes Federation (IDF) statistics, the global prevalence of diabetes among the age group of 20-79 years is 8.8%. In addition, 1 in every 2 persons is unaware of the condition. This unawareness and ignorance lead to further complications. Pre-diabetes is the preceding condition of diabetes, and in most of the cases, this ultimately leads to the development of diabetes. Diabetes can be classified into three types, namely type 1 diabetes, type 2 diabetes mellitus (T2DM) and gestational diabetes. The diagnosis of both pre-diabetes and diabetes is based on glucose criteria; the common modalities used are fasting plasma glucose (FPG) test and oral glucose tolerance test (OGTT). A glucometer is commonly used by diabetic patients to measure blood glucose levels with fast and rather accurate measurements. A few of the more advanced and minimally invasive modalities include the glucose-sensing patch, SwEatch, eyeglass biosensor, breath analysis, etc. Despite a considerable amount of data being collected and analyzed regarding diabetes, the actual molecular mechanism of developing type 2 diabetes mellitus (T2DM) is still unknown. Both genetic and epigenetic factors are associated with T2DM. The complications of diabetes can predominantly be classified into two categories: microvascular and macrovascular. Retinopathy, nephropathy, and neuropathy are grouped under microvascular complications, whereas stroke, cardiovascular disease, and peripheral artery disease (PAD) belong to macrovascular complications. Unfortunately, until now, no complete cure for diabetes has been found. However, the treatment of pre-diabetes has shown significant success in preventing the further progression of diabetes. To prevent pre-diabetes from developing into T2DM, lifestyle intervention has been found to be very promising. Various aspects of diabetes, including the aforementioned topics, have been reviewed in this paper.
Collapse
Affiliation(s)
- Radia Marium Modhumi Khan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Zoey Jia Yu Chua
- School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Jia Chi Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Yingying Yang
- Tongji University School of Medicine, Shanghai 201204, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 65 Solna, Sweden
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
- Department of Microbiology, Tumor, and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
| | - Yan Zhao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|