1
|
Jiang Y, Zhou X, He Z, Wei L, Zhu S, Zhang H, Zhang J, Chen Y, Du Y, Ding W, Wang S, Wang Z, Feng L. DHA Improves neurodevelopmental abnormalities in offspring of gestational diabetes mellitus patients via the PPAR-γ/FATP4 pathway. Biochem Pharmacol 2024; 232:116726. [PMID: 39716642 DOI: 10.1016/j.bcp.2024.116726] [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: 09/11/2024] [Revised: 12/03/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Offspring of women with gestational diabetes mellitus (GDM) face an increased risk of long-term neurodevelopmental abnormalities. This study explores the altered expression of key placental fatty acid transport proteins-FATP2, FATP4, FATP6, FABP4, and FAT/CD36-in GDM patients, and the potential of docosahexaenoic acid (DHA) to mitigate neurodevelopmental risks in offspring by enhancing their expression through activation of peroxisome proliferator-activated receptor γ (PPAR-γ). Our findings demonstrate that placental FATP4 expression is reduced in GDM patients. In HTR8/SVneo cells, PPAR-γ activation upregulated the expression of FATP4, FAT/CD36, and FABP4, while PPAR-γ inhibition only reduced FAT/CD36 expression. DHA treatment led to increased expression of FATP4, FATP/CD36 and FABP4, which was partially reversed by PPAR-γ inhibition. Consistent results were observed in an insulin-resistant cell model. Supplementing GDM mice with exogenous DHA restored placental FATP4 expression and improved offspring social behavior and cognitive function. These results suggest that DHA supplementation during pregnancy could reduce the adverse effects of GDM on placental FATP4 expression and support better neurodevelopmental outcomes in offspring by promoting essential fatty acid transport through the PPAR-γ/FATP4 pathway. This study highlights the therapeutic potential of DHA in improving fetal outcomes in GDM pregnancies.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xuan Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhenzhen He
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lijie Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shenglan Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Huiting Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jingyi Zhang
- Department of Obstetrics and Gynecology Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuting Chen
- Department of Obstetrics and Gynecology Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuanyuan Du
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wencheng Ding
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shaoshuai Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Zizhuo Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Seessle J, Liebisch G, Staffer S, Tuma-Kellner S, Merle U, Herrmann T, Chamulitrat W. Enterocyte-specific FATP4 deficiency elevates blood lipids via a shift from polar to neutral lipids in distal intestine. Am J Physiol Gastrointest Liver Physiol 2024; 327:G202-G216. [PMID: 38915276 DOI: 10.1152/ajpgi.00109.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/13/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
Fatty acid transport protein (FATP)4 was thought to mediate intestinal lipid absorption, which was disputed by a study using keratinocyte-Fatp4-rescued Fatp4-/- mice. These knockouts when fed with a Western diet showed elevated intestinal triglyceride (TG) and fatty acid levels. To investigate a possible role of FATP4 on intestinal lipid processing, ent-Fatp4 (KO) mice were generated by Villin-Cre-specific inactivation of the Fatp4 gene. We aimed to measure circulating and intestinal lipids in control and KO mice after acute or chronic fat intake or during aging. Remarkably, ent-Fatp4 mice displayed an approximately 30% decrease in ileal behenic, lignoceric, and nervonic acids, ceramides containing these FA, as well as, ileal sphingomyelin, phosphatidylcholine, and phosphatidylinositol levels. Such decreases were concomitant with an increase in jejunal cholesterol ester. After a 2-wk recovery from high lipid overload by tyloxapol and oral-lipid treatment, ent-Fatp4 mice showed an increase in plasma TG and chylomicrons. Upon overnight fasting followed by an oral fat meal, ent-Fatp4 mice showed an increase in plasma TG-rich lipoproteins and the particle number of chylomicrons and very low-density lipoproteins. During aging or after feeding with a high-fat high-cholesterol (HFHC) diet, ent-Fatp4 mice showed an increase in plasma TG, fatty acids, glycerol, and lipoproteins as well as intestinal lipids. HFHC-fed KO mice displayed an increase in body weight, the number of lipid droplets with larger sizes in the ileum, concomitant with a decrease in ileal ceramides and phosphatidylcholine. Thus, enterocyte FATP4 deficiency led to a metabolic shift from polar to neutral lipids in distal intestine rendering an increase in plasma lipids and lipoproteins.NEW & NOTEWORTHY Enterocyte-specific Fatp4 deficiency in mice increased intestinal lipid absorption with elevation of blood lipids during fasting and aging, as well as after an acute oral fat-loading or chronic HFHC feeding. Lipidomics revealed that knockout mice displayed a shift from very long-chain to long-chain fatty acids, and from polar to neutral lipids, predominantly in the ileum. Thus, FATP4 may have a physiological function in the control of blood lipids via metabolic shifts in distal intestine.
Collapse
Affiliation(s)
- Jessica Seessle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Simone Staffer
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Herrmann
- Department of Medical Clinic 1, Westkuesten Hospital, Heide, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
3
|
Pilon M, Ruiz M. PAQR proteins and the evolution of a superpower: Eating all kinds of fats: Animals rely on evolutionarily conserved membrane homeostasis proteins to compensate for dietary variation. Bioessays 2023; 45:e2300079. [PMID: 37345585 DOI: 10.1002/bies.202300079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/23/2023]
Abstract
Recently published work showed that members of the PAQR protein family are activated by cell membrane rigidity and contribute to our ability to eat a wide variety of diets. Cell membranes are primarily composed of phospholipids containing dietarily obtained fatty acids, which poses a challenge to membrane properties because diets can vary greatly in their fatty acid composition and could impart opposite properties to the cellular membranes. In particular, saturated fatty acids (SFAs) can pack tightly and form rigid membranes (like butter at room temperature) while unsaturated fatty acids (UFAs) form more fluid membranes (like vegetable oils). Proteins of the PAQR protein family, characterized by the presence of seven transmembrane domains and a cytosolic N-terminus, contribute to membrane homeostasis in bacteria, yeasts, and animals. These proteins respond to membrane rigidity by stimulating fatty acid desaturation and incorporation of UFAs into phospholipids and explain the ability of animals to thrive on diets with widely varied fat composition. Also see the video abstract here: https://youtu.be/6ckcvaDdbQg.
Collapse
Affiliation(s)
- Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Mario Ruiz
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
4
|
Göcebe D, Jansakun C, Zhang Y, Staffer S, Tuma-Kellner S, Altamura S, Muckenthaler MU, Merle U, Herrmann T, Chamulitrat W. Myeloid-specific fatty acid transport protein 4 deficiency induces a sex-dimorphic susceptibility for nonalcoholic steatohepatitis in mice fed a high-fat, high-cholesterol diet. Am J Physiol Gastrointest Liver Physiol 2023; 324:G389-G403. [PMID: 36881564 PMCID: PMC10085558 DOI: 10.1152/ajpgi.00181.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 03/08/2023]
Abstract
Newborns with FATP4 mutations exhibit ichthyosis prematurity syndrome (IPS), and adult patients show skin hyperkeratosis, allergies, and eosinophilia. We have previously shown that the polarization of macrophages is altered by FATP4 deficiency; however, the role of myeloid FATP4 in the pathogenesis of nonalcoholic steatohepatitis (NASH) is not known. We herein phenotyped myeloid-specific Fatp4-deficient (Fatp4M-/-) mice under chow and high-fat, high-cholesterol (HFHC) diet. Bone-marrow-derived macrophages (BMDMs) from Fatp4M-/- mice showed significant reduction in cellular sphingolipids in males and females, and additionally phospholipids in females. BMDMs and Kupffer cells from Fatp4M-/- mice exhibited increased LPS-dependent activation of proinflammatory cytokines and transcription factors PPARγ, CEBPα, and p-FoxO1. Correspondingly, these mutants under chow diet displayed thrombocytopenia, splenomegaly, and elevated liver enzymes. After HFHC feeding, Fatp4M-/- mice showed increased MCP-1 expression in livers and subcutaneous fat. Plasma MCP-1, IL4, and IL13 levels were elevated in male and female mutants, and female mutants additionally showed elevation of IL5 and IL6. After HFHC feeding, male mutants showed an increase in hepatic steatosis and inflammation, whereas female mutants showed a greater severity in hepatic fibrosis associated with immune cell infiltration. Thus, myeloid-FATP4 deficiency led to steatotic and inflammatory NASH in males and females, respectively. Our work offers some implications for patients with FATP4 mutations and also highlights considerations in the design of sex-targeted therapies for NASH treatment.NEW & NOTEWORTHY FATP4 deficiency in BMDMs and Kupffer cells led to increased proinflammatory response. Fatp4M-/- mice displayed thrombocytopenia, splenomegaly, and elevated liver enzymes. In response to HFHC feeding, male mutants were prone to hepatic steatosis, whereas female mutants showed exaggerated fibrosis. Our study provides insights into a sex-dimorphic susceptibility to NASH by myeloid-FATP4 deficiency.
Collapse
Affiliation(s)
- Deniz Göcebe
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Chutima Jansakun
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
- School of Allied Health Sciences, Walailak University, Nakhonsrithammarat, Thailand
| | - Yuling Zhang
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Simone Staffer
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Sandro Altamura
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL), German Centre for Cardiovascular Research, Partner Site, University of Heidelberg, Heidelberg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Walee Chamulitrat
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
5
|
Wu Z, Hu G, Zhang Y, Ao Z. IGF2 May Enhance Placental Fatty Acid Metabolism by Regulating Expression of Fatty Acid Carriers in the Growth of Fetus and Placenta during Late Pregnancy in Pigs. Genes (Basel) 2023; 14:genes14040872. [PMID: 37107630 PMCID: PMC10137774 DOI: 10.3390/genes14040872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Fatty acids (FAs) are essential substances for the growth and development of the fetus and placenta. The growing fetus and placenta must obtain adequate FAs received from the maternal circulation and facilitated by various placental FA carriers, including FA transport proteins (FATPs), FA translocase (FAT/CD36), and cytoplasmic FA binding proteins (FABPs). Placental nutrition transport was regulated by imprinted genes H19 and insulin-like growth factor 2 (IGF2). Nevertheless, the relationship between the expression patterns of H19/IGF2 and placental fatty acid metabolism throughout pig pregnancy remains poorly studied and unclear. We investigated the placental fatty acid profile, expression patterns of FA carriers, and H19/IGF2 in the placentae on Days 40 (D40), 65 (D65), and 95 (D95) of pregnancy. The results showed that the width of the placental folds and the number of trophoblast cells of D65 placentae were significantly increased than those of D40 placentae. Several important long-chain FAs (LCFAs), including oleic acid, linoleic acid, arachidonatic acid, eicosapentaenoic acid, and docosatetraenoic acid, in the pig placenta showed dramatically increased levels throughout pregnancy. The pig placenta possessed higher expression levels of CD36, FATP4, and FABP5 compared with other FA carriers, and their expression levels had significantly upregulated 2.8-, 5.6-, and 12.0-fold from D40 to D95, respectively. The transcription level of IGF2 was dramatically upregulated and there were corresponding lower DNA methylation levels in the IGF2 DMR2 in D95 placentae relative to D65 placentae. Moreover, in vitro experimentation revealed that the overexpression of IGF2 resulted in a significant increase in fatty acid uptake and expression levels of CD36, FATP4, and FABP5 in PTr2 cells. In conclusion, our results indicate that CD36, FATP4, and FABP5 may be important regulators that enhance the transport of LCFAs in the pig placenta and that IGF2 may be involved in FA metabolism by affecting the FA carriers expression to support the growth of the fetus and placenta during late pregnancy in pigs.
Collapse
Affiliation(s)
- Zhimin Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
- Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Guangling Hu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
- Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yiyu Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
- Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zheng Ao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
- Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
6
|
Li X, Liu Q, Pan Y, Chen S, Zhao Y, Hu Y. New insights into the role of dietary triglyceride absorption in obesity and metabolic diseases. Front Pharmacol 2023; 14:1097835. [PMID: 36817150 PMCID: PMC9932209 DOI: 10.3389/fphar.2023.1097835] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
The incidence of obesity and associated metabolic diseases is increasing globally, adversely affecting human health. Dietary fats, especially triglycerides, are an important source of energy for the body, and the intestine absorbs lipids through a series of orderly and complex steps. A long-term high-fat diet leads to intestinal dysfunction, inducing obesity and metabolic disorders. Therefore, regulating dietary triglycerides absorption is a promising therapeutic strategy. In this review, we will discuss diverse aspects of the dietary triglycerides hydrolysis, fatty acid uptake, triglycerides resynthesis, chylomicron assembly, trafficking, and secretion processes in intestinal epithelial cells, as well as potential targets in this process that may influence dietary fat-induced obesity and metabolic diseases. We also mention the possible shortcomings and deficiencies in modulating dietary lipid absorption targets to provide a better understanding of their administrability as drugs in obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Xiaojing Li
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiaohong Liu
- Institute of Clinical Pharmacology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing Pan
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Si Chen
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhao
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Yu Zhao, ; Yiyang Hu,
| | - Yiyang Hu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,Institute of Clinical Pharmacology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Yu Zhao, ; Yiyang Hu,
| |
Collapse
|
7
|
Shrestha N, McCarron A, Rout-Pitt N, Donnelley M, Parsons DW, Hryciw DH. Essential Fatty Acid Deficiency in Cystic Fibrosis Disease Progression: Role of Genotype and Sex. Nutrients 2022; 14:nu14214666. [PMID: 36364928 PMCID: PMC9657825 DOI: 10.3390/nu14214666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Adequate intake of nutrients such as essential fatty acids (EFA) are critical in cystic fibrosis (CF). The clinical course of deterioration of lung function in people with CF has been shown to relate to nutrition. Independent of the higher energy consumption and malabsorption due to pancreatic insufficiency, EFA deficiency is closely associated with the risk of pulmonary infection, the most significant pathology in CF. This review will focus on the EFA deficiency identified in people with CF, as well as the limited progress made in deciphering the exact metabolic pathways that are dysfunctional in CF. Specifically, people with CF are deficient in linoleic acid, an omega 6 fatty acid, and the ratio of arachidonic acid (omega 6 metabolite) and docosahexaenoic acid (omega 3 metabolite) is increased. Analysis of the molecular pathways in bronchial cells has identified changes in the enzymes that metabolise EFA. However, fatty acid metabolism primarily occurs in the liver, with EFA metabolism in CF liver not yet investigated, indicating that further research is required. Despite limited understanding in this area, it is well known that adequate EFA concentrations are critical to normal membrane structure and function, and thus are important to consider in disease processes. Novel insights into the relationship between CF genotype and EFA phenotype will be discussed, in addition to sex differences in EFA concentrations in people with CF. Collectively, investigating the specific effects of genotype and sex on fatty acid metabolism may provide support for the management of people with CF via personalised genotype- and sex-specific nutritional therapies.
Collapse
Affiliation(s)
- Nirajan Shrestha
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD 4222, Australia
| | - Alexandra McCarron
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5001, Australia
- Respiratory and Sleep Medicine, Women’s and Children’s Hospital, 72 King William Road, North Adelaide, SA 5006, Australia
| | - Nathan Rout-Pitt
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5001, Australia
- Respiratory and Sleep Medicine, Women’s and Children’s Hospital, 72 King William Road, North Adelaide, SA 5006, Australia
| | - Martin Donnelley
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5001, Australia
- Respiratory and Sleep Medicine, Women’s and Children’s Hospital, 72 King William Road, North Adelaide, SA 5006, Australia
| | - David W. Parsons
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5001, Australia
- Respiratory and Sleep Medicine, Women’s and Children’s Hospital, 72 King William Road, North Adelaide, SA 5006, Australia
| | - Deanne H. Hryciw
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3000, Australia
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
- Correspondence: ; Tel.: +61-7-3735-3601
| |
Collapse
|
8
|
Wu Z, Xu C, Zheng T, Li Q, Yang S, Shao J, Guan W, Zhang S. A critical role of AMP-activated protein kinase in regulating intestinal nutrient absorption, barrier function, and intestinal diseases. J Cell Physiol 2022; 237:3705-3716. [PMID: 35892164 DOI: 10.1002/jcp.30841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023]
Abstract
As one of the most important organs in animals, the intestine is responsible for nutrient absorption and acts as a barrier between the body and the environment. Intestinal physiology and function require the participation of energy. 5'-adenosine monophosphate-activated protein kinase (AMPK), a classical and highly expressed energy regulator in intestinal cells, regulates the process of nutrient absorption and barrier function and is also involved in the therapy of intestinal diseases. Studies have yielded findings that AMPK regulates the absorption of glucose, amino acids, and fatty acids in the intestine primarily by regulating transportation systems, as we detailed here. Moreover, AMPK is involved in the regulation of the intestinal mechanical barrier and immune barrier through manipulating the expression of tight junctions, antimicrobial peptides, and secretory immunoglobulins. In addition, AMPK also participates in the regulation of intestinal diseases, which indicates that AMPK is a promising therapeutic target for intestinal diseases and cancer. In this review, we summarized the current understanding regarding how AMPK regulates intestinal nutrient absorption, barrier function, and intestinal diseases.
Collapse
Affiliation(s)
- Zhihui Wu
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Chengfei Xu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| |
Collapse
|
9
|
Li H, Herrmann T, Seeßle J, Liebisch G, Merle U, Stremmel W, Chamulitrat W. Role of fatty acid transport protein 4 in metabolic tissues: insights into obesity and fatty liver disease. Biosci Rep 2022; 42:BSR20211854. [PMID: 35583196 PMCID: PMC9160530 DOI: 10.1042/bsr20211854] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/28/2022] Open
Abstract
Fatty acid (FA) metabolism is a series of processes that provide structural substances, signalling molecules and energy. Ample evidence has shown that FA uptake is mediated by plasma membrane transporters including FA transport proteins (FATPs), caveolin-1, fatty-acid translocase (FAT)/CD36, and fatty-acid binding proteins. Unlike other FA transporters, the functions of FATPs have been controversial because they contain both motifs of FA transport and fatty acyl-CoA synthetase (ACS). The widely distributed FATP4 is not a direct FA transporter but plays a predominant function as an ACS. FATP4 deficiency causes ichthyosis premature syndrome in mice and humans associated with suppression of polar lipids but an increase in neutral lipids including triglycerides (TGs). Such a shift has been extensively characterized in enterocyte-, hepatocyte-, and adipocyte-specific Fatp4-deficient mice. The mutants under obese and non-obese fatty livers induced by different diets persistently show an increase in blood non-esterified free fatty acids and glycerol indicating the lipolysis of TGs. This review also focuses on FATP4 role on regulatory networks and factors that modulate FATP4 expression in metabolic tissues including intestine, liver, muscle, and adipose tissues. Metabolic disorders especially regarding blood lipids by FATP4 deficiency in different cell types are herein discussed. Our results may be applicable to not only patients with FATP4 mutations but also represent a model of dysregulated lipid homeostasis, thus providing mechanistic insights into obesity and development of fatty liver disease.
Collapse
Affiliation(s)
- Huili Li
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Thomas Herrmann
- Westkuesten Hospital, Esmarchstraße 50, 25746 Heide, Germany
| | - Jessica Seeßle
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Wolfgang Stremmel
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Zhang L, Li X, Liu X, Wang X, Li X, Cheng X, Yan S, Zhu Y, Li R, Wen L, Wang J. Purified diet versus whole food diet and the inconsistent results in studies using animal models. Food Funct 2022; 13:4286-4301. [PMID: 35297926 DOI: 10.1039/d1fo04311k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In animal models, purified diets (PDs) and whole food diets (WFDs) are used for different purposes. In similar studies, different dietary patterns may lead to inconsistent results. The aim of this study was to evaluate and compare the effects of WFDs and PDs on changes in the metabolism of mice. We found that different dietary patterns produced different results in lipid metabolism experiments. Compared with those of the PD-fed mice, the WFD-fed mice had higher body weights and serum glucose, serum lipid, and liver lipid levels (p < 0.01), as well as low glucose tolerance (p < 0.01) and insulin sensitivity (p < 0.05). The body weight and fasting blood glucose increased by 20% in the WFD-fed mice, and the white adipose tissue weight increased by ∼50%. The WFD-fed mice also had a comparatively higher abundance of Lactobacillus, Turicibacter, Bifidobacterium, Desulfovibrio, and Candidatus saccharimonas (p < 0.01), which were positively correlated with lipid accumulation. Dietary patterns should be chosen cautiously in studies that use rodents as models. Inappropriate selection of animal dietary patterns may lead to experimental systematic errors and paradoxical results.
Collapse
Affiliation(s)
- Linyu Zhang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China.
| | - Xin Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China.
| | - Xiangyan Liu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China.
| | - Xianglin Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China.
| | - Xiaowen Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China.
| | - Xianyu Cheng
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China.
| | - Sisi Yan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China.
| | - Yuanyuan Zhu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China.
| | - Rongfang Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China. .,Hunan Collaborative Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Lixin Wen
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China. .,Hunan Collaborative Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Ji Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha City, 410128, China. .,Changsha Lvye Biotechnology Co., Ltd, Changsha 410100, China
| |
Collapse
|
11
|
Kappen C, Kruger C, Jones S, Salbaum JM. Nutrient Transporter Gene Expression in the Early Conceptus-Implications From Two Mouse Models of Diabetic Pregnancy. Front Cell Dev Biol 2022; 10:777844. [PMID: 35478964 PMCID: PMC9035823 DOI: 10.3389/fcell.2022.777844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/28/2022] [Indexed: 11/29/2022] Open
Abstract
Maternal diabetes in early pregnancy increases the risk for birth defects in the offspring, particularly heart, and neural tube defects. While elevated glucose levels are characteristic for diabetic pregnancies, these are also accompanied by hyperlipidemia, indicating altered nutrient availability. We therefore investigated whether changes in the expression of nutrient transporters at the conception site or in the early post-implantation embryo could account for increased birth defect incidence at later developmental stages. Focusing on glucose and fatty acid transporters, we measured their expression by RT-PCR in the spontaneously diabetic non-obese mouse strain NOD, and in pregnant FVB/N mouse strain dams with Streptozotocin-induced diabetes. Sites of expression in the deciduum, extra-embryonic, and embryonic tissues were determined by RNAscope in situ hybridization. While maternal diabetes had no apparent effects on levels or cellular profiles of expression, we detected striking cell-type specificity of particular nutrient transporters. For examples, Slc2a2/Glut2 expression was restricted to the endodermal cells of the visceral yolk sac, while Slc2a1/Glut1 expression was limited to the mesodermal compartment; Slc27a4/Fatp4 and Slc27a3/Fatp3 also exhibited reciprocally exclusive expression in the endodermal and mesodermal compartments of the yolk sac, respectively. These findings not only highlight the significance of nutrient transporters in the intrauterine environment, but also raise important implications for the etiology of birth defects in diabetic pregnancies, and for strategies aimed at reducing birth defects risk by nutrient supplementation.
Collapse
Affiliation(s)
- Claudia Kappen
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Claudia Kruger
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Sydney Jones
- Regulation of Gene Expression, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - J. Michael Salbaum
- Regulation of Gene Expression, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| |
Collapse
|
12
|
Sanches APV, de Oliveira JL, Ferreira MS, Lima BDS, Miyamoto JÉ, Simino LADP, Torsoni MA, Torsoni AS, Milanski M, Ignácio-Souza LM. Obesity phenotype induced by high-fat diet leads to maternal-fetal constraint, placental inefficiency, and fetal growth restriction in mice. J Nutr Biochem 2022; 104:108977. [PMID: 35248701 DOI: 10.1016/j.jnutbio.2022.108977] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/20/2021] [Accepted: 02/09/2022] [Indexed: 11/28/2022]
Abstract
The aim of this study was to investigate certain parameters regarding the maternal-fetal outcomes in a diet-induced obesity model. Obese, glucose-intolerant females who were exposed to a high-fat diet prior to pregnancy had lower placental efficiency and lower birth weight pups compared to the controls. Simple linear regression analyses showed that maternal obesity disrupts the proportionality between maternal and fetal outcomes during pregnancy. Maternal obesity is correlated with fetal outcomes, perhaps because of problems with hormonal signaling and exacerbation of inflammation in the maternal metabolic environment. The maternal obese phenotype altered the thickness of the placental layer, the transport of fatty acids, and the expression of growth factors. For example, lower expression of epidermal growth factor receptor (EGFR) mRNA in the obesity-prone group may have contributed to the rupture of the placental layers, leading to adverse fetal outcomes. Furthermore, maintenance of maternal glucose homeostasis and overexpression of placental growth factor (PGF) in the obesity-resistant group likely protected the placenta and fetuses from morphological and functional damage.
Collapse
Affiliation(s)
- Ana Paula Varela Sanches
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Josilene Lopes de Oliveira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Maíra Schuchter Ferreira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Bruna de Souza Lima
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Josiane Érica Miyamoto
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Laís Angélica de Paula Simino
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil
| | - Márcio Alberto Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Letícia Martins Ignácio-Souza
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, Limeira, São Paulo, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.
| |
Collapse
|
13
|
Hryciw DH, Jackson CA, Shrestha N, Parsons D, Donnelley M, McAinch AJ. Role for animal models in understanding essential fatty acid deficiency in cystic fibrosis. Cell Mol Life Sci 2021; 78:7991-7999. [PMID: 34741185 PMCID: PMC11072998 DOI: 10.1007/s00018-021-04014-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/11/2021] [Accepted: 10/25/2021] [Indexed: 10/19/2022]
Abstract
Essential fatty acid deficiency has been observed in most patients with Cystic Fibrosis (CF); however, pancreatic supplementation does not restore the deficiency, suggesting a different pathology independent of the pancreas. At this time, the underlying pathological mechanisms are largely unknown. Essential fatty acids are obtained from the diet and processed by organs including the liver and intestine, two organs significantly impacted by mutations in the cystic fibrosis transmembrane conductance regulator gene (Cftr). There are several CF animal models in a variety of species that have been developed to investigate molecular mechanisms associated with the CF phenotype. Specifically, global and systemic mutations in Cftr which mimic genotypic changes identified in CF patients have been generated in mice, rats, sheep, pigs and ferrets. These mutations produce CFTR proteins with a gating defect, trafficking defect, or an absent or inactive CFTR channel. Essential fatty acids are critical to CFTR function, with a bidirectional relationship between CFTR and essential fatty acids proposed. Currently, there are limited analyses on the essential fatty acid status in most of these animal models. Of interest, in the mouse model, essential fatty acid status is dependent on the genotype and resultant phenotype of the mouse. Future investigations should identify an optimal animal model that has most of the phenotypic changes associated with CF including the essential fatty acid deficiencies, which can be used in the development of therapeutics.
Collapse
Affiliation(s)
- Deanne H Hryciw
- School of Environment and Science, Griffith University, Nathan, QLD, Australia.
- Centre for Planetary Health and Food Security, Griffith University, Nathan, QLD, Australia.
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia.
| | - Courtney A Jackson
- School of Environment and Science, Griffith University, Nathan, QLD, Australia
| | - Nirajan Shrestha
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | - David Parsons
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, SA, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Martin Donnelley
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, SA, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Andrew J McAinch
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, VIC, Australia
| |
Collapse
|
14
|
Lee JG, Kim G, Park SG, Yon JM, Yeom J, Song HE, Cheong SA, Lim JS, Sung YH, Kim K, Yoo HJ, Hong EJ, Nam KH, Seong JK, Kim CJ, Nam SY, Baek IJ. Lipid signatures reflect the function of the murine primary placentation. Biol Reprod 2021; 106:583-596. [PMID: 34850819 DOI: 10.1093/biolre/ioab219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
The placenta regulates maternal-fetal communication, and its defect leads to significant pregnancy complications. The maternal and embryonic circulations are primitively connected in early placentation, but the function of the placenta during this developmentally essential period is relatively unknown. We thus performed a comparative proteomic analysis of the placenta before and after primary placentation and found that the metabolism and transport of lipids were characteristically activated in this period. The placental fatty acid (FA) carriers in specific placental compartments were upregulated according to gestational age, and metabolomic analysis also showed that the placental transport of FAs increased in a time-dependent manner. Further analysis of two mutant mice models with embryonic lethality revealed that lipid-related signatures could reflect the functional state of the placenta. Our findings highlight the importance of the nutrient transport function of the primary placenta in the early gestational period and the role of lipids in embryonic development.
Collapse
Affiliation(s)
- Jong Geol Lee
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Korea Mouse Phenotyping Center, Seoul, Republic of Korea
| | - Globinna Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seul Gi Park
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.,Biomedical Mouse Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongwon-Gun, Republic of Korea
| | - Jung-Min Yon
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeonghun Yeom
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ha Eun Song
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-A Cheong
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Korea Mouse Phenotyping Center, Seoul, Republic of Korea
| | - Joon Seo Lim
- Clinical Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Hoon Sung
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyun Ju Yoo
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Korea Mouse Phenotyping Center, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ki-Hoan Nam
- Korea Mouse Phenotyping Center, Seoul, Republic of Korea.,Biomedical Mouse Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongwon-Gun, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul, Republic of Korea.,College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Chong Jai Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Yoon Nam
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - In-Jeoung Baek
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Korea Mouse Phenotyping Center, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Degalez F, Jehl F, Muret K, Bernard M, Lecerf F, Lagoutte L, Désert C, Pitel F, Klopp C, Lagarrigue S. Watch Out for a Second SNP: Focus on Multi-Nucleotide Variants in Coding Regions and Rescued Stop-Gained. Front Genet 2021; 12:659287. [PMID: 34306009 PMCID: PMC8293744 DOI: 10.3389/fgene.2021.659287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/27/2021] [Indexed: 12/30/2022] Open
Abstract
Most single-nucleotide polymorphisms (SNPs) are located in non-coding regions, but the fraction usually studied is harbored in protein-coding regions because potential impacts on proteins are relatively easy to predict by popular tools such as the Variant Effect Predictor. These tools annotate variants independently without considering the potential effect of grouped or haplotypic variations, often called "multi-nucleotide variants" (MNVs). Here, we used a large RNA-seq dataset to survey MNVs, comprising 382 chicken samples originating from 11 populations analyzed in the companion paper in which 9.5M SNPs- including 3.3M SNPs with reliable genotypes-were detected. We focused our study on in-codon MNVs and evaluate their potential mis-annotation. Using GATK HaplotypeCaller read-based phasing results, we identified 2,965 MNVs observed in at least five individuals located in 1,792 genes. We found 41.1% of them showing a novel impact when compared to the effect of their constituent SNPs analyzed separately. The biggest impact variation flux concerns the originally annotated stop-gained consequences, for which around 95% were rescued; this flux is followed by the missense consequences for which 37% were reannotated with a different amino acid. We then present in more depth the rescued stop-gained MNVs and give an illustration in the SLC27A4 gene. As previously shown in human datasets, our results in chicken demonstrate the value of haplotype-aware variant annotation, and the interest to consider MNVs in the coding region, particularly when searching for severe functional consequence such as stop-gained variants.
Collapse
Affiliation(s)
- Fabien Degalez
- INRAE, INSTITUT AGRO, PEGASE UMR 1348, Saint-Gilles, France
| | - Frédéric Jehl
- INRAE, INSTITUT AGRO, PEGASE UMR 1348, Saint-Gilles, France
| | - Kévin Muret
- INRAE, INSTITUT AGRO, PEGASE UMR 1348, Saint-Gilles, France
| | - Maria Bernard
- INRAE, SIGENAE, Genotoul Bioinfo MIAT, Castanet-Tolosan, France.,INRAE, AgroParisTech, Université Paris-Saclay, GABI UMR 1313, Jouy-en-Josas, France
| | | | | | - Colette Désert
- INRAE, INSTITUT AGRO, PEGASE UMR 1348, Saint-Gilles, France
| | - Frédérique Pitel
- INRAE, INPT, ENVT, Université de Toulouse, GenPhySE UMR 1388, Castanet-Tolosan, France
| | | | | |
Collapse
|
16
|
Levy E, Beaulieu JF, Spahis S. From Congenital Disorders of Fat Malabsorption to Understanding Intra-Enterocyte Mechanisms Behind Chylomicron Assembly and Secretion. Front Physiol 2021; 12:629222. [PMID: 33584351 PMCID: PMC7873531 DOI: 10.3389/fphys.2021.629222] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/06/2021] [Indexed: 11/13/2022] Open
Abstract
During the last two decades, a large body of information on the events responsible for intestinal fat digestion and absorption has been accumulated. In particular, many groups have extensively focused on the absorptive phase in order to highlight the critical "players" and the main mechanisms orchestrating the assembly and secretion of chylomicrons (CM) as essential vehicles of alimentary lipids. The major aim of this article is to review understanding derived from basic science and clinical conditions associated with impaired packaging and export of CM. We have particularly insisted on inborn metabolic pathways in humans as well as on genetically modified animal models (recapitulating pathological features). The ultimate goal of this approach is that "experiments of nature" and in vivo model strategy collectively allow gaining novel mechanistic insight and filling the gap between the underlying genetic defect and the apparent clinical phenotype. Thus, uncovering the cause of disease contributes not only to understanding normal physiologic pathway, but also to capturing disorder onset, progression, treatment and prognosis.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU Ste-Justine, Université de Montréal, Montreal, QC, Canada
- Department of Nutrition, Université de Montréal, Montreal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
| | - Jean François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Schohraya Spahis
- Research Centre, CHU Ste-Justine, Université de Montréal, Montreal, QC, Canada
- Department of Nutrition, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
17
|
Benninghoff T, Espelage L, Eickelschulte S, Zeinert I, Sinowenka I, Müller F, Schöndeling C, Batchelor H, Cames S, Zhou Z, Kotzka J, Chadt A, Al-Hasani H. The RabGAPs TBC1D1 and TBC1D4 Control Uptake of Long-Chain Fatty Acids Into Skeletal Muscle via Fatty Acid Transporter SLC27A4/FATP4. Diabetes 2020; 69:2281-2293. [PMID: 32868338 DOI: 10.2337/db20-0180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 08/24/2020] [Indexed: 11/13/2022]
Abstract
The two closely related RabGTPase-activating proteins (RabGAPs) TBC1D1 and TBC1D4 play a crucial role in the regulation of GLUT4 translocation in response to insulin and contraction in skeletal muscle. In mice, deficiency in one or both RabGAPs leads to reduced insulin- and contraction-stimulated glucose uptake and to elevated fatty acid (FA) uptake and oxidation in both glycolytic and oxidative muscle fibers without altering mitochondrial copy number and the abundance of proteins for oxidative phosphorylation. Here we present evidence for a novel mechanism of skeletal muscle lipid utilization involving the two RabGAPs and the FA transporter SLC27A4/FATP4. Both RabGAPs control the uptake of saturated and unsaturated long-chain FAs (LCFAs) into skeletal muscle and knockdown (Kd) of a subset of RabGAP substrates, Rab8, Rab10, or Rab14, decreased LCFA uptake into these cells. In skeletal muscle from Tbc1d1 and Tbc1d4 knockout animals, SLC27A4/FATP4 abundance was increased and depletion of SLC27A4/FATP4 but not FAT/CD36 completely abrogated the enhanced FA oxidation in RabGAP-deficient skeletal muscle and cultivated C2C12 myotubes. Collectively, our data demonstrate that RabGAP-mediated control of skeletal muscle lipid metabolism converges with glucose metabolism at the level of downstream RabGTPases and involves regulated transport of LCFAs via SLC27A4/FATP4.
Collapse
Affiliation(s)
- Tim Benninghoff
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Lena Espelage
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Samaneh Eickelschulte
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Isabel Zeinert
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Isabelle Sinowenka
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Frank Müller
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Christina Schöndeling
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Hannah Batchelor
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Sandra Cames
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Zhou Zhou
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Jörg Kotzka
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| |
Collapse
|
18
|
Fernandez RF, Ellis JM. Acyl-CoA synthetases as regulators of brain phospholipid acyl-chain diversity. Prostaglandins Leukot Essent Fatty Acids 2020; 161:102175. [PMID: 33031993 PMCID: PMC8693597 DOI: 10.1016/j.plefa.2020.102175] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/22/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022]
Abstract
Each individual cell-type is defined by its distinct morphology, phenotype, molecular and lipidomic profile. The importance of maintaining cell-specific lipidomic profiles is exemplified by the numerous diseases, disorders, and dysfunctional outcomes that occur as a direct result of altered lipidome. Therefore, the mechanisms regulating cellular lipidome diversity play a role in maintaining essential biological functions. The brain is an organ particularly rich in phospholipids, the main constituents of cellular membranes. The phospholipid acyl-chain profile of membranes in the brain is rather diverse due in part to the high degree of cellular heterogeneity. These membranes and the acyl-chain composition of their phospholipids are highly regulated, but the mechanisms that confer this tight regulation are incompletely understood. A family of enzymes called acyl-CoA synthetases (ACSs) stands at a pinnacle step allowing influence over cellular acyl-chain selection and subsequent metabolic flux. ACSs perform the initial reaction for cellular fatty acid metabolism by ligating a Coenzyme A to a fatty acid which both traps a fatty acid within a cell and activates it for metabolism. The ACS family of enzymes is large and diverse consisting of 25-26 family members that are nonredundant, each with unique distribution across and within cell types, and differential fatty acid substrate preferences. Thus, ACSs confer a critical intracellular fatty acid selecting step in a cell-type dependent manner providing acyl-CoA moieties that serve as essential precursors for phospholipid synthesis and remodeling, and therefore serve as a key regulator of cellular membrane acyl-chain compositional diversity. Here we will discuss how the contribution of individual ACSs towards brain lipid metabolism has only just begun to be elucidated and discuss the possibilities for how ACSs may differentially regulate brain lipidomic diversity.
Collapse
Affiliation(s)
- Regina F Fernandez
- Department of Physiology and East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, NC, United States
| | - Jessica M Ellis
- Department of Physiology and East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, NC, United States.
| |
Collapse
|
19
|
Regulation of intestinal lipid metabolism: current concepts and relevance to disease. Nat Rev Gastroenterol Hepatol 2020; 17:169-183. [PMID: 32015520 DOI: 10.1038/s41575-019-0250-7] [Citation(s) in RCA: 258] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2019] [Indexed: 12/21/2022]
Abstract
Lipids entering the gastrointestinal tract include dietary lipids (triacylglycerols, cholesteryl esters and phospholipids) and endogenous lipids from bile (phospholipids and cholesterol) and from shed intestinal epithelial cells (enterocytes). Here, we comprehensively review the digestion, uptake and intracellular re-synthesis of intestinal lipids as well as their packaging into pre-chylomicrons in the endoplasmic reticulum, their modification in the Golgi apparatus and the exocytosis of the chylomicrons into the lamina propria and subsequently to lymph. We also discuss other fates of intestinal lipids, including intestinal HDL and VLDL secretion, cytosolic lipid droplets and fatty acid oxidation. In addition, we highlight the applicability of these findings to human disease and the development of therapeutics targeting lipid metabolism. Finally, we explore the emerging role of the gut microbiota in modulating intestinal lipid metabolism and outline key questions for future research.
Collapse
|
20
|
Skin permeability barrier formation by the ichthyosis-causative gene FATP4 through formation of the barrier lipid ω- O-acylceramide. Proc Natl Acad Sci U S A 2020; 117:2914-2922. [PMID: 31974308 DOI: 10.1073/pnas.1917525117] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The epidermis-specific lipid acylceramide plays a pivotal role in the formation of the permeability barrier in the skin; abrogation of its synthesis causes the skin disorder ichthyosis. However, the acylceramide synthetic pathway has not yet been fully elucidated: Namely, the acyl-CoA synthetase (ACS) involved in this pathway remains to be identified. Here, we hypothesized it to be encoded by FATP4/ACSVL4, the causative gene of ichthyosis prematurity syndrome (IPS). In vitro experiments revealed that FATP4 exhibits ACS activity toward an ω-hydroxy fatty acid (FA), an intermediate of the acylceramide synthetic pathway. Fatp4 knockout (KO) mice exhibited severe skin barrier dysfunction and morphological abnormalities in the epidermis. The total amount of acylceramide in Fatp4 KO mice was reduced to ∼10% of wild-type mice. Decreased levels and shortening of chain lengths were observed in the saturated, nonacylated ceramides. FA levels were not decreased in the epidermis of Fatp4 KO mice. The expression levels of the FA elongase Elovl1 were reduced in Fatp4 KO epidermis, partly accounting for the reduction and shortening of saturated, nonacylated ceramides. A decrease in acylceramide levels was also observed in human keratinocytes with FATP4 knockdown. From these results, we conclude that skin barrier dysfunction observed in IPS patients and Fatp4 KO mice is caused mainly by reduced acylceramide production. Our findings further elucidate the molecular mechanism governing acylceramide synthesis and IPS pathology.
Collapse
|
21
|
Zhu Y, Tan Q, Zhang L, Yao J, Zhou H, Hu P, Liang X, Liu H. The migration of docosahexenoic acid (DHA) to the developing ovary of female zebrafish (Danio rerio). Comp Biochem Physiol A Mol Integr Physiol 2019; 233:97-105. [PMID: 30978471 DOI: 10.1016/j.cbpa.2019.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 02/06/2023]
Abstract
Fish selectively reserves docosahexenoic acid (DHA) in ovary during gonadal development. However, no direct proof supports this. The present study tried to elucidate the DHA migration to the developing ovary of female zebrafish. An injection study of 13C-labeled DHA for DHA tracing was conducted, and another injection study of unlabeled-DHA (DHA-injected group) and BSA-saline (control group) was conducted for lipid and DHA content detection, related gene expression analyses, and histological observation. The results showed that the rapid absorption of lipid occurred at stage III with a constant accumulation of DHA in the ovary. The proportion of oocytes at stage III on day 7 and 21, and at stage IV on day 3 and 21 in DHA-injected group was significantly higher than that in control group, respectively (P < .05). The injected 13C-labeled DHA was accumulated twice in the ovary respectively on day 1 and 7, and remained at a relatively high level. In DHA-injected group, the fatp4 expression was significantly higher in ovary on day 3, 5 and 7 (P < .05), and significantly lower (P < .05) in liver on day 5, 14 and in muscle on day 1, 5 and 7 than that in control group. In conclusion, the present study suggested a migration of DHA from the liver and muscle to the gonads when necessary.
Collapse
Affiliation(s)
- Yanhong Zhu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingsong Tan
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Leisen Zhang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Junpeng Yao
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Hai Zhou
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Pengcheng Hu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xufang Liang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Hong Liu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
22
|
Zhang W, Chen R, Yang T, Xu N, Chen J, Gao Y, Stetler RA. Fatty acid transporting proteins: Roles in brain development, aging, and stroke. Prostaglandins Leukot Essent Fatty Acids 2018; 136:35-45. [PMID: 28457600 PMCID: PMC5650946 DOI: 10.1016/j.plefa.2017.04.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 04/16/2017] [Accepted: 04/20/2017] [Indexed: 12/18/2022]
Abstract
Polyunsaturated fatty acids are required for the brain development and significantly impact aging and stroke. Due to the hydrophobicity of fatty acids, fatty acids transportation related proteins that include fatty acid binding proteins (FABPs), long chain acyl-coA synthase (ACS), fatty acid transportation proteins (FATPs), fatty acid translocase (FAT/CD36) and newly reported major facilitator superfamily domain-containing protein (Mfsd2a) play critical roles in the uptake of various fatty acids, especially polyunsaturated fatty acids. They are not only involved in neurodevelopment, but also have great impact on neurological disease, such as aging related dementia and stroke.
Collapse
Affiliation(s)
- Wenting Zhang
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Ruiying Chen
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Na Xu
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - R Anne Stetler
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA.
| |
Collapse
|
23
|
Jensen VF, Mølck AM, Lykkesfeldt J, Bøgh IB. Effect of maternal hypoglycaemia during gestation on materno-foetal nutrient transfer and embryo-foetal development: Evidence from experimental studies focused primarily on the rat. Reprod Toxicol 2018; 77:1-24. [DOI: 10.1016/j.reprotox.2018.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 12/07/2017] [Accepted: 01/30/2018] [Indexed: 01/14/2023]
|
24
|
Cifarelli V, Abumrad NA. Intestinal CD36 and Other Key Proteins of Lipid Utilization: Role in Absorption and Gut Homeostasis. Compr Physiol 2018; 8:493-507. [PMID: 29687890 PMCID: PMC6247794 DOI: 10.1002/cphy.c170026] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Several proteins have been implicated in fatty acid (FA) transport by enterocytes including the scavenger receptor CD36 (SR-B2), the scavenger receptor B1 (SR-B1) a member of the CD36 family and the FA transport protein 4 (FATP4). Here, we review the regulation of enterocyte FA uptake and its function in lipid absorption including prechylomicron formation, assembly and transport. Emphasis is given to CD36, which is abundantly expressed along the digestive tract of rodents and humans and has been the most studied. We also address the pleiotropic functions of CD36 that go beyond lipid absorption and metabolism to include recent evidence of its impact on intestinal homeostasis and barrier maintenance. Areas of progress involving contribution of membrane phospholipid remodeling and of cytosolic FA-binding proteins, FABP1 and FABP2 to fat absorption will be covered. © 2018 American Physiological Society. Compr Physiol 8:493-507, 2018.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Internal Medicine, Center for Human Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| | - Nada A. Abumrad
- Department of Internal Medicine, Center for Human Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
25
|
Demenis C, McLaughlin J, Smith CP. Sulfated Cholecystokinin-8 Promotes CD36-Mediated Fatty Acid Uptake into Primary Mouse Duodenal Enterocytes. Front Physiol 2017; 8:660. [PMID: 28919867 PMCID: PMC5586203 DOI: 10.3389/fphys.2017.00660] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/18/2017] [Indexed: 11/13/2022] Open
Abstract
Cholecystokinin (CCK) is an archetypal incretin hormone secreted by intestinal enteroendocrine cells (EEC) in response to ingested nutrients. The aim of this study was to determine whether CCK modulates enterocyte fatty acid uptake by primary mouse duodenal cells. Exposure of primary mouse duodenal cells to 10 pM sulfated CCK-8 caused a two fold increase in dodecanoic acid fatty acid (FA) uptake. The selective CCK A receptor antagonist loxiglumide (100 μM) completely abolished the CCK-8 induced FA uptake. The CD36 fatty acid translocase-specific inhibitor sulfo-N-succinimidyl oleate (1 μM) also completely inhibited CCK-8 induced FA uptake, as did treatment with 200 μM phloretin. Together these data show CCK induces FA uptake into duodenal enterocytes; this action involves the CCK-RA receptor and is carrier mediated by CD36.
Collapse
Affiliation(s)
- Claire Demenis
- School of Medical Sciences, University of ManchesterManchester, United Kingdom
| | - John McLaughlin
- School of Medical Sciences, University of ManchesterManchester, United Kingdom
| | - Craig P Smith
- School of Medical Sciences, University of ManchesterManchester, United Kingdom
| |
Collapse
|
26
|
The ‘Goldilocks zone’ of fatty acid metabolism; to ensure that the relationship with cardiac function is just right. Clin Sci (Lond) 2017; 131:2079-2094. [DOI: 10.1042/cs20160671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/25/2022]
Abstract
Fatty acids (FA) are the main fuel used by the healthy heart to power contraction, supplying 60–70% of the ATP required. FA generate more ATP per carbon molecule than glucose, but require more oxygen to produce the ATP, making them a more energy dense but less oxygen efficient fuel compared with glucose. The pathways involved in myocardial FA metabolism are regulated at various subcellular levels, and can be divided into sarcolemmal FA uptake, cytosolic activation and storage, mitochondrial uptake and β-oxidation. An understanding of the critical involvement of each of these steps has been amassed from genetic mouse models, where forcing the heart to metabolize too much or too little fat was accompanied by cardiac contractile dysfunction and hypertrophy. In cardiac pathologies, such as heart disease and diabetes, aberrations in FA metabolism occur concomitantly with changes in cardiac function. In heart failure, FA oxidation is decreased, correlating with systolic dysfunction and hypertrophy. In contrast, in type 2 diabetes, FA oxidation and triglyceride storage are increased, and correlate with diastolic dysfunction and insulin resistance. Therefore, too much FA metabolism is as detrimental as too little FA metabolism in these settings. Therapeutic compounds that rebalance FA metabolism may provide a mechanism to improve cardiac function in disease. Just like Goldilocks and her porridge, the heart needs to maintain FA metabolism in a zone that is ‘just right’ to support contractile function.
Collapse
|
27
|
Lin MH, Miner JH, Turk J, Hsu FF. Linear ion-trap MS n with high-resolution MS reveals structural diversity of 1-O-acylceramide family in mouse epidermis. J Lipid Res 2017; 58:772-782. [PMID: 28154204 DOI: 10.1194/jlr.d071647] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 01/27/2017] [Indexed: 12/30/2022] Open
Abstract
1-O-acylceramide is a new class of epidermal cer-amide (Cer) found in humans and mice. Here, we report an ESI linear ion-trap (LIT) multiple-stage MS (MSn) approach with high resolution toward structural characterization of this lipid family isolated from mice. Molecular species desorbed as the [M + H]+ ions were subjected to LIT MS2 to yield predominately the [M + H - H2O]+ ions, followed by MS3 to cleave the 1-O-acyl residue to yield the [M + H - H2O - (1-O-FA)]+ ions. The structures of the N-acyl chain and long-chain base (LCB) of the molecule were determined by MS4 on [M + H - H2O - (1-O-FA)]+ ions that yielded multiple sets of specific ions. Using this approach, isomers varied in the 1-O-acyl (from 14:0- to 30:0-O-acyl) and N-acyl chains (from 14:0- to 34:1-N-acyl) with 18:1-sphingosine as the major LCB were found for the entire family. Minor isomers consisting of 16:1-, 17:1-, 18:2-, and 19:1-sphingosine LCBs with odd fatty acyl chain or with monounsaturated N- or O-fatty acyl substituents were also identified. An estimation of more than 700 1-O-acylceramide species, largely isobaric isomers, are present, underscoring the complexity of this Cer family.
Collapse
Affiliation(s)
- Meei-Hua Lin
- Division of Nephrology Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jeffrey H Miner
- Division of Nephrology Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - John Turk
- Mass Spectrometry Resource, Division of Endocrinology, Diabetes, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Fong-Fu Hsu
- Mass Spectrometry Resource, Division of Endocrinology, Diabetes, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
28
|
Ahowesso C, Black PN, Saini N, Montefusco D, Chekal J, Malosh C, Lindsley CW, Stauffer SR, DiRusso CC. Chemical inhibition of fatty acid absorption and cellular uptake limits lipotoxic cell death. Biochem Pharmacol 2015; 98:167-81. [PMID: 26394026 DOI: 10.1016/j.bcp.2015.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/03/2015] [Indexed: 12/31/2022]
Abstract
Chronic elevation of plasma free fatty acid (FFA) levels is commonly associated with obesity, type 2 diabetes, cardiovascular disease and some cancers. Experimental evidence indicates FFA and their metabolites contribute to disease development through lipotoxicity. Previously, we identified a specific fatty acid transport inhibitor CB16.2, a.k.a. Lipofermata, using high throughput screening methods. In this study, efficacy of transport inhibition was measured in four cell lines that are models for myocytes (mmC2C12), pancreatic β-cells (rnINS-1E), intestinal epithelial cells (hsCaco-2), and hepatocytes (hsHepG2), as well as primary human adipocytes. The compound was effective in inhibiting uptake with IC50s between 3 and 6μM for all cell lines except human adipocytes (39μM). Inhibition was specific for long and very long chain fatty acids but had no effect on medium chain fatty acids (C6-C10), which are transported by passive diffusion. Derivatives of Lipofermata were evaluated to understand structural contributions to activity. Lipofermata prevented palmitate-mediated oxidative stress, induction of BiP and CHOP, and cell death in a dose-dependent manner in hsHepG2 and rnINS-1E cells, suggesting it will prevent induction of fatty acid-mediated cell death pathways and lipotoxic disease by channeling excess fatty acids to adipose tissue and away from liver and pancreas. Importantly, mice dosed orally with Lipofermata were not able to absorb (13)C-oleate demonstrating utility as an inhibitor of fatty acid absorption from the gut.
Collapse
Affiliation(s)
- Constance Ahowesso
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0664, United States
| | - Paul N Black
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0664, United States
| | - Nipun Saini
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0664, United States
| | - David Montefusco
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0664, United States
| | - Jessica Chekal
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0664, United States
| | - Chrysa Malosh
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, United States
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, United States; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - Shaun R Stauffer
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Vanderbilt Specialized Chemistry Center for Probe Development (MLPCN), Nashville, TN 37232, United States; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - Concetta C DiRusso
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0664, United States.
| |
Collapse
|
29
|
Abstract
Dietary lipids are efficiently absorbed by the small intestine, incorporated into triglyceride-rich lipoproteins (chylomicrons), and transported in the circulation to various tissues. Intestinal lipid absorption and mobilization and chylomicron synthesis and secretion are highly regulated processes. Elevated chylomicron production rate contributes to the dyslipidemia seen in common metabolic disorders such as insulin-resistant states and type 2 diabetes and likely increases the risk for atherosclerosis seen in these conditions. An in-depth understanding of the regulation of chylomicron production may provide leads for the development of drugs that could be of therapeutic utility in the prevention of dyslipidemia and atherosclerosis. Chylomicron secretion is subject to regulation by various factors, including diet, body weight, genetic variants, hormones, nutraceuticals, medications, and emerging interventions such as bariatric surgical procedures. In this review we discuss the regulation of chylomicron production, mechanisms that underlie chylomicron dysregulation, and potential avenues for future research.
Collapse
Affiliation(s)
- Satya Dash
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, M5G 2C4 Canada;
| | | | | | | |
Collapse
|
30
|
Tsai CY, Peh MT, Feng W, Dymock BW, Moore PK. Hydrogen sulfide promotes adipogenesis in 3T3L1 cells. PLoS One 2015; 10:e0119511. [PMID: 25822632 PMCID: PMC4378953 DOI: 10.1371/journal.pone.0119511] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 01/14/2015] [Indexed: 12/30/2022] Open
Abstract
The effect of hydrogen sulfide (H2S) on differentiation of 3T3L1-derived adipocytes was examined. Endogenous H2S was increased after 3T3L1 differentiation. The expression of the H2S-synthesising enzymes, cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST), was increased in a time-dependent manner during 3T3L1 differentiation. Expression of genes associated with adipogenesis related genes including fatty acid binding protein 4 (FABP4/aP2), a key regulator of this process, was increased by GYY4137 (a slow-releasing H2S donor compound) and sodium hydrosulfide (NaHS, a classical H2S donor) but not by ZYJ1122 or time-expired NaHS. Furthermore expression of these genes were reduced by aminooxyacetic acid (AOAA, CBS inhibitor), DL-propargylglycine (PAG, CSE inhibitor) as well as by CSE small interference RNA (siCSE) and siCBS. The size and number of lipid droplets in mature adipocytes was significantly increased by both GYY4137 and NaHS, which also impaired the ability of CL316,243 (β3-agonist) to promote lipolysis in these cells. In contrast, AOAA and PAG had the opposite effect. Taken together, we show that the H2S-synthesising enzymes CBS, CSE and 3-MST are endogenously expressed during adipogenesis and that both endogenous and exogenous H2S modulate adipogenesis and adipocyte maturation.
Collapse
Affiliation(s)
- Chin-Yi Tsai
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
- * E-mail:
| | - Meng Teng Peh
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Wei Feng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Brian William Dymock
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Philip Keith Moore
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
| |
Collapse
|
31
|
Abstract
The intestinal production of lipoproteins is one of the key processes by which the body prepares dietary lipid for dissemination to locations throughout the body where they are required. Paramount to this is the relationship between dietary lipid and the enterocytes that line the gut, along with the processes which prepare this lipid for efficient uptake by these cells. These include those which occur in the mouth and stomach along with those which occur within the intestinal lumen itself. Additionally, the interplay between digested lipid, dual avenues for lipid uptake by enterocytes (passive and lipid transporter proteins), a system of intercellular lipid resynthesis and transport, and a complex system of lipoprotein synthesis yield a system open to significant modulation. In this review, we will attempt to outline the processes of lipid digestion, lipoprotein synthesis and the exogenous and endogenous factors which exert their influence.
Collapse
Affiliation(s)
- Alan A Hennessy
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland,
| | | | | | | | | |
Collapse
|
32
|
Levy E. Insights from human congenital disorders of intestinal lipid metabolism. J Lipid Res 2014; 56:945-62. [PMID: 25387865 DOI: 10.1194/jlr.r052415] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Indexed: 12/24/2022] Open
Abstract
The intestine must challenge the profuse daily flux of dietary fat that serves as a vital source of energy and as an essential component of cell membranes. The fat absorption process takes place in a series of orderly and interrelated steps, including the uptake and translocation of lipolytic products from the brush border membrane to the endoplasmic reticulum, lipid esterification, Apo synthesis, and ultimately the packaging of lipid and Apo components into chylomicrons (CMs). Deciphering inherited disorders of intracellular CM elaboration afforded new insight into the key functions of crucial intracellular proteins, such as Apo B, microsomal TG transfer protein, and Sar1b GTPase, the defects of which lead to hypobetalipoproteinemia, abetalipoproteinemia, and CM retention disease, respectively. These "experiments of nature" are characterized by fat malabsorption, steatorrhea, failure to thrive, low plasma levels of TGs and cholesterol, and deficiency of liposoluble vitamins and essential FAs. After summarizing and discussing the functions and regulation of these proteins for reader's comprehension, the current review focuses on their specific roles in malabsorptions and dyslipidemia-related intestinal fat hyperabsorption while dissecting the spectrum of clinical manifestations and managements. The influence of newly discovered proteins (proprotein convertase subtilisin/kexin type 9 and angiopoietin-like 3 protein) on fat absorption has also been provided. Finally, it is stressed how the overexpression or polymorphism status of the critical intracellular proteins promotes dyslipidemia and cardiometabolic disorders.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU Sainte-Justine and Department of Nutrition, Université de Montréal, Montreal, Quebec H3T 1C5, Canada
| |
Collapse
|
33
|
Abstract
Accumulation of triacylglycerols within the cytoplasm of hepatocytes to the degree that lipid droplets are visible microscopically is called liver steatosis. Most commonly, it occurs when there is an imbalance between the delivery or synthesis of fatty acids in the liver and their disposal through oxidative pathways or secretion into the blood as a component of triacylglycerols in very low density lipoprotein. This disorder is called nonalcoholic fatty liver disease (NAFLD) in the absence of alcoholic abuse and viral hepatitis, and it is often associated with insulin resistance, obesity and type 2 diabetes. Also, liver steatosis can be induced by many other causes including excessive alcohol consumption, infection with genotype 3 hepatitis C virus and certain medications. Whereas hepatic triacylglycerol accumulation was once considered the ultimate effector of hepatic lipotoxicity, triacylglycerols per se are quite inert and do not induce insulin resistance or cellular injury. Rather, lipotoxic injury in the liver appears to be mediated by the global ongoing fatty acid enrichment in the liver, paralleling the development of insulin resistance. A considerable number of fatty acid metabolites may be responsible for hepatic lipotoxicity and liver injury. Additional key contributors include hepatic cytosolic lipases and the "lipophagy" of lipid droplets, as sources of hepatic fatty acids. The specific origin of the lipids, mainly triacylglycerols, accumulating in liver has been unraveled by recent kinetic studies, and identifying the origin of the accumulated triacylglycerols in the liver of patients with NAFLD may direct the prevention and treatment of this condition.
Collapse
Affiliation(s)
- David Q-H Wang
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, Missouri
| | | | | |
Collapse
|
34
|
Wadhwani N, Patil V, Pisal H, Joshi A, Mehendale S, Gupte S, Wagh G, Joshi S. Altered maternal proportions of long chain polyunsaturated fatty acids and their transport leads to disturbed fetal stores in preeclampsia. Prostaglandins Leukot Essent Fatty Acids 2014; 91:21-30. [PMID: 24928794 DOI: 10.1016/j.plefa.2014.05.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 04/22/2014] [Accepted: 05/05/2014] [Indexed: 11/23/2022]
Abstract
Our previous cross-sectional studies have shown altered proportions of long chain polyunsaturated fatty acids (LCPUFA) in preeclampsia (PE) at the end of pregnancy when the pathology has already progressed. The present longitudinal study for the first time reports fatty acid proportions from 16th week of gestation till delivery and placental transport in PE. This is a hospital based study where women were recruited in early pregnancy. Maternal blood was collected at 3 time points i.e. T1=16-20th week, T2=26-30th week and T3=at delivery. Cord blood and placenta were collected at delivery. This study reports data on 140 normotensive control (NC) and 54 PE women. In PE we report lower proportions of DHA in maternal plasma at T1, cord plasma and placenta (p<0.05 for all). The mRNA levels of placental ∆5 desaturase, fatty acid transport proteins -1, -4, were lower (p<0.05 for all) in PE. There was also a positive association between cord and maternal plasma DHA and total omega-3 fatty acids at T1. This study demonstrates that women with PE have lower fatty acids stores at 16-20th week of gestation and lower placental synthesis and transport. It is likely that supplementation of omega-3 fatty acids during the 16-20th week of gestation may help in improving fatty acid status in infants born to mothers with PE.
Collapse
Affiliation(s)
- Nisha Wadhwani
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India
| | - Vidya Patil
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India
| | - Hemlata Pisal
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India
| | - Asmita Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India
| | - Savita Mehendale
- Department of Obstetrics and Gynecology, Bharati Medical College and Hospital, Bharati Vidyapeeth University, Pune 411043, India
| | | | - Girija Wagh
- Department of Obstetrics and Gynecology, Bharati Medical College and Hospital, Bharati Vidyapeeth University, Pune 411043, India
| | - Sadhana Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune Satara Road, Pune 411043, India.
| |
Collapse
|
35
|
Dongiovanni P, Anstee QM, Valenti L. Genetic predisposition in NAFLD and NASH: impact on severity of liver disease and response to treatment. Curr Pharm Des 2014; 19:5219-38. [PMID: 23394097 PMCID: PMC3850262 DOI: 10.2174/13816128113199990381] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 02/01/2013] [Indexed: 02/07/2023]
Abstract
Liver fat deposition related to systemic insulin resistance defines non-alcoholic fatty liver disease (NAFLD) which, when associated with oxidative hepatocellular damage, inflammation, and activation of fibrogenesis, i.e. non-alcoholic steatohepatitis (NASH), can progress towards cirrhosis and hepatocellular carcinoma. Due to the epidemic of obesity, NAFLD is now the most frequent liver disease and the leading cause of altered liver enzymes in Western countries. Epidemiological, familial, and twin studies provide evidence for an element of heritability of NAFLD. Genetic modifiers of disease severity and progression have been identified through genome-wide association studies. These include the Patatin-like phosholipase domain-containing 3 (PNPLA3) gene variant I148M as a major determinant of inter-individual and ethnicity-related differences in hepatic fat content independent of insulin resistance and serum lipid concentration. Association studies confirm that the I148M polymorphism is also a strong modifier of NASH and progressive hepatic injury. Furthermore, a few large multicentre case-control studies have demonstrated a role for genetic variants implicated in insulin signalling, oxidative stress, and fibrogenesis in the progression of NAFLD towards fibrosing NASH, and confirm that hepatocellular fat accumulation and insulin resistance are key operative mechanisms closely involved in the progression of liver damage. It is now important to explore the molecular mechanisms underlying these associations between gene variants and progressive liver disease, and to evaluate their impact on the response to available therapies. It is hoped that this knowledge will offer further insights into pathogenesis, suggest novel therapeutic targets, and could help guide physicians towards individualised therapy that improves clinical outcome.
Collapse
Affiliation(s)
- Paola Dongiovanni
- Department of Pathophysiology and Transplantation, section Internal Medicine, Università degli Studi Milano, UO Medicina Interna1B, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | |
Collapse
|
36
|
Hagberg C, Mehlem A, Falkevall A, Muhl L, Eriksson U. Endothelial fatty acid transport: role of vascular endothelial growth factor B. Physiology (Bethesda) 2014; 28:125-34. [PMID: 23455771 DOI: 10.1152/physiol.00042.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dietary lipids present in the circulation have to be transported through the vascular endothelium to be utilized by tissue cells, a vital mechanism that is still poorly understood. Vascular endothelial growth factor B (VEGF-B) regulates this process by controlling the expression of endothelial fatty acid transporter proteins (FATPs). Here, we summarize research on the role of the vascular endothelium in nutrient transport, with emphasis on VEGF-B signaling.
Collapse
|
37
|
Wang TY, Liu M, Portincasa P, Wang DQH. New insights into the molecular mechanism of intestinal fatty acid absorption. Eur J Clin Invest 2013; 43:1203-23. [PMID: 24102389 PMCID: PMC3996833 DOI: 10.1111/eci.12161] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 08/20/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND Dietary fat is one of the most important energy sources of all the nutrients. Fatty acids, stored as triacylglycerols (also called triglycerides) in the body, are an important reservoir of stored energy and derived primarily from animal fats and vegetable oils. DESIGN Although the molecular mechanisms for the transport of water-insoluble amphipathic fatty acids across cell membranes have been debated for many years, it is now believed that the dominant means for intestinal fatty acid uptake is via membrane-associated fatty acid-binding proteins, that is, fatty acid transporters on the apical membrane of enterocytes. RESULTS These findings indicate that intestinal fatty acid absorption is a multistep process that is regulated by multiple genes at the enterocyte level, and intestinal fatty acid absorption efficiency could be determined by factors influencing intraluminal fatty acid molecules across the brush border membrane of enterocytes. To facilitate research on intestinal, hepatic and plasma triacylglycerol metabolism, it is imperative to establish standard protocols for precisely and accurately measuring the efficiency of intestinal fatty acid absorption in humans and animal models. In this review, we will discuss the chemical structure and nomenclature of fatty acids and summarize recent progress in investigating the molecular mechanisms underlying the intestinal absorption of fatty acids, with a particular emphasis on the physical chemistry of intestinal lipids and the molecular physiology of intestinal fatty acid transporters. CONCLUSIONS A better understanding of the molecular mechanism of intestinal fatty acid absorption should lead to novel approaches to the treatment and the prevention of fatty acid-related metabolic diseases that are prevalent worldwide.
Collapse
Affiliation(s)
- Tony Y Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | | | | | | |
Collapse
|
38
|
Fatty acid transporters in skin development, function and disease. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:362-8. [PMID: 24120574 DOI: 10.1016/j.bbalip.2013.09.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/25/2013] [Accepted: 09/30/2013] [Indexed: 12/12/2022]
Abstract
Fatty acids in the epidermis can be incorporated into complex lipids or exist in a free form, and they are crucial to proper functions of the epidermis and its appendages, such as sebaceous glands. Epidermal fatty acids can be synthesized de novo by keratinocytes or taken up from extracutaneous sources in a process that likely involves protein transporters. Several proteins that are expressed in the epidermis have been proposed to facilitate the uptake of long-chain fatty acids (LCFA) in mammalian cells, including fatty acid translocase/CD36, fatty acid binding protein, and fatty acid transport protein (FATP)/very long-chain acyl-CoA synthetase. In this review, we will discuss the mechanisms by which these candidate transporters facilitate the uptake of fatty acids. We will then discuss the clinical implications of defects in these transporters and relevant animal models, including the FATP4 animal models and ichthyosis prematurity syndrome, a congenital ichthyosis caused by FATP4 deficiency. This article is part of a Special Issue entitled The Important Role of Lipids in the Epidermis and their Role in the Formation and Maintenance of the Cutaneous Barrier. Guest Editors: Kenneth R. Feingold and Peter Elias.
Collapse
|
39
|
Anderson CM, Stahl A. SLC27 fatty acid transport proteins. Mol Aspects Med 2013; 34:516-28. [PMID: 23506886 DOI: 10.1016/j.mam.2012.07.010] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 06/18/2012] [Indexed: 12/20/2022]
Abstract
The uptake and metabolism of long chain fatty acids (LCFA) are critical to many physiological and cellular processes. Aberrant accumulation or depletion of LCFA underlie the pathology of numerous metabolic diseases. Protein-mediated transport of LCFA has been proposed as the major mode of LCFA uptake and activation. Several proteins have been identified to be involved in LCFA uptake. This review focuses on the SLC27 family of fatty acid transport proteins, also known as FATPs, with an emphasis on the gain- and loss-of-function animal models that elucidate the functions of FATPs in vivo and how these transport proteins play a role in physiological and pathological situations.
Collapse
Affiliation(s)
- Courtney M Anderson
- Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California Berkeley, CA, USA
| | | |
Collapse
|
40
|
Buttet M, Traynard V, Tran TTT, Besnard P, Poirier H, Niot I. From fatty-acid sensing to chylomicron synthesis: role of intestinal lipid-binding proteins. Biochimie 2013; 96:37-47. [PMID: 23958439 DOI: 10.1016/j.biochi.2013.08.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/05/2013] [Indexed: 12/31/2022]
Abstract
Today, it is well established that the development of obesity and associated diseases results, in part, from excessive lipid intake associated with a qualitative imbalance. Among the organs involved in lipid homeostasis, the small intestine is the least studied even though it determines lipid bioavailability and largely contributes to the regulation of postprandial hyperlipemia (triacylglycerols (TG) and free fatty acids (FFA)). Several Lipid-Binding Proteins (LBP) are expressed in the small intestine. Their supposed intestinal functions were initially based on what was reported in other tissues, and took no account of the physiological specificity of the small intestine. Progressively, the identification of regulating factors of intestinal LBP and the description of the phenotype of their deletion have provided new insights into cellular and molecular mechanisms involved in fat absorption. This review will discuss the physiological contribution of each LBP in the main steps of intestinal absorption of long-chain fatty acids (LCFA): uptake, trafficking and reassembly into chylomicrons (CM). Moreover, current data indicate that the small intestine is able to adapt its lipid absorption capacity to the fat content of the diet, especially through the coordinated induction of LBP. This adaptation requires the existence of a mechanism of intestinal lipid sensing. Emerging data suggest that the membrane LBP CD36 may operate as a lipid receptor that triggers an intracellular signal leading to the modulation of the expression of LBP involved in CM formation. This event could be the starting point for the optimized synthesis of large CM, which are efficiently degraded in blood. Better understanding of this intestinal lipid sensing might provide new approaches to decrease the prevalence of postprandial hypertriglyceridemia, which is associated with cardiovascular diseases, insulin resistance and obesity.
Collapse
Affiliation(s)
- Marjorie Buttet
- Physiologie de la Nutrition et Toxicologie Team (NUTox), UMR U866 INSERM, Université de Bourgogne, AgroSup Dijon, 1 Esplanade Erasme, 21000 Dijon, France
| | | | | | | | | | | |
Collapse
|
41
|
Tremblay AJ, Lamarche B, Guay V, Charest A, Lemelin V, Couture P. Short-term, high-fat diet increases the expression of key intestinal genes involved in lipoprotein metabolism in healthy men. Am J Clin Nutr 2013; 98:32-41. [PMID: 23719552 DOI: 10.3945/ajcn.113.060251] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The modulation of cholesterol and fatty acid homeostasis by dietary fatty acids is thought to be mediated by changes in the expression of key intestinal genes involved in lipoprotein metabolism. However, the short-term effect of dietary fat intake on the expression of these genes has not been fully investigated in humans. OBJECTIVE To test whether short-term changes in dietary fatty acid intake affect the expression of key intestinal genes involved in lipoprotein metabolism, we conducted a randomized, double-blind, crossover study in 12 nonobese, healthy men with normal plasma lipid profiles. DESIGN Participants were subjected to the following 2 intensive 3-d dietary interventions under isocaloric conditions: 1) a high-fat diet (37% of energy from fat and 50% of energy from carbohydrates) and 2) a low-fat diet (25% of energy from fat and 62% of energy from carbohydrates). Expressions of key genes involved in lipoprotein metabolism were compared by using real-time polymerase chain reaction quantification on duodenal biopsy specimens obtained in a fasting state after each diet. RESULTS After the 3-d high-fat diet, plasma cholesterol, LDL cholesterol, and HDL cholesterol concentrations were significantly higher than concentrations observed after the low-fat diet was consumed. The high-fat diet also resulted in significant increases in the intestinal messenger RNA expression of several key genes involved in lipoprotein metabolism. Plasma triglycerides and apolipoprotein B-48 concentrations were significantly lower after the high-fat diet than after the low-fat diet. CONCLUSION These findings suggest that short-term exposure to a high-fat diet upregulates the expression of key genes involved in lipid and lipoprotein metabolism at the enterocyte level. This trial was registered at clinicaltrials.gov as NCT01806441.
Collapse
|
42
|
A spontaneous Fatp4/Scl27a4 splice site mutation in a new murine model for congenital ichthyosis. PLoS One 2012; 7:e50634. [PMID: 23226340 PMCID: PMC3511458 DOI: 10.1371/journal.pone.0050634] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 10/23/2012] [Indexed: 11/19/2022] Open
Abstract
Congenital ichthyoses are life-threatening conditions in humans. We describe here the identification and molecular characterization of a novel recessive mutation in mice that results in newborn lethality with severe congenital lamellar ichthyosis. Mutant newborns have a taut, shiny, non-expandable epidermis that resembles cornified manifestations of autosomal-recessive congenital ichthyosis in humans. The skin is stretched so tightly that the newborn mice are immobilized. The genetic defect was mapped to a region near the proximal end of chromosome 2 by SNP analysis, suggesting Fatp4/Slc27a4 as a candidate gene. FATP4 mutations in humans cause ichthyosis prematurity syndrome (IPS), and mutations of Fatp4 in mice have previously been found to cause a phenotype that resembles human congenital ichthyoses. Characterization of the Fatp4 cDNA revealed a fusion of exon 8 to exon 10, with deletion of exon 9. Genomic sequencing identified an A to T mutation in the splice donor sequence at the 3'-end of exon 9. Loss of exon 9 results in a frame shift mutation upstream from the conserved very long-chain acyl-CoA synthase (VLACS) domain. Histological studies revealed that the mutant mice have defects in keratinocyte differentiation, along with hyperproliferation of the stratum basale of the epidermis, a hyperkeratotic stratum corneum, and reduced numbers of secondary hair follicles. Since Fatp4 protein is present primarily at the stratum granulosum and the stratum spinosum, the hyperproliferation and the alterations in hair follicle induction suggest that very long chain fatty acids, in addition to being required for normal cornification, may influence signals from the stratum corneum to the basal cells that help to orchestrate normal skin differentiation.
Collapse
|
43
|
Dubé E, Gravel A, Martin C, Desparois G, Moussa I, Ethier-Chiasson M, Forest JC, Giguère Y, Masse A, Lafond J. Modulation of fatty acid transport and metabolism by maternal obesity in the human full-term placenta. Biol Reprod 2012; 87:14, 1-11. [PMID: 22553224 DOI: 10.1095/biolreprod.111.098095] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Knowledge of the consequences of maternal obesity in human placental fatty acids (FA) transport and metabolism is limited. Animal studies suggest that placental uptake of maternal FA is altered by maternal overnutrition. We hypothesized that high maternal body mass index (BMI) affects human placental FA transport by modifying expression of key transporters. Full-term placentas were obtained by vaginal delivery from normal weight (BMI, 18.5-24.9 kg/m(2)) and obese (BMI > 30 kg/m(2)) women. Blood samples were collected from the mother at each trimester and from cord blood at delivery. mRNA and protein expression levels were evaluated with real-time RT-PCR and Western blotting. Lipoprotein lipase (LPL) activity was evaluated using enzyme fluorescence. In vitro linoleic acid transport was studied with isolated trophoblasts. Our results demonstrated that maternal obesity is associated with increased placental weight, decreased gestational age, decreased maternal high-density lipoprotein (HDL) levels during the first and third trimesters, increased maternal triglyceride levels during the second and third trimesters, and increased maternal T3 levels during all trimesters, and decreased maternal cholesterol (CHOL) and low-density lipoprotein (LDL) levels during the third trimester; and increased newborn CHOL, LDL, apolipoprotein B100, and T3 levels. Increases in placental CD36 mRNA and protein expression levels, decreased SLC27A4 and FABP1 mRNA and protein and FABP3 protein expression, and increased LPL activity and decreased villus cytotrophoblast linoleic acid transport were also observed. No changes were seen in expression of PPARA, PPARD, or PPARG mRNA and protein. Overall this study demonstrated that maternal obesity impacts placental FA uptake without affecting fetal growth. These changes, however, could modify the fetus metabolism and its predisposition to develop diseases later in life.
Collapse
Affiliation(s)
- Evemie Dubé
- Laboratoire de Physiologie Materno-Fœtale, Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Frochot V, Alqub M, Cattin AL, Carrière V, Houllier A, Baraille F, Barbot L, Saint-Just S, Ribeiro A, Lacasa M, Cardot P, Chambaz J, Rousset M, Lacorte JM. The transcription factor HNF-4α: a key factor of the intestinal uptake of fatty acids in mouse. Am J Physiol Gastrointest Liver Physiol 2012; 302:G1253-63. [PMID: 22461026 DOI: 10.1152/ajpgi.00329.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
With an excessive postprandial accumulation of intestine-derived, triglyceride-rich lipoproteins being a risk factor of cardiovascular diseases, it is essential to characterize the mechanisms controlling the intestinal absorption of dietary lipids. Our aim was to investigate the role of the transcription factor hepatocyte nuclear factor (HNF)-4α in this process. We used transgenic mice with a specific and inducible intestinal knockout of Hnf-4α gene. One hour after a lipid bolus, in the presence of the lipase inhibitor tyloxapol, lower amounts of triglycerides were found in both plasma and intestinal epithelium of the intestine-specific Hnf-4α knockout (Hnf-4α(intΔ)) mice compared with the Hnf-4α(loxP/loxP) control mice. These discrepancies were due to a net decrease of the intestinal uptake of fatty acid in Hnf-4α(intΔ) mice compared with Hnf-4α(loxP/loxP) mice, as assessed by the amount of radioactivity that was recovered in intestine and plasma after gavage with labeled triolein or oleic acid, or in intestinal epithelial cells isolated from jejunum after a supply of labeled oleic acid-containing micelles. This decreased fatty acid uptake was associated with significant lower levels of the fatty acid transport protein-4 mRNA and protein along the intestinal tract and with a lower acyl-CoA synthetase activity in Hnf-4α(intΔ) mice compared with the control mice. We conclude that the transcription factor HNF-4α is a key factor of the intestinal absorption of dietary lipids, which controls this process as early as in the initial step of fatty acid uptake by enterocytes.
Collapse
Affiliation(s)
- Vincent Frochot
- Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, UMRS, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Enhanced fatty acid oxidation and FATP4 protein expression after endurance exercise training in human skeletal muscle. PLoS One 2012; 7:e29391. [PMID: 22235293 PMCID: PMC3250432 DOI: 10.1371/journal.pone.0029391] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 11/28/2011] [Indexed: 11/29/2022] Open
Abstract
FATP1 and FATP4 appear to be important for the cellular uptake and handling of long chain fatty acids (LCFA). These findings were obtained from loss- or gain of function models. However, reports on FATP1 and FATP4 in human skeletal muscle are limited. Aerobic training enhances lipid oxidation; however, it is not known whether this involves up-regulation of FATP1 and FATP4 protein. Therefore, the aim of this project was to investigate FATP1 and FATP4 protein expression in the vastus lateralis muscle from healthy human individuals and to what extent FATP1 and FATP4 protein expression were affected by an increased fuel demand induced by exercise training. Eight young healthy males were recruited to the study. All subjects were non smokers and did not participate in regular physical activity (<1 time per week for the past 6 months, VO2peak 3.4±0.1 l O2 min−1). Subjects underwent an 8 week supervised aerobic training program. Training induced an increase in VO2peak from 3.4±0.1 to 3.9±0.1 l min−1 and citrate synthase activity was increased from 53.7±2.5 to 80.8±3.7 µmol g−1 min−1. The protein content of FATP4 was increased by 33%, whereas FATP1 protein content was reduced by 20%. Interestingly, at the end of the training intervention a significant association (r2 = 0.74) between the observed increase in skeletal muscle FATP4 protein expression and lipid oxidation during a 120 min endurance exercise test was observed. In conclusion, based on the present findings it is suggested that FATP1 and FATP4 proteins perform different functional roles in handling LCFA in skeletal muscle with FATP4 apparently more important as a lipid transport protein directing lipids for lipid oxidation.
Collapse
|
46
|
Königsdorf CAI, Navarrete Santos A, Schmidt JS, Fischer S, Fischer B. Expression profile of fatty acid metabolism genes in preimplantation blastocysts of obese and non-obese mice. Obes Facts 2012; 5:575-86. [PMID: 22986646 DOI: 10.1159/000342583] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 02/09/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent studies have disclosed a close relationship between maternal obesity, fetal metabolism and pre- and postnatal development. The lipid metabolism in preimplantation embryos is a possible target of metabolic programming. METHODS 31 genes of beta-oxidation and fatty acid and cholesterol uptake, synthesis and regulation were analyzed in day 3.5 blastocysts from NZO (obese) and C57Bl/6 (normal weight) mice by RT-PCR and semiquantitative PCR. RESULTS The most obvious difference between both strains was the lack of the RXR gamma transcript in NZO blastocysts. In adult NZO mice, RXR gamma is detectable in most tissues. In a semiquantitative analysis, a higher transcription rate of fatty acid transport protein 4 (p = 0.004) and a reduced transcript number of fatty acid synthase (p = 0.049) was found in NZO blastocysts. Cholesterol synthesis regulation was modified in NZO blastocysts, as indicated by the ratio of sterol regulatory element-binding protein (SREBP) 2 / insulin-induced gene 1 (Insig 1) (p = 0.001). CONCLUSION In mouse blastocysts enzymes and signal molecules of fatty acid and cholesterol metabolism resemble those expressed postnatally. Distinct differences in transcription rates of genes between blastocysts from obese and non-obese mothers indicate that preimplantation embryo development is an early target for metabolic programming.
Collapse
|
47
|
|
48
|
The expression and function of fatty acid transport protein-2 and -4 in the murine placenta. PLoS One 2011; 6:e25865. [PMID: 22028793 PMCID: PMC3197585 DOI: 10.1371/journal.pone.0025865] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 09/13/2011] [Indexed: 11/19/2022] Open
Abstract
Background The uptake and trans-placental trafficking of fatty acids from the maternal blood into the fetal circulation are essential for embryonic development, and involve several families of proteins. Fatty acid transport proteins (FATPs) uniquely transport fatty acids into cells. We surmised that placental FATPs are germane for fetal growth, and are regulated during hypoxic stress, which is associated with reduced fat supply to the fetus. Methodology/Principal Findings Using cultured primary term human trophoblasts we found that FATP2, FATP4 and FATP6 were highly expressed in trophoblasts. Hypoxia enhanced the expression of trophoblastic FATP2 and reduced the expression of FATP4, with no change in FATP6. We also found that Fatp2 and Fatp4 are expressed in the mouse amnion and placenta, respectively. Mice deficient in Fatp2 or Fatp4 did not deviate from normal Mendelian distribution, with both embryos and placentas exhibiting normal weight and morphology, triglyceride content, and expression of genes related to fatty acid mobilization. Conclusions/Significance We conclude that even though hypoxia regulates the expression of FATP2 and FATP4 in human trophoblasts, mouse Fatp2 and Fatp4 are not essential for intrauterine fetal growth.
Collapse
|
49
|
Kazantzis M, Stahl A. Fatty acid transport proteins, implications in physiology and disease. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:852-7. [PMID: 21979150 DOI: 10.1016/j.bbalip.2011.09.010] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 09/19/2011] [Accepted: 09/19/2011] [Indexed: 01/12/2023]
Abstract
Uptake of long-chain fatty acids plays pivotal roles in metabolic homeostasis and human physiology. Uptake rates must be controlled in an organ-specific fashion to balance storage with metabolic needs during transitions between fasted and fed states. Many obesity-associated diseases, such as insulin resistance in skeletal muscle, cardiac lipotoxicity, and hepatic steatosis, are thought to be driven by the overflow of fatty acids from adipose stores and the subsequent ectopic accumulation of lipids resulting in apoptosis, ER stress, and inactivation of the insulin receptor signaling cascade. Thus, it is of critical importance to understand the components that regulate the flux of fatty acid between the different organ systems. Cellular uptake of fatty acids by key metabolic organs, including the intestine, adipose tissue, muscle, heart, and liver, has been shown to be protein mediated and various unique combinations of fatty acid transport proteins (FATPs/SLC27A1-6) are expressed by all of these tissues. Here we review our current understanding of how FATPs can contribute to normal physiology and how FATP mutations as well as hypo- and hypermorphic changes contribute to disorders ranging from cardiac lipotoxicity to hepatosteatosis and ichthyosis. Ultimately, our increasing knowledge of FATP biology has the potential to lead to the development of new diagnostic tools and treatment options for some of the most pervasive chronic human disorders. This article is part of a Special Issue entitled Triglyceride Metabolism and Disease.
Collapse
|
50
|
Khnykin D, Miner JH, Jahnsen F. Role of fatty acid transporters in epidermis: Implications for health and disease. DERMATO-ENDOCRINOLOGY 2011; 3:53-61. [PMID: 21695012 PMCID: PMC3117002 DOI: 10.4161/derm.3.2.14816] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 01/07/2011] [Accepted: 01/13/2011] [Indexed: 12/31/2022]
Abstract
Skin epidermis is an active site of lipid synthesis. The intercellular lipids of human stratum corneum (SC) are unique in composition and quite different from the lipids found in most biological membranes. The three major lipids in the SC are free fatty acids, cholesterol and ceramides. Fatty acids can be synthesized by keratinocytes de novo and, in addition, need to be taken up from the circulation. The latter process has been shown to be protein mediated, and several fatty acid transporters are expressed in skin. Recent studies of transgenic and knockout animal models for fatty acid transporters and the identification of fatty acid transport protein 4 (FATP4 or SLC27A4) mutations as causative for Ichthyosis Prematurity Syndrome highlight the vital roles of fatty acid transport and metabolism in skin homeostasis. This review provides an overview of our current understanding of the role of fatty acids and their transporters in cutaneous biology, including their involvement in epidermal barrier generation and skin inflammation.
Collapse
Affiliation(s)
- Denis Khnykin
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT); Department of Pathology; Oslo University Hospital-Rikshospitalet; Oslo, Norway
| | | | | |
Collapse
|