51
|
The impact of PUFA-enriched yogurt consumption on cardiovascular risk markers: A review. PHARMANUTRITION 2023. [DOI: 10.1016/j.phanu.2023.100330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
52
|
Omega-3 polyunsaturated fatty acids and corneal nerve health: Current evidence and future directions. Ocul Surf 2023; 27:1-12. [PMID: 36328309 DOI: 10.1016/j.jtos.2022.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
Abstract
Corneal nerves play a key role in maintaining ocular surface integrity. Corneal nerve damage, from local or systemic conditions, can lead to ocular discomfort, pain, and, if poorly managed, neurotrophic keratopathy. Omega-3 polyunsaturated fatty acids (PUFAs) are essential dietary components that play a key role in neural development, maintenance, and function. Their potential application in modulating ocular and systemic inflammation has been widely reported. Omega-3 PUFAs and their metabolites also have neuroprotective properties and can confer benefit in neurodegenerative disease. Several preclinical studies have shown that topical administration of omega-3 PUFA-derived lipid mediators promote corneal nerve recovery following corneal surgery. Dietary omega-3 PUFA supplementation can also reduce corneal epithelial nerve loss and promote corneal nerve regeneration in diabetes. Omega-3 PUFAs and their lipid mediators thus show promise as therapeutic approaches to modulate corneal nerve health in ocular and systemic disease. This review discusses the role of dietary omega-3 PUFAs in maintaining ocular surface health and summarizes the possible applications of omega-3 PUFAs in the management of ocular and systemic conditions that cause corneal nerve damage. In examining the current evidence, this review also highlights relatively underexplored applications of omega-3 PUFAs in conferring neuroprotection and addresses their therapeutic potential in mediating corneal nerve regeneration.
Collapse
|
53
|
Eccles JA, Baldwin WS. Detoxification Cytochrome P450s (CYPs) in Families 1-3 Produce Functional Oxylipins from Polyunsaturated Fatty Acids. Cells 2022; 12:82. [PMID: 36611876 PMCID: PMC9818454 DOI: 10.3390/cells12010082] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
This manuscript reviews the CYP-mediated production of oxylipins and the current known function of these diverse set of oxylipins with emphasis on the detoxification CYPs in families 1-3. Our knowledge of oxylipin function has greatly increased over the past 3-7 years with new theories on stability and function. This includes a significant amount of new information on oxylipins produced from linoleic acid (LA) and the omega-3 PUFA-derived oxylipins such as α-linolenic acid (ALA), docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA). However, there is still a lack of knowledge regarding the primary CYP responsible for producing specific oxylipins, and a lack of mechanistic insight for some clinical associations between outcomes and oxylipin levels. In addition, the role of CYPs in the production of oxylipins as signaling molecules for obesity, energy utilization, and development have increased greatly with potential interactions between diet, endocrinology, and pharmacology/toxicology due to nuclear receptor mediated CYP induction, CYP inhibition, and receptor interactions/crosstalk. The potential for diet-diet and diet-drug/chemical interactions is high given that these promiscuous CYPs metabolize a plethora of different endogenous and exogenous chemicals.
Collapse
Affiliation(s)
| | - William S. Baldwin
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
54
|
Shaikh SR, Virk R, Van Dyke TE. Potential Mechanisms by Which Hydroxyeicosapentaenoic Acids Regulate Glucose Homeostasis in Obesity. Adv Nutr 2022; 13:2316-2328. [PMID: 35709423 PMCID: PMC9776734 DOI: 10.1093/advances/nmac073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/16/2022] [Accepted: 06/13/2022] [Indexed: 01/29/2023] Open
Abstract
Dysregulation of glucose metabolism in response to diet-induced obesity contributes toward numerous complications, such as insulin resistance and hepatic steatosis. Therefore, there is a need to develop effective strategies to improve glucose homeostasis. In this review, we first discuss emerging evidence from epidemiological studies and rodent experiments that increased consumption of EPA (either as oily fish, or dietary/pharmacological supplements) may have a role in preventing impairments in insulin and glucose homeostasis. We then review the current evidence on how EPA-derived metabolites known as hydroxyeicosapentaenoic acids (HEPEs) may be a major mode of action by which EPA exerts its beneficial effects on glucose and lipid metabolism. Notably, cell culture and rodent studies show that HEPEs prevent fat accumulation in metabolic tissues through peroxisome proliferator activated receptor (PPAR)-mediated mechanisms. In addition, activation of the resolvin E1 pathway, either by administration of EPA in the diet or via intraperitoneal administration of resolvin E1, improves hyperglycemia, hyperinsulinemia, and liver steatosis through multiple mechanisms. These mechanisms include shifting immune cell phenotypes toward resolution of inflammation and preventing dysbiosis of the gut microbiome. Finally, we present the next steps for this line of research that will drive future precision randomized clinical trials with EPA and its downstream metabolites. These include dissecting the variables that drive heterogeneity in the response to EPA, such as the baseline microbiome profile and fatty acid status, circadian rhythm, genetic variation, sex, and age. In addition, there is a critical need to further investigate mechanisms of action for HEPEs and to establish the concentration of HEPEs in differing tissues, particularly in response to consumption of oily fish and EPA-enriched supplements.
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School
of Medicine, The University of North Carolina at Chapel Hill, Chapel
Hill, NC, USA
| | - Rafia Virk
- Department of Nutrition, Gillings School of Global Public Health and School
of Medicine, The University of North Carolina at Chapel Hill, Chapel
Hill, NC, USA
| | - Thomas E Van Dyke
- Center for Clinical and Translational Research, The Forsyth
Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and Immunity, Harvard School of
Dental Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
55
|
Si H, Li S, Nan W, Sang J, Xu C, Li Z. Integrated Transcriptome and Microbiota Reveal the Regulatory Effect of 25-Hydroxyvitamin D Supplementation in Antler Growth of Sika Deer. Animals (Basel) 2022; 12:ani12243497. [PMID: 36552417 PMCID: PMC9774409 DOI: 10.3390/ani12243497] [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: 10/29/2022] [Revised: 11/21/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The level of plasma 25-hydroxyvitamin D (25(OH)D) is associated with the growth of the antler, a fast-growing bone organ of Cervidae. However, the benefits of 25(OH)D supplementation on antler growth and the underlying mechanisms remain unclear. Here, the antler growth profile and transcriptome, plasma parameters, rumen bacteria, and metabolites (volatile fatty acids and amino acids) were determined in sika deer in a 25(OH)D supplementation group (25(OH)D, n = 8) and a control group (Ctrl, n = 8). 25(OH)D supplementation significantly increased the antler weight and growth rate. The levels of IGF-1,25(OH)D and 1,25-dihydroxyvitamin D were significantly higher in the 25(OH)D group than in the Ctrl group, while the levels of LDL-C were lower. The levels of valerate and branched-chain amino acids in the rumen fluid were significantly different between the 25(OH)D and Ctrl groups. The bacterial diversity indices were not significantly different between the two groups. However, the relative abundances of the butyrate-producing bacteria (families Lachnospiraceae and Succinivibrionaceae) and the pyruvate metabolism pathway were higher in the 25(OH)D group. The transcriptomic profile of the antler was significantly different between the 25(OH)D and Ctrl groups, with 356 up- and 668 down-regulated differentially expressed genes (DEGs) in the 25(OH)D group. The up-regulated DEGs were enriched in the proteinaceous extracellular matrix and collagen, while the down-regulated DEGs were enriched in the immune system and lipid metabolism pathways. Overall, these results provide novel insights into the effects of 25(OH)D supplementation on the host metabolism, rumen microbiota, and antler transcriptome of sika deer.
Collapse
Affiliation(s)
- Huazhe Si
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Songze Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Weixiao Nan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Jianan Sang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Chao Xu
- Department of Special Animal Nutrition and Feed Science, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
- Correspondence: (C.X.); (Z.L.)
| | - Zhipeng Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
- Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Jilin Agricultural University, Changchun 130118, China
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Correspondence: (C.X.); (Z.L.)
| |
Collapse
|
56
|
Zarkesh M, Safarian M, Asghari G, Daneshafrooz A, Yuzbashian E, Hedayati M, Mirmiran P, Khalaj A. Is Habitual Dietary Intake of Fats Associated with Apelin Gene Expression in Visceral and Subcutaneous Adipose Tissues and Its Serum Levels in Obese Adults? Public Health Genomics 2022; 26:16-23. [PMID: 36481653 DOI: 10.1159/000526961] [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: 09/15/2021] [Accepted: 08/04/2022] [Indexed: 12/22/2023] Open
Abstract
INTRODUCTION Apelin could be one of the last protective defenses before developing obesity-related disorders, including insulin resistance, type 2 diabetes, and hypertension, which can be modified by dietary intake. The present study investigated the association of habitual intake of total fatty acids (TFAs), saturated-, monounsaturated-, polyunsaturated FAs, n-3, and n-6 FAs with Apelin expression in visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT). METHODS We obtained VAT and SAT from 168 participants (64 nonobese and 104 obese) who had undergone open abdominal surgery. Dietary intake information was gathered with a valid and reliable food frequency questionnaire. The mRNA expression of the Apelin gene was analyzed by real-time PCR. RESULTS Apelin serum levels were increased in the obese subjects compared to the nonobese group (p = 0.016). The SAT and VAT Apelin mRNA levels were significantly elevated in the obese participants compared to the nonobese ones (p < 0.05). Based on BMI status, only obese subjects indicated a positive association between SAT and VAT Apelin expression and TFA intake (p < 0.001). However, this association was observed between SAT and VAT Apelin gene expression and polyunsaturated fatty acid (PUFA) and n-3 FA intakes in both obese and nonobese groups (p < 0.05). CONCLUSION High Apelin gene expression was associated with TFA intake in obese subjects in both fat tissues. However, habitual intake of PUFA and n-3 FA was associated with Apelin gene expression in obese and nonobese individuals. Our results indicate a determinative role of the quality and quantity of FA intake on adipose tissue.
Collapse
Affiliation(s)
- Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Safarian
- Department of Biochemistry of Nutrition Sciences, Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Golaleh Asghari
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Tehran, Iran
| | - Afsoon Daneshafrooz
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Emad Yuzbashian
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Tehran, Iran
| | - Alireza Khalaj
- Department of Surgery, Tehran Obesity Treatment Center, Shahed University, Tehran, Iran
| |
Collapse
|
57
|
Bahrami-Nejad Z, Zhang ZB, Tholen S, Sharma S, Rabiee A, Zhao ML, Kraemer FB, Teruel MN. Early enforcement of cell identity by a functional component of the terminally differentiated state. PLoS Biol 2022; 20:e3001900. [PMID: 36469503 PMCID: PMC9721491 DOI: 10.1371/journal.pbio.3001900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/02/2022] [Indexed: 12/12/2022] Open
Abstract
How progenitor cells can attain a distinct differentiated cell identity is a challenging problem given the fluctuating signaling environment in which cells exist and that critical transcription factors are often not unique to a differentiation process. Here, we test the hypothesis that a unique differentiated cell identity can result from a core component of the differentiated state doubling up as a signaling protein that also drives differentiation. Using live single-cell imaging in the adipocyte differentiation system, we show that progenitor fat cells (preadipocytes) can only commit to terminally differentiate after up-regulating FABP4, a lipid buffer that is highly enriched in mature adipocytes. Upon induction of adipogenesis in mouse preadipocyte cells, we show that after a long delay, cells first abruptly start to engage a positive feedback between CEBPA and PPARG before then engaging, after a second delay, a positive feedback between FABP4 and PPARG. These sequential positive feedbacks both need to engage in order to drive PPARG levels past the threshold for irreversible differentiation. In the last step before commitment, PPARG transcriptionally increases FABP4 expression while fatty acid-loaded FABP4 increases PPARG activity. Together, our study suggests a control principle for robust cell identity whereby a core component of the differentiated state also promotes differentiation from its own progenitor state.
Collapse
Affiliation(s)
- Zahra Bahrami-Nejad
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
| | - Zhi-Bo Zhang
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
- Department of Biochemistry and the Drukier Institute for Children’s Health, Weill Cornell Medical College of Cornell University, New York, New York, United States of America
| | - Stefan Tholen
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
| | - Sanjeev Sharma
- Department of Biochemistry and the Drukier Institute for Children’s Health, Weill Cornell Medical College of Cornell University, New York, New York, United States of America
| | - Atefeh Rabiee
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
- Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California, United States of America
| | - Michael L. Zhao
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
| | - Fredric B. Kraemer
- Department of Medicine/Division of Endocrinology, Stanford University, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Mary N. Teruel
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
- Department of Biochemistry and the Drukier Institute for Children’s Health, Weill Cornell Medical College of Cornell University, New York, New York, United States of America
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, United States of America
- Weill Center for Metabolic Health, Division of Endocrinology, Diabetes & Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College of Cornell University, New York, New York, United States of America
| |
Collapse
|
58
|
Petunina NA, Goncharova EV, Kuzina IA, Nedosugova LV, Martirosyan NS, Теlnova MЕ. The role of pioglitazone in the fight against insulin resistance, atherosclerosis, cardiovascular disease, and non-alcoholic fatty liver disease. DIABETES MELLITUS 2022. [DOI: 10.14341/dm12859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Modern strategies for the treatment of type 2 diabetes mellitus involve the use of pathogenetically based approaches aimed at achieving optimal glycemic control and its long-term retention. Timely and rational use of 9 classes of hypoglycemic drugs, including as part of combination therapy, makes it possible to achieve significant success in diabetes therapy. One of the fundamental principles in the treatment of type 2 diabetes mellitus is the effect on insulin resistance. For this purpose, two groups of drugs are used: biguanides and thiazolidinediones (glitazones). The action of glitazones is directly related to an increase in the sensitivity of insulin-dependent tissues to insulin and a pronounced decrease in hyperinsulinemia in patients with type 2 diabetes. Of particular interest are the pathways of insulin signal transduction, the mechanisms of insulin resistance, and the possibilities of pathogenetic therapy with thiazolidinediones. Pioglitazone is currently the only available member of the thiazolidinedione class in the world, allowing to expand the management of diabetes mellitus by reducing insulin resistance in muscle and adipose tissue and glucose production by the liver. Its use can have a number of pleiotropic effects, including on cardiovascular diseases and non-alcoholic fatty liver disease, which expands the priorities for choosing hypoglycemic therapy in patients with type 2 diabetes at various stages of therapy.
Collapse
Affiliation(s)
- N. A. Petunina
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. V. Goncharova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - I. A. Kuzina
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - L. V. Nedosugova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - N. S. Martirosyan
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - M. Е. Теlnova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
59
|
PPARα in the Epigenetic Driver Seat of NAFLD: New Therapeutic Opportunities for Epigenetic Drugs? Biomedicines 2022; 10:biomedicines10123041. [PMID: 36551797 PMCID: PMC9775974 DOI: 10.3390/biomedicines10123041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing epidemic and the most common cause of chronic liver disease worldwide. It consists of a spectrum of liver disorders ranging from simple steatosis to NASH which predisposes patients to further fibrosis, cirrhosis and even hepatocarcinoma. Despite much research, an approved treatment is still lacking. Finding new therapeutic targets has therefore been a main priority. Known as a main regulator of the lipid metabolism and highly expressed in the liver, the nuclear receptor peroxisome proliferator-activated receptor-α (PPARα) has been identified as an attractive therapeutic target. Since its expression is silenced by DNA hypermethylation in NAFLD patients, many research strategies have aimed to restore the expression of PPARα and its target genes involved in lipid metabolism. Although previously tested PPARα agonists did not ameliorate the disease, current research has shown that PPARα also interacts and regulates epigenetic DNMT1, JMJD3, TET and SIRT1 enzymes. Moreover, there is a growing body of evidence suggesting the orchestrating role of epigenetics in the development and progression of NAFLD. Therefore, current therapeutic strategies are shifting more towards epigenetic drugs. This review provides a concise overview of the epigenetic regulation of NAFLD with a focus on PPARα regulation and highlights recently identified epigenetic interaction partners of PPARα.
Collapse
|
60
|
da Silva Batista E, Nakandakari SCBR, Ramos da Silva AS, Pauli JR, Pereira de Moura L, Ropelle ER, Camargo EA, Cintra DE. Omega-3 pleiad: The multipoint anti-inflammatory strategy. Crit Rev Food Sci Nutr 2022; 64:4817-4832. [PMID: 36382659 DOI: 10.1080/10408398.2022.2146044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Omega 3 (ω3) fatty acids have been described since the 1980s as promising anti-inflammatory substances. Prostaglandin and leukotriene modulation were exhaustively explored as the main reason for ω3 beneficial outcomes. However, during the early 2000s, after the human genome decoding advent, the nutrigenomic approaches exhibited an impressive plethora of ω3 targets, now under the molecular point of view. Different G protein-coupled receptors (GPCRs) recognizing ω3 and its derivatives appear to be responsible for blocking inflammation and insulin-sensitizing effects. A new class of ω3-derived substances, such as maresins, resolvins, and protectins, increases ω3 actions. Inflammasome disruption, the presence of GPR120 on immune cell surfaces, and intracellular crosstalk signaling mediated by PPARγ compose the last discoveries regarding the multipoint anti-inflammatory targets for this nutrient. This review shows a detailed mechanistic proposal to understand ω3 fatty acid action over the inflammatory environment in the background of several chronic diseases.
Collapse
Affiliation(s)
- Ellencristina da Silva Batista
- Graduate Program of Health Sciences (PPGCS), Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Nutrition Department, Federal University of Sergipe, Lagarto, Sergipe, Brazil
| | - Susana Castelo Branco Ramos Nakandakari
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | | | - José Rodrigo Pauli
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Enilton A Camargo
- Graduate Program of Health Sciences (PPGCS), Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Dennys Esper Cintra
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
- OCRC - Obesity and Comorbidities Research Center, UNICAMP, São Paulo, Brazil
| |
Collapse
|
61
|
Birkic N, Azar T, Maddipati KR, Minic Z, Reynolds CA. Excessive dietary linoleic acid promotes plasma accumulation of pronociceptive fatty acyl lipid mediators. Sci Rep 2022; 12:17832. [PMID: 36284115 PMCID: PMC9596689 DOI: 10.1038/s41598-022-21823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/04/2022] [Indexed: 01/20/2023] Open
Abstract
Various fatty acyl lipid mediators are derived from dietary polyunsaturated fatty acids (PUFAs) and modulate nociception. The modern diet is rich in linoleic acid, which is associated with nociceptive hypersensitivities and may present a risk factor for developing pain conditions. Although recommendations about fatty acid intake exist for some diseases (e.g. cardiovascular disease), the role of dietary fatty acids in promoting pain disorders is not completely understood. To determine how dietary linoleic acid content influences the accumulation of pro- and anti-nociceptive fatty acyl lipid mediators, we created novel rodent diets using custom triglyceride blends rich in either linoleic acid or oleic acid. We quantified the fatty acyl lipidome in plasma of male and female rats fed these custom diets from the time of weaning through nine weeks of age. Dietary fatty acid composition determined circulating plasma fatty acyl lipidome content. Exposure to a diet rich in linoleic acid was associated with accumulation of linoleic and arachidonic acid-derived pro-nociceptive lipid mediators and reduction of anti-nociceptive lipid mediators derived from the omega-3 PUFAs. Our findings provide mechanistic insights into exaggerated nociceptive hypersensitivity associated with excessive dietary linoleic acid intake and highlight potential biomarkers for pain risk stratification.
Collapse
Affiliation(s)
- Nada Birkic
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Toni Azar
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Krishna Rao Maddipati
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zeljka Minic
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Christian A Reynolds
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia.
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
62
|
Han M, Chen Z, He P, Li Z, Chen Q, Tong Z, Wang M, Du H, Zhang H. YgiM may act as a trigger in the sepsis caused by Klebsiella pneumoniae through the membrane-associated ceRNA network. Front Genet 2022; 13:973145. [PMID: 36212144 PMCID: PMC9537587 DOI: 10.3389/fgene.2022.973145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/07/2022] [Indexed: 11/27/2022] Open
Abstract
Sepsis is one of the diseases that can cause serious mortality. In E. coli, an inner membrane protein YgiM encoded by gene ygiM can target the eukaryotic peroxisome. Peroxisome is a membrane-enclosed organelle associated with the ROS metabolism and was reported to play the key role in immune responses and inflammation during the development of sepsis. Klebsiella pneumoniae (K. pneumoniae) is one of the important pathogens causing sepsis. However, the function of gene vk055_4013 which is highly homologous to ygiM of E. coli has not been demonstrated in K. pneumoniae. In this study, we prepared ΔygiM of K. pneumoniae ATCC43816, and found that the deletion of ygiM did not affect bacterial growth and mouse mortality in the mouse infection model. Interestingly, ΔygiM not only resulted in reduced bacterial resistance to macrophages, but also attenuated pathological manifestations in mouse organs. Furthermore, based on the data of Gene Expression Omnibus, the expression profiles of micro RNAs (miRNAs) and messenger RNAs (mRNAs) in the serum of 44 sepsis patients caused by K. pneumoniae infection were analyzed, and 11 differently expressed miRNAs and 8 DEmRNAs associated with the membrane function were found. Finally, the membrane-associated competing endogenous RNAs (ceRNAs) network was constructed. In this ceRNAs network, DEmiRNAs (hsa-miR-7108-5p, hsa-miR-6780a-5p, hsa-miR-6756-5p, hsa-miR-4433b-3p, hsa-miR-3652, hsa-miR-342-3p, hsa-miR-32-5p) and their potential downstream target DEmRNAs (VNN1, CEACAM8, PGLYRP1) were verified in the cell model infected by wild type and ΔygiM of K. pneumoniae, respectively. Taken together, YgiM may trigger the sepsis caused by K. pneumoniae via membrane-associated ceRNAs. This study provided new insights into the role of YgiM in the process of K. pneumoniae induced sepsis.
Collapse
Affiliation(s)
- Mingxiao Han
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ping He
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Clinical Laboratory, Sichuan Province Science City Hospital, Chengdu, China
| | - Ziyuan Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zelei Tong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
63
|
Pavithra V, Janhavi P, Natasha J, Neelam R, Mrityunjaya M, Selvi MK, Ravindra PV. A blend of cod liver oil and virgin coconut oil improves the endurance performance in mice. SPORT SCIENCES FOR HEALTH 2022. [DOI: 10.1007/s11332-022-01001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
64
|
Duan Y, Zhang X, Zhang X, Lin J, Shu X, Man W, Jiang M, Zhang Y, Wu D, Zhao Z, Sun D. Inhibition of macrophage-derived foam cells by Adipsin attenuates progression of atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166533. [PMID: 36064133 DOI: 10.1016/j.bbadis.2022.166533] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/03/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022]
Abstract
Phagocytosis of oxidized low-density lipoprotein (OxLDL) by macrophages yields "foam cells" and serves as a hallmark of atherosclerotic lesion. Adipsin is a critical component of the complement activation pathway. Recent evidence has indicated an obligatory role for Adipsin in pathological models including ischemia-reperfusion and sepsis. Adipsin levels are significantly decreased in patients with asymptomatic carotid atherosclerosis, implying the role for Adipsin as a potential marker of asymptomatic carotid atherosclerosis. This study was designed to evaluate the role for Adipsin in atherosclerosis and the mechanisms involved using both in vivo and in vitro experiments. ApoE-/-/AdipsinTg mice were constructed and were fed a high-fat diet for 12 weeks. Compared with ApoE-/- mice, area of the sclerotic plaques was reduced, along with lower macrophage deposition within the plaque in ApoE-/-/AdipsinTg mice. RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were stimulated with oxLDL (50 μg/ml). Adenovirus vectors containing the Adipsin gene were transfected into macrophages. Lipid accumulation was observed by Oil red O staining. Western blot and reverse transcription-polymerase chain reaction data revealed that Adipsin overexpression inhibited oxLDL-induced lipid uptake and foam cell formation and upregulation of CD36 and PPARγ in Ad-Adipsin-transfected macrophages. In addition, the PPARγ-specific agonist GW1929 reversed Adipsin overexpression-evoked inhibitory effect on lipid uptake. These results demonstrate unequivocally that Adipsin inhibits lipid uptake in a PPARγ/CD36-dependent manner and prevents the formation of foam cells, implying that Adipsin may be a potential therapeutic target against atherosclerosis.
Collapse
Affiliation(s)
- Yu Duan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaofei Shu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wanrong Man
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dexi Wu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhijing Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
65
|
Stifel U, Caratti G, Tuckermann J. Novel insights into the regulation of cellular catabolic metabolism in macrophages through nuclear receptors. FEBS Lett 2022; 596:2617-2629. [PMID: 35997656 DOI: 10.1002/1873-3468.14474] [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: 02/15/2022] [Revised: 06/14/2022] [Accepted: 07/04/2022] [Indexed: 11/07/2022]
Abstract
Regulation of cellular catabolic metabolism in immune cells has recently become a major concept for resolution of inflammation. Nuclear receptors (NRs), including peroxisome proliferator activator receptors (PPARs), 1,25-dihydroxyvitamin D(3) receptor (VDR), liver X receptors (LXRs), glucocorticoid receptors (GRs), estrogen-related receptor α (ERRα) and Nur77, have been identified as major modulators of inflammation, affecting innate immune cells, such as macrophages. Evidence emerges on how NRs regulate cellular metabolism in macrophages during inflammatory processes and contribute to the resolution of inflammation. This could have new implications for our understanding of how NRs shape immune responses and inform anti-inflammatory drug design. This review will highlight the recent developments about NRs and their role in cellular metabolism in macrophages.
Collapse
Affiliation(s)
- Ulrich Stifel
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany
| | - Giorgio Caratti
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany.,NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany
| |
Collapse
|
66
|
Przybycień P, Gąsior-Perczak D, Placha W. Cannabinoids and PPAR Ligands: The Future in Treatment of Polycystic Ovary Syndrome Women with Obesity and Reduced Fertility. Cells 2022; 11:cells11162569. [PMID: 36010645 PMCID: PMC9406585 DOI: 10.3390/cells11162569] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 11/21/2022] Open
Abstract
Cannabinoids (CBs) are used to treat chronic pain, chemotherapy-induced nausea and vomiting, and multiple sclerosis spasticity. Recently, the medicinal use of CBs has attracted increasing interest as a new therapeutic in many diseases. Data indicate a correlation between CBs and PPARs via diverse mechanisms. Both the endocannabinoid system (ECS) and peroxisome proliferator-activated receptors (PPARs) may play a significant role in PCOS and PCOS related disorders, especially in disturbances of glucose-lipid metabolism as well as in obesity and fertility. Taking into consideration the ubiquity of PCOS in the human population, it seems indispensable to search for new potential therapeutic targets for this condition. The aim of this review is to examine the relationship between metabolic disturbances and obesity in PCOS pathology. We discuss current and future therapeutic interventions for PCOS and related disorders, with emphasis on the metabolic pathways related to PCOS pathophysiology. The link between the ECS and PPARs is a promising new target for PCOS, and we examine this relationship in depth.
Collapse
Affiliation(s)
- Piotr Przybycień
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 31-034 Krakow, Poland
- Endocrinology Clinic, Holycross Cancer Centre, 25-734 Kielce, Poland
| | - Danuta Gąsior-Perczak
- Endocrinology Clinic, Holycross Cancer Centre, 25-734 Kielce, Poland
- Collegium Medicum, Jan Kochanowski University, 25-317 Kielce, Poland
| | - Wojciech Placha
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 31-034 Krakow, Poland
- Correspondence: ; Tel.: +48-12-422-74-00
| |
Collapse
|
67
|
Evans N, Conley JM, Cardon M, Hartig P, Medlock-Kakaley E, Gray LE. In vitro activity of a panel of per- and polyfluoroalkyl substances (PFAS), fatty acids, and pharmaceuticals in peroxisome proliferator-activated receptor (PPAR) alpha, PPAR gamma, and estrogen receptor assays. Toxicol Appl Pharmacol 2022; 449:116136. [PMID: 35752307 PMCID: PMC9341220 DOI: 10.1016/j.taap.2022.116136] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/03/2022] [Accepted: 06/16/2022] [Indexed: 10/17/2022]
Abstract
Data demonstrate numerous per- and polyfluoroalkyl substances (PFAS) activate peroxisome proliferator-activated receptor alpha (PPARα), however, additional work is needed to characterize PFAS activity on PPAR gamma (PPARγ) and other nuclear receptors. We utilized in vitro assays with either human or rat PPARα or PPARγ ligand binding domains to evaluate 16 PFAS (HFPO-DA, HFPO-DA-AS, NBP2, PFMOAA, PFHxA, PFOA, PFNA, PFDA, PFOS, PFBS, PFHxS, PFOSA, EtPFOSA, and 4:2, 6:2 and 8:2 FTOH), 3 endogenous fatty acids (oleic, linoleic, and octanoic), and 3 pharmaceuticals (WY14643, clofibrate, and the metabolite clofibric acid). We also tested chemicals for human estrogen receptor (hER) transcriptional activation. Nearly all compounds activated both PPARα and PPARγ in both human and rat ligand binding domain assays, except for the FTOH compounds and PFOSA. Receptor activation and relative potencies were evaluated based on effect concentration 20% (EC20), top percent of max fold induction (pmaxtop), and area under the curve (AUC). HFPO-DA and HFPO-DA-AS were the most potent (lowest EC20, highest pmaxtop and AUC) of all PFAS in rat and human PPARα assays, being slightly less potent than oleic and linoleic acid, while NBP2 was the most potent in rat and human PPARγ assays. Only PFHxS, 8:2 and 6:2 FTOH exhibited hER agonism >20% pmax. In vitro measures of human and rat PPARα and PPARγ activity did not correlate with oral doses or serum concentrations of PFAS that induced increases in male rat liver weight from the National Toxicology Program 28-d toxicity studies. Data indicate that both PPARα and PPARγ activation may be molecular initiating events that contribute to the in vivo effects observed for many PFAS.
Collapse
Affiliation(s)
- Nicola Evans
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment/Public Health and Integrated Toxicology Division, Research Triangle Park, NC 27711, USA.
| | - Justin M Conley
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment/Public Health and Integrated Toxicology Division, Research Triangle Park, NC 27711, USA.
| | - Mary Cardon
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment/Public Health and Integrated Toxicology Division, Research Triangle Park, NC 27711, USA.
| | - Phillip Hartig
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment/Public Health and Integrated Toxicology Division, Research Triangle Park, NC 27711, USA.
| | - Elizabeth Medlock-Kakaley
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment/Public Health and Integrated Toxicology Division, Research Triangle Park, NC 27711, USA.
| | - L Earl Gray
- U.S. Environmental Protection Agency/Office of Research & Development/Center for Public Health and Environmental Assessment/Public Health and Integrated Toxicology Division, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
68
|
Chen L, Yang CS, Chen SD, Zhou QX, Wang GQ, Cai SL, Li WH, Luo HZ. Multi-omics characterization of the unsaturated fatty acid biosynthesis pathway in colon cancer. Am J Cancer Res 2022; 12:3985-4000. [PMID: 36119831 PMCID: PMC9442000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/30/2022] [Indexed: 06/15/2023] Open
Abstract
The biosynthesis of unsaturated fatty acids is involved in the initiation and progression of colon adenocarcinoma (COAD). In this study, we aimed to investigate the multi-omics characteristics of unsaturated fatty acid biosynthesis-related genes and explore their prognostic value in colon cancer by analyzing the data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. An unsaturated fatty acid biosynthesis pathway related-genes enrichment score (BUFAS) was constructed utilizing the single sample gene set enrichment analysis (ssGSEA). We discovered that a high BUFAS was associated with longer overall survival (OS) in both the training and the validation sets. Multivariable analysis including the clinical characteristics further verified the independent prognostic value of the BUFAS in both the TCGA-COAD and the GSE39582 datasets. In addition, GSEA analysis revealed that BUFAS was positively associated with several signaling pathways, including MTORC1, peroxisome, and pathways related to fatty acid metabolism, while was negatively associated with other signaling pathways, such as hedgehog, NOTCH, and Wnt/beta-catenin pathway. Furthermore, in the COAD cell lines of the Genomics of Drug Sensitivity in Cancer (GDSC) database, we found that BUFAS was positively correlated with the drug sensitivities of cisplatin, gemcitabine, camptothecin, lapatinib, and afatinib, while was negatively correlated with that of ponatinib. Moreover, in the COAD single-cell transcriptomic dataset (GSE146771), the BUFAS varied among different cell types and was enriched in mast cells and fibroblasts. Taken together, the BUFAS we constructed could be used as an independent prognostic signature in predicting the OS and drug resistance of colon cancer. Unsaturated fatty acid biosynthesis pathway might serve as potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Ling Chen
- Department of Gastrointestinal Surgery, Xiangya HospitalNo. 87 Xiangya Road, Changsha, Hunan, China
| | - Chang-Shun Yang
- Department of Surgical Oncology, Shengli Clinical Medical College of Fujian Medical UniversityNo. 134 East Street, Fuzhou, Fujian, China
| | - Si-Dong Chen
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Qiao-Xia Zhou
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Guo-Qiang Wang
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Shang-Li Cai
- Burning Rock Biotech, Building 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou, Guangdong, China
| | - Wei-Hua Li
- Department of Surgical Oncology, Shengli Clinical Medical College of Fujian Medical UniversityNo. 134 East Street, Fuzhou, Fujian, China
| | - Hong-Zhi Luo
- Department of Tumor Surgery, Zhongshan City People’s HospitalNo. 2 Sunwen Middle Road, Zhongshan, Guangdong, China
| |
Collapse
|
69
|
Liu J, Sahin C, Ahmad S, Magomedova L, Zhang M, Jia Z, Metherel AH, Orellana A, Poda G, Bazinet RP, Attisano L, Cummins CL, Peng H, Krause HM. The omega-3 hydroxy fatty acid 7( S)-HDHA is a high-affinity PPARα ligand that regulates brain neuronal morphology. Sci Signal 2022; 15:eabo1857. [PMID: 35857636 DOI: 10.1126/scisignal.abo1857] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
The nuclear receptor peroxisome proliferator-activated receptor alpha (PPARα) is emerging as an important target in the brain for the treatment or prevention of cognitive disorders. The identification of high-affinity ligands for brain PPARα may reveal the mechanisms underlying the synaptic effects of this receptor and facilitate drug development. Here, using an affinity purification-untargeted mass spectrometry (AP-UMS) approach, we identified an endogenous, selective PPARα ligand, 7(S)-hydroxy-docosahexaenoic acid [7(S)-HDHA]. Results from mass spectrometric detection of 7(S)-HDHA in mouse and rat brain tissues, time-resolved FRET analyses, and thermal shift assays collectively revealed that 7(S)-HDHA potently activated PPARα with an affinity greater than that of other ligands identified to date. We also found that 7(S)-HDHA activation of PPARα in cultured mouse cortical neurons stimulated neuronal growth and arborization, as well as the expression of genes associated with synaptic plasticity. The findings suggest that this DHA derivative supports and enhances neuronal synaptic capacity in the brain.
Collapse
Affiliation(s)
- Jiabao Liu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Cigdem Sahin
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Samar Ahmad
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 3E2
| | - Lilia Magomedova
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Minhao Zhang
- Department of Chemistry, York University, Toronto, ON M3J 1P3, Canada
| | - Zhengping Jia
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Adam H Metherel
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Arturo Orellana
- Department of Chemistry, York University, Toronto, ON M3J 1P3, Canada
| | - Gennady Poda
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
- Drug Discovery, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 3E2
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Hui Peng
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
- School of the Environment, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Henry M Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
70
|
Ruscica M, Sirtori CR, Carugo S, Calder PC, Corsini A. OMEGA-3 AND CARDIOVASCULAR PREVENTION – IS THIS STILL A CHOICE? Pharmacol Res 2022; 182:106342. [DOI: 10.1016/j.phrs.2022.106342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023]
|
71
|
Ghaddar B, Gence L, Veeren B, Bringart M, Bascands JL, Meilhac O, Diotel N. Aqueous Extract of Psiloxylon mauritianum, Rich in Gallic Acid, Prevents Obesity and Associated Deleterious Effects in Zebrafish. Antioxidants (Basel) 2022; 11:antiox11071309. [PMID: 35883799 PMCID: PMC9312056 DOI: 10.3390/antiox11071309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/26/2022] [Accepted: 06/26/2022] [Indexed: 12/04/2022] Open
Abstract
Obesity has reached epidemic proportions, and its prevalence tripled worldwide between 1975 and 2016, especially in Reunion Island, a French overseas region. Psiloxylon mauritianum, an endemic medicinal plant from Reunion Island registered in the French pharmacopeia, has recently gained interest in combating metabolic disorders because of its traditional lipid-lowering and “anti-diabetic” use. However, scientific data are lacking regarding its toxicity and its real benefits on metabolic diseases. In this study, we aim to determine the toxicity of an aqueous extract of P. mauritianum on zebrafish eleutheroembryos following the OECD toxicity assay (Organization for Economic Cooperation and Development, guidelines 36). After defining a non-toxic dose, we determined by liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) that this extract is rich in gallic acid but contains also caffeoylquinic acid, kaempferol and quercetin, as well as their respective derivatives. We also showed that the non-toxic dose exhibits lipid-lowering effects in a high-fat-diet zebrafish larvae model. In a next step, we demonstrated its preventive effects on body weight gain, hyperglycemia and liver steatosis in a diet-induced obesity model (DIO) performed in adults. It also limited the deleterious effects of overfeeding on the central nervous system (i.e., cerebral oxidative stress, blood-brain barrier breakdown, neuro-inflammation and blunted neurogenesis). Interestingly, adult DIO fish treated with P. mauritianum display normal feeding behavior but higher feces production. This indicates that the “anti-weight-gain” effect is probably due to the action of P. mauritianum on the intestinal lipid absorption and/or on the microbiota, leading to the increase in feces production. Therefore, in our experimental conditions, the aqueous extract of P. mauritianum exhibited “anti-weight-gain” properties, which prevented the development of obesity and its deleterious effects at the peripheral and central levels. These effects should be further investigated in preclinical models of obese/diabetic mice, as well as the impact of P. mauritianum on the gut microbiota.
Collapse
Affiliation(s)
- Batoul Ghaddar
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97400 Saint-Denis, La Réunion, France; (B.G.); (L.G.); (B.V.); (M.B.); (J.-L.B.); (O.M.)
| | - Laura Gence
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97400 Saint-Denis, La Réunion, France; (B.G.); (L.G.); (B.V.); (M.B.); (J.-L.B.); (O.M.)
| | - Bryan Veeren
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97400 Saint-Denis, La Réunion, France; (B.G.); (L.G.); (B.V.); (M.B.); (J.-L.B.); (O.M.)
| | - Matthieu Bringart
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97400 Saint-Denis, La Réunion, France; (B.G.); (L.G.); (B.V.); (M.B.); (J.-L.B.); (O.M.)
| | - Jean-Loup Bascands
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97400 Saint-Denis, La Réunion, France; (B.G.); (L.G.); (B.V.); (M.B.); (J.-L.B.); (O.M.)
| | - Olivier Meilhac
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97400 Saint-Denis, La Réunion, France; (B.G.); (L.G.); (B.V.); (M.B.); (J.-L.B.); (O.M.)
- CHU de La Réunion, 97400 Saint-Denis, La Réunion, France
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97400 Saint-Denis, La Réunion, France; (B.G.); (L.G.); (B.V.); (M.B.); (J.-L.B.); (O.M.)
- Correspondence:
| |
Collapse
|
72
|
Targeting Nuclear Receptors in Lung Cancer—Novel Therapeutic Prospects. Pharmaceuticals (Basel) 2022; 15:ph15050624. [PMID: 35631448 PMCID: PMC9145966 DOI: 10.3390/ph15050624] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023] Open
Abstract
Lung cancer, the second most commonly diagnosed cancer, is the major cause of fatalities worldwide for both men and women, with an estimated 2.2 million new incidences and 1.8 million deaths, according to GLOBOCAN 2020. Although various risk factors for lung cancer pathogenesis have been reported, controlling smoking alone has a significant value as a preventive measure. In spite of decades of extensive research, mechanistic cues and targets need to be profoundly explored to develop potential diagnostics, treatments, and reliable therapies for this disease. Nuclear receptors (NRs) function as transcription factors that control diverse biological processes such as cell growth, differentiation, development, and metabolism. The aberrant expression of NRs has been involved in a variety of disorders, including cancer. Deregulation of distinct NRs in lung cancer has been associated with numerous events, including mutations, epigenetic modifications, and different signaling cascades. Substantial efforts have been made to develop several small molecules as agonists or antagonists directed to target specific NRs for inhibiting tumor cell growth, migration, and invasion and inducing apoptosis in lung cancer, which makes NRs promising candidates for reliable lung cancer therapeutics. The current work focuses on the importance of various NRs in the development and progression of lung cancer and highlights the different small molecules (e.g., agonist or antagonist) that influence NR expression, with the goal of establishing them as viable therapeutics to combat lung cancer.
Collapse
|
73
|
Søderstrøm S, Lille-Langøy R, Yadetie F, Rauch M, Milinski A, Dejaegere A, Stote RH, Goksøyr A, Karlsen OA. Agonistic and potentiating effects of perfluoroalkyl substances (PFAS) on the Atlantic cod (Gadus morhua) peroxisome proliferator-activated receptors (Ppars). ENVIRONMENT INTERNATIONAL 2022; 163:107203. [PMID: 35364415 DOI: 10.1016/j.envint.2022.107203] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
Toxicity mediated by per- and polyfluoroalkyl substances (PFAS), and especially perfluoroalkyl acids (PFAAs), has been linked to activation of peroxisome proliferator-activated receptors (Ppar) in many vertebrates. Here, we present the primary structures, phylogeny, and tissue-specific distributions of the Atlantic cod (Gadus morhua) gmPpara1, gmPpara2, gmPparb, and gmPparg, and demonstrate that the carboxylic acids PFHxA, PFOA, PFNA, as well as the sulfonic acid PFHxS, activate gmPpara1 in vitro, which was also supported by in silico analyses. Intriguingly, a binary mixture of PFOA and the non-activating PFOS produced a higher activation of gmPpara1 compared to PFOA alone, suggesting that PFOS has a potentiating effect on receptor activation. Supporting the experimental data, docking and molecular dynamics simulations of single and double-ligand complexes led to the identification of a putative allosteric binding site, which upon binding of PFOS stabilizes an active conformation of gmPpara1. Notably, binary exposures of gmPpara1, gmPpara2, and gmPparb to model-agonists and PFAAs produced similar potentiating effects. This study provides novel mechanistic insights into how PFAAs may modulate the Ppar signaling pathway by either binding the canonical ligand-binding pocket or by interacting with an allosteric binding site. Thus, individual PFAAs, or mixtures, could potentially modulate the Ppar-signaling pathway in Atlantic cod by interfering with at least one gmPpar subtype.
Collapse
Affiliation(s)
- Sofie Søderstrøm
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway; Institute of Marine Research, Nordnesgaten 50, NO-5005 Bergen, Norway(1)
| | - Roger Lille-Langøy
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway; Institute of Marine Research, Nordnesgaten 50, NO-5005 Bergen, Norway(1)
| | - Fekadu Yadetie
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway
| | - Mateusz Rauch
- Department of Integrated Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, Illkirch, France
| | - Ana Milinski
- Department of Integrated Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, Illkirch, France
| | - Annick Dejaegere
- Department of Integrated Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, Illkirch, France
| | - Roland H Stote
- Department of Integrated Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, Illkirch, France
| | - Anders Goksøyr
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway
| | - Odd André Karlsen
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway.
| |
Collapse
|
74
|
Johnson WT, Dorn NC, Ogbonna DA, Bottini N, Shah NJ. Lipid-based regulators of immunity. Bioeng Transl Med 2022; 7:e10288. [PMID: 35600637 PMCID: PMC9115682 DOI: 10.1002/btm2.10288] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 11/22/2022] Open
Abstract
Lipids constitute a diverse class of molecular regulators with ubiquitous physiological roles in sustaining life. These carbon-rich compounds are primarily sourced from exogenous sources and may be used directly as structural cellular building blocks or as a substrate for generating signaling mediators to regulate cell behavior. In both of these roles, lipids play a key role in both immune activation and suppression, leading to inflammation and resolution, respectively. The simple yet elegant structural properties of lipids encompassing size, hydrophobicity, and molecular weight enable unique biodistribution profiles that facilitate preferential accumulation in target tissues to modulate relevant immune cell subsets. Thus, the structural and functional properties of lipids can be leveraged to generate new materials as pharmacological agents for potently modulating the immune system. Here, we discuss the properties of three classes of lipids: polyunsaturated fatty acids, short-chain fatty acids, and lipid adjuvants. We describe their immunoregulatory functions in modulating disease pathogenesis in preclinical models and in human clinical trials. We conclude with an outlook on harnessing the diverse and potent immune modulating properties of lipids for immunoregulation.
Collapse
Affiliation(s)
- Wade T. Johnson
- Department of NanoengineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Nicholas C. Dorn
- Department of NanoengineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
- Chemical Engineering ProgramUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Dora A. Ogbonna
- Department of NanoengineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
- Chemical Engineering ProgramUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Nunzio Bottini
- Division of Rheumatology, Allergy and Immunology, Department of MedicineUniversity of California, San DiegoLa JollaCaliforniaUSA
- Program in ImmunologyUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Nisarg J. Shah
- Department of NanoengineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
- Chemical Engineering ProgramUniversity of California, San DiegoLa JollaCaliforniaUSA
- Program in ImmunologyUniversity of California, San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
75
|
Guo YX, Wang BY, Gao H, Hua RX, Gao L, He CW, Wang Y, Xu JD. Peroxisome Proliferator–Activated Receptor-α: A Pivotal Regulator of the Gastrointestinal Tract. Front Mol Biosci 2022; 9:864039. [PMID: 35558563 PMCID: PMC9086433 DOI: 10.3389/fmolb.2022.864039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/14/2022] [Indexed: 11/15/2022] Open
Abstract
Peroxisome proliferator–activated receptor (PPAR)-α is a ligand-activated transcription factor distributed in various tissues and cells. It regulates lipid metabolism and plays vital roles in the pathology of the cardiovascular system. However, its roles in the gastrointestinal tract (GIT) are relatively less known. In this review, after summarizing the expression profile of PPAR-α in the GIT, we analyzed its functions in the GIT, including physiological control of the lipid metabolism and pathologic mediation in the progress of inflammation. The mechanism of this regulation could be achieved via interactions with gut microbes and further impact the maintenance of body circadian rhythms and the secretion of nitric oxide. These are also targets of PPAR-α and are well-described in this review. In addition, we also highlighted the potential use of PPAR-α in treating GIT diseases and the inadequacy of clinical trials in this field.
Collapse
Affiliation(s)
- Yue-Xin Guo
- Department of Oral Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bo-Ya Wang
- Eight Program of Clinical Medicine, Peking University Health Science Center, Beijing, China
| | - Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Rong-Xuan Hua
- Clinical Medicine of “5+3” Program, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Gao
- Department of Biomedical Informatics, Faculty of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Cheng-Wei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Dermatology, Tongren Hospital, Capital Medical University, Beijing, China
| | - Jing-Dong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- *Correspondence: Jing-Dong Xu,
| |
Collapse
|
76
|
Kitahata K, Matsuo K, Sato M, Susami Y, Hara Y, Morikawa T, Oiso N, Kawada A, Otsuka A, Nakayama T. Anti-allergic effect of ascorbic acid derivative DDH-1 in a mouse model of atopic dermatitis. Exp Dermatol 2022; 31:1234-1242. [PMID: 35441724 DOI: 10.1111/exd.14578] [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: 09/03/2021] [Revised: 03/24/2022] [Accepted: 04/17/2022] [Indexed: 11/28/2022]
Abstract
Atopic dermatitis (AD) is the most common inflammatory skin disease, which is characterized by excessive Th2 immune responses. In AD patients, the expression of the chemokines CCL17 and CCL22 is increased in skin lesions, leading to the infiltration of Th2 cells. In addition, typical pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6, have also been shown to be associated with the pathogenesis of AD. Recently, DDH-1, an ascorbic acid derivative, has been synthesized and demonstrated to have a more stabilized structure and better skin penetrability. Furthermore, DDH-1 has been shown to suppress pro-inflammatory cytokine expression in vitro and in vivo. Therefore, using an AD mouse model, we evaluated the effect of DDH-1 to reduce allergic skin inflammation. We found that cutaneous administration of DDH-1 significantly reduced the expression levels of TNF-α, IL-1β, and IL-6 in the skin lesions of AD-like mice. Additionally, DDH-1 administration also significantly reduced the expression levels of CCL17 and CCL22, resulting in decreased skin infiltration of Th2 cells. Consequently, DDH-1 reduced ear and epidermal thickness, the serum IgE levels, and the number of infiltrating inflammatory cells and mast cells into the AD-like skin lesions. Combination treatment with DDH-1 and corticosteroid more efficiently improved the skin lesions compared to corticosteroid alone. Collectively, our results suggest that DDH-1 has an anti-allergic effect in an AD mouse model by reducing not only the pro-inflammatory cytokine expression but also the Th2-associated chemokine expression. Thus, DDH-1 may be beneficial for AD treatment and prevention as a monotherapy or in combination with corticosteroids.
Collapse
Affiliation(s)
- Kosuke Kitahata
- Division of Chemotherapy, Kindai University, Faculty of Pharmacy, Higashi-Osaka, Kowakae 3-4-1, Osaka, 577-8502, Japan
| | - Kazuhiko Matsuo
- Division of Chemotherapy, Kindai University, Faculty of Pharmacy, Higashi-Osaka, Kowakae 3-4-1, Osaka, 577-8502, Japan
| | - Masako Sato
- Department of Dermatology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osaka-sayama, Osaka, Japan
| | - Yoko Susami
- Division of Chemotherapy, Kindai University, Faculty of Pharmacy, Higashi-Osaka, Kowakae 3-4-1, Osaka, 577-8502, Japan
| | - Yuta Hara
- Division of Chemotherapy, Kindai University, Faculty of Pharmacy, Higashi-Osaka, Kowakae 3-4-1, Osaka, 577-8502, Japan
| | - Toshio Morikawa
- Department of Pharmaceutical Food Sciences, Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka, 577-8502, Japan
| | - Naoki Oiso
- Department of Dermatology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osaka-sayama, Osaka, Japan
| | - Akira Kawada
- Department of Dermatology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osaka-sayama, Osaka, Japan
| | - Atsushi Otsuka
- Department of Dermatology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osaka-sayama, Osaka, Japan
| | - Takashi Nakayama
- Division of Chemotherapy, Kindai University, Faculty of Pharmacy, Higashi-Osaka, Kowakae 3-4-1, Osaka, 577-8502, Japan
| |
Collapse
|
77
|
Tachibana K, Fukuda S, Fukushima J, Ishimoto K, Sakata M, Nishimori Y, Doi T. Exploring Compounds to be used as Cosmetic Agents that Activate Peroxisome Proliferator-Activated Receptor Alpha. Int J Cosmet Sci 2022; 44:189-200. [PMID: 35244215 PMCID: PMC9321093 DOI: 10.1111/ics.12767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The human epidermis is formed by the proliferation and differentiation of keratinocytes adjacent to the basement membrane. The outermost layer, the stratum corneum, is equipped with a barrier function that prevents water evaporation, and intercellular lipids play an important role in this barrier function. When the barrier is functioning normally, evaporation is prevented; however, when barrier function is impaired, moisture evaporates, resulting in dry and rough skin. Therefore, maintenance of normal barrier function is critical for maintaining normal skin function. Peroxisome proliferator-activated receptor α (PPARα) is mainly involved in lipid metabolism in the liver but is also expressed in the epidermis and is involved in inducing keratinocyte differentiation, promoting lipid production, maintaining barrier function, and suppressing skin inflammation. Hence, compounds that activate PPARα are expected to control skin function. Therefore, we identified PPARα activators from among extracts of natural resources that have been approved for use in humans and analyzed the effects of these extracts on skin function. METHODS First, extracts of 474 natural resources were screened using a PPARα activator screening cell line independently constructed in our laboratory. Next, reporter assays were performed using the Gal4-chimera system to evaluate whether these extracts act as ligands for PPARα. We then analyzed their effect on primary normal human epidermal keratinocyte cells by using real-time RT-PCR. Finally, we evaluated PPARα activation effect by the combination of these extracts. RESULTS We identified 36 extracts having the effect of activating PPARα. In particular, #419, a Typha angustifolia spike extract, showed concentration-dependent transcriptional activation through PPARα-LBD and was considered to be likely to contain a compound that is a ligand of PPARα. #419 increased the expression of PPARα target genes as well as genes related to skin function in primary cultured human epidermal keratinocytes. Finally, the use of #419 in combination with nine extracts increased PPAR activity more than twice as much as #419 alone treatment. CONCLUSIONS These results showed that the reporter cell line could be useful for discovering extracts of natural resources and that the identified Typha angustifolia spike extract could be used in cosmetics that activate PPARα, which expected to improve skin function.
Collapse
Affiliation(s)
- Keisuke Tachibana
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Syohei Fukuda
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Jun Fukushima
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Maruho Co., Ltd., 1-11-1 Nakatsu, Kita-ku, Osaka, 531-0071, Japan
| | - Kenji Ishimoto
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Sakata
- Maruho Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa, Kanagawa, 251-8555, Japan
| | | | - Takefumi Doi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
78
|
Gangwar SK, Kumar A, Jose S, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Nuclear receptors in oral cancer-emerging players in tumorigenesis. Cancer Lett 2022; 536:215666. [DOI: 10.1016/j.canlet.2022.215666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 12/24/2022]
|
79
|
Atefi M, Entezari MH, Vahedi H, Hassanzadeh A. The effects of sesame oil on metabolic biomarkers: a systematic review and meta-analysis of clinical trials. J Diabetes Metab Disord 2022; 21:1065-1080. [PMID: 35673414 PMCID: PMC9167273 DOI: 10.1007/s40200-022-00997-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/30/2022] [Indexed: 11/26/2022]
Abstract
Background Clinical evidences showing the effects of sesame oil on metabolic biomarkers led to inconsistent results. Propose This meta-analysis was designed to examine the effects of sesame oil on metabolic biomarkers in adults, including the maximum number of clinical trials. Methods Google Scholar, PubMed, Web of Science, and Scopus were systematically searched to date up to July 2021 to identify eligible clinical trial studies. We obtained the pooled estimates of weighted mean differences (WMDs) with their 95% confidence intervals (CIs) using random-effects meta-analysis. Result Meta-analysis showed that sesame oil consumption significantly lowered the levels of fasting blood glucose (FBG) (WMD: -3.268 mg/dl; 95% CI: -4.677, -1.86; P < 0.001), and malondialdehyde (MDA) (WMD: -4.847 nmol/dL; 95% CI: -7.051, -2.698; P < 0.001) between the intervention and control groups. Also, this study showed sesame oil consumption significantly decreased HbA1C (WMD: -2.057%; 95% CI: -3.467, -0.646; P = 0.004), systolic blood pressure (SBP) (WMD: -2.679 mmHg; 95% CI: -5.257, -0.101; P < 0.001), diastolic blood pressure (DBP) (WMD: -1.981 mmHg; 95% CI: -3.916, -0.046; P = 0.045), body weight (WMD: -0.346 kg; 95% CI: -0.641, -0.051; P = 0.021), and body mass index (BMI) (WMD: -0.385 kg/m2; 95% CI:-0.721, -0.049; P = 0.025) after intervention. No significant effect was seen in serum insulin levels (p > .05). Conclusions The current study provided some evidence regarding the beneficial effects of sesame oil on metabolic biomarkers. Further studies are still required to confirm our results. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-022-00997-2.
Collapse
Affiliation(s)
- Masoumeh Atefi
- Department of Clinical Nutrition, School of Nutrition & Food Science, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohammad Hassan Entezari
- Food Security Research Center and Department of Clinical Nutrition, School of Nutrition & Food Science, Isfahan University of Medical Sciences, PO Box 81745, Isfahan, I.R Iran
| | - Hamid Vahedi
- Department of Gastroenterology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, I.R Iran
| | - Akbar Hassanzadeh
- Department of Epidemiology and Biostatistics, School of Health, Isfahan University of Medical Sciences, Isfahan, I.R Iran
| |
Collapse
|
80
|
Sedghi M, Mohammadi I, Sarrami Z, Ghasemi R, Azarfar A. Effects of a yeast cell wall product on the performance of broiler chickens and PGC-1α, TLR4, IL-10 and PPARγ genes expression. ITALIAN JOURNAL OF ANIMAL SCIENCE 2022. [DOI: 10.1080/1828051x.2021.2025161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Mohammad Sedghi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Ishmael Mohammadi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Zahra Sarrami
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Razie Ghasemi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Arash Azarfar
- Department of Animal Science, Faculty of Agriculture, Lorestan University, Khorramabad, Iran
| |
Collapse
|
81
|
Bian C, Sun J, Huang X, Ji S, Ji H. Endoplasmic reticulum stress is involved in lipid accumulation induced by oleic acid in adipocytes of grass carp (Ctenopharyngodon idella): focusing on the transcriptional level. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:275-284. [PMID: 35091868 DOI: 10.1007/s10695-021-01031-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/21/2021] [Indexed: 06/14/2023]
Abstract
It has been extensively claimed that endoplasmic reticulum stress (ER stress) is related to lipid accumulation in mammals, but little is known in fish. This study aims at elucidating the role of ER stress in mediating lipid accumulation induced by monounsaturated oleic acid (OA) with a focus on the transcriptional level. We treated the adipocytes of grass carp with 200 μM and 400 μM OA, respectively, while the control group was treated with 2% bovine serum albumin (BSA). The results showed that cell viability was significantly improved, while 400 μM OA treatment promoted neutral lipid accumulation along with stimulating ER stress more obviously. Although lipolysis and fatty acid β-oxidation were activated simultaneously, the primary effect of OA seems to be promotion of lipid accumulation. To further explore whether ER stress affects lipid accumulation, 4-phenyl butyric acid (4-PBA), an effective inhibitor of ER stress, was used to pretreat the cells for 4 h. Unsurprisingly, it was found that the mRNA expressions of genes linked with ER stress were decreased. Intracellular triglyceride (TG) content was also decreased, which was in accordance with the mRNA expressions of adipogenic and lipogenic transcription factors as well as their target genes. Collectively, our data shows that ER stress may take part in OA-induced lipid accumulation in adipocytes via activating adipogenesis and lipogenesis. Based on this, strategies for protecting ER could be used to alleviate excessive accumulation of lipid in grass carp adipose tissue.
Collapse
Affiliation(s)
- Chenchen Bian
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Jian Sun
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Xiaocheng Huang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Shanghong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China.
| |
Collapse
|
82
|
Sun J, Zhang L, Zhou F, Shaw S, Roos A, Berger M, Bäcklin BM, Huang Y, Zheng X, Wang X, Chen D. Hepatic Fatty Acid Profiles Associated with Exposure to Emerging and Legacy Halogenated Contaminants in Two Harbor Seal Populations across the North Atlantic. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:1830-1840. [PMID: 35068154 DOI: 10.1021/acs.est.1c06512] [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] [Indexed: 06/14/2023]
Abstract
Fatty acids (FAs) have been extensively used as indicators of foraging ecology in marine mammals, yet their association with exposure to contaminants has rarely been investigated. The present study provided the first characterization of the relationship between hepatic FA profiles and exposure to a suite of contaminants in a sentinel species─the harbor seal (Phoca vitulina)─from the Gulf of Maine and the south coast of Sweden. FA profiles differed in the two seal populations, and the levels of legacy and alternative brominated flame retardants and polyhalogenated carbazoles were significantly elevated in Maine seals. Correlations between individual FAs and multiple flame retardants (FRs) and poly- and perfluoroalkyl substances (PFASs) were found in seals from both populations. Moreover, several FR and PFAS chemicals were significantly associated with the estimated desaturating enzyme activity inferred from the FA profiles. The ratios of poly to monounsaturated FAs (∑PUFAs/∑MUFAs) and those of unsaturated to saturated FAs (∑UFAs/∑SFAs) were significantly associated with HBBZ, PFHxS, or BDE 47 in seals from Maine and Sweden, whereas ∑n - 6/∑n - 3 PUFAs was significantly associated with BDE 154 and 36-CCZ in Swedish and Maine seals, respectively. Our results suggest the lipid metabolism-disrupting potential of these contaminants in marine mammals and warrant continuous biomonitoring and risk assessment, considering the critical role of PUFAs in vital biological processes.
Collapse
Affiliation(s)
- Jiachen Sun
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, CN-510632 Guangzhou, Guangdong, China
| | - Long Zhang
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, CN-510632 Guangzhou, Guangdong, China
| | - Fengli Zhou
- Research Center of Harmful Algae and Marine Biology, Jinan University, CN-510632 Guangzhou, Guangdong, China
| | - Susan Shaw
- Shaw Institute, Blue Hill Research Center, Blue Hill, Maine 04614, United States
| | - Anna Roos
- Department of Contaminant Research and Monitoring, Swedish Museum of Natural History, Box 5007, Stockholm SE-10405, Sweden
| | - Michelle Berger
- Shaw Institute, Blue Hill Research Center, Blue Hill, Maine 04614, United States
| | - Britt-Marie Bäcklin
- Department of Contaminant Research and Monitoring, Swedish Museum of Natural History, Box 5007, Stockholm SE-10405, Sweden
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, CN-230032 Hefei, Anhui, China
| | - Xiaoshi Zheng
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, CN-510632 Guangzhou, Guangdong, China
| | - Xiaodong Wang
- Research Center of Harmful Algae and Marine Biology, Jinan University, CN-510632 Guangzhou, Guangdong, China
| | - Da Chen
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, CN-510632 Guangzhou, Guangdong, China
| |
Collapse
|
83
|
Sahin C, Magomedova L, Ferreira TAM, Liu J, Tiefenbach J, Alves PS, Queiroz FJG, Oliveira ASD, Bhattacharyya M, Grouleff J, Nogueira PCN, Silveira ER, Moreira DC, Leite JRSDA, Brand GD, Uehling D, Poda G, Krause H, Cummins CL, Romeiro LAS. Phenolic Lipids Derived from Cashew Nut Shell Liquid to Treat Metabolic Diseases. J Med Chem 2022; 65:1961-1978. [PMID: 35089724 DOI: 10.1021/acs.jmedchem.1c01542] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metabolic diseases are increasing at staggering rates globally. The peroxisome proliferator-activated receptors (PPARα/γ/δ) are fatty acid sensors that help mitigate imbalances between energy uptake and utilization. Herein, we report compounds derived from phenolic lipids present in cashew nut shell liquid (CNSL), an abundant waste byproduct, in an effort to create effective, accessible, and sustainable drugs. Derivatives of anacardic acid and cardanol were tested for PPAR activity in HEK293 cell co-transfection assays, primary hepatocytes, and 3T3-L1 adipocytes. In vivo studies using PPAR-expressing zebrafish embryos identified CNSL derivatives with varying tissue-specific activities. LDT409 (23) is an analogue of cardanol with partial agonist activity for PPARα and PPARγ. Pharmacokinetic profiling showed that 23 is orally bioavailable with a half-life of 4 h in mice. CNSL derivatives represent a sustainable source of selective PPAR modulators with balanced intermediate affinities (EC50 ∼ 100 nM to 10 μM) that provide distinct and favorable gene activation profiles for the treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Cigdem Sahin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Lilia Magomedova
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Thais A M Ferreira
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Jiabao Liu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Jens Tiefenbach
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Priscilla S Alves
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Fellipe J G Queiroz
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Andressa S de Oliveira
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Mousumi Bhattacharyya
- Ontario Institute for Cancer Research, MaRS Centre, Toronto, Ontario M5G 0A3, Canada
| | - Julie Grouleff
- Ontario Institute for Cancer Research, MaRS Centre, Toronto, Ontario M5G 0A3, Canada
| | - Patrícia C N Nogueira
- CENAUREMN, Federal University of Ceará, Campus do Pici, Fortaleza, CE 60020-181, Brazil
| | - Edilberto R Silveira
- CENAUREMN, Federal University of Ceará, Campus do Pici, Fortaleza, CE 60020-181, Brazil
| | - Daniel C Moreira
- Faculty of Medicine, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | | | - Guilherme D Brand
- Chemistry Institute, University of Brasília, Campus Universitário Darcy Ribeiro, Brasília, DF 70910-900, Brazil
| | - David Uehling
- Ontario Institute for Cancer Research, MaRS Centre, Toronto, Ontario M5G 0A3, Canada
| | - Gennady Poda
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada.,Ontario Institute for Cancer Research, MaRS Centre, Toronto, Ontario M5G 0A3, Canada
| | - Henry Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Luiz A S Romeiro
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| |
Collapse
|
84
|
Fernández-Díaz J, Beteta-Göbel R, Torres M, Cabot J, Fernández-García P, Lladó V, Escribá PV, Busquets X. Tri-2-Hydroxyarachidonein Induces Cytocidal Autophagy in Pancreatic Ductal Adenocarcinoma Cancer Cell Models. Front Physiol 2022; 12:782525. [PMID: 35126175 PMCID: PMC8811354 DOI: 10.3389/fphys.2021.782525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Cell proliferation in pancreatic cancer is determined by a complex network of signaling pathways. Despite the extensive understanding of these protein-mediated signaling processes, there are no significant drug discoveries that could considerably improve a patient’s survival. However, the recent understanding of lipid-mediated signaling gives a new perspective on the control of the physiological state of pancreatic cells. Lipid signaling plays a major role in the induction of cytocidal autophagy and can be exploited using synthetic lipids to induce cell death in pancreatic cancer cells. In this work, we studied the activity of a synthetic lipid, tri-2-hydroxyarachidonein (TGM4), which is a triacylglycerol mimetic that contains three acyl moieties with four double bonds each, on cellular and in vivo models of pancreatic cancer. We demonstrated that TGM4 inhibited proliferation of Mia-PaCa-2 (human pancreatic carcinoma) and PANC-1 (human pancreatic carcinoma of ductal cells) in in vitro models and in an in vivo xenograft model of Mia-PaCa-2 cells. In vitro studies demonstrated that TGM4 induced cell growth inhibition paralleled with an increased expression of PARP and CHOP proteins together with the presence of sub-G0 cell cycle events, indicating cell death. This cytocidal effect was associated with elevated ER stress or autophagy markers such as BIP, LC3B, and DHFR. In addition, TGM4 activated peroxisome proliferator-activated receptor gamma (PPAR-γ), which induced elevated levels of p-AKT and downregulation of p-c-Jun. We conclude that TGM4 induced pancreatic cell death by activation of cytocidal autophagy. This work highlights the importance of lipid signaling in cancer and the use of synthetic lipid structures as novel and potential approaches to treat pancreatic cancer and other neoplasias.
Collapse
Affiliation(s)
- Javier Fernández-Díaz
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain
| | - Roberto Beteta-Göbel
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain
| | - Manuel Torres
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain
| | - Joan Cabot
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain
| | | | - Victoria Lladó
- Laminar Pharmaceuticals, Department of R&D, Palma de Mallorca, Spain
| | - Pablo V. Escribá
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain
| | - Xavier Busquets
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain
- *Correspondence: Xavier Busquets,
| |
Collapse
|
85
|
Robinson GF, Sooda KKY, Phillips RM, Allison SJ, Javid FA. Investigation of the cytotoxicity induced by didocosahexaenoin, an omega 3 derivative, in human prostate carcinoma cell lines. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100085. [PMID: 35112078 PMCID: PMC8790608 DOI: 10.1016/j.crphar.2022.100085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 10/29/2022] Open
Abstract
The aim of the present study was to investigate the cytotoxicity induced by an omega-3 derivative, didocosahexaenoin (Dido) on human prostate carcinoma cells and to compare the cytotoxicity to that of docosahexaenoic acid (DHA). Different carcinoma- and non-carcinoma cells were exposed to various concentrations of omega-3 compounds at varying exposure times and the cytotoxicity was measured by MTT assay. The mechanism of Dido-induced apoptosis was investigated in prostate carcinoma cells. Dido induced stronger cytotoxicity than DHA in human prostate carcinoma cells in a dose- and time-dependent manner. Dido was also more selective and potent in inducing cytotoxicity in prostate carcinoma cells than other carcinoma cell lines tested. Pre-treatment with Dido increased the level of reactive oxygen species (ROS) in prostate carcinoma cells. Pre-treatment with various antioxidants reduced the cytotoxicity induced by Dido. Pre-treatment with Dido ≥30 μM also induced apoptosis which was suggested to involve an externalisation of phosphatidyl serine, a significant increase in the mitochondrial membrane potential (p < 0.01) and the level of activated caspase 3/7 (p < 0.05) in prostate carcinoma cells. This study is the first to show that Dido induced cytotoxicity with high selectivity and higher potency than DHA in human prostate carcinoma cells. The mechanism of action is likely to involve an increase in the level of ROS, loss in the mitochondrial membrane potential as well as externalisation of phosphatidyl serine and increase in the caspase 3/7 activity. Dido may have potential to be used for the adjuvant therapy or combination therapy with conventional chemotherapeutic drugs.
Collapse
Affiliation(s)
- Glenn F. Robinson
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Kartheek KY. Sooda
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Roger M. Phillips
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Simon J. Allison
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Farideh A. Javid
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| |
Collapse
|
86
|
The Preservation of PPARγ Genome Duplicates in Some Teleost Lineages: Insights into Lipid Metabolism and Xenobiotic Exploitation. Genes (Basel) 2022; 13:genes13010107. [PMID: 35052447 PMCID: PMC8774674 DOI: 10.3390/genes13010107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/28/2021] [Accepted: 12/31/2021] [Indexed: 01/10/2023] Open
Abstract
Three peroxisome proliferator-activated receptor paralogues (PPARα, -β and -γ) are currently recognized in vertebrate genomes. PPARγ is known to modulate nutrition, adipogenesis and immunity in vertebrates. Natural ligands of PPARγ have been proposed; however, the receptor also binds synthetic ligands such as endocrine disruptors. Two paralogues of PPARα and PPARβ have been documented in teleost species, a consequence of the 3R WGD. Recently, two PPARγ paralogue genes were also identified in Astyanax mexicanus. We aimed to determine whether the presence of two PPARγ paralogues is prevalent in other teleost genomes, through genomic and phylogenetic analysis. Our results showed that besides Characiformes, two PPARγ paralogous genes were also identified in other teleost taxa, coinciding with the teleost-specific, whole-genome duplication and with the retention of both genes prior to the separation of the Clupeocephala. To functionally characterize these genes, we used the European sardine (Sardina pilchardus) as a model. PPARγA and PPARγB display a different tissue distribution, despite the similarity of their functional profiles: they are unresponsive to tested fatty acids and other human PPARγ ligands yet yield a transcriptional response in the presence of tributyltin (TBT). This observation puts forward the relevance of comparative analysis to decipher alternative binding architectures and broadens the disruptive potential of man-made chemicals for aquatic species.
Collapse
|
87
|
Wang Y, Wang J, Zhao Y, Liu P, Cai D, Zhang X, Gao L. Regulatory mechanisms of Beta-carotene and BCMO1 in adipose tissues: A gene enrichment-based bioinformatics analysis. Hum Exp Toxicol 2022; 41:9603271211072871. [PMID: 35306905 DOI: 10.1177/09603271211072871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Beta-carotene (β-carotene, BC) is one of the carotenoids most commonly consumed by humans. BCMO1 is expressed in various human tissues and is considered to be a key enzyme that converts BC into vitamin A. Studies indicated that BC-derived carotenoid signaling molecules affected the physiological functions of fat cells. In order to investigate the role and possible molecular mechanism of BC in mouse adipocytes, we conducted 4-group and 2-group difference analysis based on the data of GSE27271 chip in the Gene Expression Omnibus database. Genes differentially expressed in the inguinal white adipose tissue of mice were screened out and combined with the STRING database to construct protein-protein interaction (PPI) networks. Among them, Alb (albumin), Mug1 (murinoglobulin-1) and Uox (urate oxidase) genes were at relatively key positions and may affect the action of BC. Besides, Ppara (peroxisome proliferator-activated receptor alpha), Acly (ATP-citrate lyase) and Fabp5 (fatty acid-binding protein 5) genes constituted functional partners with many genes in the PPI network, and these genes may be Bcmo1 targeting molecules. Gene Ontology (GO) function and signaling pathways enrichment analysis were performed on the genes with protein interaction relationship in the PPI network. Fatty acid binding, cholesterol metabolic process, and regulation of fatty acid metabolic process were significantly enriched, and PPAR signaling pathway showed the most significant, indicating that BC and Bcmo1 might synergistically affect body metabolic functions such as fat metabolism. In general, BC and Bcmo1 may play a role in fat metabolism in mice, thereby affecting other functions or diseases.
Collapse
Affiliation(s)
- Yutao Wang
- Institute of Quality Standard and Testing Technology for Agro-Products, 74641Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Test Technology on Food Quality and Safety, Jinan, China
| | - Jinshen Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuhua Zhao
- Institute of Quality Standard and Testing Technology for Agro-Products, 74641Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Test Technology on Food Quality and Safety, Jinan, China
| | - Pingxiang Liu
- Institute of Quality Standard and Testing Technology for Agro-Products, 74641Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Test Technology on Food Quality and Safety, Jinan, China
| | - Da Cai
- Institute of Quality Standard and Testing Technology for Agro-Products, 74641Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Test Technology on Food Quality and Safety, Jinan, China
| | - Xiao Zhang
- Institute of Quality Standard and Testing Technology for Agro-Products, 74641Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Test Technology on Food Quality and Safety, Jinan, China
| | - Lei Gao
- Institute of Quality Standard and Testing Technology for Agro-Products, 74641Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Test Technology on Food Quality and Safety, Jinan, China
| |
Collapse
|
88
|
Chang RC, Thangavelu CS, Joloya EM, Kuo A, Li Z, Blumberg B. Cannabidiol Promotes Adipogenesis of Human and Mouse Mesenchymal Stem Cells via PPARγ by Inducing Lipogenesis but Not Lipolysis. Biochem Pharmacol 2022; 197:114910. [DOI: 10.1016/j.bcp.2022.114910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 11/02/2022]
|
89
|
Qi Y, Zhang H, Fan H, Wang X, Zhao A, Tian Y, Yang G, Li C, Wei J, Yao W, Hao C. PPARγ/LXRα axis mediated phenotypic plasticity of lung fibroblasts in silica-induced experimental silicosis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 292:118272. [PMID: 34718086 DOI: 10.1016/j.envpol.2021.118272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 09/05/2021] [Accepted: 09/29/2021] [Indexed: 06/13/2023]
Abstract
Silicosis is a disease mainly caused by pulmonary interstitial fibrosis caused by long-term inhalation of dust with excessively high content of free SiO2. Transdifferentiation of lung fibroblasts into myofibroblasts is an important cellular basis for silicosis, but the key transcription factors (TFs) involved in this process are still unclear. In order to explore the biological regulation of transcription factor PPARγ/LXRα in silica-induced pulmonary fibrosis, this study explored the molecular mechanism of PPARγ/LXRα involved in regulating transcription factors related to SiO2-induced lung injury at the cellular level and in animal models. ChIP-qPCR detected that PPARγ directly regulated the transcriptional activity of the LXRα gene promoter, while the PPARγ agonist RSG increased the expression of LXRα. In addition, we demonstrated in the cell model that upregulation of LXRα can inhibit silica-mediated fibroblast transdifferentiation, accompanied by an increase in the expression of SREBF1, PLTP and ABCA1. The results of LXRα silencing experiment matched those of overexpression experiment. These studies explored the role of LXRα in plasticity and phenotypic transformation between lung fibroblasts and myofibroblasts. Therefore, inhibiting or reversing the transdifferentiation of lung fibroblasts to myofibroblasts by intervening PPARγ/LXRα may provide a new therapeutic target for the treatment of silicosis.
Collapse
Affiliation(s)
- Yuanmeng Qi
- School of Public Health, Zhengzhou University, Henan, China
| | - Haichen Zhang
- School of Pharmacy, Zhengzhou University, Henan, China
| | - Hui Fan
- Department of Ultrasound, The Third Affiliated Hospital of Zhengzhou University, Henan, China
| | - Xinyu Wang
- School of Public Health, Zhengzhou University, Henan, China
| | - Ahui Zhao
- Henan Disease Control and Prevention Center, Henan, China
| | - Yangyang Tian
- School of Public Health, Zhengzhou University, Henan, China
| | - Guo Yang
- School of Public Health, Zhengzhou University, Henan, China
| | - Chao Li
- School of Public Health, Zhengzhou University, Henan, China
| | - Jingjing Wei
- School of Public Health, Zhengzhou University, Henan, China
| | - Wu Yao
- School of Public Health, Zhengzhou University, Henan, China
| | - Changfu Hao
- School of Public Health, Zhengzhou University, Henan, China.
| |
Collapse
|
90
|
Baek J, He C, Afshinnia F, Michailidis G, Pennathur S. Lipidomic approaches to dissect dysregulated lipid metabolism in kidney disease. Nat Rev Nephrol 2022; 18:38-55. [PMID: 34616096 PMCID: PMC9146017 DOI: 10.1038/s41581-021-00488-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2021] [Indexed: 01/03/2023]
Abstract
Dyslipidaemia is a hallmark of chronic kidney disease (CKD). The severity of dyslipidaemia not only correlates with CKD stage but is also associated with CKD-associated cardiovascular disease and mortality. Understanding how lipids are dysregulated in CKD is, however, challenging owing to the incredible diversity of lipid structures. CKD-associated dyslipidaemia occurs as a consequence of complex interactions between genetic, environmental and kidney-specific factors, which to understand, requires an appreciation of perturbations in the underlying network of genes, proteins and lipids. Modern lipidomic technologies attempt to systematically identify and quantify lipid species from biological systems. The rapid development of a variety of analytical platforms based on mass spectrometry has enabled the identification of complex lipids at great precision and depth. Insights from lipidomics studies to date suggest that the overall architecture of free fatty acid partitioning between fatty acid oxidation and complex lipid fatty acid composition is an important driver of CKD progression. Available evidence suggests that CKD progression is associated with metabolic inflexibility, reflecting a diminished capacity to utilize free fatty acids through β-oxidation, and resulting in the diversion of accumulating fatty acids to complex lipids such as triglycerides. This effect is reversed with interventions that improve kidney health, suggesting that targeting of lipid abnormalities could be beneficial in preventing CKD progression.
Collapse
Affiliation(s)
- Judy Baek
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Chenchen He
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Farsad Afshinnia
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Subramaniam Pennathur
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
91
|
Smolič T, Zorec R, Vardjan N. Pathophysiology of Lipid Droplets in Neuroglia. Antioxidants (Basel) 2021; 11:22. [PMID: 35052526 PMCID: PMC8773017 DOI: 10.3390/antiox11010022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, increasing evidence regarding the functional importance of lipid droplets (LDs), cytoplasmic storage organelles in the central nervous system (CNS), has emerged. Although not abundantly present in the CNS under normal conditions in adulthood, LDs accumulate in the CNS during development and aging, as well as in some neurologic disorders. LDs are actively involved in cellular lipid turnover and stress response. By regulating the storage of excess fatty acids, cholesterol, and ceramides in addition to their subsequent release in response to cell needs and/or environmental stressors, LDs are involved in energy production, in the synthesis of membranes and signaling molecules, and in the protection of cells against lipotoxicity and free radicals. Accumulation of LDs in the CNS appears predominantly in neuroglia (astrocytes, microglia, oligodendrocytes, ependymal cells), which provide trophic, metabolic, and immune support to neuronal networks. Here we review the most recent findings on the characteristics and functions of LDs in neuroglia, focusing on astrocytes, the key homeostasis-providing cells in the CNS. We discuss the molecular mechanisms affecting LD turnover in neuroglia under stress and how this may protect neural cell function. We also highlight the role (and potential contribution) of neuroglial LDs in aging and in neurologic disorders.
Collapse
Affiliation(s)
- Tina Smolič
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.S.); (R.Z.)
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.S.); (R.Z.)
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.S.); (R.Z.)
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
| |
Collapse
|
92
|
Role of Phytoconstituents as PPAR Agonists: Implications for Neurodegenerative Disorders. Biomedicines 2021; 9:biomedicines9121914. [PMID: 34944727 PMCID: PMC8698906 DOI: 10.3390/biomedicines9121914] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPAR-γ, PPAR-α, and PPAR-β/δ) are ligand-dependent nuclear receptors that play a critical role in the regulation of hundreds of genes through their activation. Their expression and targeted activation play an important role in the treatment of a variety of diseases, including neurodegenerative, cardiovascular, diabetes, and cancer. In recent years, several reviews have been published describing the therapeutic potential of PPAR agonists (natural or synthetic) in the disorders listed above; however, no comprehensive report defining the role of naturally derived phytoconstituents as PPAR agonists targeting neurodegenerative diseases has been published. This review will focus on the role of phytoconstituents as PPAR agonists and the relevant preclinical studies and mechanistic insights into their neuroprotective effects. Exemplary research includes flavonoids, fatty acids, cannabinoids, curcumin, genistein, capsaicin, and piperine, all of which have been shown to be PPAR agonists either directly or indirectly. Additionally, a few studies have demonstrated the use of clinical samples in in vitro investigations. The role of the fruit fly Drosophila melanogaster as a potential model for studying neurodegenerative diseases has also been highlighted.
Collapse
|
93
|
Amoroso M, Langgartner D, Lowry CA, Reber SO. Rapidly Growing Mycobacterium Species: The Long and Winding Road from Tuberculosis Vaccines to Potent Stress-Resilience Agents. Int J Mol Sci 2021; 22:ijms222312938. [PMID: 34884743 PMCID: PMC8657684 DOI: 10.3390/ijms222312938] [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] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory diseases and stressor-related psychiatric disorders, for which inflammation is a risk factor, are increasing in modern Western societies. Recent studies suggest that immunoregulatory approaches are a promising tool in reducing the risk of suffering from such disorders. Specifically, the environmental saprophyte Mycobacterium vaccae National Collection of Type Cultures (NCTC) 11659 has recently gained attention for the prevention and treatment of stress-related psychiatric disorders. However, effective use requires a sophisticated understanding of the effects of M. vaccae NCTC 11659 and related rapidly growing mycobacteria (RGMs) on microbiome–gut–immune–brain interactions. This historical narrative review is intended as a first step in exploring these mechanisms and provides an overview of preclinical and clinical studies on M. vaccae NCTC 11659 and related RGMs. The overall objective of this review article is to increase the comprehension of, and interest in, the mechanisms through which M. vaccae NCTC 11659 and related RGMs promote stress resilience, with the intention of fostering novel clinical strategies for the prevention and treatment of stressor-related disorders.
Collapse
Affiliation(s)
- Mattia Amoroso
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, University of Ulm, 89081 Ulm, Germany; (M.A.); (D.L.)
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, University of Ulm, 89081 Ulm, Germany; (M.A.); (D.L.)
| | - Christopher A. Lowry
- Department of Integrative Physiology, Center for Neuroscience and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA;
- Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), The Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, USA
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO 80045, USA
- Senior Fellow, inVIVO Planetary Health, of the Worldwide Universities Network (WUN), West New York, NJ 07093, USA
| | - Stefan O. Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, University of Ulm, 89081 Ulm, Germany; (M.A.); (D.L.)
- Correspondence:
| |
Collapse
|
94
|
Capitão AMF, Lopes-Marques M, Páscoa I, Sainath SB, Hiromori Y, Matsumaru D, Nakanishi T, Ruivo R, Santos MM, Castro LFC. An ancestral nuclear receptor couple, PPAR-RXR, is exploited by organotins. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 797:149044. [PMID: 34303232 DOI: 10.1016/j.scitotenv.2021.149044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/06/2021] [Accepted: 07/10/2021] [Indexed: 06/13/2023]
Abstract
Environmental chemicals have been reported to greatly disturb the endocrine and metabolic systems of multiple animal species. A recent example involves the exploitation of the nuclear receptor (NR) heterodimeric pair composed by PPAR/RXR (peroxisome proliferator-activated receptor/retinoid X receptor), which shows lipid perturbation in mammalian species. While gene orthologues of both of these receptors have been described outside vertebrates, no functional characterization of PPAR has been carried in protostome lineages. We provide the first functional analysis of PPAR in Patella sp. (Mollusca), using model obesogens such as tributyltin (TBT), triphenyltin (TPT), and proposed natural ligands (fatty acid molecules). To gain further insights, we used site-directed mutagenesis to PPAR and replaced the tyrosine 277 by a cysteine (the human homologous amino acid and TBT anchor residue) and an alanine. Additionally, we explored the alterations in the fatty acid profiles after an exposure to the model obesogen TBT, in vivo. Our results show that TBT and TPT behave as an antagonist of Patella sp. PPAR/RXR and that the tyrosine 277 is important, but not essential in the response to TBT. Overall, these results suggest a relation between the response of the mollusc PPAR-RXR to TBT and the lipid profile alterations reported at environmentally relevant concentrations. Our findings highlight the importance of comparative analysis between protostome and deuterostome lineages to decipher the differential impact of environmental chemicals.
Collapse
Affiliation(s)
- Ana M F Capitão
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), University of Porto (U.Porto), Department of Biology, Porto, Portugal
| | - Mónica Lopes-Marques
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Inês Páscoa
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - S B Sainath
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Department of Biotechnology, Vikrama Simhapuri University, Nellore 524 003, AP, India
| | - Youhei Hiromori
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan; Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan
| | - Daisuke Matsumaru
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan
| | - Raquel Ruivo
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal
| | - Miguel M Santos
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), University of Porto (U.Porto), Department of Biology, Porto, Portugal.
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto (U.Porto), Matosinhos, Portugal; Faculty of Sciences (FCUP), University of Porto (U.Porto), Department of Biology, Porto, Portugal.
| |
Collapse
|
95
|
Betz IR, Qaiyumi SJ, Goeritzer M, Thiele A, Brix S, Beyhoff N, Grune J, Klopfleisch R, Greulich F, Uhlenhaut NH, Kintscher U, Foryst-Ludwig A. Cardioprotective Effects of Palmitoleic Acid (C16:1n7) in a Mouse Model of Catecholamine-Induced Cardiac Damage Are Mediated by PPAR Activation. Int J Mol Sci 2021; 22:ijms222312695. [PMID: 34884498 PMCID: PMC8657733 DOI: 10.3390/ijms222312695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 01/25/2023] Open
Abstract
Palmitoleic acid (C16:1n7) has been identified as a regulator of physiological cardiac hypertrophy. In the present study, we aimed to investigate the molecular pathways involved in C16:1n7 responses in primary murine cardiomyocytes (PCM) and a mouse model of isoproterenol (ISO)-induced cardiac damage. PCMs were stimulated with C16:1n7 or a vehicle. Afterwards, RNA sequencing was performed using an Illumina HiSeq sequencer. Confirmatory analysis was performed in PCMs and HL-1 cardiomyocytes. For an in vivo study, 129 sv mice were orally treated with a vehicle or C16:1n7 for 22 days. After 5 days of pre-treatment, the mice were injected with ISO (25 mg/kg/d s. c.) for 4 consecutive days. Cardiac phenotyping was performed using echocardiography. In total, 129 genes were differentially expressed in PCMs stimulated with C16:1n7, including Angiopoietin-like factor 4 (Angptl4) and Pyruvate Dehydrogenase Kinase 4 (Pdk4). Both Angptl4 and Pdk4 are proxisome proliferator-activated receptor α/δ (PPARα/δ) target genes. Our in vivo results indicated cardioprotective and anti-fibrotic effects of C16:1n7 application in mice. This was associated with the C16:1n7-dependent regulation of the cardiac PPAR-specific signaling pathways. In conclusion, our experiments demonstrated that C16:1n7 might have protective effects on cardiac fibrosis and inflammation. Our study may help to develop future lipid-based therapies for catecholamine-induced cardiac damage.
Collapse
Affiliation(s)
- Iris Rosa Betz
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- Berlin Institute of Health, Emergency Department Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Sarah Julia Qaiyumi
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
| | - Madeleine Goeritzer
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Arne Thiele
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Sarah Brix
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Niklas Beyhoff
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Jana Grune
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany;
| | - Franziska Greulich
- German Center for Environmental Health GmbH, Institute for Diabetes and Cancer (IDC), 85764 Munich, Germany; (F.G.); (N.H.U.)
- Metabolic Programming, School of Life Sciences Weihenstephan, Technische Universitaet Muenchen (TUM), 85354 Freising, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, 13125 Berlin, Germany
| | - Nina Henriette Uhlenhaut
- German Center for Environmental Health GmbH, Institute for Diabetes and Cancer (IDC), 85764 Munich, Germany; (F.G.); (N.H.U.)
- Metabolic Programming, School of Life Sciences Weihenstephan, Technische Universitaet Muenchen (TUM), 85354 Freising, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, 13125 Berlin, Germany
| | - Ulrich Kintscher
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Anna Foryst-Ludwig
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence:
| |
Collapse
|
96
|
Dixit G, Prabhu A. The pleiotropic peroxisome proliferator activated receptors: Regulation and therapeutics. Exp Mol Pathol 2021; 124:104723. [PMID: 34822814 DOI: 10.1016/j.yexmp.2021.104723] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
The Peroxisome proliferator-activated receptors (PPARs) are key regulators of metabolic events in our body. Owing to their implication in maintenance of homeostasis, both PPAR agonists and antagonists assume therapeutic significance. Understanding the molecular mechanisms of each of the PPAR isotypes in the healthy body and during disease is crucial to exploiting their full therapeutic potential. This article is an attempt to present a rational analysis of the multifaceted therapeutic effects and underlying mechanisms of isotype-specific PPAR agonists, dual PPAR agonists, pan PPAR agonists as well as PPAR antagonists. A holistic understanding of the mechanistic dimensions of these key metabolic regulators will guide future efforts to identify novel molecules in the realm of metabolic, inflammatory and immunotherapeutic diseases.
Collapse
Affiliation(s)
- Gargi Dixit
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
97
|
Jeong DW, Lee S, Chun YS. How cancer cells remodel lipid metabolism: strategies targeting transcription factors. Lipids Health Dis 2021; 20:163. [PMID: 34775964 PMCID: PMC8590761 DOI: 10.1186/s12944-021-01593-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Reprogramming of lipid metabolism has received increasing recognition as a hallmark of cancer cells because lipid dysregulation and the alteration of related enzyme profiles are closely correlated with oncogenic signals and malignant phenotypes, such as metastasis and therapeutic resistance. In this review, we describe recent findings that support the importance of lipids, as well as the transcription factors involved in cancer lipid metabolism. With recent advances in transcription factor analysis, including computer-modeling techniques, transcription factors are emerging as central players in cancer biology. Considering the limited number and the crucial role of transcription factors associated with lipid rewiring in cancers, transcription factor targeting is a promising potential strategy for cancer therapy.
Collapse
Affiliation(s)
- Do-Won Jeong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Seulbee Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Yang-Sook Chun
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea. .,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea. .,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
98
|
Blaess M, Kaiser L, Sommerfeld O, Csuk R, Deigner HP. Drug triggered pruritus, rash, papules, and blisters - is AGEP a clash of an altered sphingolipid-metabolism and lysosomotropism of drugs accumulating in the skin? Lipids Health Dis 2021; 20:156. [PMID: 34743684 PMCID: PMC8573906 DOI: 10.1186/s12944-021-01552-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 09/09/2021] [Indexed: 11/26/2022] Open
Abstract
Rash, photosensitivity, erythema multiforme, and the acute generalized exanthematous pustulosis (AGEP) are relatively uncommon adverse reactions of drugs. To date, the etiology is not well understood and individual susceptibility still remains unknown. Amiodarone, chlorpromazine, amitriptyline, and trimipramine are classified lysosomotropic as well as photosensitizing, however, they fail to trigger rash and pruritic papules in all individuals. Lysosomotropism is a common charcteristic of various drugs, but independent of individuals. There is evidence that the individual ability to respond to external oxidative stress is crosslinked with the elongation of long-chain fatty acids to very long-chain fatty acids by ELOVLs. ELOVL6 and ELOVL7 are sensitive to ROS induced depletion of cellular NADPH and insufficient regeneration via the pentose phosphate pathway and mitochondrial fatty acid oxidation. Deficiency of NADPH in presence of lysosomotropic drugs promotes the synthesis of C16-ceramide in lysosomes and may contribute to emerging pruritic papules of AGEP. However, independently from a lysosomomotropic drug, severe depletion of ATP and NAD(P)H, e.g., by UV radiation or a potent photosensitizer can trigger likewise the collapse of the lysosomal transmembrane proton gradient resulting in lysosomal C16-ceramide synthesis and pruritic papules. This kind of papules are equally present in polymorphous light eruption (PMLE/PLE) and acne aestivalis (Mallorca acne). The suggested model of a compartmentalized ceramide metabolism provides a more sophisticated explanation of cutaneous drug adverse effects and the individual sensitivity to UV radiation. Parameters such as pKa and ClogP of the triggering drug, cutaneous fatty acid profile, and ceramide profile enables new concepts in risk assessment and scoring of AGEP as well as prophylaxis outcome.
Collapse
Affiliation(s)
- Markus Blaess
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Str. 17, D-78054, Villingen-Schwenningen, Germany
| | - Lars Kaiser
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Str. 17, D-78054, Villingen-Schwenningen, Germany
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstraße 25, D-79104, Freiburg, Germany
| | - Oliver Sommerfeld
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, D-07747, Jena, Germany
| | - René Csuk
- Organic Chemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Straße 2, D-06120, Halle (Saale), Germany
| | - Hans-Peter Deigner
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Str. 17, D-78054, Villingen-Schwenningen, Germany.
- EXIM Department, Fraunhofer Institute IZI, Schillingallee 68, D-18057, Leipzig, Rostock, Germany.
- Faculty of Science, Associated member of Tuebingen University, Auf der Morgenstelle 8, D- 72076, Tübingen, Germany.
| |
Collapse
|
99
|
Lee D, Tomita Y, Allen W, Tsubota K, Negishi K, Kurihara T. PPARα Modulation-Based Therapy in Central Nervous System Diseases. Life (Basel) 2021; 11:life11111168. [PMID: 34833044 PMCID: PMC8622664 DOI: 10.3390/life11111168] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 12/11/2022] Open
Abstract
The burden of neurodegenerative diseases in the central nervous system (CNS) is increasing globally. There are various risk factors for the development and progression of CNS diseases, such as inflammatory responses and metabolic derangements. Thus, curing CNS diseases requires the modulation of damaging signaling pathways through a multitude of mechanisms. Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors (PPARα, PPARβ/δ, and PPARγ), and they work as master sensors and modulators of cellular metabolism. In this regard, PPARs have recently been suggested as promising therapeutic targets for suppressing the development of CNS diseases and their progressions. While the therapeutic role of PPARγ modulation in CNS diseases has been well reviewed, the role of PPARα modulation in these diseases has not been comprehensively summarized. The current review focuses on the therapeutic roles of PPARα modulation in CNS diseases, including those affecting the brain, spinal cord, and eye, with recent advances. Our review will enable more comprehensive therapeutic approaches to modulate PPARα for the prevention of and protection from various CNS diseases.
Collapse
Affiliation(s)
- Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Yohei Tomita
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
- Correspondence: (Y.T.); (T.K.); Tel.: +1-617-919-2533 (Y.T.); +81-3-5636-3204 (T.K.)
| | - William Allen
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | | | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Correspondence: (Y.T.); (T.K.); Tel.: +1-617-919-2533 (Y.T.); +81-3-5636-3204 (T.K.)
| |
Collapse
|
100
|
Raeisi-Dehkordi H, Amiri M, Moghtaderi F, Zimorovat A, Rahmanian M, Mozaffari-Khosravi H, Salehi-Abargouei A. Effects of sesame, canola and sesame-canola oils on body weight and composition in adults with type 2 diabetes mellitus: a randomized, triple-blind, cross-over clinical trial. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:6083-6092. [PMID: 33899246 DOI: 10.1002/jsfa.11265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/23/2021] [Accepted: 04/25/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Recent investigations have proposed that sesame and canola oils might affect body fat distribution. The present study aimed to examine the effects of sesame, canola and sesame-canola (a blend of sesame and canola oils) oils on body weight and composition in adults with type 2 diabetes mellitus in the context of a randomized, triple-blind, three-way, cross-over clinical trial. RESULTS Eligible participants were randomized to replace their regular dietary oil with sesame oil (SO), canola oil (CO) and sesame-canola oil (SCO) (with 40% SO and 60% CO). Treatment periods lasted 9 weeks and were separated by 4-week wash-out periods. Body weight and composition were measured at the beginning, in the middle and at the end of each intervention phase. In total, 93 participants completed the study. After adjustment for confounders, within-period changes were observed following SO and CO intake for body weight (0.34 ± 0.16 kg and 0.33 ± 0.17 kg) and visceral fat (0.13 ± 0.06% and 0.13 ± 0.05%, P < 0.05), respectively. Body mass index was increased within SO intake (0.13 ± 0.05 kg m-2 , P = 0.031). All of the treatment oils resulted in reduced waist circumference and index of central obesity (P < 0.05). A significant difference in change values was observed for visceral fat between SCO (-0.14 ± 0.07%) and SO (0.12 ± 0.08%) treatment periods in females (P = 0.02). CONCLUSION Sesame and canola oils might lead to a modest favorable body fat redistribution by reducing central adiposity, particularly in females; however, the changes were of little clinical importance. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Hamidreza Raeisi-Dehkordi
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mojgan Amiri
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Moghtaderi
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Zimorovat
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Rahmanian
- Diabetes Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hassan Mozaffari-Khosravi
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Diabetes Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amin Salehi-Abargouei
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|