1
|
Deng X, Liu H, Zhao W, Wu R, Chen K, Li Q, Yazlık MO, Özkan H, Ren J, Zhang J, Liu S, Mei L, Li S, Zhan J, Hu B. Expression of AMPK and PLIN2 in the regulation of lipid metabolism and oxidative stress in bitches with open cervix pyometra. BMC Vet Res 2025; 21:164. [PMID: 40082867 PMCID: PMC11905669 DOI: 10.1186/s12917-025-04622-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/24/2025] [Indexed: 03/16/2025] Open
Abstract
The pathogenesis of canine pyometra is multifactorial, involving hormonal imbalances, aberrant immune responses, and metabolic dysregulation includes lipid metabolism and oxidative stress. This study focuses on lipid metabolism and oxidative stress, revealing the key regulatory role of AMPK and PLIN2 in canine pyometra. Bitches with open cervix pyometra (n:8) and healthy bitches undergoing elective ovariohysterectomy (n:4) were enrolled to the study. In experiment one, the serum and tissue levels of Malondialdehyde (MDA) and Superoxide Dismutase (SOD) activity were assessed. Additionally, uterine histopathological analysis, AMPK and PLIN2 expressions were determined through immunohistochemistry. Furthermore, inflammation, oxidative stress, and lipid metabolism-related factors were evaluated using Western blot analysis. In experiment two, primary cell cultures were prepared from healthy uterine endometrial cells of the dogs in control group. Cultured canine endometrial epithelial cells were treated with lipopolysaccharide (LPS) along with oleic acid (OA) to induce an inflammatory response. Tissue and serum MDA and SOD levels were greater in dogs with pyometra. Accumulated lipid droplets were observed in the uterine tissue of bitches with pyometra. The phosphorylation of AMPK and the expression of PLIN2 significantly increased in the pyometra group. The expression of related lipid synthesis proteins such as ACC1, FASN, SREBP-1c, and PLIN2 was upregulated, while PPARα and PGC1α were downregulated in bitches with pyometra. In experiment two, activation of AMPK and PLIN2 not only restores the expression of PGC1α, but also effectively alleviates inflammation and oxidative stress. The role of lipid metabolism and oxidative stress in canine pyometra is elucidated, thus contributing to the pathogenesis of pyometra in dogs.
Collapse
Affiliation(s)
- Xin Deng
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Hui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Wei Zhao
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Rui Wu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Kuo Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Qing Li
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Murat Onur Yazlık
- Department of Obstetrics and Gynecology, Ankara University Faculty of Veterinary Medicine, Ankara, 06070, Turkey
| | - Hüseyin Özkan
- Faculty of Veterinary Medicine, Department of Genetics, Hatay Mustafa Kemal University, Hatay, 31060, Turkey
| | - Jingyuan Ren
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Jiacheng Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Shiyi Liu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Ling Mei
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Shangfeng Li
- Zhi Pet Animal Hospital, Chengdu, Sichuan, 611830, PR China
| | - Jiasui Zhan
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, SE-75007, Sweden
| | - Binhong Hu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China.
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, SE-75007, Sweden.
- Zhi Pet Animal Hospital, Chengdu, Sichuan, 611830, PR China.
| |
Collapse
|
2
|
Valdivia-Padilla AV, Sharma A, Zegbe JA, Morales-Domínguez JF. Metabolomic Characterization and Bioinformatic Studies of Bioactive Compounds in Two Varieties of Psidium guajava L. Leaf by GC-MS Analysis. Int J Mol Sci 2025; 26:2530. [PMID: 40141181 PMCID: PMC11942350 DOI: 10.3390/ijms26062530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The guava tree (Psidium guajava L.) is a tropical plant from the Myrtaceae family. Leaf extracts from this plant have been used in traditional medicine to treat gastrointestinal disorders and exhibit several functional activities that benefit human health. Different varieties of guava trees produce fruits in colors ranging from white to red and present a characteristic metabolic profile in both their leaves and fruits. This study presents a metabolomic characterization of the leaves from two guava varieties: the Caxcana cultivar with yellow fruits and the S-56 accession with pink fruits. Metabolite profiling was conducted using Gas Chromatography-Mass Spectrometry (GC-MS) on methanol extracts, followed by multivariate statistical analysis, including Principal Component Analysis (PCA), and a heat map visualization of compound concentrations in the two varieties. The results identified β-caryophyllene as the major secondary metabolite present in both varieties, with a relative abundance of 16.46% in the Caxcana variety and 23.06% in the S-56 cultivar. Furthermore, in silico analyses, such as network pharmacology and molecular docking, revealed key interactions with proteins such as CB2, PPARα, BAX, BCL2, and AKT1, suggesting potential therapeutic relevance. These findings highlight the pharmacological potential of guava leaf metabolites in natural product chemistry and drug discovery.
Collapse
Affiliation(s)
- Ana Victoria Valdivia-Padilla
- Centro de Ciencias Básicas, Departamento de Química, Universidad Autónoma de Aguascalientes, Aguascalientes 20131, Mexico;
| | - Ashutosh Sharma
- Centre of Bioengineering, NatProLab, School of Engineering and Sciences, Tecnologico de Monterrey, Querétaro 76130, Mexico;
| | - Jorge A. Zegbe
- Campo Experimental Pabellón, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Km 32.5 Carretera Aguascalientes-Zacatecas, Pabellón de Arteaga, Aguascalientes 20668, Mexico;
| | | |
Collapse
|
3
|
Huang Z, Shi M, Zhang C, Deng Z, Qin T, Wu J, Zhang X, Han W, Li S, Gao B, Xiao Y, Huang D, Ye W. Meteorin-like protein alleviates intervertebral disc degeneration by suppressing lipid accumulation in nucleus pulposus cells via PPARα-CPT1A activation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167635. [PMID: 39706351 DOI: 10.1016/j.bbadis.2024.167635] [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: 09/01/2024] [Revised: 11/19/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Disturbances in lipid metabolism are closely related to intervertebral disc degeneration (IDD). However, the lipid metabolism characteristics of nucleus pulposus (NP) cells during IDD are unclear. Exercise protects against IDD and acts as a potent mediator of organ metabolism, in which muscle-secreted myokines actively participate. However, whether exercise-induced myokines alleviate IDD by regulating lipid metabolism in NP cells remains unknown. The present study revealed that lipid accumulation is the metabolic reprogramming phenotype in NP cells during IDD, which was attributed to an imbalance between increased fatty acid/triglyceride synthesis and diminished utilization, and was further associated with extracellular matrix (ECM) degradation and cell senescence. To explore the interaction between exercise and IDD, Sprague-Dawley rats were subjected to five weeks of treadmill running exercise, and rats in the exercise group exhibited less severe IDD than did those in the sedentary group. The expression of meteorin-like protein (Metrnl), a newly-discovered myokine that participates in lipid metabolism regulation, was observed to increase in muscle, serum and NP tissue after exercise. Moreover, Metrnl ameliorated lipid accumulation in NP cells and further alleviated ECM degradation and cell senescence. Mechanistically, Metrnl activated the fatty acid β-oxidation rate-limiting enzyme carnitine palmitoyltransferase 1A (CPT1A) via peroxisome proliferator-activated receptor α (PPARα) to increase lipid utilization in NP cells. This study provides insight into the lipid metabolic features of NP cells in IDD and reveals the intrinsic connections among exercise, metabolism and IDD, with the myokine Metrnl emerging as a pivotal mediator with therapeutic potential.
Collapse
Affiliation(s)
- Zhengqi Huang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Shi
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chao Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihuai Deng
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianyu Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jiajun Wu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Orthopedics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaohe Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weitao Han
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuangxing Li
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bo Gao
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yin Xiao
- School of Medicine and Dentistry & Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| | - Dongsheng Huang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Wei Ye
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
4
|
Kąpa M, Koryciarz I, Kustosik N, Jurowski P, Pniakowska Z. Future Directions in Diabetic Retinopathy Treatment: Stem Cell Therapy, Nanotechnology, and PPARα Modulation. J Clin Med 2025; 14:683. [PMID: 39941353 PMCID: PMC11818668 DOI: 10.3390/jcm14030683] [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: 11/06/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
This narrative review focuses on innovative treatment approaches to diabetic retinopathy to meet the urgent demand for advancements in managing both the early and late stages of the disease. Recent studies highlight the potential of adipose stem cells and their secreted factors in mitigating the retinal complications of diabetes, with promising results in improving visual acuity and reducing inflammation and angiogenesis in diabetic retinopathy. However, caution is warranted regarding the safety and long-term therapeutic effects of adipose stem cells transplantation. Bone marrow mesenchymal stem cells can also mitigate retinal damage in diabetic retinopathy. Studies demonstrate that bone marrow mesenchymal stem cells-derived exosomes can suppress the Wnt/β-catenin pathway, reducing oxidative stress, inflammation, and angiogenesis in the diabetic retina, offering promise for future diabetic retinopathy treatments. Nanotechnology has the ability to precisely target the retina and minimize systemic side effects. Nanoparticles and nanocarriers offer improved bioavailability, sustained release of therapeutics, and potential for synergistic effects. They can be a new way of effective treatment and prevention of diabetic retinopathy. Activation and modulation of PPARα as a means for diabetic retinopathy treatment has been widely investigated in recent years and demonstrated promising effects in clinical trials. PPARα activation turned out to be a promising therapeutic method for treating dyslipidemia, inflammation, and insulin sensitivity. The combination of PPARα modulators with small molecules offers an interesting perspective for retinal diseases' therapy.
Collapse
Affiliation(s)
- Maria Kąpa
- Department of Ophthalmology and Vision Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland; (M.K.); (N.K.); (P.J.); (Z.P.)
| | - Iga Koryciarz
- Department of Ophthalmology and Vision Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland; (M.K.); (N.K.); (P.J.); (Z.P.)
| | - Natalia Kustosik
- Department of Ophthalmology and Vision Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland; (M.K.); (N.K.); (P.J.); (Z.P.)
| | - Piotr Jurowski
- Department of Ophthalmology and Vision Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland; (M.K.); (N.K.); (P.J.); (Z.P.)
| | - Zofia Pniakowska
- Department of Ophthalmology and Vision Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland; (M.K.); (N.K.); (P.J.); (Z.P.)
- Optegra Eye Clinic, 90-127 Lodz, Poland
| |
Collapse
|
5
|
Schiel A, Tosso RD, Angelina E, García A, Hennuyer N, Cortes D, Cabedo N, Enriz RD. N-Tosyl Hydrazone Benzopyran, a New Ligand of PPARα Obtained from Mapping the Conformational Space of Its Active Site. J Chem Inf Model 2025; 65:298-311. [PMID: 39720903 DOI: 10.1021/acs.jcim.4c01887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
We report here a new ligand for the peroxisome-proliferator-activated receptor type α (PPARα), an N-tosyl hydrazone benzopyran that was designed throughout the mapping of the polar zone of the binding site of PPARα; such a compound displays a strong activity on this receptor that is comparable to that of the reference compound WY-14643. For the design of the N-tosyl hydrazone benzopyran, we have carried out an exhaustive conformational study of WY-14643 and a previously reported hydrazine benzopyran derivative using conformational potential energy surfaces (PES). This study allowed us to map in a systematic way the entire binding site of the PPARα. PESs allowed us to evaluate all of the critical points on the surface (minimum, transition states, and maxima) and determine the different conformational interconversion paths. Once the geometries of the different complexes were determined, we carried out the study of the different molecular interactions that stabilize these complexes through the use of QTAIM calculations. We report here for the first time the molecular behavior of WY-14643 and two compounds synthesized in our lab interacting in the active site of the PPARα providing all of the details about the different interactions that stabilize the formation of these complexes. On the basis of such information, we were able to design and synthesize a new N-tosyl hydrazone benzopyran possessing a strong agonist effect on PPARα. The information provided by this study is very useful to get a better understanding of the behavior with this type of ligand on the PPARα, giving also interesting information as a guide for the design of new ligands for this receptor.
Collapse
Affiliation(s)
- Ayelén Schiel
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL). CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina
- Departamento de Química, Universidad Ncaional del Sur, Instituto de Química del Sur (INQUISUR-UNS). CONICET, Av. Alem 1253, B800CPB Bahía Blanca, Argentina
| | - Rodrigo D Tosso
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL). CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina
| | - Emilio Angelina
- Laboratorio de Estructura Molecular y Propiedades, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Instituto de Química Básica y Aplicada (IQUIBA-NEA). CONICET, Avda. Libertad 5460, 3400 Corrientes, Argentina
| | - Ainhoa García
- Departamento de Farmacología, Universidad de Valencia, Burjassot, 46100 Valencia, Spain
- Instituto de Investigación Sanitaria-INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Nathalie Hennuyer
- Univ Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U-1011-EGID, F-59000 Lille, France
| | - Diego Cortes
- Departamento de Farmacología, Universidad de Valencia, Burjassot, 46100 Valencia, Spain
- Instituto de Investigación Sanitaria-INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Nuria Cabedo
- Departamento de Farmacología, Universidad de Valencia, Burjassot, 46100 Valencia, Spain
- Instituto de Investigación Sanitaria-INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL). CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina
| |
Collapse
|
6
|
Niu QQ, Xi YT, Zhang CR, Li XY, Li CZ, Wang HD, Li P, Yin YL. Potential mechanism of perillaldehyde in the treatment of nonalcoholic fatty liver disease based on network pharmacology and molecular docking. Eur J Pharmacol 2024; 985:177092. [PMID: 39510336 DOI: 10.1016/j.ejphar.2024.177092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic metabolic liver diseases worldwide. Perillaldehyde (4-propyl-1-en-2-ylcyclohexene-1-aldehyde, PA) is a terpenoid compound extracted from Perilla, which has effective pharmacological activities such as anti-inflammatory, antidepressant, and anticancer. This study aimed to explore the pharmacological effects of PA in intervening with NAFLD and reveal its potential mechanisms. Firstly, we identified the core targets of PA intervention therapy for NAFLD through network pharmacology and molecular docking techniques. After that, in vitro animal experiments such as H&E and Masson staining, immunofluorescence, immunohistochemistry, and Western blot were conducted to validate the results network effectively pharmacology predicted. Network pharmacology analysis suggested that PPAR-α may be the core target of PA intervention in NAFLD. H&E and Masson staining showed that after low-dose (50 mg/kg) PA administration, there was a noticeable improvement in fat deposition in the livers of NAFLD mice, and liver tissue fibrosis was alleviated. Immunohistochemical and immunofluorescence analysis showed that low dose (50 mg/kg) PA could reduce hepatocyte apoptosis, decrease the content of pro-apoptosis protein Bax, and increase the expression of anti-apoptosis protein Bcl-2 in NAFLD mice. Western blot results confirmed that low-dose (50 mg/kg) PA could increase the expression of PPAR-α and inhibit the expression of NF-κB in NAFLD mice. Our study indicated that PA could enhance the activity of PPAR-α and reduce the level of NF-κB in NAFLD mice, which may positively affect the prevention of NAFLD.
Collapse
Affiliation(s)
- Qian-Qian Niu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China; Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, 13200, Malaysia
| | - Yu-Ting Xi
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Chun-Rui Zhang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Xi-Yue Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Cheng-Zhi Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Hui-Dan Wang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China.
| | - Ya-Ling Yin
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
7
|
Ortiz-Barragán E, Estrada-Soto S, Giacoman-Martínez A, Alarcón-Aguilar FJ, Fortis-Barrera Á, Marquina-Rodríguez H, Gaona-Tovar E, Lazzarini-Lechuga R, Suárez-Alonso A, Almanza-Pérez JC. Antihyperglycemic and Hypolipidemic Activities of Flavonoids Isolated from Smilax Dominguensis Mediated by Peroxisome Proliferator-Activated Receptors. Pharmaceuticals (Basel) 2024; 17:1451. [PMID: 39598363 PMCID: PMC11597028 DOI: 10.3390/ph17111451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/29/2024] Open
Abstract
Background/objetives: Mexican people use Smilax dominguensis as a traditional medicine for diabetes control. Some reports have shown an anti-hyperglycemic effect in animal models. In the current research, a chemical bio-guided fractionation in vitro and in silico was performed to identify compounds with anti-hyperglycemic and hypolipidemic effects through PPARγ/α dual agonist activity because they regulate genes involved in energy storage and burning, such as GLUT4 and FATP. Methods: The S. dominguensis extract was evaluated in mice through oral glucose tolerance tests. The bioactive extract was fractionated by open-column chromatography, and seven final fractions (F1-F7) were obtained and evaluated. C2C12 myoblasts were treated with the fractions, and the mRNA expression levels of PPARs, GLUT-4, and FATP were quantified. The most active fractions were evaluated on GLUT-4 translocation and lipid storage in C2C12 cells and 3T3-L1 adipocytes, respectively. Results: The F3 fraction increased the expressions of PPARγ, GLUT-4, PPARα, and FATP, and it induced GLUT-4 translocation and decreased lipid storage. F3 was then analyzed by NMR, identifying three flavonoids: luteolin, apigenin, and kaempferol. These compounds were analyzed by molecular docking and on PPAR expressions. Luteolin, apigenin, and kaempferol produced a discrete increase in the mRNA expression of PPARs. Luteolin and kaempferol also decreased lipid storage. Conclusions: Our findings indicate that the compounds identified in S. dominguensis exhibit dual agonist activity on PPARγ/PPARα and have the potential for the development of new therapeutic agents helpful in diabetes, obesity, or metabolic syndrome.
Collapse
Affiliation(s)
- Erandi Ortiz-Barragán
- Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09310, Mexico;
| | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (S.E.-S.)
| | - Abraham Giacoman-Martínez
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocaril San Rafael Atlixco 186, Col. Leyes de Reforma 1a Secc. Iztapalapa, Ciudad de México 09310, Mexico; (A.G.-M.); (F.J.A.-A.); (Á.F.-B.); (A.S.-A.)
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de México 11340, Mexico
| | - Francisco J. Alarcón-Aguilar
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocaril San Rafael Atlixco 186, Col. Leyes de Reforma 1a Secc. Iztapalapa, Ciudad de México 09310, Mexico; (A.G.-M.); (F.J.A.-A.); (Á.F.-B.); (A.S.-A.)
| | - Ángeles Fortis-Barrera
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocaril San Rafael Atlixco 186, Col. Leyes de Reforma 1a Secc. Iztapalapa, Ciudad de México 09310, Mexico; (A.G.-M.); (F.J.A.-A.); (Á.F.-B.); (A.S.-A.)
| | - Hugo Marquina-Rodríguez
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (S.E.-S.)
| | - Emmanuel Gaona-Tovar
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (S.E.-S.)
| | - Roberto Lazzarini-Lechuga
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09310, Mexico;
| | - Alfredo Suárez-Alonso
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocaril San Rafael Atlixco 186, Col. Leyes de Reforma 1a Secc. Iztapalapa, Ciudad de México 09310, Mexico; (A.G.-M.); (F.J.A.-A.); (Á.F.-B.); (A.S.-A.)
| | - Julio César Almanza-Pérez
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocaril San Rafael Atlixco 186, Col. Leyes de Reforma 1a Secc. Iztapalapa, Ciudad de México 09310, Mexico; (A.G.-M.); (F.J.A.-A.); (Á.F.-B.); (A.S.-A.)
| |
Collapse
|
8
|
Park J, Song H, Moon S, Kim Y, Cho S, Han K, Park CY, Cho SW, Oh CM. Cardiometabolic benefits of fenofibrate in heart failure related to obesity and diabetes. Cardiovasc Diabetol 2024; 23:343. [PMID: 39285303 PMCID: PMC11406805 DOI: 10.1186/s12933-024-02417-6] [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: 05/13/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a serious and common condition affecting millions of people worldwide, with obesity being a major cause of metabolic disorders such as diabetes and cardiovascular disease. This study aimed to investigate the effects of fenofibrate, a peroxisome proliferator-activated receptor alpha (PPARα) agonist, on the obese- and diabetes-related cardiomyopathy. METHODS AND RESULTS We used db/db mice and high fat diet-streptozotocin induced diabetic mice to investigate the underlying mechanisms of fenofibrate's beneficial effects on heart function. Fenofibrate reduced fibrosis, and lipid accumulation, and suppressed inflammatory and immunological responses in the heart via TNF signaling. In addition, we investigated the beneficial effects of fenofibrate on HF hospitalization. The Korean National Health Insurance database was used to identify 427,154 fenofibrate users and 427,154 non-users for comparison. During the 4.22-year follow-up, fenofibrate use significantly reduced the risk of HF hospitalization (hazard ratio, 0.907; 95% CI 0.824-0.998). CONCLUSIONS The findings suggest that fenofibrate may be a useful therapeutic agent for obesity- and diabetes-related cardiomyopathy.
Collapse
Affiliation(s)
- Jiwon Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Hangyul Song
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Shinje Moon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Sungsoo Cho
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Cheol-Young Park
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje Univeristy Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Gyeonggi-Do, Korea.
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea.
| |
Collapse
|
9
|
Wang J, Zhang Q, Fu H, Han Y, Li X, Zou Q, Yuan S, Sun L. ASCT2 Regulates Fatty Acid Metabolism to Trigger Glutamine Addiction in Basal-like Breast Cancer. Cancers (Basel) 2024; 16:3028. [PMID: 39272886 PMCID: PMC11394221 DOI: 10.3390/cancers16173028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
As a crucial amino acid, glutamine can provide the nitrogen and carbon sources needed to support cancer cell proliferation, invasion, and metastasis. Interestingly, different types of breast cancer have different dependences on glutamine. This research shows that basal-like breast cancer depends on glutamine, while the other types of breast cancer may be more dependent on glucose. Glutamine transporter ASCT2 is highly expressed in various cancers and significantly promotes the growth of breast cancer. However, the key regulatory mechanism of ASCT2 in promoting basal-like breast cancer progression remains unclear. Our research demonstrates the significant change in fatty acid levels caused by ASCT2, which may be a key factor in glutamine sensitivity. This phenomenon results from the mutual activation between ASCT2-mediated glutamine transport and lipid metabolism via the nuclear receptor PPARα. ASCT2 cooperatively promoted PPARα expression, leading to the upregulation of lipid metabolism. Moreover, we also found that C118P could inhibit lipid metabolism by targeting ASCT2. More importantly, this research identifies a potential avenue of evidence for the prevention and early intervention of basal-like breast cancer by blocking the glutamine-lipid feedback loop.
Collapse
Affiliation(s)
- Jia Wang
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Qian Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Huaizi Fu
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Han
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Xue Li
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Qianlin Zou
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Shengtao Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Li Sun
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
10
|
Habib S. Team players in the pathogenesis of metabolic dysfunctions-associated steatotic liver disease: The basis of development of pharmacotherapy. World J Gastrointest Pathophysiol 2024; 15:93606. [PMID: 39220834 PMCID: PMC11362842 DOI: 10.4291/wjgp.v15.i4.93606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
Nutrient metabolism is regulated by several factors. Social determinants of health with or without genetics are the primary regulator of metabolism, and an unhealthy lifestyle affects all modulators and mediators, leading to the adaptation and finally to the exhaustion of cellular functions. Hepatic steatosis is defined by presence of fat in more than 5% of hepatocytes. In hepatocytes, fat is stored as triglycerides in lipid droplet. Hepatic steatosis results from a combination of multiple intracellular processes. In a healthy individual nutrient metabolism is regulated at several steps. It ranges from the selection of nutrients in a grocery store to the last step of consumption of ATP as an energy or as a building block of a cell as structural component. Several hormones, peptides, and genes have been described that participate in nutrient metabolism. Several enzymes participate in each nutrient metabolism as described above from ingestion to generation of ATP. As of now several publications have revealed very intricate regulation of nutrient metabolism, where most of the regulatory factors are tied to each other bidirectionally, making it difficult to comprehend chronological sequence of events. Insulin hormone is the primary regulator of all nutrients' metabolism both in prandial and fasting states. Insulin exerts its effects directly and indirectly on enzymes involved in the three main cellular function processes; metabolic, inflammation and repair, and cell growth and regeneration. Final regulators that control the enzymatic functions through stimulation or suppression of a cell are nuclear receptors in especially farnesoid X receptor and peroxisome proliferator-activated receptor/RXR ligands, adiponectin, leptin, and adiponutrin. Insulin hormone has direct effect on these final modulators. Whereas blood glucose level, serum lipids, incretin hormones, bile acids in conjunction with microbiota are intermediary modulators which are controlled by lifestyle. The purpose of this review is to overview the key players in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) that help us understand the disease natural course, risk stratification, role of lifestyle and pharmacotherapy in each individual patient with MASLD to achieve personalized care and target the practice of precision medicine. PubMed and Google Scholar databases were used to identify publication related to metabolism of carbohydrate and fat in states of health and disease states; MASLD, cardiovascular disease and cancer. More than 1000 publications including original research and review papers were reviewed.
Collapse
Affiliation(s)
- Shahid Habib
- Department of Hepatology, Liver Institute PLLC, Tucson, AZ 85712, United States
| |
Collapse
|
11
|
Dey AD, Mannan A, Dhiman S, Singh TG. Unlocking new avenues for neuropsychiatric disease therapy: the emerging potential of Peroxisome proliferator-activated receptors as promising therapeutic targets. Psychopharmacology (Berl) 2024; 241:1491-1516. [PMID: 38801530 DOI: 10.1007/s00213-024-06617-6] [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: 01/29/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
RATIONALE Peroxisome proliferator-activated receptors (PPARs) are transcription factors that regulate various physiological processes such as inflammation, lipid metabolism, and glucose homeostasis. Recent studies suggest that targeting PPARs could be beneficial in treating neuropsychiatric disorders by modulating neuronal function and signaling pathways in the brain. PPAR-α, PPAR-δ, and PPAR-γ have been found to play important roles in cognitive function, neuroinflammation, and neuroprotection. Dysregulation of PPARs has been associated with neuropsychiatric disorders like bipolar disorder, schizophrenia, major depression disorder, and autism spectrum disorder. The limitations and side effects of current treatments have prompted research to target PPARs as a promising novel therapeutic strategy. Preclinical and clinical studies have shown the potential of PPAR agonists and antagonists to improve symptoms associated with these disorders. OBJECTIVE This review aims to provide an overview of the current understanding of PPARs in neuropsychiatric disorders, their potential as therapeutic targets, and the challenges and future directions for developing PPAR-based therapies. METHODS An extensive literature review of various search engines like PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out with the keywords "PPAR, Neuropsychiatric disorders, Oxidative stress, Inflammation, Bipolar Disorder, Schizophrenia, Major depression disorder, Autism spectrum disorder, molecular pathway". RESULT & CONCLUSION Although PPARs present a hopeful direction for innovative therapeutic approaches in neuropsychiatric conditions, additional research is required to address obstacles and convert this potential into clinically viable and individualized treatments.
Collapse
Affiliation(s)
- Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | | |
Collapse
|
12
|
Zheng Y, Xu Y, Ji L, San W, Shen D, Zhou Q, Meng G, Shi J, Chen Y. Roles of distinct nuclear receptors in diabetic cardiomyopathy. Front Pharmacol 2024; 15:1423124. [PMID: 39114353 PMCID: PMC11303215 DOI: 10.3389/fphar.2024.1423124] [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: 04/25/2024] [Accepted: 06/21/2024] [Indexed: 08/10/2024] Open
Abstract
Diabetes mellitus induces a pathophysiological disorder known as diabetic cardiomyopathy and may eventually cause heart failure. Diabetic cardiomyopathy is manifested with systolic and diastolic contractile dysfunction along with alterations in unique cardiomyocyte proteins and diminished cardiomyocyte contraction. Multiple mechanisms contribute to the pathology of diabetic cardiomyopathy, mainly including abnormal insulin metabolism, hyperglycemia, glycotoxicity, cardiac lipotoxicity, endoplasmic reticulum stress, oxidative stress, mitochondrial dysfunction, calcium treatment damage, programmed myocardial cell death, improper Renin-Angiotensin-Aldosterone System activation, maladaptive immune modulation, coronary artery endothelial dysfunction, exocrine dysfunction, etc. There is an urgent need to investigate the exact pathogenesis of diabetic cardiomyopathy and improve the diagnosis and treatment of this disease. The nuclear receptor superfamily comprises a group of transcription factors, such as liver X receptor, retinoid X receptor, retinoic acid-related orphan receptor-α, retinoid receptor, vitamin D receptor, mineralocorticoid receptor, estrogen-related receptor, peroxisome proliferatoractivated receptor, nuclear receptor subfamily 4 group A 1(NR4A1), etc. Various studies have reported that nuclear receptors play a crucial role in cardiovascular diseases. A recently conducted work highlighted the function of the nuclear receptor superfamily in the realm of metabolic diseases and their associated complications. This review summarized the available information on several important nuclear receptors in the pathophysiology of diabetic cardiomyopathy and discussed future perspectives on the application of nuclear receptors as targets for diabetic cardiomyopathy treatment.
Collapse
Affiliation(s)
- Yangyang Zheng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Yongji Xu
- School of Medicine, Nantong University, Nantong, China
| | - Li Ji
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Wenqing San
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Danning Shen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Qianyou Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Guoliang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Jiahai Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yun Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| |
Collapse
|
13
|
Żulińska S, Strosznajder AK, Strosznajder JB. Current View on PPAR-α and Its Relation to Neurosteroids in Alzheimer's Disease and Other Neuropsychiatric Disorders: Promising Targets in a Therapeutic Strategy. Int J Mol Sci 2024; 25:7106. [PMID: 39000217 PMCID: PMC11241121 DOI: 10.3390/ijms25137106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) may play an important role in the pathomechanism/pathogenesis of Alzheimer's disease (AD) and several other neurological/neuropsychiatric disorders. AD leads to progressive alterations in the redox state, ion homeostasis, lipids, and protein metabolism. Significant alterations in molecular processes and the functioning of several signaling pathways result in the degeneration and death of synapses and neuronal cells, leading to the most severe dementia. Peroxisome proliferator-activated receptor alpha (PPAR-α) is among the processes affected by AD; it regulates the transcription of genes related to the metabolism of cholesterol, fatty acids, other lipids and neurotransmission, mitochondria biogenesis, and function. PPAR-α is involved in the cholesterol transport to mitochondria, the substrate for neurosteroid biosynthesis. PPAR-α-coding enzymes, such as sulfotransferases, which are responsible for neurosteroid sulfation. The relation between PPAR-α and cholesterol/neurosteroids may have a significant impact on the course and progression of neurodegeneration/neuroprotection processes. Unfortunately, despite many years of intensive studies, the pathogenesis of AD is unknown and therapy for AD and other neurodegenerative diseases is symptomatic, presenting a significant goal and challenge today. This review presents recent achievements in therapeutic approaches for AD, which are targeting PPAR-α and its relation to cholesterol and neurosteroids in AD and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sylwia Żulińska
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| | - Anna K. Strosznajder
- Department of Psychiatry, Medical University of Warsaw, Nowowiejska St. 27, 00-665 Warsaw, Poland;
| | - Joanna B. Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
14
|
Miao L, Zhou Y, Tan D, Zhou C, Ruan CC, Wang S, Wang Y, Vong CT, Cheang WS. Ginsenoside Rk1 improves endothelial function in diabetes through activating peroxisome proliferator-activated receptors. Food Funct 2024; 15:5485-5495. [PMID: 38690748 DOI: 10.1039/d3fo05222b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Ginsenoside Rk1, one kind of ginsenoside, is a minor ginsenoside found in Panax ginseng and used as traditional Chinese medicine for centuries. It exhibits anti-tumor and anti-aggregation effects. However, little research has been done on its effect on endothelial function. This study investigated whether ginsenoside Rk1 improved endothelial dysfunction in diabetes and the underlying mechanisms in vivo and in vitro. Male C57BL/6 mice were fed with a 12 week high-fat diet (60% kcal % fat), whereas treatment groups were orally administered with ginsenoside Rk1 (10 and 20 mg per kg per day) in the last 4 weeks. Aortas isolated from C57BL/6 mice were induced by high glucose (HG; 30 mM) and co-treated with or without ginsenoside Rk1 (1 and 10 μM) for 48 h ex vivo. Moreover, primary rat aortic endothelial cells (RAECs) were cultured and stimulated by HG (44 mM) to mimic hyperglycemia, with or without the co-treatment of ginsenoside Rk1 (10 μM) for 48 h. Endothelium-dependent relaxations of mouse aortas were damaged with elevated oxidative stress and downregulation of three isoforms of peroxisome proliferator-activated receptors (PPARs), PPAR-α, PPAR-β/δ, and PPAR-γ, as well as endothelial nitric oxide synthase (eNOS) phosphorylation due to HG or high-fat diet stimulation, which also existed in RAECs. However, after the treatment with ginsenoside Rk1, these impairments were all ameliorated significantly. Moreover, the vaso-protective and anti-oxidative effects of ginsenoside Rk1 were abolished by PPAR antagonists (GSK0660, GW9662 or GW6471). In conclusion, this study reveals that ginsenoside Rk1 ameliorates endothelial dysfunction and suppresses oxidative stress in diabetic vasculature through activating the PPAR/eNOS pathway.
Collapse
Affiliation(s)
- Lingchao Miao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| | - Yan Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| | - Dechao Tan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
- Macau Centre for Research and Development in Chinese Medicine, University of Macau, Macau SAR, China
| | - Chunxiu Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
- Macau Centre for Research and Development in Chinese Medicine, University of Macau, Macau SAR, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
- Macau Centre for Research and Development in Chinese Medicine, University of Macau, Macau SAR, China
| | - Chi Teng Vong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
- Macau Centre for Research and Development in Chinese Medicine, University of Macau, Macau SAR, China
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
15
|
Ramamurthy K, Priya PS, Murugan R, Arockiaraj J. Hues of risk: investigating genotoxicity and environmental impacts of azo textile dyes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:33190-33211. [PMID: 38676865 DOI: 10.1007/s11356-024-33444-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
The textile industry, with its extensive use of dyes and chemicals, stands out as a significant source of water pollution. Exposure to certain textile dyes, such as azo dyes and their breakdown products like aromatic amines, has been associated with health concerns like skin sensitization, allergic reactions, and even cancer in humans. Annually, the worldwide production of synthetic dyes approximates 7 × 107 tons, of which the textile industry accounts for over 10,000 tons. Inefficient dyeing procedures result in the discharge of 15-50% of azo dyes, which do not adequately bind to fibers, into wastewater. This review delves into the genotoxic impact of azo dyes, prevalent in the textile industry, on aquatic ecosystems and human health. Examining different families of textile dye which contain azo group in their structure such as Sudan I and Sudan III Sudan IV, Basic Red 51, Basic Violet 14, Disperse Yellow 7, Congo Red, Acid Red 26, and Acid Blue 113 reveals their carcinogenic potential, which may affect both industrial workers and aquatic life. Genotoxic and carcinogenic characteristics, chromosomal abnormalities, induced physiological and neurobehavioral changes, and disruptions to spermatogenesis are evident, underscoring the harmful effects of these dyes. The review calls for comprehensive investigations into the toxic profile of azo dyes, providing essential insights to safeguard the aquatic ecosystem and human well-being. The importance of effective effluent treatment systems is underscored to mitigate adverse impacts on agricultural lands, water resources, and the environment, particularly in regions heavily reliant on wastewater irrigation for food production.
Collapse
Affiliation(s)
- Karthikeyan Ramamurthy
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur, 603203, Tamil Nadu, India
| | - Peter Snega Priya
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur, 603203, Tamil Nadu, India
| | - Raghul Murugan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur, 603203, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur, 603203, Tamil Nadu, India.
| |
Collapse
|
16
|
Sharma S, Sharma D, Dhobi M, Wang D, Tewari D. An insight to treat cardiovascular diseases through phytochemicals targeting PPAR-α. Mol Cell Biochem 2024; 479:707-732. [PMID: 37171724 DOI: 10.1007/s11010-023-04755-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Peroxisome proliferator-activated receptor-α (PPAR-α) belonging to the nuclear hormone receptor superfamily is a promising target for CVDs which mechanistically improves the production of high-density lipid as well as inhibit vascular smooth muscle cell proliferation. PPAR-α mainly interferes with adenosine monophosphate-activated protein kinase, transforming growth factor-β-activated kinase, and nuclear factor-κB pathways to protect against cardiac complications. Natural products/extracts could serve as a potential therapeutic strategy in CVDs for targeting PPAR-α with broad safety margins. In recent years, the understanding of naturally derived PPAR-α agonists has considerably improved; however, the information is scattered. In vitro and in vivo studies on acacetin, apigenin, arjunolic acid, astaxanthin, berberine, resveratrol, vaticanol C, hispidulin, ginsenoside Rb3, and genistein showed significant effects in CVDs complications by targeting PPAR-α. With the aim of demonstrating the tremendous chemical variety of natural products targeting PPAR-α in CVDs, this review provides insight into various natural products that can work to prevent CVDs by targeting the PPAR-α receptor along with their detailed mechanism.
Collapse
Affiliation(s)
- Supriya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Divya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Mahaveer Dhobi
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| |
Collapse
|
17
|
Zhang Y, Xiao B, Liu Y, Wu S, Xiang Q, Xiao Y, Zhao J, Yuan R, Xie K, Li L. Roles of PPAR activation in cancer therapeutic resistance: Implications for combination therapy and drug development. Eur J Pharmacol 2024; 964:176304. [PMID: 38142851 DOI: 10.1016/j.ejphar.2023.176304] [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: 09/17/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 12/26/2023]
Abstract
Therapeutic resistance is a major obstacle to successful treatment or effective containment of cancer. Peroxisome proliferator-activated receptors (PPARs) play an essential role in regulating energy homeostasis and determining cell fate. Despite of the pleiotropic roles of PPARs in cancer, numerous studies have suggested their intricate relationship with therapeutic resistance in cancer. In this review, we provided an overview of the roles of excessively activated PPARs in promoting resistance to modern anti-cancer treatments, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy. The mechanisms through which activated PPARs contribute to therapeutic resistance in most cases include metabolic reprogramming, anti-oxidant defense, anti-apoptosis signaling, proliferation-promoting pathways, and induction of an immunosuppressive tumor microenvironment. In addition, we discussed the mechanisms through which activated PPARs lead to multidrug resistance in cancer, including drug efflux, epithelial-to-mesenchymal transition, and acquisition and maintenance of the cancer stem cell phenotype. Preliminary studies investigating the effect of combination therapies with PPAR antagonists have suggested the potential of these antagonists in reversing resistance and facilitating sustained cancer management. These findings will provide a valuable reference for further research on and clinical translation of PPAR-targeting treatment strategies.
Collapse
Affiliation(s)
- Yanxia Zhang
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China; Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yunduo Liu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Qin Xiang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yuhan Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Junxiu Zhao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Ruanfei Yuan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Keping Xie
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China.
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| |
Collapse
|
18
|
Wang F, Liu Y, Dong Y, Zhao M, Huang H, Jin J, Fan L, Xiang R. Haploinsufficiency of Lipin3 leads to hypertriglyceridemia and obesity by disrupting the expression and nucleocytoplasmic localization of Lipin1. Front Med 2024; 18:180-191. [PMID: 37776435 DOI: 10.1007/s11684-023-1003-0] [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: 12/16/2022] [Accepted: 04/27/2023] [Indexed: 10/02/2023]
Abstract
Lipin proteins including Lipin 1-3 act as transcriptional co-activators and phosphatidic acid phosphohydrolase enzymes, which play crucial roles in lipid metabolism. However, little is known about the function of Lipin3 in triglyceride (TG) metabolism. Here, we identified a novel mutation (NM_001301860: p.1835A>T/p.D612V) of Lipin3 in a large family with hypertriglyceridemia (HTG) and obesity through whole-exome sequencing and Sanger sequencing. Functional studies revealed that the novel variant altered the half-life and stability of the Lipin3 protein. Hence, we generated Lipin3 heterozygous knockout (Lipin3-heKO) mice and cultured primary hepatocytes to explore the pathophysiological roles of Lipin3 in TG metabolism. We found that Lipin3-heKO mice exhibited obvious obesity, HTG, and non-alcoholic fatty liver disorder. Mechanistic study demonstrated that the haploinsufficiency of Lipin3 in primary hepatocytes may induce the overexpression and abnormal distribution of Lipin1 in cytosol and nucleoplasm. The increased expression of Lipin1 in cytosol may contribute to TG anabolism, and the decreased Lipin1 in nucleoplasm can reduce PGC1α, further leading to mitochondrial dysfunction and reduced TG catabolism. Our study suggested that Lipin3 was a novel disease-causing gene inducing obesity and HTG. We also established a relationship between Lipin3 and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Fang Wang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Yuxing Liu
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
- Department of Cellular Biology, School of Life Sciences, Key Laboratory of Pediatric Rare Diseases, Ministry of Education, Central South University, Changsha, 410013, China
| | - Yi Dong
- Department of Cellular Biology, School of Life Sciences, Key Laboratory of Pediatric Rare Diseases, Ministry of Education, Central South University, Changsha, 410013, China
| | - Meifang Zhao
- Department of Cellular Biology, School of Life Sciences, Key Laboratory of Pediatric Rare Diseases, Ministry of Education, Central South University, Changsha, 410013, China
| | - Hao Huang
- Department of Cellular Biology, School of Life Sciences, Key Laboratory of Pediatric Rare Diseases, Ministry of Education, Central South University, Changsha, 410013, China
| | - Jieyuan Jin
- Department of Cellular Biology, School of Life Sciences, Key Laboratory of Pediatric Rare Diseases, Ministry of Education, Central South University, Changsha, 410013, China
| | - Liangliang Fan
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
- Department of Cellular Biology, School of Life Sciences, Key Laboratory of Pediatric Rare Diseases, Ministry of Education, Central South University, Changsha, 410013, China.
| | - Rong Xiang
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
- Department of Cellular Biology, School of Life Sciences, Key Laboratory of Pediatric Rare Diseases, Ministry of Education, Central South University, Changsha, 410013, China.
| |
Collapse
|
19
|
Adarthaiya S, Sehgal A. Moringa oleifera Lam. as a potential plant for alleviation of the metabolic syndrome-A narrative review based on in vivo and clinical studies. Phytother Res 2024; 38:755-775. [PMID: 38015048 DOI: 10.1002/ptr.8079] [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: 04/27/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023]
Abstract
The metabolic syndrome (MetS) refers to the co-occurrence of risk factors, including hyperglycaemia, increased body weight, hypertension and dyslipidemia, which eventually lead to diabetes and cardiovascular disease, a common health problem worldwide. Recently, there has been an increasing interest in the use of plant-based products for the management of MetS, because of their less detrimental and more beneficial effects. Moringa oleifera (Moringaceae), commonly known as drumstick, is cultivated worldwide for its nutritional and medicinal properties. This review focuses on the in vivo and human studies concerning the potential of M. oleifera in the alleviation of MetS and its comorbidities. The search for relevant articles was carried out in PubMed and Google Scholar databases. Randomised controlled and clinical trials from the PubMed database were included in this review. The results suggested that the administration of M. oleifera, in vivo, shows clear signs of improvement in MetS indices. Despite fewer human studies, the existing data documented convincing results that uphold the potential of M. oleifera against MetS. Therefore, future research discussing the probable mechanism of action is much needed which could further assure the usage of M. oleifera in the treatment regimen of MetS.
Collapse
Affiliation(s)
- Saikrupa Adarthaiya
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| | - Amit Sehgal
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| |
Collapse
|
20
|
Xia J, Wang Z, Yu P, Yan X, Zhao J, Zhang G, Gong D, Zeng Z. Effect of Different Medium-Chain Triglycerides on Glucose Metabolism in High-Fat-Diet Induced Obese Rats. Foods 2024; 13:241. [PMID: 38254542 PMCID: PMC10815142 DOI: 10.3390/foods13020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Obesity can be associated with significant metabolic disorders. Our previous study found that medium-chain triglycerides (MCTs) improved lipid metabolism in obese rats. However, scant attention has been given to exploring the impact of MCTs on glucose metabolism in obese rats. This study is designed to examine the effects and mechanisms of three distinct MCTs on glucose metabolism in obese rats. To induce obesity, Sprague-Dawley (SD) rats were fed a high-fat diet, followed by a 12-week treatment with caprylic triglyceride (CYT), capric triglyceride (CT), and lauric triglyceride (LT). The results showed that three types of MCT intervention reduced the levels of lipids (TC, TG, LDL-c, and HDL-c), hyperglycemia, insulin resistance (insulin, OGTT, HOMA-IR, and ISI), and inflammatory markers (IL-4, IL-1β, and TNF-α) in obese rats (p < 0.01), The above parameters have been minimally improved in the high-fat restoring group (HR) group. MCTs can modulate the PI3K/AKT signaling pathways to alleviate insulin resistance and improve glucose metabolism in obese rats. Furthermore, MCTs can activate peroxisome proliferator-activated receptor (PPAR) γ and reduce the phosphorylation of PPARγser237 mediated by CDK5, which can improve insulin sensitivity without lipid deposition in obese rats. Among the MCT group, CT administration performed the best in the above pathways, with the lowest blood glucose level and insulin resistance. These findings contribute to a deeper understanding of the connection between health benefits and the specific type of MCT employed.
Collapse
Affiliation(s)
- Jiaheng Xia
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, China; (J.X.); (Z.W.)
| | - Zhixin Wang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, China; (J.X.); (Z.W.)
| | - Ping Yu
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, China; (J.X.); (Z.W.)
- Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, Nanchang University, Nanchang 330031, China
| | - Xianghui Yan
- School of Resources and Environment, Nanchang University, Nanchang 330031, China;
| | - Junxin Zhao
- School of Food Science and Technology, Nanchang University, Nanchang 330031, China;
| | - Guohua Zhang
- Institute of Biological Resources, Jiangxi Academy of Sciences, Nanchang 330096, China;
| | - Deming Gong
- New Zealand Institute of Natural Medicine Research, 8 Ha Crescent, Auckland 2104, New Zealand;
| | - Zheling Zeng
- Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, Nanchang University, Nanchang 330031, China
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang 330047, China
| |
Collapse
|
21
|
Paneth A, Kaproń B, Plech T, Paduch R, Trotsko N, Paneth P. Combined In Silico and In Vitro Analyses to Assess the Anticancer Potential of Thiazolidinedione-Thiosemicarbazone Hybrid Molecules. Int J Mol Sci 2023; 24:17521. [PMID: 38139350 PMCID: PMC10743653 DOI: 10.3390/ijms242417521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
The number of people affected by cancer and antibiotic-resistant bacterial infections has increased, such that both diseases are already seen as current and future leading causes of death globally. To address this issue, based on a combined in silico and in vitro approach, we explored the anticancer potential of known antibacterials with a thiazolidinedione-thiosemicarbazone (TZD-TSC) core structure. A cytotoxicity assessment showed encouraging results for compounds 2-4, with IC50 values against T98G and HepG2 cells in the low micromolar range. TZD-TSC 3 proved to be most toxic to cancer cell lines, with IC50 values of 2.97 ± 0.39 µM against human hepatoma HepG2 cells and IC50 values of 28.34 ± 2.21 µM against human glioblastoma T98G cells. Additionally, compound 3 induced apoptosis and showed no specific hemolytic activity. Furthermore, treatment using 3 on cancer cell lines alters these cells' morphology and further suppresses migratory activity. Molecular docking, in turn, suggests that 3 would have the capacity to simultaneously target HDACs and PPARγ, by the activation of PPARγ and the inhibition of both HDAC4 and HDAC8. Thus, the promising preliminary results obtained with TZD-TSC 3 represent an encouraging starting point for the rational design of novel chemotherapeutics with dual antibacterial and anticancer activities.
Collapse
Affiliation(s)
- Agata Paneth
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Barbara Kaproń
- Department of Clinical Genetics, Faculty of Medicine, Medical University of Lublin, 20-080 Lublin, Poland
| | - Tomasz Plech
- Department of Pharmacology, Faculty of Health Sciences, Medical University of Lublin, 20-080 Lublin, Poland;
| | - Roman Paduch
- Department of Virology and Immunology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, 20-033 Lublin, Poland;
| | - Nazar Trotsko
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Piotr Paneth
- Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, 90-924 Lodz, Poland
| |
Collapse
|
22
|
Wu J, Fan Q, He Q, Zhong Q, Zhu X, Cai H, He X, Xu Y, Huang Y, Di X. Potential drug targets for myocardial infarction identified through Mendelian randomization analysis and Genetic colocalization. Medicine (Baltimore) 2023; 102:e36284. [PMID: 38065874 PMCID: PMC10713171 DOI: 10.1097/md.0000000000036284] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Myocardial infarction (MI) is a major cause of death and disability worldwide, but current treatments are limited by their invasiveness, side effects, and lack of efficacy. Novel drug targets for MI prevention are urgently needed. In this study, we used Mendelian randomization to identify potential therapeutic targets for MI using plasma protein quantitative trait loci as exposure variables and MI as the outcome variable. We further validated our findings using reverse causation analysis, Bayesian co-localization analysis, and external datasets. We also constructed a protein-protein interaction network to explore the relationships between the identified proteins and known MI targets. Our analysis revealed 2 proteins, LPA and APOA5, as potential drug targets for MI, with causal effects on MI risk confirmed by multiple lines of evidence. LPA and APOA5 are involved in lipid metabolism and interact with target proteins of current MI medications. We also found 4 other proteins, IL1RN, FN1, NT5C, and SEMA3C, that may have potential as drug targets but require further confirmation. Our study demonstrates the utility of Mendelian randomization and protein quantitative trait loci in discovering novel drug targets for complex diseases such as MI. It provides insights into the underlying mechanisms of MI pathology and treatment.
Collapse
Affiliation(s)
- Jiayu Wu
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiaoming Fan
- Clifford Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi He
- The Eighth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Zhong
- The First Affiliated Hospital of Jinzhou Medical University, China
| | - Xianqiong Zhu
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, China
| | - Huilian Cai
- Clifford Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaolin He
- Clifford Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Xu
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuxuan Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xingwei Di
- The First Affiliated Hospital of Jinzhou Medical University, China
| |
Collapse
|
23
|
Liu X, Xu X, Zhang T, Xu L, Tao H, Liu Y, Zhang Y, Meng X. Fatty acid metabolism disorders and potential therapeutic traditional Chinese medicines in cardiovascular diseases. Phytother Res 2023; 37:4976-4998. [PMID: 37533230 DOI: 10.1002/ptr.7965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/13/2023] [Accepted: 07/07/2023] [Indexed: 08/04/2023]
Abstract
Cardiovascular diseases are currently the primary cause of mortality in the whole world. Growing evidence indicated that the disturbances in cardiac fatty acid metabolism are crucial contributors in the development of cardiovascular diseases. The abnormal cardiac fatty acid metabolism usually leads to energy deficit, oxidative stress, excessive apoptosis, and inflammation. Targeting fatty acid metabolism has been regarded as a novel approach to the treatment of cardiovascular diseases. However, there are currently no specific drugs that regulate fatty acid metabolism to treat cardiovascular diseases. Many traditional Chinese medicines have been widely used to treat cardiovascular diseases in clinics. And modern studies have shown that they exert a cardioprotective effect by regulating the expression of key proteins involved in fatty acid metabolism, such as peroxisome proliferator-activated receptor α and carnitine palmitoyl transferase 1. Hence, we systematically reviewed the relationship between fatty acid metabolism disorders and four types of cardiovascular diseases including heart failure, coronary artery disease, cardiac hypertrophy, and diabetic cardiomyopathy. In addition, 18 extracts and eight monomer components from traditional Chinese medicines showed cardioprotective effects by restoring cardiac fatty acid metabolism. This work aims to provide a reference for the finding of novel cardioprotective agents targeting fatty acid metabolism.
Collapse
Affiliation(s)
- Xianfeng Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Xinmei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Tao Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Lei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Honglin Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Yue Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, Sichuan, People's Republic of China
| |
Collapse
|
24
|
Lin C, Li ZL, Cai XL, Hu SY, Lv F, Yang WJ, Ji LN. Indirect comparison of efficacy and safety of chiglitazar and thiazolidinedione in patients with type 2 diabetes: A meta-analysis. World J Diabetes 2023; 14:1573-1584. [PMID: 37970134 PMCID: PMC10642417 DOI: 10.4239/wjd.v14.i10.1573] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/22/2023] [Accepted: 08/17/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND Chiglitazar is an emerging pan-agonist of all peroxisome proliferator activated receptors (PPAR)-α, δ and γ, and has therapeutic potential for type 2 diabetes (T2D). However, to date, no clinical studies or meta-analyses have compared the efficacy and safety of chiglitazar and traditional PPAR-γ agonist thiazolidinediones (TZDs). A meta-analysis concerning this topic is therefore required. AIM To compare the efficacy and safety of chiglitazar and TZD in patients with T2D. METHODS PubMed, Medline, Embase, the Cochrane Central Register of Controlled Trials, Reference Citation Analysis and Clinicaltrial.gov websites were searched from August 1994 to March 2022. Randomized controlled trials (RCTs) of chiglitazar or TZD vs placebo in patients with T2D were included. Indirect comparisons and sensitivity analyses were implemented to evaluate multiple efficacy and safety endpoints of interest. RESULTS We included 93 RCTs that compared TZD with placebo and one that compared chiglitazar with placebo. For efficacy endpoints, the augmented dose of chig-litazar resulted in greater reductions in hemoglobin (Hb)A1c [weighted mean difference (WMD) = -0.15%, 95% confidence interval (CI): -0.27 to -0.04%], triglycerides (WMD = -0.17 mmol/L, 95%CI: -0.24 to -0.11 mmol/L) and alanine aminotransferase (WMD = -5.25 U/L, 95%CI: -8.50 to -1.99 U/L), and a greater increase in homeostasis model assessment-β (HOMA-β) (WMD = 17.75, 95%CI: 10.73-24.77) when compared with TZD treatment. For safety endpoints, the risks of hypoglycemia, edema, bone fractures, upper respiratory tract infection, urinary tract infection, and weight gain were all comparable between the augmented dose of chiglitazar and TZD. In patients with baseline HbA1c ≥ 8.5%, body mass index ≥ 30 kg/m2 or diabetes duration < 10 years, the HbA1c reduction and HOMA-β increase were more conspicuous for the augmented dose of chiglitazar compared with TZD. CONCLUSION Augmented dose of chiglitazar, a pan-activator of PPARs, may serve as an antidiabetic agent with preferable glycemic and lipid control, better β-cell function preserving capacity, and does not increase the risk of safety concerns when compared with TZD.
Collapse
Affiliation(s)
- Chu Lin
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Zong-Lin Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Xiao-Ling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Sui-Yuan Hu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Fang Lv
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Wen-Jia Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| | - Li-Nong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
25
|
Fuior EV, Zvintzou E, Filippatos T, Giannatou K, Mparnia V, Simionescu M, Gafencu AV, Kypreos KE. Peroxisome Proliferator-Activated Receptor α in Lipoprotein Metabolism and Atherosclerotic Cardiovascular Disease. Biomedicines 2023; 11:2696. [PMID: 37893070 PMCID: PMC10604751 DOI: 10.3390/biomedicines11102696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are a group of ligand-binding transcription factors with pivotal action in regulating pleiotropic signaling pathways of energetic metabolism, immune responses and cell proliferation and differentiation. A significant body of evidence indicates that the PPARα receptor is an important modulator of plasma lipid and lipoprotein metabolism, with pluripotent effects influencing the lipid and apolipoprotein cargo of both atherogenic and antiatherogenic lipoproteins and their functionality. Clinical evidence supports an important role of PPARα agonists (fibric acid derivatives) in the treatment of hypertriglyceridemia and/or low high-density lipoprotein (HDL) cholesterol levels, although the effects of clinical trials are contradictory and point to a reduction in the risk of nonfatal and fatal myocardial infarction events. In this manuscript, we provide an up-to-date critical review of the existing relevant literature.
Collapse
Affiliation(s)
- Elena Valeria Fuior
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Evangelia Zvintzou
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Theodosios Filippatos
- Internal Medicine Clinic, Department of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Katerina Giannatou
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Victoria Mparnia
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Anca Violeta Gafencu
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
| | - Kyriakos E. Kypreos
- Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (E.V.F.); (E.Z.); (M.S.)
- Pharmacology Laboratory, Department of Medicine, University of Patras, 26500 Rio Achaias, Greece; (K.G.); (V.M.)
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
| |
Collapse
|
26
|
Jurgelewicz A, Nault R, Harkema J, Zacharewski TR, LaPres JJ. Characterizing the impact of simvastatin co-treatment of cell specific TCDD-induced gene expression and systemic toxicity. Sci Rep 2023; 13:16598. [PMID: 37789023 PMCID: PMC10547718 DOI: 10.1038/s41598-023-42972-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/17/2023] [Indexed: 10/05/2023] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is associated with metabolic syndrome (MetS) in humans and elicits pathologies in rodents that resemble non-alcoholic fatty liver disease (NAFLD) in humans through activation of the aryl hydrocarbon receptor (AHR) pathway. Dysregulation of cholesterol homeostasis, an aspect of MetS, is linked to NAFLD pathogenesis. TCDD exposure is also linked to the suppression of genes that encode key cholesterol biosynthesis steps and changes in serum cholesterol levels. In a previous experiment, treating mice with TCDD in the presence of simvastatin, a 3-Hydroxy-3-Methylglutaryl-CoA Reductase competitive inhibitor, altered lipid and glycogen levels, AHR-battery gene expression, and liver injury in male mice compared to TCDD alone. The aim of this study was to deduce a possible mechanism(s) for the metabolic changes and increased injury using single-nuclei RNA sequencing in mouse liver. We demonstrated that co-treated mice experienced wasting and increased AHR activation compared to TCDD alone. Furthermore, relative proportions of cell (sub)types were different between TCDD alone and co-treated mice including important mediators of NAFLD progression like hepatocytes and immune cell populations. Analysis of non-overlapping differentially expressed genes identified several pathways where simvastatin co-treatment significantly impacted TCDD-induced changes, which may explain the differences between treatments. Overall, these results demonstrate a connection between dysregulation of cholesterol homeostasis and toxicant-induced metabolic changes.
Collapse
Affiliation(s)
- Amanda Jurgelewicz
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicological Sciences, Michigan State University, East Lansing, MI, USA
| | - Rance Nault
- Institute for Integrative Toxicological Sciences, Michigan State University, East Lansing, MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, 602 Wilson Rd, East Lansing, MI, 48824, USA
| | - Jack Harkema
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Timothy R Zacharewski
- Institute for Integrative Toxicological Sciences, Michigan State University, East Lansing, MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, 602 Wilson Rd, East Lansing, MI, 48824, USA
| | - John J LaPres
- Institute for Integrative Toxicological Sciences, Michigan State University, East Lansing, MI, USA.
- Department of Biochemistry and Molecular Biology, Michigan State University, 602 Wilson Rd, East Lansing, MI, 48824, USA.
| |
Collapse
|
27
|
YIN Y, TANG H, FANG Y, LIU W, WANG J, HU Y, PENG J. Hepatic transcriptome delineates the therapeutic effects of Sanren Tang on high-fat diet-induced non-alcoholic fatty liver disease. J TRADIT CHIN MED 2023; 43:1092-1102. [PMID: 37946471 PMCID: PMC10623253 DOI: 10.19852/j.cnki.jtcm.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 12/25/2022] [Indexed: 11/12/2023]
Abstract
OBJECTIVE To evaluate the effects of Sanren Tang (SRT, ) on a high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) in mice and to investigate the hepatic transcriptome regulated by SRT. METHODS The primary SRT components were identified using ultra-high-performance liquid chromatography-high-resolution accurate mass spectrometry. The SRT-induced pharmacological effects on HFD-induced NAFLD were evaluated in mice for 16 weeks. Obeticholic acid was used as a control drug. Body weight, food intake, and homeostatic model assessment for insulin resistance (HOMA-IR) index were analysed. Hepatic histological changes were observed in haematoxylin and eosin-stained sections and quantified using the NAFLD activity score (NAS). Serum alanine aminotransferase (ALT) and hepatic triglyceride (TG) levels were measured. Lipids in hepatocytes were visualised by Oil red staining. RNA-sequencing was performed to determine the transcriptome profile of the liver tissue. The differentially expressed genes were validated using real-time polymerase chain reaction and Western blotting. RESULTS Four principal compounds were identified in the SRT: adenosine, amygdalin, luteoloside, and magnolol. SRT ameliorated hepatic histology and lipid deposition in the NAFLD mice, and decreased HOMA-IR, NAS and ALT, and hepatic TG levels. Hepatic transcriptome analysis revealed 232 HFD-regulated genes that were reversed by SRT simultaneously. Retinol metabolism, cytokine-cytokine receptor interaction, and peroxisome proliferator-activated receptor (PPAR) γ signalling were the top three SRT-regulated pathways in NAFLD. CONCLUSIONS SRT significantly ameliorated HFD-induced NAFLD, which was correlated with the regulation of genes enriched in the retinol metabolism, cytokine-cytokine receptor interaction, and PPARγ signalling pathways.
Collapse
Affiliation(s)
- Yixiao YIN
- 1 Institute of Liver diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- 2 Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai 201203, China
- 3 Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Hao TANG
- 1 Institute of Liver diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- 2 Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai 201203, China
- 3 Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Yi FANG
- 1 Institute of Liver diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- 2 Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai 201203, China
- 3 Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Wei LIU
- 1 Institute of Liver diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- 2 Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai 201203, China
- 3 Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Jun WANG
- 1 Institute of Liver diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- 2 Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai 201203, China
- 3 Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Yiyang HU
- 4 Institute of Clinical Pharmacology, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- 5 Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai 201203, China
- 6 Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| | - Jinghua PENG
- 1 Institute of Liver diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- 2 Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai 201203, China
- 3 Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, China
| |
Collapse
|
28
|
Chambers TL, Stroh AM, Chavez C, Brandt AR, Claiborne A, Fountain WA, Gries KJ, Jones AM, Kuszmaul DJ, Lee GA, Lester BE, Lynch CE, Minchev K, Montenegro CF, Naruse M, Raue U, Trappe TA, Trappe S. Multitissue responses to exercise: a MoTrPAC feasibility study. J Appl Physiol (1985) 2023; 135:302-315. [PMID: 37318985 PMCID: PMC10393343 DOI: 10.1152/japplphysiol.00210.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023] Open
Abstract
We assessed the feasibility of the Molecular Transducers of Physical Activity Consortium (MoTrPAC) human adult clinical exercise protocols, while also documenting select cardiovascular, metabolic, and molecular responses to these protocols. After phenotyping and familiarization sessions, 20 subjects (25 ± 2 yr, 12 M, 8 W) completed an endurance exercise bout (n = 8, 40 min cycling at 70% V̇o2max), a resistance exercise bout (n = 6, ∼45 min, 3 sets of ∼10 repetition maximum, 8 exercises), or a resting control period (n = 6, 40 min rest). Blood samples were taken before, during, and after (10 min, 2 h, and 3.5 h) exercise or rest for levels of catecholamines, cortisol, glucagon, insulin, glucose, free fatty acids, and lactate. Heart rate was recorded throughout exercise (or rest). Skeletal muscle (vastus lateralis) and adipose (periumbilical) biopsies were taken before and ∼4 h following exercise or rest for mRNA levels of genes related to energy metabolism, growth, angiogenesis, and circadian processes. Coordination of the timing of procedural components (e.g., local anesthetic delivery, biopsy incisions, tumescent delivery, intravenous line flushes, sample collection and processing, exercise transitions, and team dynamics) was reasonable to orchestrate while considering subject burden and scientific objectives. The cardiovascular and metabolic alterations reflected a dynamic and unique response to endurance and resistance exercise, whereas skeletal muscle was transcriptionally more responsive than adipose 4 h postexercise. In summary, the current report provides the first evidence of protocol execution and feasibility of key components of the MoTrPAC human adult clinical exercise protocols. Scientists should consider designing exercise studies in various populations to interface with the MoTrPAC protocols and DataHub.NEW & NOTEWORTHY This study highlights the feasibility of key aspects of the MoTrPAC adult human clinical protocols. This initial preview of what can be expected from acute exercise trial data from MoTrPAC provides an impetus for scientists to design exercise studies to interlace with the rich phenotypic and -omics data that will populate the MoTrPAC DataHub at the completion of the parent protocol.
Collapse
Affiliation(s)
- Toby L Chambers
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Andrew M Stroh
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Clarisa Chavez
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Anna R Brandt
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Alex Claiborne
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - William A Fountain
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Kevin J Gries
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Andrew M Jones
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Dillon J Kuszmaul
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Gary A Lee
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Bridget E Lester
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Colleen E Lynch
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Kiril Minchev
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | | | - Masatoshi Naruse
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Ulrika Raue
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Todd A Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Scott Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| |
Collapse
|
29
|
Apaza Ticona L, Sánchez Sánchez-Corral J, Flores Sepúlveda A, Soriano Vázquez C, Hernán Vieco C, Rumbero Sánchez Á. Novel 1,2,4-oxadiazole compounds as PPAR-α ligand agonists: a new strategy for the design of antitumour compounds. RSC Med Chem 2023; 14:1377-1388. [PMID: 37484563 PMCID: PMC10357926 DOI: 10.1039/d3md00063j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/22/2023] [Indexed: 07/25/2023] Open
Abstract
Modulation of PPAR-α by natural ligands is a novel strategy for the development of anticancer therapies. A series of 16 compounds based on the structure of 3-(pyridin-3-yl)-5-(thiophen-3-yl)-1,2,4-oxadiazole (natural compound) with antitumour potential were designed and synthesised. The cytotoxicity and PPAR agonist activity of these synthetic 1,2,4-oxadiazoles were evaluated in the A-498 and DU 145 tumour cell lines. Preliminary biological evaluation showed that most of these synthetic 1,2,4-oxadiazoles are less cytotoxic (sulforhodamine B assay) than the positive control WY-14643. Regarding the PPAR-α modulation, compound 16 was the most active, with EC50 = 0.23-0.83 μM (PPAR-α). Additionally, compound 16 had a similar activity to the natural compound (EC50 = 0.18-0.77 μM) and was less toxic in the RPTEC and WPMY-1 cell lines (non-tumour cells) (CC50 = 81.66-92.67 μM) than the natural compound. Looking at the link between chemical structure and activity, our study demonstrates that changes to the natural 1,2,4-oxadiazole at the level of the thiophenyl residue can lead to new agonists of PPAR-α with promising anti-tumour activity.
Collapse
Affiliation(s)
- Luis Apaza Ticona
- Organic Chemistry Unit, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Universidad Complutense of Madrid Plaza Ramón y Cajal s/n 28040 Madrid Spain
- Department of Organic Chemistry, Faculty of Sciences, University Autónoma of Madrid Cantoblanco 28040 Madrid Spain
| | | | | | - Carmen Soriano Vázquez
- Faculty of Pharmacy, Universidad Complutense of Madrid Plaza Ramón y Cajal s/n 28040 Madrid Spain
| | - Carmen Hernán Vieco
- Faculty of Pharmacy, Universidad Complutense of Madrid Plaza Ramón y Cajal s/n 28040 Madrid Spain
| | - Ángel Rumbero Sánchez
- Department of Organic Chemistry, Faculty of Sciences, University Autónoma of Madrid Cantoblanco 28040 Madrid Spain
| |
Collapse
|
30
|
Messina A, Concerto C, Rodolico A, Petralia A, Caraci F, Signorelli MS. Is It Time for a Paradigm Shift in the Treatment of Schizophrenia? The Use of Inflammation-Reducing and Neuroprotective Drugs-A Review. Brain Sci 2023; 13:957. [PMID: 37371435 DOI: 10.3390/brainsci13060957] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/03/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Comprehending the pathogenesis of schizophrenia represents a challenge for global mental health. To date, although it is evident that alterations in dopaminergic, serotonergic, and glutamatergic neurotransmission underlie the clinical expressiveness of the disease, neuronal disconnections represent only an epiphenomenon. In recent years, several clinical studies have converged on the hypothesis of microglia hyperactivation and a consequent neuroinflammatory state as a pathogenic substrate of schizophrenia. Prenatal, perinatal, and postnatal factors can cause microglia to switch from M2 anti-inflammatory to M1 pro-inflammatory states. A continuous mild neuroinflammatory state progressively leads to neuronal loss, a reduction in dendritic spines, and myelin degeneration. The augmentation of drugs that reduce neuroinflammation to antipsychotics could be an effective therapeutic modality in managing schizophrenia. This review will consider studies in which drugs with anti-inflammatory and neuroprotective properties have been used in addition to antipsychotic treatment in patients with schizophrenia.
Collapse
Affiliation(s)
- Antonino Messina
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Carmen Concerto
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Alessandro Rodolico
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Antonino Petralia
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95123 Catania, Italy
- Unit of Translational Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Maria Salvina Signorelli
- Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
| |
Collapse
|
31
|
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
|
32
|
Kiełbowski K, Bakinowska E, Ostrowski P, Pala B, Gromowska E, Gurazda K, Dec P, Modrzejewski A, Pawlik A. The Role of Adipokines in the Pathogenesis of Psoriasis. Int J Mol Sci 2023; 24:ijms24076390. [PMID: 37047363 PMCID: PMC10094354 DOI: 10.3390/ijms24076390] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Psoriasis is a chronic and immune-mediated skin condition characterized by pro-inflammatory cytokines and keratinocyte hyperproliferation. Dendritic cells, T lymphocytes, and keratinocytes represent the main cell subtypes involved in the pathogenesis of psoriasis, while the interleukin-23 (IL-23)/IL-17 pathway enhances the disease progression. Human adipose tissue is an endocrine organ, which secretes multiple proteins, known as adipokines, such as adiponectin, leptin, visfatin, or resistin. Current evidence highlights the immunomodulatory roles of adipokines, which may contribute to the progression or suppression of psoriasis. A better understanding of the complexity of psoriasis pathophysiology linked with adipokines could result in developing novel diagnostic or therapeutic strategies. This review aims to present the pathogenesis of psoriasis and the roles of adipokines in this process.
Collapse
|
33
|
Wen ZJ, Wei YJ, Zhang YF, Zhang YF. A review of cardiovascular effects and underlying mechanisms of legacy and emerging per- and polyfluoroalkyl substances (PFAS). Arch Toxicol 2023; 97:1195-1245. [PMID: 36947184 DOI: 10.1007/s00204-023-03477-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
Cardiovascular disease (CVD) poses the leading threats to human health and life, and their occurrence and severity are associated with exposure to environmental pollutants. Per- and polyfluoroalkyl substances (PFAS), a group of widely used industrial chemicals, are characterized by persistence, long-distance migration, bioaccumulation, and toxicity. Some PFAS, particularly perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA) and perfluorohexanesulfonic acid (PFHxS), have been banned, leaving only legacy exposure to the environment and human body, while a number of novel PFAS alternatives have emerged and raised concerns, such as polyfluoroalkyl ether sulfonic and carboxylic acid (PFESA and PFECA) and sodium p-perfluorous nonenoxybenzene sulfonate (OBS). Overall, this review systematically elucidated the adverse cardiovascular (CV) effects of legacy and emerging PFAS, emphasized the dose/concentration-dependent, time-dependent, carbon chain length-dependent, sex-specific, and coexposure effects, and discussed the underlying mechanisms and possible prevention and treatment. Extensive epidemiological and laboratory evidence suggests that accumulated serum levels of legacy PFAS possibly contribute to an increased risk of CVD and its subclinical course, such as cardiac toxicity, vascular disorder, hypertension, and dyslipidemia. The underlying biological mechanisms may include oxidative stress, signaling pathway disturbance, lipid metabolism disturbance, and so on. Various emerging alternatives to PFAS also play increasingly prominent toxic roles in CV outcomes that are milder, similar to, or more severe than legacy PFAS. Future research is recommended to conduct more in-depth CV toxicity assessments of legacy and emerging PFAS and explore more effective surveillance, prevention, and treatment strategies, accordingly.
Collapse
Affiliation(s)
- Zeng-Jin Wen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yi-Jing Wei
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yi-Fei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
34
|
Dong Q, Guo Y, Yuan J, Zhong S, Ni H, Liu J, Zhang M, Sun J, Yuan S, Yu H, Zhong Y, Jiang Q. Hexafluoropropylene oxide tetramer acid (HFPO-TeA)-induced developmental toxicities in chicken embryo: Peroxisome proliferator-activated receptor Alpha (PPARα) is involved. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114671. [PMID: 36822062 DOI: 10.1016/j.ecoenv.2023.114671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/07/2023] [Accepted: 02/19/2023] [Indexed: 06/18/2023]
Abstract
Hexafluoropropylene oxide tetramer acid (HFPO-TeA) is an emerging environmental contaminant, with environmental presence but limited toxicological information. To investigate its potential developmental toxicities, various doses of HFPO-TeA exposure were achieved in chicken embryos via air cell injection, and the exposed embryos were incubated until hatch. Within 24 h of hatch, the hatchling chickens were assessed with electrocardiography and histopathology for toxicological evaluation. For mechanistic investigation, in ovo silencing of PPARα was achieved via lentivirus microinjection, then the morphological/functional endpoints along with protein expression levels of PPARα-regulated genes were assessed. HFPO-TeA exposure in chicken embryo resulted in developmental cardiotoxicity and hepatotoxicity. Specifically, decreased right ventricular wall thickness, increased heart rate and hepatic steatosis were observed, whereas silencing of PPARα resulted in alleviation of observed toxicities. Western blotting for EHHADH and FABPs suggested that developmental exposure to HFPO-TeA effectively increased the expression levels of both targets in hatchling chicken heart and liver tissue samples, while PPARα silencing prevented such changes, suggesting that PPARα and its downstream genes are playing critical roles in HFPO-TeA induced developmental toxicities.
Collapse
Affiliation(s)
- Qixuan Dong
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Yajie Guo
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Junhua Yuan
- Department of Special Medicine, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Shuping Zhong
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Hao Ni
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Jingyi Liu
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Mengzhen Zhang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Jiaqi Sun
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Shuqi Yuan
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Huan Yu
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Yuxu Zhong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, China.
| | - Qixiao Jiang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China.
| |
Collapse
|
35
|
Qian Z, Chen L, Liu J, Jiang Y, Zhang Y. The emerging role of PPAR-alpha in breast cancer. Biomed Pharmacother 2023; 161:114420. [PMID: 36812713 DOI: 10.1016/j.biopha.2023.114420] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Breast cancer has been confirmed to have lipid disorders in the tumour microenvironment. Peroxisome proliferator-activated receptor alpha (PPARα) is a ligand-activated transcriptional factor that belongs to the family of nuclear receptors. PPARα regulates the expression of genes involved in fatty acid homeostasis and is a major regulator of lipid metabolism. Because of its effects on lipid metabolism, an increasing number of studies have investigated the relationship of PPARα with breast cancer. PPARα has been shown to impact the cell cycle and apoptosis in normal cells and tumoral cells through regulating genes of the lipogenic pathway, fatty acid oxidation, fatty acid activation, and uptake of exogenous fatty acids. Besides, PPARα is involved in the regulation of the tumour microenvironment (anti-inflammation and inhibition of angiogenesis) by modulating different signal pathways such as NF-κB and PI3K/AKT/mTOR. Some synthetic PPARα ligands are used in adjuvant therapy for breast cancer. PPARα agonists are reported to reduce the side effects of chemotherapy and endocrine therapy. In addition, PPARα agonists enhance the curative effects of targeted therapy and radiation therapy. Interestingly, with the emerging role of immunotherapy, attention has been focused on the tumour microenvironment. The dual functions of PPARα agonists in immunotherapy need further research. This review aims to consolidate the operations of PPARα in lipid-related and other ways, as well as discuss the current and potential applications of PPARα agonists in tackling breast cancer.
Collapse
Affiliation(s)
- Zhiwen Qian
- Department of Oncology, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Lingyan Chen
- Department of Oncology, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Jiayu Liu
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China.
| | - Ying Jiang
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China.
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China; Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China.
| |
Collapse
|
36
|
Meyer-Lindemann U, Moggio A, Dutsch A, Kessler T, Sager HB. The Impact of Exercise on Immunity, Metabolism, and Atherosclerosis. Int J Mol Sci 2023; 24:3394. [PMID: 36834808 PMCID: PMC9967592 DOI: 10.3390/ijms24043394] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Physical exercise represents an effective preventive and therapeutic strategy beneficially modifying the course of multiple diseases. The protective mechanisms of exercise are manifold; primarily, they are elicited by alterations in metabolic and inflammatory pathways. Exercise intensity and duration strongly influence the provoked response. This narrative review aims to provide comprehensive up-to-date insights into the beneficial effects of physical exercise by illustrating the impact of moderate and vigorous exercise on innate and adaptive immunity. Specifically, we describe qualitative and quantitative changes in different leukocyte subsets while distinguishing between acute and chronic exercise effects. Further, we elaborate on how exercise modifies the progression of atherosclerosis, the leading cause of death worldwide, representing a prime example of a disease triggered by metabolic and inflammatory pathways. Here, we describe how exercise counteracts causal contributors and thereby improves outcomes. In addition, we identify gaps that still need to be addressed in the future.
Collapse
Affiliation(s)
- Ulrike Meyer-Lindemann
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Aldo Moggio
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
| | - Alexander Dutsch
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
37
|
Pregnancy Toxemia in Ewes: A Review of Molecular Metabolic Mechanisms and Management Strategies. Metabolites 2023; 13:metabo13020149. [PMID: 36837768 PMCID: PMC9961611 DOI: 10.3390/metabo13020149] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Pregnancy toxemia is a nutritional metabolic disease during late gestation in small ruminants. The condition is characterized by disorders in carbohydrate and fat metabolism. Obese and multiparous ewes are particularly susceptible to pregnancy toxemia, which may lead to maternal death, abortion, or premature birth. Highly productive multiparous meat ewes are major breeding animals, which has led to an increased incidence of the disease. However, the pathogenesis of pregnancy toxemia remains unclear and adequate disease prevention and treatment strategies are absent. Investigating the pathogenesis of pregnancy toxemia, especially the metabolic pathways of hepatic lipids, is key to an improved understanding of the condition. This review provides a snapshot of the genes that are associated with lipid metabolism in the ovine liver, including genes involved in fatty acid oxidation, acetyl coenzyme metabolism, and triglyceride synthesis; describes the interrelationships between these genes; and summarizes the diagnosis, prevention, and treatment of pregnancy toxemia.
Collapse
|
38
|
Xu H, Gao K, Liu C, Li T, Ding Y, Ma J. Pathological mechanism of heart failure with preserved ejection fraction in rats based on iTRAQ technology. PeerJ 2023; 11:e15280. [PMID: 37159835 PMCID: PMC10163871 DOI: 10.7717/peerj.15280] [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: 02/15/2023] [Accepted: 04/03/2023] [Indexed: 05/11/2023] Open
Abstract
Objective Heart failure with preserved ejection fraction (HFpEF) is a public health problem worldwide. Treatments for the patients with HFpEF are not satisfactory because there is no unified understanding of the pathological mechanism of HFpEF. This study aims at investigating the potential pathological mechanism for the effective diagnosis and treatment of HFpEF. Methods Ten adult male Dahl salt sensitive rats (180-200 g) were divided into control and model groups. The rats in model group were fed with high salt diet (8% NaCl) to induce HFpEF for this comparative study. Behavioral changes, biochemical parameters, and histopathological changes of the rats were detected. iTRAQ technology combined with bioinformatics analysis was employed to study the differentially expressed proteins (DEPs) and their enrichment in signaling pathways. Results Echocardiography detection showed decreased LVEF, indicating impaired cardiac function (P < 0.01), increased LVPWd, indicating ventricular wall hypertrophy (P < 0.05), prolonged duration of IVRT and decreased E/A ratio, indicating diastolic dysfunction (P < 0.05) of the rats in model group. 563 DEPs were identified in the rats of both groups, with 243 up-regulated and 320 down-regulated. The expression of PPAR signaling pathway in the rats of model group was down-regulated, with PPARα most significantly decreased (91.2%) (P < 0.01), PPARγ obviously decreased (63.60%) (P < 0.05), and PPARβ/δ decreased (45.33%) (P < 0.05). The DEPs enriched in PPAR signaling pathway were mainly related to such biological processes as fatty acid beta-oxidation, such cellular components as peroxisome, and such molecular functions as lipid binding. Conclusions NaCl high salt diet is one of the factors to increase the incidence of HFpEF in rats. PPARα, PPARγ and PPAR β/δ might be the targets of HFpEF. The findings may provide a theoretical basis for the treatment of HFpEF in clinical practice.
Collapse
Affiliation(s)
- Hang Xu
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Kai Gao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Chao Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jing Ma
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
39
|
The Role of PPARs in Breast Cancer. Cells 2022; 12:cells12010130. [PMID: 36611922 PMCID: PMC9818187 DOI: 10.3390/cells12010130] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/07/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is a malignant tumor with high morbidity and lethality. Its pathogenesis is related to the abnormal expression of many genes. The peroxisome proliferator-activated receptors (PPARs) are a class of ligand-dependent transcription factors in the nuclear receptor superfamily. They can regulate the transcription of a large number of target genes, which are involved in life activities such as cell proliferation, differentiation, metabolism, and apoptosis, and regulate physiological processes such as glucose metabolism, lipid metabolism, inflammation, and wound healing. Further, the changes in its expression are associated with various diseases, including breast cancer. The experimental reports related to "PPAR" and "breast cancer" were retrieved from PubMed since the discovery of PPARs and summarized in this paper. This review (1) analyzed the roles and potential molecular mechanisms of non-coordinated and ligand-activated subtypes of PPARs in breast cancer progression; (2) discussed the correlations between PPARs and estrogen receptors (ERs) as the nuclear receptor superfamily; and (3) investigated the interaction between PPARs and key regulators in several signaling pathways. As a result, this paper identifies PPARs as targets for breast cancer prevention and treatment in order to provide more evidence for the synthesis of new drugs targeting PPARs or the search for new drug combination treatments.
Collapse
|
40
|
Wagner KD. Editorial (Preface) "Cells/Cells of the Cardiovascular System-Editorial Highlights 2020-2021: The Book Selection". Cells 2022; 11:cells11233898. [PMID: 36497157 PMCID: PMC9735509 DOI: 10.3390/cells11233898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
This introduction provides a preface to the section on "Cells of the Cardiovascular System" in the book entitled "Editor's Choice Articles in 2020-2021" [...].
Collapse
|
41
|
Zeng W, Yin X, Jiang Y, Jin L, Liang W. PPARα at the crossroad of metabolic-immune regulation in cancer. FEBS J 2022; 289:7726-7739. [PMID: 34480827 DOI: 10.1111/febs.16181] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/04/2021] [Accepted: 09/03/2021] [Indexed: 01/14/2023]
Abstract
Rewiring metabolism to sustain cell growth, division, and survival is the most prominent feature of cancer cells. In particular, dysregulated lipid metabolism in cancer has received accumulating interest, since lipid molecules serve as cell membrane structure components, secondary signaling messengers, and energy sources. Given the critical role of immune cells in host defense against cancer, recent studies have revealed that immune cells compete for nutrients with cancer cells in the tumor microenvironment and accordingly develop adaptive metabolic strategies for survival at the expense of compromised immune functions. Among these strategies, lipid metabolism reprogramming toward fatty acid oxidation is closely related to the immunosuppressive phenotype of tumor-infiltrated immune cells, including macrophages and dendritic cells. Therefore, it is important to understand the lipid-mediated crosstalk between cancer cells and immune cells in the tumor microenvironment. Peroxisome proliferator-activated receptors (PPARs) consist of a nuclear receptor family for lipid sensing, and one of the family members PPARα is responsible for fatty acid oxidation, energy homeostasis, and regulation of immune cell functions. In this review, we discuss the emerging role of PPARα-associated metabolic-immune regulation in tumor-infiltrated immune cells, and key metabolic events and pathways involved, as well as their influences on antitumor immunity.
Collapse
Affiliation(s)
- Wenfeng Zeng
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaozhe Yin
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| | - Yunhan Jiang
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lingtao Jin
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Wei Liang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
42
|
Liu Y, Duan Y, Zhao N, Zhu X, Yu X, Jiao S, Song Y, Shi L, Ma Y, Wang X, Yu B, Qu A. Peroxisome Proliferator-Activated Receptor α Attenuates Hypertensive Vascular Remodeling by Protecting Vascular Smooth Muscle Cells from Angiotensin II-Induced ROS Production. Antioxidants (Basel) 2022; 11:antiox11122378. [PMID: 36552585 PMCID: PMC9774484 DOI: 10.3390/antiox11122378] [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: 10/16/2022] [Revised: 11/14/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Vascular remodeling is the fundamental basis for hypertensive disease, in which vascular smooth muscle cell (VSMC) dysfunction plays an essential role. Previous studies suggest that the activation of peroxisome proliferator-activated receptor α (PPARα) by fibrate drugs has cardiovascular benefits independent of the lipid-lowering effects. However, the underlying mechanism remains incompletely understood. This study explored the role of PPARα in angiotensin II (Ang II)-induced vascular remodeling and hypertension using VSMC-specific Ppara-deficient mice. The PPARα expression was markedly downregulated in the VSMCs upon Ang II treatment. A PPARα deficiency in the VSMC significantly aggravated the Ang II-induced hypertension and vascular stiffness, with little influence on the cardiac function. The morphological analyses demonstrated that VSMC-specific Ppara-deficient mice exhibited an aggravated vascular remodeling and oxidative stress. In vitro, a PPARα deficiency dramatically increased the production of mitochondrial reactive oxidative species (ROS) in Ang II-treated primary VSMCs. Finally, the PPARα activation by Wy14643 improved the Ang II-induced ROS production and vascular remodeling in a VSMC PPARα-dependent manner. Taken together, these data suggest that PPARα plays a critical protective role in Ang II-induced hypertension via attenuating ROS production in VSMCs, thus providing a potential therapeutic target for hypertensive diseases.
Collapse
Affiliation(s)
- Ye Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yan Duan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Nan Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Xinxin Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Xiaoting Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Shiyu Jiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Yanting Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
- Department of Pathology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Li Shi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Yutao Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, China
- Correspondence:
| |
Collapse
|
43
|
Oh KK. Network pharmacology-based analysis of signaling pathways of an anti-osteoporotic triterpenoid from Acyranthes bidentata Blume root. 3 Biotech 2022; 12:312. [PMID: 36276446 PMCID: PMC9537396 DOI: 10.1007/s13205-022-03362-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/12/2022] [Indexed: 11/01/2022] Open
Abstract
In Korea folk remedies, Acyranthes bidentata Blume is a functional food plant to treat bone diseases; especially, its roots have been used to alleviate osteoporosis (OP), but its key chemical compound(s) and mechanism of action against osteoporosis have not reported yet. This study suggests that Acyranthes bidentata Blume root (ABBR) has promising compound(s) against OP. We utilized network pharmacology to evaluate the therapeutic value. The chemical compounds from Acyranthes bidentata Blume root (ABBR) were identified by gas chromatography-mass spectrum (GC-MS); their physicochemical properties have been evaluated by SwissADME. Next, the target(s) related to a triterpenoid or OP-related targets were investigated by public databases. The signaling pathways from final targets were visualized, constructed, and analyzed by RPackage. Finally, we performed a molecular docking (MD) to explore key target(s) and compound(s) by employing AutoDockVina tools; the residues of amino acids interacted with ligands were identified by LigPlot + v.22. A total of 24 chemicals were accepted by the Lipinski's rules. We found a sole triterpenoid from ABBR via GC-MS, suggesting that might be a potent ligand to alleviate OP. Thereby, the 42 targets were associated with the triterpenoid; the 19 targets among them were connected to OP-targets (1426). The final 19 targets were related directly to 8 signaling pathways on STRING database. On Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and a key signaling pathway (PPAR signaling pathway), four key targets (PPARA, PPARD, FABP3, and FABP4) and a key compound (Methyl 3β-hydroxyolean-18-en-28-oate) were selected via MD. Collectively, the triterpenoid from ABBR might have potent anti-osteoporotic efficacy by activating PPARA, PPARD, FABP3, and FABP4 on PPAR signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03362-5.
Collapse
Affiliation(s)
- Ki Kwang Oh
- Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341 South Korea
| |
Collapse
|
44
|
Fatty Acid Amide Hydrolase Deficiency Is Associated with Deleterious Cardiac Effects after Myocardial Ischemia and Reperfusion in Mice. Int J Mol Sci 2022; 23:ijms232012690. [PMID: 36293543 PMCID: PMC9604059 DOI: 10.3390/ijms232012690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Ischemic cardiomyopathy leads to inflammation and left ventricular (LV) dysfunction. Animal studies provided evidence for cardioprotective effects of the endocannabinoid system, including cardiomyocyte adaptation, inflammation, and remodeling. Cannabinoid type-2 receptor (CB2) deficiency led to increased apoptosis and infarctions with worsened LV function in ischemic cardiomyopathy. The aim of our study was to investigate a possible cardioprotective effect of endocannabinoid anandamide (AEA) after ischemia and reperfusion (I/R). Therefore, fatty acid amide hydrolase deficient (FAAH)−/− mice were subjected to repetitive, daily, 15 min, left anterior descending artery (LAD) occlusion over 3 and 7 consecutive days. Interestingly, FAAH−/− mice showed stigmata such as enhanced inflammation, cardiomyocyte loss, stronger remodeling, and persistent scar with deteriorated LV function compared to wild-type (WT) littermates. As endocannabinoids also activate PPAR-α (peroxisome proliferator-activated receptor), PPAR-α mediated effects of AEA were eliminated with PPAR-α antagonist GW6471 i.v. in FAAH−/− mice. LV function was assessed using M-mode echocardiography. Immunohistochemical analysis revealed apoptosis, macrophage accumulation, collagen deposition, and remodeling. Hypertrophy was determined by cardiomyocyte area and heart weight/tibia length. Molecular analyses involved Taqman® RT-qPCR and immune cells were analyzed with fluorescence-activated cell sorting (FACS). Most importantly, collagen deposition was reduced to WT levels when FAAH−/− mice were treated with GW6471. Chemokine ligand-2 (CCL2) expression was significantly higher in FAAH−/− mice compared to WT, followed by higher macrophage infiltration in infarcted areas, both being reversed by GW6471 treatment. Besides restoring antioxidative properties and contractile elements, PPAR-α antagonism also reversed hypertrophy and remodeling in FAAH−/− mice. Finally, FAAH−/−-mice showed more substantial downregulation of PPAR-α compared to WT, suggesting a compensatory mechanism as endocannabinoids are also ligands for PPAR-α, and its activation causes lipotoxicity leading to cardiomyocyte apoptosis. Our study gives novel insights into the role of endocannabinoids acting via PPAR-α. We hypothesize that the increase in endocannabinoids may have partially detrimental effects on cardiomyocyte survival due to PPAR-α activation.
Collapse
|
45
|
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: 5.0] [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
|
46
|
Gao H, Li Y, Chen X. Interactions between nuclear receptors glucocorticoid receptor α and peroxisome proliferator-activated receptor α form a negative feedback loop. Rev Endocr Metab Disord 2022; 23:893-903. [PMID: 35476174 DOI: 10.1007/s11154-022-09725-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2022] [Indexed: 02/05/2023]
Abstract
Both nuclear receptors glucocorticoid receptor α (GRα) and peroxisome proliferator-activated receptor α (PPARα) are involved in energy and lipid metabolism, and possess anti-inflammation effects. Previous studies indicate that a regulatory loop may exist between them. In vivo and in vitro studies showed that glucocorticoids stimulate hepatic PPARα expression via GRα at the transcriptional level. This stimulation of PPARα by GRα has physiological relevance and PPARα is involved in many glucocorticoid-induced pathophysiological processes, including gluconeogenesis and ketogenesis during fasting, insulin resistance, hypertension and anti-inflammatory effects. PPARα also synergizes with GRα to promote erythroid progenitor self-renewal. As the feedback, PPARα inhibits glucocorticoid actions at pre-receptor and receptor levels. PPARα decreases glucocorticoid production through inhibiting the expression and activity of type-1 11β-hydroxysteroid dehydrogenase, which converts inactive glucocorticoids to active glucocorticoids at local tissues, and also down-regulates hepatic GRα expression, thus forming a complete and negative feedback loop. This negative feedback loop sheds light on prospective multi-drug therapeutic treatments in inflammatory diseases through a combination of glucocorticoids and PPARα agonists. This combination may potentially enhance the anti-inflammatory effects while alleviating side effects on glucose and lipid metabolism due to GRα activation. More investigations are needed to clarify the underlying mechanism and the relevant physiological or pathological significance of this regulatory loop.
Collapse
Affiliation(s)
- Hongjiao Gao
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Zunyi Medical University (the First People's Hospital of Zunyi), 563002, Zunyi, China
| | - Yujue Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Xiang Chen
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
47
|
Perry RJ. Regulation of Hepatic Lipid and Glucose Metabolism by INSP3R1. Diabetes 2022; 71:1834-1841. [PMID: 35657697 PMCID: PMC9450566 DOI: 10.2337/dbi22-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022]
Abstract
With the rising epidemics of obesity and nonalcoholic fatty liver disease (NAFLD) and its downstream consequences including steatohepatitis, cirrhosis, and type 2 diabetes in the U.S. and worldwide, new therapeutic approaches are urgently needed to treat these devastating conditions. Glucagon, known for a century to be a glucose-raising hormone and clearly demonstrated to contribute to fasting and postprandial hyperglycemia in both type 1 and type 2 diabetes, represents an unlikely target to improve health in those with metabolic syndrome. However, recent work from our group and others' identifies an unexpected role for glucagon as a potential means of treating NAFLD, improving insulin sensitivity, and improving the lipid profile. We propose a unifying, calcium-dependent mechanism for glucagon's effects both to stimulate hepatic gluconeogenesis and to enhance hepatic mitochondrial oxidation: signaling through the inositol 1,4,5-trisphosphate receptor type 1 (INSP3R1), glucagon activates phospholipase C (PKC)/protein kinase A (PKA) signaling to enhance adipose triglyceride lipase (ATGL)-dependent intrahepatic lipolysis and, in turn, increase cytosolic gluconeogenesis by allosteric activation of pyruvate carboxylase. Simultaneously in the mitochondria, calcium transferred through mitochondria-associated membranes activates several dehydrogenases in the tricarboxylic acid cycle, correlated with an increase in mitochondrial energy expenditure and reduction in ectopic lipid. This model suggests that short-term, cyclic treatment with glucagon or other INSP3R1 antagonists could hold promise as a means to reset lipid homeostasis in patients with NAFLD.
Collapse
Affiliation(s)
- Rachel J. Perry
- Section of Endocrinology & Metabolism, Department of Internal Medicine, and Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
48
|
Qiu H, Song E, Hu Y, Li T, Ku KC, Wang C, Cheung BMY, Cheong LY, Wang Q, Wu X, Hoo RLC, Wang Y, Xu A. Hepatocyte-Secreted Autotaxin Exacerbates Nonalcoholic Fatty Liver Disease Through Autocrine Inhibition of the PPARα/FGF21 Axis. Cell Mol Gastroenterol Hepatol 2022; 14:1003-1023. [PMID: 35931383 PMCID: PMC9490100 DOI: 10.1016/j.jcmgh.2022.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS The prevalence of nonalcoholic fatty liver disease (NAFLD) has reached epidemic proportions globally as a result of the rapid increase in obesity. However, there is no Food and Drug Administration-approved pharmacotherapy available for NAFLD. This study investigated the role of autotaxin, a secreted enzyme that hydrolyzes lysophosphatidylcholine to produce lysophosphatidic acid (LPA), in the pathogenesis of NAFLD and to explore whether genetic or pharmacologic interventions targeting autotaxin ameliorate NAFLD. METHODS The clinical association of autotaxin with the severity of NAFLD was analyzed in 125 liver biopsy-proven NAFLD patients. C57BL/6N mice or fibroblast growth factor 21 (FGF21)-null mice were fed a high-fat diet or a choline-deficient diet to investigate the role of the autotaxin-FGF21 axis in NAFLD development by hepatic knockdown and antibody neutralization. Huh7 cells were used to investigate the autocrine effects of autotaxin. RESULTS Serum autotaxin levels were associated positively with histologic scores and NAFLD severity. Hepatocytes, but not adipocytes, were the major contributor to increased circulating autotaxin in both patients and mouse models with NAFLD. In mice, knocking-down hepatic autotaxin or treatment with a neutralizing antibody against autotaxin significantly reduced high-fat diet-induced NAFLD and high fat- and choline-deficient diet-induced nonalcoholic steatohepatitis and fibrosis, accompanied by a marked increase of serum FGF21. Mechanistically, autotaxin inhibited the transcriptional activity of peroxisome proliferator-activated receptor α through LPA-induced activation of extracellular signal-regulated kinas, thereby leading to suppression of hepatic FGF21 production. The therapeutic benefit of anti-autotaxin neutralizing antibody against NAFLD was abrogated in FGF21-null mice. CONCLUSIONS Liver-secreted autotaxin acts in an autocrine manner to exacerbate NAFLD through LPA-induced suppression of the peroxisome proliferator-activated receptor α-FGF21 axis and is a promising therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Han Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Erfei Song
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yue Hu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Tengfei Li
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kam Ching Ku
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bernard M Y Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaoping Wu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Ruby L C Hoo
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Yong Wang
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China.
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
49
|
The Editor’s Choice Articles—Section “Cells of the Cardiovascular System” 2020–2021. Cells 2022; 11:cells11142173. [PMID: 35883616 PMCID: PMC9323559 DOI: 10.3390/cells11142173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
Cells is experiencing a rapid increase in attractiveness and impact [...]
Collapse
|
50
|
Intervention of Shugan Xiaozhi Decoction on Nonalcoholic Fatty Liver Disease via Mediating Gut-Liver Axis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4801695. [PMID: 35837380 PMCID: PMC9276511 DOI: 10.1155/2022/4801695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease with an increasing incidence rate but few therapies. Shugan Xiaozhi decoction (SX) has demonstrated beneficial effects in treating NAFLD with an unclear mechanism. This study was aimed at investigating the therapeutic mechanism of SX on high-fat diet-induced NAFLD rats via the gut-liver axis. Hepatic steatosis and integrity of intestinal mucosa in NAFLD rats were assessed by histopathological staining. The level of lipid and inflammation were estimated by enzyme-linked immunosorbent assay. Western Blotting was used to detect apolipoprotein (apo) B48 expression. 16S rRNA analysis was used to measure the changes of gut microbial composition after SX treatment. The expressions of zona occludens 1 protein (ZO-1), occludin, and secretory immunoglobulin A (sIgA) in the colon were detected by immunostaining to investigate the intestinal barrier function. Our study found that SX reduced hepatic steatosis, the levels of alanine aminotransferase, aspartate aminotransferase, total cholesterol, and triglyceride and apoB48 expression but increased peroxisome proliferator activated receptor α (PPARα) level. Moreover, SX altered the diversity of gut microbiota, upregulating the relative abundance of f_Prevotellaceae, while downregulating f_Bacteroidales_ S24-7, f_Lachnospiraceae, f_Ruminococcaceae, f_Erysipelotrichaceae, and f_Desulfovibrionaceae. By increasing the expression of ZO-1 and occludin and decreasing the level of proinflammatory factors, including sIgA, lipopolysaccharide, tumor necrosis factor-α, interleukin-1β, monocyte chemotactic protein-1, and transforming growth factor-β1, SX improved intestinal mucosal integrity and barrier function. Our study illustrated that the gut-liver axis was a potential way for SX to ameliorate NAFLD, that is, by regulating the expression of PPARα, apoB48, and modulating gut microbiota to protect the intestinal barrier function, and thus alleviate lipid deposition and inflammatory response in the liver.
Collapse
|