1
|
Tang J, Yang N, Pan S, Ren P, Chen M, Jin J, He Q, Zeng Y. The renal damage and mechanisms relevant to antitumoral drugs. Front Oncol 2023; 13:1331671. [PMID: 38148845 PMCID: PMC10749913 DOI: 10.3389/fonc.2023.1331671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Over the past few decades, significant progress has been made in the development of drugs to combat cancer. It is unfortunate that these drugs can also lead to various kidney injuries and imbalances in electrolyte levels. Nephrotoxicity caused by chemotherapy drugs can impact different parts of the kidneys, including the glomeruli, renal tubules, interstitium, or renal microvessels. Despite the existing knowledge, our understanding of the mechanisms underlying the renal damage caused by antitumoral drugs remains incomplete. In this review, we aim to provide a comprehensive overview of the specific types of kidney injury and the mechanisms responsible for the drug-mediated renal damage, and briefly discuss possible prevention and treatment measures. Sensitive blood and urine biomarkers can provide clinicians with more information about kidney injury detection and reference value for subsequent treatment options. In addition, we emphasize that both oncologists and nephrologists have a responsibility to remain vigilant against the potential nephrotoxicity of the drugs. It's crucial for experts in both fields to collaborate in early detection, monitoring and prevention of kidney damage.
Collapse
Affiliation(s)
- Jiyu Tang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Nan Yang
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
- Jinzhou Medical University, Graduate School of Clinical Medicine, Jinzhou, China
| | - Shujun Pan
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Peiyao Ren
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Maosheng Chen
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| | - Juan Jin
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Qiang He
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Yuqun Zeng
- Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
2
|
Takeda S, Ahhmed AM, Sogawa K, Mouri S, Kaneko S, Sakata R, Mizunoya W. Antioxidant activity of venison subjected to in vitro cooking and gastrointestinal digestion and isolation of its 2,2-diphenyl-1-picrylhydrazyl radical scavenging peptides. Anim Sci J 2023; 94:e13870. [PMID: 37743546 DOI: 10.1111/asj.13870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/14/2023] [Accepted: 08/16/2023] [Indexed: 09/26/2023]
Abstract
Venison, a type of game meat, has several health benefits because it contains not only high protein and low fat but also bioactive peptides with several physiological properties, including antioxidative and angiotensin-converting enzyme inhibitory properties. The aim of the present study was to investigate the antioxidant activity of venison treated by in vitro cooking and gastrointestinal digestion. We subjected venison along with pork and beef to in vitro cooking and digestion and assessed their antioxidant activity via 2,2-diphenyl-1-picrylhydrazyl radical scavenging (DPPH-RS) and hydrophilic oxygen radical absorbance capacity (H-ORAC) assays. The peptide contents of all types of cooked and digested meat samples were higher than those of the untreated and cooked samples. The DPPH-RS activities and H-ORAC of digested venison, pork, and beef were increased compared with those of untreated samples. DPPH-RS activity was significantly higher in the digested venison samples than in the digested pork and beef samples. In this study, several fractions of digested venison from the chromatography exhibited DPPH-RS activity. Peptide analysis, using liquid-chromatography with tandem mass spectrometry, unveiled two peptides DIDDLELTLAK and TQTVCNFTDGALVQHQEWDGK with high DPPH-RS activities. Thus, venison is a rich source of antioxidant peptides and potentially demonstrate an antioxidation ability by digestive enzymes in vivo.
Collapse
Affiliation(s)
- Shiro Takeda
- School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
- Graduate School of Veterinary Science, Azabu University, Sagamihara, Kanagawa, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Abdulatef M Ahhmed
- Department of Nutritional Therapy, Graduate School of Medical Science, the Libyan Academy, Tripoli, Libya
| | - Kazuki Sogawa
- School of Life and Environmental Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Saki Mouri
- Graduate School of Veterinary Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Sakurako Kaneko
- Graduate School of Veterinary Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Ryoichi Sakata
- School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
- Graduate School of Veterinary Science, Azabu University, Sagamihara, Kanagawa, Japan
| | - Wataru Mizunoya
- School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
- Graduate School of Veterinary Science, Azabu University, Sagamihara, Kanagawa, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Kanagawa, Japan
| |
Collapse
|
3
|
On the Sensitivity of the Virion Envelope to Lipid Peroxidation. Microbiol Spectr 2022; 10:e0300922. [PMID: 36125312 PMCID: PMC9603946 DOI: 10.1128/spectrum.03009-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Emerging viruses are a public health threat best managed with broad spectrum antivirals. Common viral structures, like capsids or virion envelopes, have been proposed as targets for broadly active antiviral drugs. For example, a number of lipoperoxidators have been proposed to preferentially affect viral infectivity by targeting metabolically inactive enveloped virions while sparing metabolically active cells. However, this presumed preferential virion sensitivity to lipoperoxidation remains untested. To test whether virions are indeed more sensitive to lipoperoxidation than are cells, we analyzed the effects of two classic generic lipoperoxidators: lipophilic 2,2'-azobis(2,4-dimethylvaleronitrile) (AMVN) and hydrophilic 2,2'-azobis(2-methylpropionamidine) dihydrochloride (AAPH) on Vero and human foreskin fibroblasts (HFF) cell viability, HSV-1 plaquing efficiency, and virion and cell lipoperoxidation. Cells or virions were incubated with the lipoperoxidators at 37°C for 2 h or incubated in atmospheric O2, and dose responses (half maximal cytotoxic and effective concentration [CC50 and EC50]) were evaluated by three or four parameter regression. The HSV-1 virions were slightly more sensitive to lipoperoxidators than were the cells (selectivity index [SI], 3.3 to 7.4). The effects of the lipophilic AMVN on both cell and virion viability directly correlated with the extent of membrane lipoperoxidation as evaluated by two different probes, C11-Bodipy and liperfluo. Moreover, the hydrophilic AAPH-induced virion inactivation at lower concentrations than did lipoperoxidation. Known lipoperoxidators inhibit infectivity via lipoperoxidation-independent mechanisms. Antioxidants protected against a loss of viral infectivity by less than 5-fold. Carrier bovine serum albumin (BSA) protected against both peroxidators to a similar extent when present together with the lipoperoxidating agents, suggesting that BSA quenches them as expected. Virions incubated in atmospheric oxidative conditions suffered losses of infectivity that were similar to those of chemically peroxidated virions, and they were protected by water soluble vitamin C and BSA with no evident lipoperoxidation, indicating predominant peroxidative damage to nonlipid virion components. Thus, lipoperoxidation is not a mechanism by which to specifically inhibit the infectivity of enveloped viruses, and the effects of known lipoperoxidators on virion infectivity are not solely mediated by lipoperoxidation. IMPORTANCE Small molecules that induce lipoperoxidation have been proposed repeatedly as potential antiviral drugs based on a presumed unique sensitivity of virions to this type of damage. Several small molecules that inactivate virions without affecting cells have been proposed to act primarily by inducing lipoperoxidation. However, the preferential sensitivity of virions to lipoperoxidators had not been experimentally evaluated. Using two of the best characterized small molecule lipoperoxidators, which are widely considered to be the prototypical water soluble and liposoluble lipoperoxidators, we show that lipoperoxidators have no preference for virions over cells. Moreover, they also inactivate virions by mechanisms other than the induction of lipoperoxidation. Therefore, the general induction of lipoperoxidation is not a path by which to develop antivirals. Moreover, molecules with specific antiviral activity which are not cytotoxic and have no preference to localize to virions over cells are unlikely to act primarily by inducing lipoperoxidation.
Collapse
|
4
|
Yu J, Hu D, Cheng Y, Guo J, Wang Y, Tan Z, Peng J, Zhou H. Lipidomics and transcriptomics analyses of altered lipid species and pathways in oxaliplatin-treated colorectal cancer cells. J Pharm Biomed Anal 2021; 200:114077. [PMID: 33892396 DOI: 10.1016/j.jpba.2021.114077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/13/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022]
Abstract
Drug resistance and adverse reactions to oxaliplatin remain a considerable issue in clinical practice. Emerging evidence has suggested that alterations in the lipid metabolism during drug therapy affect cancer cells. To gain insight into the important process of lipid metabolism, we investigated the lipid and gene expression profile changes in HT29 cells treated with oxaliplatin. A total of 1403 lipid species from 16 lipid classes were identified by UHPLC-MS. Interestingly, phospholipids, including phosphatidylglycerol (PG), phosphatidic acid (PA), phosphatidylcholine (PC), and most of phosphatidylethanolamine (PE) with polyunsaturated fatty acid (PUFA) chains, were significantly higher due to oxaliplatin treatment, while triacylglycerols (TAGs) with a saturated fatty acid chain or monounsaturated fatty acid were significantly downregulated. Gene Set Enrichment Analysis (GSEA) based on RNA sequencing data suggested that neutral lipid metabolism was enriched in the control group, whereas the phospholipid metabolic process was enriched in the oxaliplatin-treated group. We observed that altered lipid metabolism enzyme genes were involved in the synthesis and lipolysis of TAGs and the Lands cycle pathway based on the network between the core lipid-related gene and lipid species, which was further verified by qRT-PCR. In summary, our findings revealed that oxaliplatin impressed a specific lipid profile signature and lipid transcriptional reprogramming in HT29 cells, which provides new insights into biomarker discovery and pathways for overcoming drug resistance and adverse reactions.
Collapse
Affiliation(s)
- Jing Yu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Dongli Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Yu Cheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Jiwei Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Yicheng Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Zhirong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Jingbo Peng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China.
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China.
| |
Collapse
|
5
|
Zhu D, Rostami MR, Zuo WL, Leopold PL, Crystal RG. Single-Cell Transcriptome Analysis of Mouse Liver Cell-Specific Tropism and Transcriptional Dysregulation Following Intravenous Administration of AAVrh.10 Vectors. Hum Gene Ther 2020; 31:590-604. [PMID: 32143547 PMCID: PMC7232697 DOI: 10.1089/hum.2019.366] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/16/2020] [Indexed: 01/20/2023] Open
Abstract
Capitalizing on liver tropism of adeno-associated viral (AAV) vectors, intravenous vector administration is commonly used to genetically modify hepatocytes, a strategy currently in clinical trials for a number of liver-based hereditary disorders. Although hepatocytes are known to exhibit extensive phenotypic heterogeneity influenced by liver zonation and dietary cycle, there is little data available for the tropism capacity, as well as the potential transcriptional dysregulation, of AAV vectors for specific liver cell types. To assess these issues, we employed single-cell RNA sequencing of the mouse liver after intravenous administration of the liver tropic AAVrh.10 vector to characterize cell-specific AAV-mediated transgene expression and transcriptome dysregulation. Wild-type 8-week-old male C57Bl/6 mice under normal feed cycle were randomly divided into three groups and intravenously administered phosphate-buffered saline (PBS), AAVrh.10Null (no transgene), or AAVrh.10mCherry (marker gene). Overall, a total of 46,500 liver cells were sequenced. The single-cell transcriptomic profiles were grouped into three separate clusters of hepatocytes (Ttr-enriched "Hep1," Tat-enriched "Hep2," and Alb-enriched "Hep3") and multiple other cell types. The hepatocyte diversity was driven by glucose and lipid homeostasis signaling. Assessment of the transgene expression demonstrated that AAVrh.10 is primarily Hep1-tropic, with a 10-gene signature positively correlated with AAVrh.10-mediated transgene expression. The transgene expression was less in Hep2 and Hep3 cells with a high receptor tyrosine kinase phenotype. Importantly, AAVrh.10 vector interactions with the liver markedly altered the transcriptional patterns of all cell types, with modified genes enriched in pathways of complement and coagulation cascade, cytochrome P450, peroxisome, antigen processing and presentation, and endoplasmic reticulum protein processing. These observations provide insights into the liver cell-specific consequences of AAV-mediated liver gene transfer, far beyond the well-known organ-specific expression of the vector-delivered transgene.
Collapse
Affiliation(s)
- Detu Zhu
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Mahboubeh R. Rostami
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Wu-lin Zuo
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Philip L. Leopold
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
6
|
Lu YC, Chang CC, Wang CP, Hung WC, Tsai IT, Tang WH, Wu CC, Wei CT, Chung FM, Lee YJ, Hsu CC. Circulating fatty acid-binding protein 1 (FABP1) and nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus. Int J Med Sci 2020; 17:182-190. [PMID: 32038102 PMCID: PMC6990891 DOI: 10.7150/ijms.40417] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/19/2019] [Indexed: 12/30/2022] Open
Abstract
Background: Fatty acid-binding protein 1 (FABP1) (also known as liver-type fatty acid-binding protein or LFABP) is a protein that is mainly expressed in the liver, and is associated with hepatocyte injury in acute transplant rejection. Reduced levels of FABP1 in mice livers have been shown to be effective against nonalcoholic fatty liver disease (NAFLD). In this study, we investigated the association between plasma FABP1 levels and NAFLD in patients with type 2 diabetes mellitus (T2DM). Methods: We enrolled 267 T2DM patients. Clinical and biochemical parameters were measured. The severity of NAFLD was assessed by ultrasound. FABP1 levels were determined using by enzyme-linked immunosorbent assays. Results: FABP1 levels were higher in patients with overt NAFLD, defined as more than a moderate degree of fatty liver compared to those without NAFLD. Age- and sex-adjusted analysis of FABP1 showed positive associations with body mass index (BMI), waist circumference, homeostasis model assessment estimate of β-cell function, creatinine, and fatty liver index, but showed negative associations with albumin and estimated glomerular filtration rate (eGFR). The odds ratio (OR) for the risk of overt NAFLD with increasing levels of sex-specific FABP1 was significantly increased (OR 2.63 [95% CI 1.30-5.73] vs. 4.94 [2.25-11.48]). The OR in the second and third tertiles of FABP1 remained significant after adjustments for BMI, triglycerides, high-density lipoprotein cholesterol, HbA1C, homeostasis model assessment estimate of insulin resistance, white blood cell count, hepatic enzymes, and eGFR. Conclusion: Our results indicate that FABP1 may play a role in the pathogenesis of NAFLD in patients with T2DM.
Collapse
Affiliation(s)
- Yung-Chuan Lu
- Division of Endocrinology and Metabolism, E-Da Hospital, Kaohsiung, 82445 Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan
| | - Chi-Chang Chang
- Department of Obstetrics & Gynecology, E-Da Hospital, Kaohsiung, 82445 Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan.,Department of Obstetrics & Gynecology, E-Da Dachang Hospital, Kaohsiung 80794 Taiwan
| | - Chao-Ping Wang
- Division of Cardiology, E-Da Hospital, Kaohsiung, 82445 Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan
| | - Wei-Chin Hung
- Division of Cardiology, E-Da Hospital, Kaohsiung, 82445 Taiwan.,The School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan
| | - I-Ting Tsai
- Departmen of Emergency, E-Da Hospital, Kaohsiung, 82445 Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan
| | - Wei-Hua Tang
- Lee's Endocrinology Clinic, Pingtung, 90000 Taiwan
| | - Cheng-Ching Wu
- Division of Cardiology, E-Da Hospital, Kaohsiung, 82445 Taiwan.,The School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan.,Division of Cardiology, Department of Internal Medicine, E-Da Cancer Hospital, Kaohsiung 82445 Taiwan
| | - Ching-Ting Wei
- Division of General Surgery, Department of Surgery, E-Da Hospital, Kaohsiung, 82445 Taiwan
| | - Fu-Mei Chung
- Division of Cardiology, E-Da Hospital, Kaohsiung, 82445 Taiwan
| | | | - Chia-Chang Hsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, 82445 Taiwan.,The School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan.,Health Examination Center, E-Da Dachang Hospital, Kaohsiung, Taiwan
| |
Collapse
|
7
|
Nerstedt A, Kurhe Y, Cansby E, Caputo M, Gao L, Vorontsov E, Ståhlman M, Nuñez-Durán E, Borén J, Marschall HU, Mashek DG, Saunders DN, Sihlbom C, Hoy AJ, Mahlapuu M. Lipid droplet-associated kinase STK25 regulates peroxisomal activity and metabolic stress response in steatotic liver. J Lipid Res 2019; 61:178-191. [PMID: 31857389 DOI: 10.1194/jlr.ra119000316] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/05/2019] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are emerging as leading causes of liver disease worldwide and have been recognized as one of the major unmet medical needs of the 21st century. Our recent translational studies in mouse models, human cell lines, and well-characterized patient cohorts have identified serine/threonine kinase (STK)25 as a protein that coats intrahepatocellular lipid droplets (LDs) and critically regulates liver lipid homeostasis and progression of NAFLD/NASH. Here, we studied the mechanism-of-action of STK25 in steatotic liver by relative quantification of the hepatic LD-associated phosphoproteome from high-fat diet-fed Stk25 knockout mice compared with their wild-type littermates. We observed a total of 131 proteins and 60 phosphoproteins that were differentially represented in STK25-deficient livers. Most notably, a number of proteins involved in peroxisomal function, ubiquitination-mediated proteolysis, and antioxidant defense were coordinately regulated in Stk25 -/- versus wild-type livers. We confirmed attenuated peroxisomal biogenesis and protection against oxidative and ER stress in STK25-deficient human liver cells, demonstrating the hepatocyte-autonomous manner of STK25's action. In summary, our results suggest that regulation of peroxisomal function and metabolic stress response may be important molecular mechanisms by which STK25 controls the development and progression of NAFLD/NASH.
Collapse
Affiliation(s)
- Annika Nerstedt
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Yeshwant Kurhe
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Emmelie Cansby
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mara Caputo
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lei Gao
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Egor Vorontsov
- Proteomics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Esther Nuñez-Durán
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Borén
- Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN
| | - Darren N Saunders
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Carina Sihlbom
- Proteomics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Margit Mahlapuu
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
8
|
Tang W, Chen O, Yao F, Cui L. miR‑455 targets FABP4 to protect human endometrial stromal cells from cytotoxicity induced by hydrogen peroxide. Mol Med Rep 2019; 20:4781-4790. [PMID: 31638263 PMCID: PMC6854537 DOI: 10.3892/mmr.2019.10727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 04/04/2019] [Indexed: 01/03/2023] Open
Abstract
Oxidative stress and dysregulation of antioxidant systems are associated with various complications in pregnancy. Endometriosis is a common gynecologic disease that affects women of reproductive age. Recent studies have indicated that oxidative stress may be involved in the pathophysiology of endometriosis. It has been reported that microRNAs can regulate the cellular response to oxidative stress, and mounting evidence indicates that fatty acid binding protein 4 (FABP4) plays an essential role in the regulation of systemic redox capacity. In the present study, we demonstrated that miR-455 is a putative FABP4-targeting miRNA. A luciferase activity assay revealed that miR-455 can successfully bind to the 3′-UTR of FABP4. Overexpression of miR-455 led to the downregulation of FABP4 at both the mRNA and protein levels in a human endometrial stromal cell line. Then, the roles of miR-455 and FABP4 in oxidative stress induced by hydrogen peroxide (H2O2) in human endometrial stromal cells were examined. We found that ectopic expression of miR-455 protected cells from damage caused by H2O2. Further investigation found that forced expression of miR-455 reduced the level of reactive oxygen species (ROS) and malondialdehyde (MDA), while the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) were promoted. Silencing of FABP4 also generated cytoprotective effects against H2O2 in human endometrial stromal cells. Moreover, overexpression FABP4 abrogated the miR-455-mediated antioxidative stress effects in cells. Taken together, we propose that miR-455 protects human endometrial stromal cells from oxidative stress at least partly via regulation of FABP4.
Collapse
Affiliation(s)
- Wenbo Tang
- Department of Obstetrics and Gynecology, Ningbo First Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Oujing Chen
- Department of Obstetrics and Gynecology, Ningbo First Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Fengxiang Yao
- Department of Obstetrics and Gynecology, Ningbo First Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Lining Cui
- Department of Obstetrics and Gynecology, Ningbo First Hospital, Ningbo, Zhejiang 315000, P.R. China
| |
Collapse
|
9
|
Chen W, Wang K, Liu S. Molecular cloning and tissue distribution of fatty acid binding protein-3 in goldfish (Carassius auratus) and its mRNA expression in response to cadmium and PAMPs. Comp Biochem Physiol A Mol Integr Physiol 2018; 224:68-75. [PMID: 30008387 DOI: 10.1016/j.cbpa.2018.06.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 06/25/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
Fatty acid binding proteins (FABPs) are members of the conserved, multigene family of intracellular lipid binding proteins. In this study, the full-length cDNA of goldfish (Carassius auratus) FABP-3 (gfFABP-3) was successfully cloned. gfFABP-3 had an open reading frame of 402 bp and encoded a 133 amino acid polypeptide. The predicted gfFABP-3 protein included a lipocalin domain and displayed typical conserved FABP tertiary structures. Reverse transcription-PCR (RT-PCR) revealed that the gfFABP-3 gene was expressed in all tested tissues, with higher levels of expression in the testis, liver, heart, fat and kidney. After 24 h of cadmium exposure, gfFABP-3 was significantly upregulated in the gill, liver and spleen, but downregulated in the intestine, as compared to unexposed controls. gfFABP-3 expression was significantly downregulated in the spleen in goldfish challenged with LPS and Poly I:C. Our study provides a molecular characterization of goldfish FABP-3 and indicated that gfFABP-3 was potentially associated with the toxic effects of cadmium on lipid metabolism, and with the immune response to pathogenic infection.
Collapse
Affiliation(s)
- Wenbo Chen
- Department of Biology, Institute of Resources and Environment, Henan Polytechnic University, Jiaozuo 454000, Henan, China.
| | - Kaimeng Wang
- Department of Biology, Institute of Resources and Environment, Henan Polytechnic University, Jiaozuo 454000, Henan, China
| | - Shiyu Liu
- Department of Biology, Institute of Resources and Environment, Henan Polytechnic University, Jiaozuo 454000, Henan, China
| |
Collapse
|
10
|
Liang T, Jinglong X, Shusheng D, Aiyou W. Maternal obesity stimulates lipotoxicity and up-regulates inflammatory signaling pathways in the full-term swine placenta. Anim Sci J 2018; 89:1310-1322. [PMID: 29947166 DOI: 10.1111/asj.13064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/22/2018] [Indexed: 01/03/2023]
Abstract
This study aimed to investigate the effects of back-fat thickness (BF), at mating of sows, on placental lipotoxicity, oxidative stress, and inflammation. We performed iTRAQ labeling-based proteomic analysis on term placentas obtained by vaginal delivery from BFI (15-20 mm, control) and BFII (21-27 mm, obese) sows formed according to BF at mating. Proteomic analysis revealed 413 proteins to be significantly different in placenta from BFII sows by ≥1.2-fold. Gene ontology (GO) analysis identified proteins related to lipid metabolism and inflammatory response to be altered in placenta from obese sows. Indicative of a lipotoxic placental environment, increased placental lipid, and up-regulated mRNA expression of lipogenic genes, including ADRP (p = .06), PPARD, FASN, ACACA, DGAT1, and LIPIN3, were associated with decreased AMPK and increased activation of WNT signaling in placenta from BFII group (p < .05). Furthermore, we observed a 18% decrease in total antioxidant capacity (TAC), increased mRNA content of pro-inflammatory cytokines IL-6, IL-18, and TNF-α, and increased activation of inflammatory NF-κB and JNK signaling in placenta from BFII sows that was significantly associated with macrophage accumulation (p < .05). These findings suggest that maternal obesity aggravates a lipotoxic environment in pig term placenta that may be associated with placental dysfunction and impaired fetal growth.
Collapse
Affiliation(s)
- Tian Liang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xie Jinglong
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Dong Shusheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wen Aiyou
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
| |
Collapse
|
11
|
George B, Joy MS, Aleksunes LM. Urinary protein biomarkers of kidney injury in patients receiving cisplatin chemotherapy. Exp Biol Med (Maywood) 2017; 243:272-282. [PMID: 29231123 DOI: 10.1177/1535370217745302] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite recent progress in the development of novel approaches to treat cancer, traditional antineoplastic drugs, such as cisplatin, remain a mainstay of regimens targeting solid tumors. Use of cisplatin is limited by acute kidney injury, which occurs in approximately 30% of patients. Current clinical measures, such as serum creatinine and estimated glomerular filtration rate, are inadequate in their ability to detect acute kidney injury, particularly when there is only a moderate degree of injury. Thus, there is an urgent need for improved diagnostic biomarkers to predict nephrotoxicity. There is also interest by the U.S. Food and Drug Administration to validate and implement new biomarkers to identify clinical and subclinical acute kidney injury in patients during the drug approval process. This minireview provides an overview of the current literature regarding the utility of urinary proteins (albumin, beta-2-microglobulin, N-acetyl-D-glucosaminidase, kidney injury molecule-1, neutrophil gelatinase-associated lipocalin, and cystatin C) as biomarkers for cisplatin-induced AKI. Many of the well-studied urinary proteins (KIM-1, NGAL, B2M, albumin) as well as emerging biomarkers (calbindin, monocyte chemotactic protein-1, and trefoil factor 3) display distinct patterns of time-dependent excretion after cisplatin administration. Implementation of these biomarker proteins in the oncology clinic has been hampered by a lack of validation studies. To address these issues, large head-to-head studies are needed to fully characterize time-dependent responses and establish accurate cutoff values and ranges, particularly in cancer patients. Impact statement There is growing interest in using urinary protein biomarkers to detect acute kidney injury in oncology patients prescribed the nephrotoxic anticancer drug cisplatin. We aim to synthesize and organize the existing literature on biomarkers examined clinically in patients receiving cisplatin-containing chemotherapy regimens. This minireview highlights several proteins (kidney injury molecule-1, beta-2-microglobulin, neutrophil gelatinase-associated lipocalin, calbindin, monocyte chemotactic protein-1, trefoil factor 3) with the greatest promise for detecting cisplatin-induced acute kidney injury in humans. A comprehensive review of the existing literature may aid in the design of larger studies needed to implement the clinical use of these urinary proteins as biomarkers of kidney injury.
Collapse
Affiliation(s)
- Blessy George
- 1 Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Ernest Mario School of Pharmacy, Piscataway, NJ 08854, USA
| | - Melanie S Joy
- 2 Department of Pharmaceutical Sciences, 129263 University of Colorado at Denver - Anschutz Medical Campus , Skaggs School of Pharmacy and Pharmaceutical Sciences, 129263 University of Colorado at Denver - Anschutz Medical Campus , University of Colorado, Aurora, CO 80045, USA
| | - Lauren M Aleksunes
- 1 Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Ernest Mario School of Pharmacy, Piscataway, NJ 08854, USA.,3 Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,4 Lipid Center, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
12
|
Hisamichi M, Kamijo-Ikemori A, Sugaya T, Hoshino S, Kimura K, Shibagaki Y. Role of bardoxolone methyl, a nuclear factor erythroid 2-related factor 2 activator, in aldosterone- and salt-induced renal injury. Hypertens Res 2017; 41:8-17. [PMID: 28978980 DOI: 10.1038/hr.2017.83] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/06/2017] [Accepted: 07/31/2017] [Indexed: 01/09/2023]
Abstract
The aim of this study was to investigate the renoprotective effect of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator with an antioxidant effect, in a salt-sensitive hypertension model induced by aldosterone (Ald) and salt. Tubulointerstitial damage with urinary liver-type fatty acid-binding protein (L-FABP) was evaluated using human L-FABP chromosomal transgenic (L-FABP+/-) male mice. The mice in the Ald group (n=7) received systemic Ald infusions via an osmotic minipump and were given 1% NaCl water for 35 days. Those in the Ald-BM group (n=8) were administered BM intraperitoneally in addition to an injection of Ald and salt. The dose of BM was gradually increased every 7 days up to 10 mg kg-1 per day, which was maintained for 14 days. The administration of BM significantly increased renal expression of the Nrf2 target antioxidant gene. Tubulointerstitial damage was significantly ameliorated in the Ald-BM group compared to the Ald group. The increase in reactive oxygen species (ROS) and upregulation of angiotensinogen expression in the kidneys of the Ald group was significantly prevented in the Ald-BM group. The upregulation of human L-FABP expression induced in the kidneys and increase in urinary L-FABP in the Ald group were significantly suppressed by BM administration. In conclusion, BM ameliorated tubulointerstitial damage in the Ald- and salt-induced hypertension model through suppression of both ROS production and intrarenal renin-angiotensin system activation. Urinary L-FABP may be a useful marker reflecting the therapeutic efficacy of BM.
Collapse
Affiliation(s)
| | - Atsuko Kamijo-Ikemori
- Department of Anatomy, Kanagawa, Japan.,Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | | |
Collapse
|
13
|
Mukai T, Egawa M, Takeuchi T, Yamashita H, Kusudo T. Silencing of FABP1 ameliorates hepatic steatosis, inflammation, and oxidative stress in mice with nonalcoholic fatty liver disease. FEBS Open Bio 2017; 7:1009-1016. [PMID: 28680813 PMCID: PMC5494302 DOI: 10.1002/2211-5463.12240] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/22/2017] [Accepted: 05/02/2017] [Indexed: 12/21/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasing in prevalence worldwide and has been identified as a risk factor for cirrhosis and hepatocellular carcinoma. However, there is no effective pharmacologic treatment for NAFLD. FABP1 is a liver‐specific fatty acid‐binding protein (FABP) that plays important roles in intracellular lipid metabolism in the liver. We investigated the effect of repression of FABP1 expression on NAFLD, using adenovirus‐mediated silencing of FABP1. FABP1 knockdown in the liver decreased the liver weight and hepatic triglyceride (TG) accumulation. The expression of inflammatory and oxidative stress markers in the liver was also reduced. The level of thiobarbituric acid‐reactive substances, a marker of lipid peroxidation, in the liver of FABP1 knockdown mice was significantly decreased. These results suggest that FABP1 reduction in the liver is an effective approach against NAFLD.
Collapse
Affiliation(s)
- Takako Mukai
- Faculty of Human Sciences Tezukayama Gakuin University Sakai Japan
| | - Miki Egawa
- Department of Biomedical Sciences College of Life and Health Sciences Chubu University Kasugai Japan
| | - Tamaki Takeuchi
- Department of Biomedical Sciences College of Life and Health Sciences Chubu University Kasugai Japan
| | - Hitoshi Yamashita
- Department of Biomedical Sciences College of Life and Health Sciences Chubu University Kasugai Japan
| | - Tatsuya Kusudo
- Faculty of Human Sciences Tezukayama Gakuin University Sakai Japan.,Department of Biomedical Sciences College of Life and Health Sciences Chubu University Kasugai Japan
| |
Collapse
|
14
|
Zou Y, Zhang Y, Han L, He Q, Hou H, Han J, Wang X, Li C, Cen J, Liu K. Oxidative stress-mediated developmental toxicity induced by isoniazide in zebrafish embryos and larvae. J Appl Toxicol 2017; 37:842-852. [DOI: 10.1002/jat.3432] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/22/2016] [Accepted: 12/06/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Yu Zou
- Key Laboratory of Natural Medicine and Immuno-Engineering; Henan University; Kaifeng 475004 Henan Province People's Republic of China
- Biology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| | - Yun Zhang
- Biology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| | - Liwen Han
- Biology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| | - Qiuxia He
- Biology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| | - Hairong Hou
- Biology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
- Shandong Provincial Engineering Laboratory for Biological Testing Technology; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| | - Jian Han
- Biology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
- Key Laboratory for Biosensor of Shandong Province; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| | - Ximin Wang
- Biology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
- Shandong Provincial Engineering Laboratory for Biological Testing Technology; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
- Key Laboratory for Biosensor of Shandong Province; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| | - Chengyun Li
- Ecology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| | - Juan Cen
- Key Laboratory of Natural Medicine and Immuno-Engineering; Henan University; Kaifeng 475004 Henan Province People's Republic of China
| | - Kechun Liu
- Biology Institute of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
- Key Laboratory for Biosensor of Shandong Province; 19 Keyuan Road, Lixia District Jinan 250014 Shandong Province People's Republic of China
| |
Collapse
|
15
|
Liver fatty acid-binding protein (L-FABP) promotes cellular angiogenesis and migration in hepatocellular carcinoma. Oncotarget 2017; 7:18229-46. [PMID: 26919097 PMCID: PMC4951284 DOI: 10.18632/oncotarget.7571] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 01/29/2016] [Indexed: 01/18/2023] Open
Abstract
Liver fatty acid-binding protein (L-FABP) is abundant in hepatocytes and known to be involved in lipid metabolism. Overexpression of L-FABP has been reported in various cancers; however, its role in hepatocellular carcinoma (HCC) remains unclear. In this study, we investigated L-FABP and its association with vascular endothelial growth factors (VEGFs) in 90 HCC patients. We found that L-FABP was highly expressed in their HCC tissues, and that this expression was positively correlated with that of VEGF-A. Additionally, L-FABP significantly promoted tumor growth and metastasis in a xenograft mouse model. We also assessed the mechanisms of L-FABP activity in tumorigenesis; L-FABP was found to associate with VEGFR2 on membrane rafts and subsequently activate the Akt/mTOR/P70S6K/4EBP1 and Src/FAK/cdc42 pathways, which resulted in up-regulation of VEGF-A accompanied by an increase in both angiogenic potential and migration activity. Our results thus suggest that L-FABP could be a potential target for HCC chemotherapy.
Collapse
|
16
|
Wood SM, Gill AJ, Brodsky AS, Lu S, Friedman K, Karashchuk G, Lombardo K, Yang D, Resnick MB. Fatty acid-binding protein 1 is preferentially lost in microsatellite instable colorectal carcinomas and is immune modulated via the interferon γ pathway. Mod Pathol 2017; 30:123-133. [PMID: 27687006 PMCID: PMC5218856 DOI: 10.1038/modpathol.2016.170] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/22/2016] [Accepted: 08/23/2016] [Indexed: 02/07/2023]
Abstract
Fatty acid-binding protein 1 (FABP1) is an intracellular protein responsible for the transportation of long chain fatty acids. Aside from its functions in lipid metabolism and cellular differentiation, FABP1 also plays a role in inflammation through its interaction with peroxisome proliferator-activated receptors (PPARs). Previously, we compared expression of colonic epithelium genes in a subset of microsatellite instable (MSI) colorectal carcinomas (medullary carcinomas) to normal colonic mucosa and found that FABP1 expression was markedly decreased in the tumors. Further analysis of RNA expression in the colorectal subtypes and The Cancer Genome Atlas data set found that FABP1 expression is decreased in the CMS1 subset of colorectal carcinomas, which is characterized by microsatellite instability. As MSI colorectal carcinomas are known for their robust immune response, we then aimed to link FABP1 to the immune microenvironment of MSI carcinomas. To confirm the gene expression results, we performed immunohistochemical analysis of a cohort of colorectal carcinomas. FABP1 was preferentially lost in MSI carcinomas (123/133, 93%) compared with microsatellite stable carcinomas (240/562, 43%, P<0.0001). In addition, higher numbers of tumor-infiltrating lymphocytes were present in tumors with loss of FABP1 (P<0.0001). Decreased expression of the fatty acid storage and glucose regulator, PPARγ, was associated with the loss of FABP1 (P<0.0001). Colorectal cancer cell lines treated with interferon γ exhibited decreased expression of FABP1. FABP1 expression was partially recovered with the treatment of the cell lines with rosiglitazone, a PPARγ agonist. This study demonstrated that the loss of FABP1 expression is associated with MSI carcinomas and that interferon γ stimulation plays a role in this process via its interaction with PPARγ.
Collapse
Affiliation(s)
- Stephanie M Wood
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Anthony J Gill
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, Sydney NSW 2006 Australia
| | - Alexander S Brodsky
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Kenneth Friedman
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Galina Karashchuk
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Kara Lombardo
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Dongfang Yang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Murray B Resnick
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
17
|
Schroeder F, McIntosh AL, Martin GG, Huang H, Landrock D, Chung S, Landrock KK, Dangott LJ, Li S, Kaczocha M, Murphy EJ, Atshaves BP, Kier AB. Fatty Acid Binding Protein-1 (FABP1) and the Human FABP1 T94A Variant: Roles in the Endocannabinoid System and Dyslipidemias. Lipids 2016; 51:655-76. [PMID: 27117865 PMCID: PMC5408584 DOI: 10.1007/s11745-016-4155-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/11/2016] [Indexed: 01/01/2023]
Abstract
The first discovered member of the mammalian FABP family, liver fatty acid binding protein (FABP1, L-FABP), occurs at high cytosolic concentration in liver, intestine, and in the case of humans also in kidney. While the rat FABP1 is well studied, the extent these findings translate to human FABP1 is not clear-especially in view of recent studies showing that endocannabinoids and cannabinoids represent novel rat FABP1 ligands and FABP1 gene ablation impacts the hepatic endocannabinoid system, known to be involved in non-alcoholic fatty liver (NAFLD) development. Although not detectable in brain, FABP1 ablation nevertheless also impacts brain endocannabinoids. Despite overall tertiary structure similarity, human FABP1 differs significantly from rat FABP1 in secondary structure, much larger ligand binding cavity, and affinities/specificities for some ligands. Moreover, while both mouse and human FABP1 mediate ligand induction of peroxisome proliferator activated receptor-α (PPARα), they differ markedly in pattern of genes induced. This is critically important because a highly prevalent human single nucleotide polymorphism (SNP) (26-38 % minor allele frequency and 8.3 ± 1.9 % homozygous) results in a FABP1 T94A substitution that further accentuates these species differences. The human FABP1 T94A variant is associated with altered body mass index (BMI), clinical dyslipidemias (elevated plasma triglycerides and LDL cholesterol), atherothrombotic cerebral infarction, and non-alcoholic fatty liver disease (NAFLD). Resolving human FABP1 and the T94A variant's impact on the endocannabinoid and cannabinoid system is an exciting challenge due to the importance of this system in hepatic lipid accumulation as well as behavior, pain, inflammation, and satiety.
Collapse
Affiliation(s)
- Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA.
| | - Avery L McIntosh
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Huan Huang
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Sarah Chung
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Kerstin K Landrock
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Lawrence J Dangott
- Department of Biochemistry and Biophysics, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Shengrong Li
- Avanti Polar Lipids, 700 Industrial Park Dr., Alabaster, AL, 35007-9105, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Eric J Murphy
- Department of Pharmacology, Physiology, and Therapeutics and Chemistry, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Barbara P Atshaves
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| |
Collapse
|
18
|
Adil M, Kandhare AD, Ghosh P, Venkata S, Raygude KS, Bodhankar SL. Ameliorative effect of naringin in acetaminophen-induced hepatic and renal toxicity in laboratory rats: role of FXR and KIM-1. Ren Fail 2016; 38:1007-20. [DOI: 10.3109/0886022x.2016.1163998] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
19
|
Chen X, Li R, Geng Z. Cold stress initiates the Nrf2/UGT1A1/L-FABP signaling pathway in chickens. Poult Sci 2015; 94:2597-603. [DOI: 10.3382/ps/pev253] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2015] [Indexed: 12/15/2022] Open
|
20
|
Wang G, Bonkovsky HL, de Lemos A, Burczynski FJ. Recent insights into the biological functions of liver fatty acid binding protein 1. J Lipid Res 2015; 56:2238-47. [PMID: 26443794 DOI: 10.1194/jlr.r056705] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Indexed: 12/18/2022] Open
Abstract
Over four decades have passed since liver fatty acid binding protein (FABP)1 was first isolated. There are few protein families for which most of the complete tertiary structures, binding properties, and tissue occurrences are described in such detail and yet new functions are being uncovered for this protein. FABP1 is known to be critical for fatty acid uptake and intracellular transport and also has an important role in regulating lipid metabolism and cellular signaling pathways. FABP1 is an important endogenous cytoprotectant, minimizing hepatocyte oxidative damage and interfering with ischemia-reperfusion and other hepatic injuries. The protein may be targeted for metabolic activation through the cross-talk among many transcriptional factors and their activating ligands. Deficiency or malfunction of FABP1 has been reported in several diseases. FABP1 also influences cell proliferation during liver regeneration and may be considered as a prognostic factor for hepatic surgery. FABP1 binds and modulates the action of many molecules such as fatty acids, heme, and other metalloporphyrins. The ability to bind heme is another cytoprotective property and one that deserves closer investigation. The role of FABP1 in substrate availability and in protection from oxidative stress suggests that FABP1 plays a pivotal role during intracellular bacterial/viral infections by reducing inflammation and the adverse effects of starvation (energy deficiency).
Collapse
Affiliation(s)
- GuQi Wang
- Jiangxi Normal University, Nanchang, Jiangxi, People's Republic of China Department of Biology, University of North Carolina at Charlotte, Charlotte, NC Carolinas HealthCare System, Charlotte, NC
| | - Herbert L Bonkovsky
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC Carolinas HealthCare System, Charlotte, NC Wake Forest Baptist Medical Center, Winston-Salem, NC
| | - Andrew de Lemos
- Carolinas HealthCare System, Charlotte, NC Wake Forest Baptist Medical Center, Winston-Salem, NC
| | | |
Collapse
|
21
|
Martin GG, Atshaves BP, Landrock KK, Landrock D, Schroeder F, Kier AB. Loss of L-FABP, SCP-2/SCP-x, or both induces hepatic lipid accumulation in female mice. Arch Biochem Biophys 2015; 580:41-9. [PMID: 26116377 DOI: 10.1016/j.abb.2015.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 06/08/2015] [Accepted: 06/17/2015] [Indexed: 02/06/2023]
Abstract
Although roles for both sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) and liver fatty acid binding protein (L-FABP) have been proposed in hepatic lipid accumulation, individually ablating these genes has been complicated by concomitant alterations in the other gene product(s). For example, ablating SCP2/SCP-x induces upregulation of L-FABP in female mice. Therefore, the impact of ablating SCP-2/SCP-x (DKO) or L-FABP (LKO) individually or both together (TKO) was examined in female mice. Loss of SCP-2/SCP-x (DKO, TKO) more so than loss of L-FABP alone (LKO) increased hepatic total lipid and total cholesterol content, especially cholesteryl ester. Hepatic accumulation of nonesterified long chain fatty acids (LCFA) and phospholipids occurred only in DKO and TKO mice. Loss of SCP-2/SCP-x (DKO, TKO) increased serum total lipid primarily by increasing triglycerides. Altered hepatic level of proteins involved in cholesterol uptake, efflux, and/or secretion was observed, but did not compensate for the loss of L-FABP, SCP-2/SCP-x or both. However, synergistic responses were not seen with the combinatorial knock out animals-suggesting that inhibiting SCP-2/SCP-x is more correlative with hepatic dysfunction than L-FABP. The DKO- and TKO-induced hepatic accumulation of cholesterol and long chain fatty acids shared significant phenotypic similarities with non-alcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, United States
| | - Barbara P Atshaves
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States
| | - Kerstin K Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, United States
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, United States
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, United States
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, United States.
| |
Collapse
|
22
|
Baek JH, Zhang X, Williams MC, Hicks W, Buehler PW, D'Agnillo F. Sodium nitrite potentiates renal oxidative stress and injury in hemoglobin exposed guinea pigs. Toxicology 2015; 333:89-99. [PMID: 25891524 DOI: 10.1016/j.tox.2015.04.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/17/2015] [Accepted: 04/14/2015] [Indexed: 12/18/2022]
Abstract
Methemoglobin-forming drugs, such as sodium nitrite (NaNO2), may exacerbate oxidative toxicity under certain chronic or acute hemolytic settings. In this study, we evaluated markers of renal oxidative stress and injury in guinea pigs exposed to extracellular hemoglobin (Hb) followed by NaNO2 at doses sufficient to simulate clinically relevant acute methemoglobinemia. NaNO2 induced rapid and extensive oxidation of plasma Hb in this model. This was accompanied by increased renal expression of the oxidative response effectors nuclear factor erythroid 2-derived-factor 2 (Nrf-2) and heme oxygenase-1 (HO-1), elevated non-heme iron deposition, lipid peroxidation, interstitial inflammatory cell activation, increased expression of tubular injury markers kidney injury-1 marker (KIM-1) and liver-fatty acid binding protein (L-FABP), podocyte injury, and cell death. Importantly, these indicators of renal oxidative stress and injury were minimal or absent following infusion of Hb or NaNO2 alone. Together, these results suggest that the exposure to NaNO2 in settings associated with increased extracellular Hb may potentiate acute renal toxicity via processes that are independent of NaNO2 induced erythrocyte methemoglobinemia.
Collapse
Affiliation(s)
- Jin Hyen Baek
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Xiaoyuan Zhang
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Matthew C Williams
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Wayne Hicks
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Paul W Buehler
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Felice D'Agnillo
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA.
| |
Collapse
|
23
|
Tan SJ, Zhang X, Jin XK, Li WW, Li JY, Wang Q. Fatty acid binding protein FABP3 from Chinese mitten crab Eriocheir sinensis participates in antimicrobial responses. FISH & SHELLFISH IMMUNOLOGY 2015; 43:264-274. [PMID: 25559445 DOI: 10.1016/j.fsi.2014.12.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 06/04/2023]
Abstract
Intracellular fatty acid-binding proteins (FABPs) are members of the lipid-binding protein superfamily. Aside from the main functions of FABPs in the uptake and transport of fatty acids, they are also critical in innate immunology. In this work, the full-length cDNA for a Chinese mitten crab Eriocheir sinensis FABP (Es-FABP3) was cloned with an open reading frame of 402 bp encoding a 133 amino acid polypeptide. Analysis using quantitative real-time PCR (qPCR) revealed that Es-FABP3 transcripts were widely distributed in gills, muscle, intestine, hepatopancreas, eyestalk, heart, stomach, brain, thoracic ganglia and hemocytes. After challenge with pathogen associated molecular pattern molecules (PAMPs), the relative mRNA expression levels of Es-FABP3 increased in hepatopancreas, gills and hemocytes. Moreover, the mature recombinant Es-FABP3 protein exhibited different binding activities to bacteria and fungus and inhibited the growth of different microbes. These collective results demonstrated the role of Es-FABP3 in the immunoreactions of E. sinensis to PAMPs.
Collapse
Affiliation(s)
- Shang-Jian Tan
- School of Life Science, East China Normal University, Shanghai, China
| | - Xing Zhang
- School of Life Science, East China Normal University, Shanghai, China
| | - Xing-Kun Jin
- School of Life Science, East China Normal University, Shanghai, China
| | - Wei-Wei Li
- School of Life Science, East China Normal University, Shanghai, China
| | - Jia-Yao Li
- Aquatic Animal Breeding Center of Shanghai University Knowledge Service Platform, Shanghai Ocean University, Shanghai, China.
| | - Qun Wang
- School of Life Science, East China Normal University, Shanghai, China.
| |
Collapse
|
24
|
Huang H, McIntosh AL, Landrock KK, Landrock D, Storey SM, Martin GG, Gupta S, Atshaves BP, Kier AB, Schroeder F. Human FABP1 T94A variant enhances cholesterol uptake. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:946-55. [PMID: 25732850 DOI: 10.1016/j.bbalip.2015.02.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 02/19/2015] [Accepted: 02/20/2015] [Indexed: 01/09/2023]
Abstract
Although expression of the human liver fatty acid binding protein (FABP1) T94A variant alters serum lipoprotein cholesterol levels in human subjects, nothing is known whereby the variant elicits these effects. This issue was addressed by in vitro cholesterol binding assays using purified recombinant wild-type (WT) FABP1 T94T and T94A variant proteins and in cultured primary human hepatocytes expressing the FABP1 T94T (genotyped as TT) or T94A (genotyped as CC) proteins. The human FABP1 T94A variant protein had 3-fold higher cholesterol-binding affinity than the WT FABP1 T94T as shown by NBD-cholesterol fluorescence binding assays and by cholesterol isothermal titration microcalorimetry (ITC) binding assays. CC variant hepatocytes also exhibited 30% higher total FABP1 protein. HDL- and LDL-mediated NBD-cholesterol uptake was faster in CC variant than TT WT human hepatocytes. VLDL-mediated uptake of NBD-cholesterol did not differ between CC and TT human hepatocytes. The increased HDL- and LDL-mediated NBD-cholesterol uptake was not associated with any significant change in mRNA levels of SCARB1, LDLR, CETP, and LCAT encoding the key proteins in lipoprotein cholesterol uptake. Thus, the increased HDL- and LDL-mediated NBD-cholesterol uptake by CC hepatocytes may be associated with higher affinity of T94A protein for cholesterol and/or increased total T94A protein level.
Collapse
Affiliation(s)
- Huan Huang
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX 77843-4466, USA
| | - Avery L McIntosh
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX 77843-4466, USA
| | - Kerstin K Landrock
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX 77843-4467, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX 77843-4467, USA
| | - Stephen M Storey
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX 77843-4466, USA
| | - Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX 77843-4466, USA
| | - Shipra Gupta
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Barbara P Atshaves
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX 77843-4467, USA
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX 77843-4466, USA.
| |
Collapse
|
25
|
Ichikawa D, Kamijo-Ikemori A, Sugaya T, Shibagaki Y, Yasuda T, Hoshino S, Katayama K, Igarashi-Migitaka J, Hirata K, Kimura K. Human liver-type fatty acid–binding protein protects against tubulointerstitial injury in aldosterone-induced renal injury. Am J Physiol Renal Physiol 2015; 308:F114-21. [DOI: 10.1152/ajprenal.00469.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
To demonstrate the renoprotective function of human liver-type fatty acid–binding protein (hL-FABP) expressed in proximal tubules in aldosterone (Aldo)-induced renal injury, hL-FABP chromosomal transgenic (Tg) and wild-type (WT) mice received systemic Aldo infusions (Tg-Aldo and WT-Aldo, respectively) were given 1% NaCl water for 28 days. In this model, elevation of systolic blood pressure, monocyte chemoattractant protein-1 expression, macrophage infiltration in the interstitium, tubulointerstitial damage, and depositions of type I and III collagens were observed. Elevation of systolic blood pressure did not differ in WT-Aldo vs. Tg-Aldo animals, however, renal injury was suppressed in Tg-Aldo compared with WT-Aldo mice. Dihydroethidium fluorescence was used to evaluate reactive oxidative stress, which was suppressed in Tg-Aldo compared with WT-Aldo mice. Gene expression of angiotensinogen in the kidney was upregulated, and excretion of urinary angiotensinogen was increased in WT-Aldo mice. This exacerbation was suppressed in Tg-Aldo mice. Expression of hL-FABP was upregulated in proximal tubules of Tg-Aldo mice. Urinary excretion of hL-FABP was significantly greater in Tg-Aldo than in Tg-control mice. In conclusion, hL-FABP ameliorated the tubulointerstitial damage in Aldo-induced renal injury via reducing oxidative stress and suppressing activation of the intrarenal renin-angiotensin system.
Collapse
Affiliation(s)
- Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Takashi Yasuda
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kimie Katayama
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | | | - Kazuaki Hirata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kenjiro Kimura
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| |
Collapse
|
26
|
Martin GG, Atshaves BP, Landrock KK, Landrock D, Storey SM, Howles PN, Kier AB, Schroeder F. Ablating L-FABP in SCP-2/SCP-x null mice impairs bile acid metabolism and biliary HDL-cholesterol secretion. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1130-43. [PMID: 25277800 PMCID: PMC4254959 DOI: 10.1152/ajpgi.00209.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/28/2014] [Indexed: 01/31/2023]
Abstract
On the basis of their abilities to bind bile acids and/or cholesterol, the physiological role(s) of liver fatty acid-binding protein (L-FABP) and sterol carrier protein (SCP) 2/SCP-x (SCP-2/SCP-x) gene products in biliary bile acid and cholesterol formation was examined in gene-ablated male mice. L-FABP (LKO) or L-FABP/SCP-2/SCP-x [triple-knockout (TKO)] ablation markedly decreased hepatic bile acid concentration, while SCP-2/SCP-x [double-knockout (DKO)] ablation alone had no effect. In contrast, LKO increased biliary bile acid, while DKO and TKO had no effect on biliary bile acid levels. LKO and DKO also altered biliary bile acid composition to increase bile acid hydrophobicity. Furthermore, LKO and TKO decreased hepatic uptake and biliary secretion of high-density lipoprotein (HDL)-derived 22-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-23,24-bisnor-5-cholen-3β-ol (NBD-cholesterol), while DKO alone had no effect. Finally, LKO and, to a lesser extent, DKO decreased most indexes contributing to cholesterol solubility in biliary bile. These results suggest different, but complementary, roles for L-FABP and SCP-2/SCP-x in biliary bile acid and cholesterol formation. L-FABP appears to function more in hepatic retention of bile acids as well as hepatic uptake and biliary secretion of HDL-cholesterol. Conversely, SCP-2/SCP-x may function more in formation and biliary secretion of bile acid, with less impact on hepatic uptake or biliary secretion of HDL-cholesterol.
Collapse
Affiliation(s)
- Gregory G Martin
- Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas
| | - Barbara P Atshaves
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Kerstin K Landrock
- Department of Pathobiology, Texas A & M University, College Station, Texas; and
| | - Danilo Landrock
- Department of Pathobiology, Texas A & M University, College Station, Texas; and
| | - Stephen M Storey
- Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas
| | - Philip N Howles
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Ann B Kier
- Department of Pathobiology, Texas A & M University, College Station, Texas; and
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas;
| |
Collapse
|
27
|
Liu JW, Montero M, Bu L, De Leon M. Epidermal fatty acid-binding protein protects nerve growth factor-differentiated PC12 cells from lipotoxic injury. J Neurochem 2014; 132:85-98. [PMID: 25147052 PMCID: PMC4270845 DOI: 10.1111/jnc.12934] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 08/01/2014] [Accepted: 08/13/2014] [Indexed: 12/11/2022]
Abstract
Epidermal fatty acid-binding protein (E-FABP/FABP5/DA11) binds and transport long-chain fatty acids in the cytoplasm and may play a protecting role during neuronal injury. We examined whether E-FABP protects nerve growth factor-differentiated PC12 cells (NGFDPC12 cells) from lipotoxic injury observed after palmitic acid (C16:0; PAM) overload. NGFDPC12 cells cultures treated with PAM/bovine serum albumin at 0.3 mM/0.15 mM show PAM-induced lipotoxicity (PAM-LTx) and apoptosis. The apoptosis was preceded by a cellular accumulation of reactive oxygen species (ROS) and higher levels of E-FABP. Antioxidants MCI-186 and N-acetyl cysteine prevented E-FABP's induction in expression by PAM-LTx, while tert-butyl hydroperoxide increased ROS and E-FABP expression. Non-metabolized methyl ester of PAM, methyl palmitic acid (mPAM), failed to increase cellular ROS, E-FABP gene expression, or trigger apoptosis. Treatment of NGFDPC12 cultures with siE-FABP showed reduced E-FABP levels correlating with higher accumulation of ROS and cell death after exposure to PAM. In contrast, increasing E-FABP cellular levels by pre-loading the cells with recombinant E-FABP diminished the PAM-induced ROS and cell death. Finally, agonists for PPARβ (GW0742) or PPARγ (GW1929) increased E-FABP expression and enhanced the resistance of NGFDPC12 cells to PAM-LTx. We conclude that E-FABP protects NGFDPC12 cells from lipotoxic injury through mechanisms that involve reduction of ROS. Epidermal fatty acid-binding protein (E-FABP) may protect nerve cells from the damaging exposure to high levels of free fatty acids (FA). We show that E-FABP can neutralize the effects of reactive oxygen species (ROS) generated by the high levels of FA in the cell and protect PC12 cells from lipotoxic injuries common in Type 2 diabetes neuropathy. Potentially, E-FABP gene up-regulation may be mediated through the NFkB pathway and future studies are needed to further evaluate this proposition.
Collapse
Affiliation(s)
- Jo-Wen Liu
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California, USA
| | | | | | | |
Collapse
|
28
|
Kajimoto K, Minami Y, Harashima H. Cytoprotective role of the fatty acid binding protein 4 against oxidative and endoplasmic reticulum stress in 3T3-L1 adipocytes. FEBS Open Bio 2014; 4:602-10. [PMID: 25161868 PMCID: PMC4141204 DOI: 10.1016/j.fob.2014.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 06/20/2014] [Accepted: 06/30/2014] [Indexed: 01/27/2023] Open
Abstract
Oxidative stress in 3T3-L1 adipocytes was elevated by silencing of FABP4. FABP4 silencing did not alter levels of glutathione or superoxide dismutase. The recombinant FABP4 significantly reduced levels of hydrogen peroxide. The resistance of adipocytes to oxidative stress was decreased by FABP4 knockdown. Silencing of FABP4 elevated the endoplasmic reticulum stress in adipocytes.
The fatty acid binding protein 4 (FABP4), one of the most abundant proteins in adipocytes, has been reported to have a proinflammatory function in macrophages. However, the physiological role of FABP4, which is constitutively expressed in adipocytes, has not been fully elucidated. Previously, we demonstrated that FABP4 was involved in the regulation of interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) production in 3T3-L1 adipocytes. In this study, we examined the effects of FABP4 silencing on the oxidative and endoplasmic reticulum (ER) stress in 3T3-L1 adipocytes. We found that the cellular reactive oxygen species (ROS) and 8-nitro-cyclic GMP levels were significantly elevated in the differentiated 3T3-L1 adipocytes transfected with a small interfering RNA (siRNA) against Fabp4, although the intracellular levels or enzyme activities of antioxidants including reduced glutathione (GSH), superoxide dismutase (SOD) and glutathione S-transferase A4 (GSTA4) were not altered. An in vitro evaluation using the recombinant protein revealed that FABP4 itself functions as a scavenger protein against hydrogen peroxide (H2O2). FABP4-knockdown resulted in a significant lowering of cell viability of 3T3-L1 adipocytes against H2O2 treatment. Moreover, four kinds of markers related to the ER stress response including the endoplasmic reticulum to nucleus signaling 1 (Ern1), the signal sequence receptor α (Ssr1), the ORM1-like 3 (Ormdl3), and the spliced X-box binding protein 1 (Xbp1s), were all elevated as the result of the knockdown of FABP4. Consequently, FABP4 might have a new role as an antioxidant protein against H2O2 and contribute to cytoprotection against oxidative and ER stress in adipocytes.
Collapse
Key Words
- Adipocyte
- Antioxidant
- ER stress
- ER, endoplasmic reticulum
- Ern1, endoplasmic reticulum to nucleus signaling 1
- FABP, fatty acid binding protein
- FABP4
- GSH, reduced glutathione
- GSTA4, glutathione S-transferase A4
- H2O2, hydrogen peroxide
- Ormdl3, ORM1-like 3
- Oxidative stress
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- Ssr1, signal sequence receptor α
- UPR, unfolded protein response
- VEGF, vascular endothelial growth factor
- Xbp1, X-box binding protein 1.
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Kazuaki Kajimoto
- Corresponding author. Tel.: +81 11 706 2197; fax: +81 11 706 4879.
| | | | | |
Collapse
|
29
|
Huang H, McIntosh AL, Martin GG, Landrock KK, Landrock D, Gupta S, Atshaves BP, Kier AB, Schroeder F. Structural and functional interaction of fatty acids with human liver fatty acid-binding protein (L-FABP) T94A variant. FEBS J 2014; 281:2266-83. [PMID: 24628888 DOI: 10.1111/febs.12780] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/17/2014] [Accepted: 03/11/2014] [Indexed: 12/13/2022]
Abstract
The human liver fatty acid-binding protein (L-FABP) T94A variant, the most common in the FABP family, has been associated with elevated liver triglyceride levels. How this amino acid substitution elicits these effects is not known. This issue was addressed using human recombinant wild-type (WT) and T94A variant L-FABP proteins as well as cultured primary human hepatocytes expressing the respective proteins (genotyped as TT, TC and CC). The T94A substitution did not alter or only slightly altered L-FABP binding affinities for saturated, monounsaturated or polyunsaturated long chain fatty acids, nor did it change the affinity for intermediates of triglyceride synthesis. Nevertheless, the T94A substitution markedly altered the secondary structural response of L-FABP induced by binding long chain fatty acids or intermediates of triglyceride synthesis. Finally, the T94A substitution markedly decreased the levels of induction of peroxisome proliferator-activated receptor α-regulated proteins such as L-FABP, fatty acid transport protein 5 and peroxisome proliferator-activated receptor α itself meditated by the polyunsaturated fatty acids eicosapentaenoic acid and docosahexaenoic acid in cultured primary human hepatocytes. Thus, although the T94A substitution did not alter the affinity of human L-FABP for long chain fatty acids, it significantly altered human L-FABP structure and stability, as well as the conformational and functional response to these ligands.
Collapse
Affiliation(s)
- Huan Huang
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ichikawa D, Kamijo-Ikemori A, Sugaya T, Shibagaki Y, Yasuda T, Katayama K, Hoshino S, Igarashi-Migitaka J, Hirata K, Kimura K. Renoprotective effect of renal liver-type fatty acid binding protein and angiotensin II type 1a receptor loss in renal injury caused by RAS activation. Am J Physiol Renal Physiol 2014; 306:F655-63. [DOI: 10.1152/ajprenal.00460.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to assess the renoprotective effect of renal human liver-type fatty acid binding protein (hL-FABP) and angiotensin II (ANG II) type 1A receptor (AT1a) loss in renal injury caused by renin-angiotensin system (RAS) activation. We established hL-FABP chromosomal transgenic mice (L-FABP+/−AT1a+/+), crossed the L-FABP+/−AT1a+/+ with AT1a knockdown homo mice (L-FABP−/−AT1a−/−), and generated L-FABP+/−AT1a hetero mice (L-FABP+/−AT1a+/−). After the back-cross of these cubs, L-FABP+/−AT1a−/− were obtained. To activate the renal RAS, wild-type mice (L-FABP−/−AT1a+/+), L-FABP+/−AT1a+/+, L-FABP−/−AT1a+/−, L-FABP+/−AT1a+/−, L-FABP−/−AT1a−/−, and L-FABP+/−AT1a−/− were administered high-dose systemic ANG II infusion plus a high-salt diet for 28 days. In the L-FABP−/−AT1a+/+, RAS activation (L-FABP−/−AT1a+/+RAS) caused hypertension and tubulointerstitial damage. In the L-FABP+/−AT1a+/+RAS, tubulointerstitial damage was significantly attenuated compared with L-FABP−/−AT1a+/+RAS. In the AT1a partial knockout (AT1a+/−) or complete knockout (AT1a−/−) mice, reduction of AT1a expression led to a significantly lower degree of renal injury compared with L-FABP−/−AT1a+/+RAS or L-FABP+/−AT1a+/+RAS mice. Renal injury in L-FABP+/−AT1a+/−RAS mice was significantly attenuated compared with L-FABP−/−AT1a+/−RAS mice. In both L-FABP−/−AT1a−/−RAS and L-FABP+/−AT1a−/−RAS mice, renal damage was rarely found. The degrees of renal hL-FABP expression and urinary hL-FABP levels increased by RAS activation and gradually decreased along with reduction of AT1a expression levels. In conclusion, in this mouse model, renal hL-FABP expression and a decrease in AT1a expression attenuated tubulointerstitial damage due to RAS activation.
Collapse
Affiliation(s)
- Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Takashi Yasuda
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Kimie Katayama
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | - Kazuaki Hirata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kenjiro Kimura
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| |
Collapse
|
31
|
Gong Y, Wang G, Gong Y, Yan J, Chen Y, Burczynski FJ. Hepatoprotective role of liver fatty acid binding protein in acetaminophen induced toxicity. BMC Gastroenterol 2014; 14:44. [PMID: 24606952 PMCID: PMC3975289 DOI: 10.1186/1471-230x-14-44] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 03/03/2014] [Indexed: 02/07/2023] Open
Abstract
Background FABP1 has been reported to possess strong antioxidant properties. Upon successful transfection of the Chang cell line, which has undetectable FABP1 mRNA levels, with human FABP1 cDNA, the Chang cells were shown to express FABP1. Using the transfected and control (normal) Chang cells and subjecting them to oxidative stress, transfected cells were reported to be associated with enhanced cell viability. This study extends those observations by investigating the effect of FABP1 on acetaminophen (AAP)-induced hepatotoxicity. We hypothesized that presence of FABP1 would enhance cell viability compared to control cells (vector transfected cells). Methods Following AAP treatment of Chang FABP1 transfected and control cells, cell viability, oxidative stress, and apoptosis were evaluated using lactate dehydrogenase (LDH) release, the fluorescent probe DCF, and Bax expression, respectively. Results FABP1 cDNA transfected cells showed greater resistance against AAP toxicity than vector transfected cells. Significantly lower LDH levels (p < 0.05) were observed as were lower DCF fluorescence intensity (p < 0.05) in FABP1 cDNA transfected cells compared to vector transfected cells. FABP1 expression also attenuated the expression of Bax following AAP induced toxicity. Conclusion FABP1 attenuated AAP-induced toxicity and may be considered a cytoprotective agent in this in vitro model of drug induced oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | | | - Frank J Burczynski
- Faculty of Pharmacy, University of Manitoba, 750 McDermot Avenue, Winnipeg, MB R3E 0T5, Canada.
| |
Collapse
|
32
|
Yamamoto Y, Hiyama S, Takase Y, Kadowaki A, Hara S. Effects of Antioxidants and Additional Emulsifiers on the Stability of Emulsified Milk Fat in the Photo/Radical Oxidation System. J Oleo Sci 2014; 63:893-901. [DOI: 10.5650/jos.ess14111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
33
|
Osaki K, Suzuki Y, Sugaya T, Tanifuji C, Nishiyama A, Horikoshi S, Tomino Y. Amelioration of Angiotensin II–Induced Salt-Sensitive Hypertension by Liver-Type Fatty Acid–Binding Protein in Proximal Tubules. Hypertension 2013; 62:712-8. [DOI: 10.1161/hypertensionaha.113.01203] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Inappropriate activation of the intrarenal renin–angiotensin system induces generation of reactive oxygen species and tubulointerstitial inflammation, which contribute to salt-sensitive hypertension (SSHT). Liver-type fatty acid–binding protein is expressed in proximal tubules in humans, but not in rodents, and may play an endogenous antioxidative role. The objective of the present study was to examine the antioxidative effect of liver-type fatty acid–binding protein on post–angiotensin II SSHT model in transgenic mice with selective overexpression of human liver-type fatty acid–binding protein in the proximal tubules. The transgenic mice showed marked protection against angiotensin II–induced SSHT. Overexpression of tubular liver-type fatty acid–binding protein prevented intrarenal T-cell infiltration and also reduced reactive oxygen species generation, intrarenal renin–angiotensin system activation, and monocyte chemotactic protein-1 expression. We also performed an in vitro study using the murine proximal tubular cell lines with or without recombinant liver-type fatty acid–binding protein and murine proximal tubular cell lines transfected with human liver-type fatty acid–binding protein, and found that gene transfection of liver-type fatty acid–binding protein and, in part, recombinant liver-type fatty acid–binding protein administration had significantly attenuated angiotensin II–induced reactive oxygen species generation and the expression of angiotensinogen and monocyte chemotactic protein-1 in murine proximal tubular cell lines. These findings indicated that liver-type fatty acid–binding protein in the proximal tubules may protect against angiotensin II–induced SSHT by attenuating activation of the intrarenal renin–angiotensin system and reducing oxidative stress and tubulointerstitial inflammation. Present data suggest that liver-type fatty acid–binding protein in the proximal tubules may be a novel therapeutic target for SSHT.
Collapse
Affiliation(s)
- Ken Osaki
- From the Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan (K.O., Y.S., C.T., S.H., Y.T.); L-FABP Department, CMIC Holdings Co., Ltd. Tokyo, Japan (T.S.); and Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan (A.N.)
| | - Yusuke Suzuki
- From the Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan (K.O., Y.S., C.T., S.H., Y.T.); L-FABP Department, CMIC Holdings Co., Ltd. Tokyo, Japan (T.S.); and Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan (A.N.)
| | - Takeshi Sugaya
- From the Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan (K.O., Y.S., C.T., S.H., Y.T.); L-FABP Department, CMIC Holdings Co., Ltd. Tokyo, Japan (T.S.); and Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan (A.N.)
| | - Chiaki Tanifuji
- From the Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan (K.O., Y.S., C.T., S.H., Y.T.); L-FABP Department, CMIC Holdings Co., Ltd. Tokyo, Japan (T.S.); and Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan (A.N.)
| | - Akira Nishiyama
- From the Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan (K.O., Y.S., C.T., S.H., Y.T.); L-FABP Department, CMIC Holdings Co., Ltd. Tokyo, Japan (T.S.); and Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan (A.N.)
| | - Satoshi Horikoshi
- From the Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan (K.O., Y.S., C.T., S.H., Y.T.); L-FABP Department, CMIC Holdings Co., Ltd. Tokyo, Japan (T.S.); and Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan (A.N.)
| | - Yasuhiko Tomino
- From the Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan (K.O., Y.S., C.T., S.H., Y.T.); L-FABP Department, CMIC Holdings Co., Ltd. Tokyo, Japan (T.S.); and Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan (A.N.)
| |
Collapse
|
34
|
Smathers RL, Galligan JJ, Shearn CT, Fritz KS, Mercer K, Ronis M, Orlicky DJ, Davidson NO, Petersen DR. Susceptibility of L-FABP-/- mice to oxidative stress in early-stage alcoholic liver. J Lipid Res 2013; 54:1335-45. [PMID: 23359610 DOI: 10.1194/jlr.m034892] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic ethanol consumption is a prominent cause of liver disease worldwide. Dysregulation of an important lipid uptake and trafficking gene, liver-fatty acid binding protein (L-FABP), may contribute to alterations in lipid homeostasis during early-stage alcoholic liver. We have reported the detrimental effects of ethanol on the expression of L-FABP and hypothesize this may deleteriously impact metabolic networks regulating fatty acids. Male wild-type (WT) and L-FABP(-/-) mice were fed a modified Lieber-DeCarli liquid diet for six weeks. To assess the response to chronic ethanol ingestion, standard biochemical indicators for alcoholic liver disease (ALD) and oxidative stress were measured. Ethanol ingestion resulted in attenuation of hepatic triglyceride accumulation and elevation of cholesterol in L-FABP(-/-) mice. Lipidomics analysis validated multiple alterations in hepatic lipids resulting from ethanol treatment. Increased immunohistochemical staining for the reactive aldehydes 4-hydroxynonenal and malondialdehyde were observed in WT mice ingesting ethanol; however, L-FABP(-/-) mice displayed prominent protein adducts in liver sections evaluated from pair-fed and ethanol-fed mice. Likewise, alterations in glutathione, thiobarbituric acid reactive substances (TBARS), 8-isoprostanes, and protein carbonyl content all indicated L-FABP(-/-) mice exhibit high sustained oxidative stress in the liver. These data establish that L-FABP is an indirect antioxidant protein essential for sequestering FFA and that its impairment could contribute to in the pathogenesis of ALD.
Collapse
Affiliation(s)
- Rebecca L Smathers
- Skaggs School of Pharmaceutical Sciences and Pharmacy and University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Comparative study of the fatty acid binding process of a new FABP from Cherax quadricarinatus by fluorescence intensity, lifetime and anisotropy. PLoS One 2012; 7:e51079. [PMID: 23284658 PMCID: PMC3528769 DOI: 10.1371/journal.pone.0051079] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 10/29/2012] [Indexed: 12/31/2022] Open
Abstract
Fatty acid-binding proteins (FABPs) are small cytosolic proteins, largely distributed in invertebrates and vertebrates, which accomplish uptake and intracellular transport of hydrophobic ligands such as fatty acids. Although long chain fatty acids play multiple crucial roles in cellular functions (structural, energy metabolism, regulation of gene expression), the precise functions of FABPs, especially those of invertebrate species, remain elusive. Here, we have identified and characterized a novel FABP family member, Cq-FABP, from the hepatopancreas of red claw crayfish Cherax quadricarinatus. We report the characterization of fatty acid-binding affinity of Cq-FABP by four different competitive fluorescence-based assays. In the two first approaches, the fluorescent probe 8-Anilino-1-naphthalenesulfonate (ANS), a binder of internal cavities of protein, was used either by directly monitoring its fluorescence emission or by monitoring the fluorescence resonance energy transfer occurring between the single tryptophan residue of Cq-FABP and ANS. The third and the fourth approaches were based on the measurement of the fluorescence emission intensity of the naturally fluorescent cis-parinaric acid probe or the steady-state fluorescence anisotropy measurements of a fluorescently labeled fatty acid (BODIPY-C16), respectively. The four methodologies displayed consistent equilibrium constants for a given fatty acid but were not equivalent in terms of analysis. Indeed, the two first methods were complicated by the existence of non specific binding modes of ANS while BODIPY-C16 and cis-parinaric acid specifically targeted the fatty acid binding site. We found a relationship between the affinity and the length of the carbon chain, with the highest affinity obtained for the shortest fatty acid, suggesting that steric effects primarily influence the interaction of fatty acids in the binding cavity of Cq-FABP. Moreover, our results show that the binding affinities of several fatty acids closely parallel their prevalences in the hepatopancreas of C. quadricarinatus as measured under specific diet conditions.
Collapse
|
36
|
Storey SM, McIntosh AL, Huang H, Martin GG, Landrock KK, Landrock D, Payne HR, Kier AB, Schroeder F. Loss of intracellular lipid binding proteins differentially impacts saturated fatty acid uptake and nuclear targeting in mouse hepatocytes. Am J Physiol Gastrointest Liver Physiol 2012; 303:G837-50. [PMID: 22859366 PMCID: PMC3469595 DOI: 10.1152/ajpgi.00489.2011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The liver expresses high levels of two proteins with high affinity for long-chain fatty acids (LCFAs): liver fatty acid binding protein (L-FABP) and sterol carrier protein-2 (SCP-2). Real-time confocal microscopy of cultured primary hepatocytes from gene-ablated (L-FABP, SCP-2/SCP-x, and L-FABP/SCP-2/SCP-x null) mice showed that the loss of L-FABP reduced cellular uptake of 12-N-methyl-(7-nitrobenz-2-oxa-1,3-diazo)-aminostearic acid (a fluorescent-saturated LCFA analog) by ∼50%. Importantly, nuclear targeting of the LCFA was enhanced when L-FABP was upregulated (SCP-2/SCP-x null) but was significantly reduced when L-FABP was ablated (L-FABP null), thus impacting LCFA nuclear targeting. These effects were not associated with a net decrease in expression of key membrane proteins involved in LCFA or glucose transport. Since hepatic LCFA uptake and metabolism are closely linked to glucose uptake, the effect of glucose on L-FABP-mediated LCFA uptake and nuclear targeting was examined. Increasing concentrations of glucose decreased cellular LCFA uptake and even more extensively decreased LCFA nuclear targeting. Loss of L-FABP exacerbated the decrease in LCFA nuclear targeting, while loss of SCP-2 reduced the glucose effect, resulting in enhanced LCFA nuclear targeting compared with control. Simply, ablation of L-FABP decreases LCFA uptake and even more extensively decreases its nuclear targeting.
Collapse
Affiliation(s)
- Stephen M. Storey
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Avery L. McIntosh
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Huan Huang
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Gregory G. Martin
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Kerstin K. Landrock
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Danilo Landrock
- 2Department of Pathobiology, Texas A & M University, College Station, Texas
| | - H. Ross Payne
- 2Department of Pathobiology, Texas A & M University, College Station, Texas
| | - Ann B. Kier
- 2Department of Pathobiology, Texas A & M University, College Station, Texas
| | - Friedhelm Schroeder
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| |
Collapse
|
37
|
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: 9.7] [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
|
38
|
Qi ZH, Liu YF, Wang WN, Xin Y, Xie FX, Wang AL. Fatty acid binding protein 10 in the orange-spotted grouper (Epinephelus coioides): characterization and regulation under pH and temperature stress. Comp Biochem Physiol C Toxicol Pharmacol 2012; 155:447-55. [PMID: 22182678 DOI: 10.1016/j.cbpc.2011.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 12/02/2011] [Accepted: 12/04/2011] [Indexed: 11/29/2022]
Abstract
We have isolated and characterized a fatty acid binding protein from the liver of the orange-spotted grouper (Epinephelus coioides). Amino acid sequence similarity of the Ec-FABP (E. coioides-FABP) was highest to FABP10s isolated from the livers of catfish, chicken, salamander and iguana. The open-reading frame of the Ec-FABP codes for a protein of 14.0 kDa with a calculated isoelectric point of 8.5. We first expressed a FABP10 protein from orange-spotted grouper (E. coioides) in Pichia pastoris, and then characterized the antioxidative potential of our recombinant Ec-FABP by DCF fluorescence assay. RT-PCR assays showed that endogenous Ec-FABP mRNA is most strongly expressed in liver with the most abundance and intestine. Change in the groupers' blood cells respiratory burst activity was examined during and after exposure to the pH and temperature stress using flow cytometry. Further analysis of Ec-FABP gene expression in liver tissue by quantitative real-time PCR demonstrated that Ec-FABP transcript levels increased when the fish were exposed to both pH and temperature stress, but the time when its mRNA expression level peaked differed under these stresses. Western blot analyses confirmed that the Ec-FABP protein was strongly expressed in the liver after exposure to the pH and temperature stress. These results suggest that Ec-FABP expression is stimulated by pH and temperature stress and that it may play important roles in general adaptive and antioxidant responses.
Collapse
Affiliation(s)
- Zeng-hua Qi
- Key Laboratory of Ecology and Environmental Science in Guangdong Higher Education, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou 510631, PR China
| | | | | | | | | | | |
Collapse
|
39
|
Tomaselli S, Assfalg M, Pagano K, Cogliati C, Zanzoni S, Molinari H, Ragona L. A Disulfide Bridge Allows for Site-Selective Binding in Liver Bile Acid Binding Protein Thereby Stabilising the Orientation of Key Amino Acid Side Chains. Chemistry 2012; 18:2857-66. [DOI: 10.1002/chem.201102203] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 12/05/2011] [Indexed: 11/08/2022]
|
40
|
Janevski M, Antonas KN, Sullivan-Gunn MJ, McGlynn MA, Lewandowski PA. The effect of cocoa supplementation on hepatic steatosis, reactive oxygen species and LFABP in a rat model of NASH. COMPARATIVE HEPATOLOGY 2011; 10:10. [PMID: 22081873 PMCID: PMC3227569 DOI: 10.1186/1476-5926-10-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 11/14/2011] [Indexed: 02/05/2023]
Abstract
Background Non alcoholic steatohepatitis is hypothesised to develop via a mechanism involving fat accumulation and oxidative stress. The current study aimed to investigate if an increase in oxidative stress was associated with changes in the expression of liver fatty acid binding protein in a rat model of non alcoholic steatohepatitis and whether cocoa supplementation attenuated those changes. Methods Female Sprague Dawley rats were fed a high fat control diet, a high fat methionine choline deficient diet, or one of four 12.5% cocoa supplementation regimes in combination with the high fat methionine choline deficient diet. Results Liver fatty acid binding protein mRNA and protein levels were reduced in the liver of animals with fatty liver disease when compared to controls. Increased hepatic fat content was accompanied by higher levels of oxidative stress in animals with fatty liver disease when compared to controls. An inverse association was found between the levels of hepatic liver fatty acid binding protein and the level of hepatic oxidative stress in fatty liver disease. Elevated NADPH oxidase protein levels were detected in the liver of animals with increased severity in inflammation and fibrosis. Cocoa supplementation was associated with partial attenuation of these pathological changes, although the severity of liver disease induced by the methionine choline deficient diet prevented complete reversal of any disease associated changes. Red blood cell glutathione was increased by cocoa supplementation, whereas liver glutathione was reduced by cocoa compared to methionine choline deficient diet fed animals. Conclusion These findings suggest a potential role for liver fatty acid binding protein and NADPH oxidase in the development of non alcoholic steatohepatitis. Furthermore, cocoa supplementation may have be of therapeutic benefit in less sever forms of NASH.
Collapse
Affiliation(s)
- Mile Janevski
- School of Medicine, Deakin University, Waurn Ponds, Australia.
| | | | | | | | | |
Collapse
|
41
|
McIntosh AL, Huang H, Atshaves BP, Wellberg E, Kuklev DV, Smith WL, Kier AB, Schroeder F. Fluorescent n-3 and n-6 very long chain polyunsaturated fatty acids: three-photon imaging in living cells expressing liver fatty acid-binding protein. J Biol Chem 2010; 285:18693-708. [PMID: 20382741 PMCID: PMC2881794 DOI: 10.1074/jbc.m109.079897] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 03/10/2010] [Indexed: 01/24/2023] Open
Abstract
Despite the considerable beneficial effects of n-3 and n-6 very long chain polyunsaturated fatty acids (VLC-PUFAs), very little is known about the factors that regulate their uptake and intracellular distribution in living cells. This issue was addressed in cells expressing liver-type fatty acid-binding protein (L-FABP) by real time multiphoton laser scanning microscopy of novel fluorescent VLC-PUFAs containing a conjugated tetraene fluorophore near the carboxyl group and natural methylene-interrupted n-3 or n-6 grouping. The fluorescent VLC-PUFAs mimicked many properties of their native nonfluorescent counterparts, including uptake, distribution, and metabolism in living cells. The unesterified fluorescent VLC-PUFAs distributed either equally in nuclei versus cytoplasm (22-carbon n-3 VLC-PUFA) or preferentially to cytoplasm (20-carbon n-3 and n-6 VLC-PUFAs). L-FABP bound fluorescent VLC-PUFA with affinity and specificity similar to their nonfluorescent natural counterparts. Regarding n-3 and n-6 VLC-PUFA, L-FABP expression enhanced uptake into the cell and cytoplasm, selectively altered the pattern of fluorescent n-6 and n-3 VLC-PUFA distribution in cytoplasm versus nuclei, and preferentially distributed fluorescent VLC-PUFA into nucleoplasm versus nuclear envelope, especially for the 22-carbon n-3 VLC-PUFA, correlating with its high binding by L-FABP. Multiphoton laser scanning microscopy data showed for the first time VLC-PUFA in nuclei of living cells and suggested a model, whereby L-FABP facilitated VLC-PUFA targeting to nuclei by enhancing VLC-PUFA uptake and distribution into the cytoplasm and nucleoplasm.
Collapse
Affiliation(s)
| | - Huan Huang
- From the Departments of Physiology and Pharmacology and
| | - Barbara P. Atshaves
- the Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, and
| | | | - Dmitry V. Kuklev
- the Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - William L. Smith
- the Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Ann B. Kier
- Pathobiology, Texas A & M University, Texas Veterinary Medical Center, College Station, Texas 77843-4466
| | | |
Collapse
|
42
|
|