1
|
Mattingly JR, Wu A, York AG. Regulation of Adaptive Immunity by Lipid Post-translational Modifications. Immune Netw 2025; 25:e11. [PMID: 40078786 PMCID: PMC11896658 DOI: 10.4110/in.2025.25.e11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/14/2025] Open
Abstract
The burgeoning field of immunometabolism highlights the interdependence between metabolic programs and efficacious immune responses. The current understanding that cellular metabolic remodeling is necessary for a competent adaptive immune response, along with acutely sensitive methodologies such as high-performance liquid chromatography/mass spectrometry and advanced proteomics, have ushered in a renaissance of lipid- and metabolic-based scientific inquiries. One facet of recent interest examines how lipids function as post-translational modifications (PTMs) and their resulting effects on adaptive immune responses. The goal of this review is to establish a fundamental understanding of these protein modifications and highlight recent findings that underscore the importance of continued investigation into lipids as PTMs.
Collapse
Affiliation(s)
- Jonathan R. Mattingly
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Aimee Wu
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Autumn G. York
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
2
|
Wang L, Cheng C, Yu X, Guo L, Wan X, Xu J, Xiang X, Yang J, Kang J, Deng Q. Conversion of α-linolenic acid into n-3 long-chain polyunsaturated fatty acids: bioavailability and dietary regulation. Crit Rev Food Sci Nutr 2024:1-33. [PMID: 39686568 DOI: 10.1080/10408398.2024.2442064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
N-3 long-chain polyunsaturated fatty acids (n-3 LCPUFAs) are essential for physiological requirements and disease prevention throughout life but are not adequately consumed worldwide. Dietary supplementation with plant-derived α-linolenic acid (ALA) has the potential to rebalance the fatty acid profile and enhance health benefits but faces challenges such as high β-oxidation consumption, low hepatic conversion efficiency, and high oxidative susceptibility under stress. This review focuses on the metabolic fate and potential regulatory targets of ALA-containing lipids in vivo, specifically the pathway from the gastrointestinal tract to the lymph, blood circulation, and liver. We propose a hypothesis that positively regulates the conversion of ALA into n-3 LCPUFAs based on the model of "fast" or "slow" absorption, transport, and hepatic metabolic fate. Furthermore, the potential effects of dietary nutrients on the metabolic conversion of ALA into n-3 LCPUFAs are discussed. The conversion of ALA is differentially regulated by structured lipids, phospholipids, other lipids, carbohydrates, specific proteins, amino acids, polyphenols, vitamins, and minerals. Future research should focus on designing a steady-state and precise delivery system for ALA, coupled with specific nutrients or phytochemicals, to effectively improve its metabolic conversion and ultimately achieve synergistic regulation of nutrition and health effects.
Collapse
Affiliation(s)
- Lei Wang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Chen Cheng
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Xiao Yu
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou, Henan, China
| | - Liang Guo
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xia Wan
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jiqu Xu
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Xia Xiang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jing Yang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jingxuan Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Qianchun Deng
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| |
Collapse
|
3
|
Steinhauser ML, Maron BA. Viewing Pulmonary Arterial Hypertension Pathogenesis and Opportunities for Disease-Modifying Therapy Through the Lens of Biomass. JACC Basic Transl Sci 2024; 9:1252-1263. [PMID: 39534642 PMCID: PMC11551874 DOI: 10.1016/j.jacbts.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 11/16/2024]
Abstract
Fibroproliferative remodeling of distal pulmonary arterioles is a cornerstone characteristic of pulmonary arterial hypertension (PAH). Data from contemporary quantitative imaging suggest that anabolic synthesis of macromolecular substrate, defined here as biomass, is the proximate event that causes vascular remodeling via pathogenic changes to DNA, collagen, cytoskeleton, and lipid membranes. Modifying biomass is achievable but requires tilting the balance in favor of endogenous degradation over synthetic pathways in order to advance the first-ever disease-modifying PAH pharmacotherapy. Viewing PAH pathobiology through the lens of biomass represents an opportunity to decipher novel determinants of disease inception and inform interventions that induce reverse remodeling.
Collapse
Affiliation(s)
- Matthew L. Steinhauser
- Division of Cardiovascular Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bradley A. Maron
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- University of Maryland-Institute for Health Computing, Bethesda, Maryland, USA
| |
Collapse
|
4
|
Li K, Lv J, Wang J, Wei Y, Zhang Y, Lin J, Zhu Q. CircZNF609 inhibited bladder cancer immunotherapy sensitivity via enhancing fatty acid uptake through IGF2BP2/CD36 pathway. Int Immunopharmacol 2024; 137:112485. [PMID: 38878487 DOI: 10.1016/j.intimp.2024.112485] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024]
Abstract
Circular RNAs (circRNAs) are gaining attention for their involvement in immune escape and immunotherapy sensitivity regulation. CircZNF609 is a well-known oncogene in various solid tumours. Our previous research revealed its role in reducing the chemosensitivity of bladder cancer (BCa) to cisplatin. However, the underlying role of circZNF609 in BCa immune escape and immunotherapy sensitivity remains unknown. We conducted BCa cells-CD8 + T cells co-culture assays, cell line-derived xenograft and patient-derived xenograft mouse models with human immune reconstitution to further confirm the role of circZNF609 in BCa immune escape and immunotherapy sensitivity. Overexpression of circZNF609 promoted BCa immune escape in vitro and in vivo. Mechanistically, circZNF609 was bound to IGF2BP2, enhancing its interaction with the 3'-untranslated region of CD36. This increased the stability of the CD36 mRNA, leading to enhanced fatty acid uptake by BCa cells and fatty acid depletion within the tumour microenvironment. Additionally, the nuclear export of circZNF609 was regulated by DDX39B. CircZNF609 promoted immune escape and suppressed BCa immunotherapy sensitivity by regulating the newly identified circZNF609/IGF2BP2/CD36 cascade. Therefore, circZNF609 holds potential as both a biomarker and therapeutic target in BCa immunotherapy.
Collapse
Affiliation(s)
- Kai Li
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiancheng Lv
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Wang
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yong Wei
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yetao Zhang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianzhong Lin
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Qingyi Zhu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
5
|
Porter NH, Clark KL, Rebelo LR, Copelin JE, Kwon IH, Lee C. Effects of saturated fatty acids with lysophospholipids on production and nutrient digestibility in lactating cows. J Dairy Sci 2024; 107:4524-4536. [PMID: 38395396 DOI: 10.3168/jds.2023-24457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/19/2024] [Indexed: 02/25/2024]
Abstract
The objective of the experiment was to determine the effects of supplemental SFA sources, lysophospholipids (LPL), and their interaction on production and nutrient digestibility in lactating dairy cows. The experiment was conducted with 48 cows in a randomized complete block design. Cows were blocked (12 blocks total) by parity and days in milk and randomly assigned to 4 dietary treatments in each block (2 × 2 factorial arrangement), i.e., 2 sources of fat supplements, C16:0 (PA)- or C18:0 (SA)-enriched fat, and with or without LPL. The experiment was conducted for 6 wk to measure daily dry matter intake, milk yield, and weekly milk composition. During the last week of the experiment, spot fecal and urine samples were collected to determine total-tract nutrient digestibility. Milk samples in the last week were also collected to analyze the milk fatty acid (FA) profile. All data were analyzed using the MIXED procedure of SAS, where block was used as a random effect and FA, LPL, and the interaction of FA by LPL were used as fixed effects. Week and interactions of week by FA or LPL were included for production measures. Different sources of SFA did not affect dry matter intake and milk yield. However, the PA treatment increased (39.7 vs. 36.8 kg) energy-corrected milk compared with SA due to increased milk fat yield. No effect of LPL on production measures was observed. Total-tract digestibilities of dry matter, organic matter, crude protein, and total FA were not different between the PA and SA groups, but PA increased (41.4% vs. 38.8%) neutral detergent fiber digestibility compared with SA. Supplementation of LPL increased (64.7% vs. 60.5%) total FA digestibility, especially 18-carbon FA (74.1% vs. 68.2%). An interaction of SFA by LPL was found for 16-carbon FA digestibility. The PA diet increased the concentration of 16-carbon FA in milk fat and SA increased the concentration of preformed FA (≥18 carbons). Supplementation of LPL decreased the concentration of trans-10 C18:1. No difference in N utilization and excretion among treatments was observed. In conclusion, the PA diet was more effective in improving milk fat yield of lactating cows compared with SA. Supplementation of LPL increased digestibility of total FA, especially 18-carbon FA but did not affect production.
Collapse
Affiliation(s)
- N H Porter
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691
| | - K L Clark
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691
| | - L R Rebelo
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691
| | - J E Copelin
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691
| | - I H Kwon
- Easy Bio Inc., Seoul 06253, South Korea
| | - C Lee
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691.
| |
Collapse
|
6
|
Mariam I, Krikigianni E, Rantzos C, Bettiga M, Christakopoulos P, Rova U, Matsakas L, Patel A. Transcriptomics aids in uncovering the metabolic shifts and molecular machinery of Schizochytrium limacinum during biotransformation of hydrophobic substrates to docosahexaenoic acid. Microb Cell Fact 2024; 23:97. [PMID: 38561811 PMCID: PMC10983653 DOI: 10.1186/s12934-024-02381-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Biotransformation of waste oil into value-added nutraceuticals provides a sustainable strategy. Thraustochytrids are heterotrophic marine protists and promising producers of omega (ω) fatty acids. Although the metabolic routes for the assimilation of hydrophilic carbon substrates such as glucose are known for these microbes, the mechanisms employed for the conversion of hydrophobic substrates are not well established. Here, thraustochytrid Schizochytrium limacinum SR21 was investigated for its ability to convert oils (commercial oils with varying fatty acid composition and waste cooking oil) into ω-3 fatty acid; docosahexaenoic acid (DHA). RESULTS Within 72 h SR21 consumed ~ 90% of the oils resulting in enhanced biomass (7.5 g L- 1) which was 2-fold higher as compared to glucose. Statistical analysis highlights C16 fatty acids as important precursors of DHA biosynthesis. Transcriptomic data indicated the upregulation of multiple lipases, predicted to possess signal peptides for secretory, membrane-anchored and cytoplasmic localization. Additionally, transcripts encoding for mitochondrial and peroxisomal β-oxidation along with acyl-carnitine transporters were abundant for oil substrates that allowed complete degradation of fatty acids to acetyl CoA. Further, low levels of oxidative biomarkers (H2O2, malondialdehyde) and antioxidants were determined for hydrophobic substrates, suggesting that SR21 efficiently mitigates the metabolic load and diverts the acetyl CoA towards energy generation and DHA accumulation. CONCLUSIONS The findings of this study contribute to uncovering the route of assimilation of oil substrates by SR21. The thraustochytrid employs an intricate crosstalk among the extracellular and intracellular molecular machinery favoring energy generation. The conversion of hydrophobic substrates to DHA can be further improved using synthetic biology tools, thereby providing a unique platform for the sustainable recycling of waste oil substrates.
Collapse
Affiliation(s)
- Iqra Mariam
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental, and Natural Resources Engineering, Luleå University of Technology, Luleå, SE-971 87, Sweden
| | - Eleni Krikigianni
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental, and Natural Resources Engineering, Luleå University of Technology, Luleå, SE-971 87, Sweden
| | - Chloe Rantzos
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental, and Natural Resources Engineering, Luleå University of Technology, Luleå, SE-971 87, Sweden
| | - Maurizio Bettiga
- Department of Life Sciences - LIFE, Division of Industrial Biotechnology, Chalmers University of Technology, Gothenburg, SE-412 96, Sweden
- Innovation Unit, Italbiotec Srl Società Benefit, Milan, Italy
| | - Paul Christakopoulos
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental, and Natural Resources Engineering, Luleå University of Technology, Luleå, SE-971 87, Sweden
| | - Ulrika Rova
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental, and Natural Resources Engineering, Luleå University of Technology, Luleå, SE-971 87, Sweden
| | - Leonidas Matsakas
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental, and Natural Resources Engineering, Luleå University of Technology, Luleå, SE-971 87, Sweden
| | - Alok Patel
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental, and Natural Resources Engineering, Luleå University of Technology, Luleå, SE-971 87, Sweden.
| |
Collapse
|
7
|
Wang J, Xi Y, Sun B, Deng J, Ai N. Utilization of low-temperature heating method to improve skim milk production: Microstructure, stability, and constituents of milk fat globule membrane. Food Chem X 2024; 21:101187. [PMID: 38370307 PMCID: PMC10869298 DOI: 10.1016/j.fochx.2024.101187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/20/2024] Open
Abstract
In the process of defatting milk, preheating treatment is an important factor affecting the flavor of skim milk. Here, raw milk was preheated at different times and temperatures. Then laser confocal microscopy, multiple-light scattering instrument, and sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) were used to analyze the microstructure of milk fat globule membrane (MFGM), milk stability, and MFGM protein (MFGMP) components. Results showed that phospholipid domain of MFGM changed from an ordered state (Lo) to a disordered state (Ld) with increase in treatment temperature and time, leading to an increase in MFGMP content in skim milk. During the stability test, the stability of raw milk decreased significantly with increase in preheating temperature, while the opposite was true for skim milk. Finally, the results of MFGMP differentiation analysis showed that, the content of ten taste-related MFGMPs in the control group samples was significantly lower compared to the optimal group (P < 0.05).
Collapse
Affiliation(s)
- Juan Wang
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Beijing 100048, China
| | - Yanmei Xi
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Beijing 100048, China
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Beijing 100048, China
| | - Jianjun Deng
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Nasi Ai
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Beijing 100048, China
| |
Collapse
|
8
|
Deng L, Kersten S, Stienstra R. Triacylglycerol uptake and handling by macrophages: From fatty acids to lipoproteins. Prog Lipid Res 2023; 92:101250. [PMID: 37619883 DOI: 10.1016/j.plipres.2023.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Macrophages are essential innate immune cells and form our first line of immune defense. Also known as professional phagocytes, macrophages interact and take up various particles, including lipids. Defective lipid handling can drive excessive lipid accumulation leading to foam cell formation, a key feature of various cardiometabolic conditions such as atherosclerosis, non-alcoholic fatty liver disease, and obesity. At the same time, intracellular lipid storage and foam cell formation can also be viewed as a protective and anti-lipotoxic mechanism against a lipid-rich environment and associated elevated lipid uptake. Traditionally, foam cell formation has primarily been linked to cholesterol uptake via native and modified low-density lipoproteins. However, other lipids, including non-esterified fatty acids and triacylglycerol (TAG)-rich lipoproteins (very low-density lipoproteins and chylomicrons), can also interact with macrophages. Recent studies have identified multiple pathways mediating TAG uptake and processing by macrophages, including endocytosis and receptor/transporter-mediated internalization and transport. This review will present the current knowledge of how macrophages take up different lipids and lipoprotein particles and address how TAG-rich lipoproteins are processed intracellularly. Understanding how macrophages take up and process different lipid species such as TAG is necessary to design future therapeutic interventions to correct excessive lipid accumulation and associated co-morbidities.
Collapse
Affiliation(s)
- Lei Deng
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Rinke Stienstra
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
9
|
Iizuka D, Sasatani M, Ishikawa A, Daino K, Hirouchi T, Kamiya K. Newly discovered genomic mutation patterns in radiation-induced small intestinal tumors of ApcMin/+ mice. PLoS One 2023; 18:e0292643. [PMID: 37824459 PMCID: PMC10569626 DOI: 10.1371/journal.pone.0292643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 08/28/2023] [Indexed: 10/14/2023] Open
Abstract
Among the small intestinal tumors that occur in irradiated mice of the established mouse model B6/B6-Chr18MSM-F1 ApcMin/+, loss of heterozygosity analysis can be utilized to estimate whether a deletion in the wild-type allele containing the Adenomatous polyposis coli (Apc) region (hereafter referred to as Deletion), a duplication in the mutant allele with a nonsense mutation at codon 850 of Apc (Duplication), or no aberration (Unidentified) has occurred. Previous research has revealed that the number of Unidentified tumors tends to increase with the radiation dose. In the present study, we investigated the molecular mechanisms underlying the development of an Unidentified tumor type in response to radiation exposure. The mRNA expression levels of Apc were significantly lower in Unidentified tumors than in normal tissues. We focused on epigenetic suppression as the mechanism underlying this decreased expression; however, hypermethylation of the Apc promoter region was not observed. To investigate whether deletions occur that cannot be captured by loss of heterozygosity analysis, we analyzed chromosome 18 using a customized array comparative genomic hybridization approach designed to detect copy-number changes in chromosome 18. However, the copy number of the Apc region was not altered in Unidentified tumors. Finally, gene mutation analysis of the Apc region using next-generation sequencing suggested the existence of a small deletion (approximately 3.5 kbp) in an Unidentified tumor from a mouse in the irradiated group. Furthermore, nonsense and frameshift mutations in Apc were found in approximately 30% of the Unidentified tumors analyzed. These results suggest that radiation-induced Unidentified tumors arise mainly due to decreased Apc expression of an unknown regulatory mechanism that does not depend on promoter hypermethylation, and that some tumors may result from nonsense mutations which are as-yet undefined point mutations.
Collapse
Affiliation(s)
- Daisuke Iizuka
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Megumi Sasatani
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Atsuko Ishikawa
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Kazuhiro Daino
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Tokuhisa Hirouchi
- Department of Radiobiology, Institute for Environmental Sciences, Rokkasho, Japan
| | - Kenji Kamiya
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
10
|
Zheng Y, Sun L, Guo J, Ma J. The crosstalk between ferroptosis and anti-tumor immunity in the tumor microenvironment: molecular mechanisms and therapeutic controversy. Cancer Commun (Lond) 2023; 43:1071-1096. [PMID: 37718480 PMCID: PMC10565387 DOI: 10.1002/cac2.12487] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/13/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023] Open
Abstract
The advent of immunotherapy has significantly reshaped the landscape of cancer treatment, greatly enhancing therapeutic outcomes for multiple types of cancer. However, only a small subset of individuals respond to it, underscoring the urgent need for new methods to improve its response rate. Ferroptosis, a recently discovered form of programmed cell death, has emerged as a promising approach for anti-tumor therapy, with targeting ferroptosis to kill tumors seen as a potentially effective strategy. Numerous studies suggest that inducing ferroptosis can synergistically enhance the effects of immunotherapy, paving the way for a promising combined treatment method in the future. Nevertheless, recent research has raised concerns about the potential negative impacts on anti-tumor immunity as a consequence of inducing ferroptosis, leading to conflicting views within the scientific community about the interplay between ferroptosis and anti-tumor immunity, thereby underscoring the necessity of a comprehensive review of the existing literature on this relationship. Previous reviews on ferroptosis have touched on related content, many focusing primarily on the promoting role of ferroptosis on anti-tumor immunity while overlooking recent evidence on the inhibitory effects of ferroptosis on immunity. Others have concentrated solely on discussing related content either from the perspective of cancer cells and ferroptosis or from immune cells and ferroptosis. Given that both cancer cells and immune cells exist in the tumor microenvironment, a one-sided discussion cannot comprehensively summarize this topic. Therefore, from the perspectives of both tumor cells and tumor-infiltrating immune cells, we systematically summarize the current conflicting views on the interplay between ferroptosis and anti-tumor immunity, intending to provide potential explanations and identify the work needed to establish a translational basis for combined ferroptosis-targeted therapy and immunotherapy in treating tumors.
Collapse
Affiliation(s)
- Yichen Zheng
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Lingqi Sun
- Department of NeurologyAir Force Hospital of the Western Theater of the Chinese People's Liberation ArmyChengduSichuanP. R. China
| | - Jiamin Guo
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ji Ma
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
11
|
Dauchy RT, Sauer LA, Blask DE. Dietary Linoleic Acid: An Omega-6 Fatty Acid Essential for Liver Regeneration in Buffalo Rats. Comp Med 2023; 73:295-311. [PMID: 37652672 PMCID: PMC10702281 DOI: 10.30802/aalas-cm-23-000004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 09/02/2023]
Abstract
Rodents are currently the most common animals used for hepatic surgical resection studies that investigate liver regeneration, chronic liver disease, acute liver failure, hepatic metastasis, hepatic function, and hepatic cancer. Our previous work has shown that dietary consumption of linoleic acid (LA) stimulates the growth of rodent and human tumors in vivo. Here we compared 3 diets - a 5% corn oil diet (control), a diet deficient in essential fatty acids (EFAD), and an EFAD supplemented with LA in amounts equal to those in the control diet (EFAD+LA). We hypothesized that consumption of the LA provided in the EFAD+LA diet would elevate plasma levels of LA and stimulate regeneration in rats after a 70% hepatectomy (HPX), and that regeneration would not occur in the EFAD rats. Each diet group was comprised of 30 male and 30 female Buffalo rats (BUFF/CrCrl). Rats were fed one of the 3 diets and water ad libitum. After 8 wk on the assigned diet, rats were underwent a 70% HPX. On days 4 and 21 after HPX, 30 male and 30 female rats from each diet group were anesthetized for in vivo study and then were euthanized for tissue collection. For the in vivo study, arterial and venous blood samples were collected from the liver. LA-, glucose-, and O₂ -uptake, and lactate- and CO₂ -output were significantly higher in LA-replete rats as compared with LA-deficient rats. After a 70% HPX, the remaining liver mass in control and EFAD+LA groups had doubled at day 4, reaching 60% of the original total weight, and had regenerated completely at day 21. However, no regeneration occurred in the EFAD group. At day 4 the portions of livers removed from the control and EFAD+LA groups had significantly higher content of LA, protein, cAMP, and DNA as compared with their livers on day 21. [³ H]thymidine incorporation into liver DNA was significantly higher in the 2 LA-replete groups, with male values greater than female values, as compared with LA-deficient group. These data indicate that liver regeneration after HPX is dependent on dietary LA. Understanding the mechanisms of LA-dependent liver regeneration in rats supports our current efforts to enhance successful surgical resection therapies in humans.
Collapse
Key Words
- akt, serine-threonine protein kinase
- a-v, arterial-venous
- ce, cholesterol esters
- cl, caudate lobe
- cp, caudate process
- icl, inferior caudate lobe
- irll, inferior right lateral lobe
- ivc, inferior vena cava
- efad, essential fatty acid deficient
- egfr, epithelial growth factor receptor
- erk1/2, extracellular signal regulated kinase p44/46 (mapk, mitogen-activated protein kinase)
- fa, fatty acid
- ffar, free fatty acid receptor
- ffa, free fatty acids
- g protein, guanine nucleotide binding protein
- hpx, 70% partial hepatectomy
- la, linoleic acid
- lll, left lateral lobe
- lml, left median lobe
- ml, middle or median lobe
- rll, right lateral lobe
- rml, right median lobe
- scl, superior caudate lobe
- srll, superior right lateral lobe
- pi3k, phosphatidylinositol-3-kinase/akt
- pl, phospholipids
- tfa, total fatty acids
- tgl, triglycerides
- wnt/β-catenin, wingless and int-1/β catenin
Collapse
Affiliation(s)
- Robert T Dauchy
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane, Louisiana
| | | | - David E Blask
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane, Louisiana
| |
Collapse
|
12
|
Fu Q, Cheung WA, Majnik AV, Ke X, Pastinen T, Lane RH. Adverse Maternal Environments Perturb Hepatic DNA Methylome and Transcriptome Prior to the Adult-Onset Non-Alcoholic Fatty Liver Disease in Mouse Offspring. Nutrients 2023; 15:2167. [PMID: 37432267 DOI: 10.3390/nu15092167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 07/12/2023] Open
Abstract
Exposure to adverse early-life environments (AME) increases the incidence of developing adult-onset non-alcoholic fatty liver disease (NAFLD). DNA methylation has been postulated to link AME and late-onset diseases. This study aimed to investigate whether and to what extent the hepatic DNA methylome was perturbed prior to the development of NAFLD in offspring exposed to AME in mice. AME constituted maternal Western diet and late-gestational stress. Male offspring livers at birth (d0) and weaning (d21) were used for evaluating the DNA methylome and transcriptome using the reduced representation of bisulfite sequencing and RNA-seq, respectively. We found AME caused 5879 differentially methylated regions (DMRs) and zero differentially expressed genes (DEGs) at d0 and 2970 and 123, respectively, at d21. The majority of the DMRs were distal to gene transcription start sites and did not correlate with DEGs. The DEGs at d21 were significantly enriched in GO biological processes characteristic of liver metabolic functions. In conclusion, AME drove changes in the hepatic DNA methylome, which preceded perturbations in the hepatic metabolic transcriptome, which preceded the onset of NAFLD. We speculate that subtle impacts on dynamic enhancers lead to long-range regulatory changes that manifest over time as gene network alternations and increase the incidence of NAFLD later in life.
Collapse
Affiliation(s)
- Qi Fu
- Department of Research Administration, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Warren A Cheung
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Amber V Majnik
- Department of Pediatrics, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Xingrao Ke
- Department of Research Administration, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Tomi Pastinen
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Robert H Lane
- Department of Administration, Children's Mercy Hospital, Kansas City, MO 64108, USA
| |
Collapse
|
13
|
Wang Y, Wang Y, Wu Y, Suo Y, Guo H, Yu Y, Yin R, Xi R, Wu J, Hua N, Zhang Y, Zhang S, Jin Z, He L, Ma G. Using the inner membrane of Escherichia coli as a scaffold to anchor enzymes for metabolic flux enhancement. Eng Life Sci 2023; 23:e2200034. [PMID: 36751472 PMCID: PMC9893748 DOI: 10.1002/elsc.202200034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/11/2023] Open
Abstract
Clustering enzymes in the same metabolic pathway is a natural strategy to enhance productivity. Synthetic protein, RNA and DNA scaffolds have been designed to artificially cluster multiple enzymes in the cell, which require complex construction processes and possess limited slots for target enzymes. We utilized the Escherichia coli inner cell membrane as a native scaffold to cluster four fatty acid synthases (FAS) and achieved to improve the efficiency of fatty acid synthesis in vivo. The construction strategy is as simple as fusing target enzymes to the N-terminus or C-terminus of the membrane anchor protein (Lgt), and the number of anchored enzymes is not restricted. This novel device not only presents a similar efficiency in clustering multiple enzymes to that of other artificial scaffolds but also promotes the product secretion, driving the entire metabolic flux forward and further increasing the gross yield compared with that in a cytoplasmic scaffold system.
Collapse
Affiliation(s)
- You Wang
- Bio‐X‐Renji Hospital Research CenterRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Yushu Wang
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Yuqi Wu
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Yang Suo
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Huaqing Guo
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Yineng Yu
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Ruonan Yin
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Rui Xi
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Jiajie Wu
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Nan Hua
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Yuehan Zhang
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Shaobo Zhang
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Zhenming Jin
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
- 2012 SJTU‐BioX‐Shanghai Team for The International Genetically Engineered Machine Competition (iGEM)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Lin He
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
| | - Gang Ma
- Bio‐X‐Renji Hospital Research CenterRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
- Bio‐X InstitutesKey Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiP.R. China
| |
Collapse
|
14
|
Huang X, Zhou Y, Sun Y, Wang Q. Intestinal fatty acid binding protein: A rising therapeutic target in lipid metabolism. Prog Lipid Res 2022; 87:101178. [PMID: 35780915 DOI: 10.1016/j.plipres.2022.101178] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 10/17/2022]
Abstract
Fatty acid binding proteins (FABPs) are key proteins in lipid transport, and the isoforms are segregated according to their tissue origins. Several isoforms, such as adipose-FABP and epidermal-FABP, have been shown to participate in multiple pathologic processes due to their ubiquitous expression. Intestinal fatty acid binding protein, also termed FABP2 or I-FABP, is specifically expressed in the small intestine. FABP2 can traffic lipids from the intestinal lumen to enterocytes and bind superfluous fatty acids to maintain a steady pool of fatty acids in the epithelium. As a lipid chaperone, FABP2 can also carry lipophilic drugs to facilitate targeted transport. When the integrity of the intestinal epithelium is disrupted, FABP2 is released into the circulation. Thus, it can potentially serve as a clinical biomarker. In this review, we discuss the pivotal role of FABP2 in intestinal lipid metabolism. We also summarize the molecular interactions that have been reported to date, highlighting the clinical prospects of FABP2 research.
Collapse
Affiliation(s)
- Xi Huang
- Shanghai Institute of Immunology, Department of Gastroenterology of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Youci Zhou
- Shanghai Institute of Immunology, Department of Gastroenterology of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yunwei Sun
- Shanghai Institute of Immunology, Department of Gastroenterology of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qijun Wang
- Shanghai Institute of Immunology, Department of Gastroenterology of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
15
|
Yang Q, Hinkle J, Reed JN, Aherrahrou R, Xu Z, Harris TE, Stephenson EJ, Musunuru K, Keller SR, Civelek M. Adipocyte-Specific Modulation of KLF14 Expression in Mice Leads to Sex-Dependent Impacts on Adiposity and Lipid Metabolism. Diabetes 2022; 71:677-693. [PMID: 35081256 PMCID: PMC8965685 DOI: 10.2337/db21-0674] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022]
Abstract
Genome-wide association studies identified single nucleotide polymorphisms on chromosome 7 upstream of KLF14 to be associated with metabolic syndrome traits and increased risk for type 2 diabetes (T2D). The associations were more significant in women than in men. The risk allele carriers expressed lower levels of the transcription factor KLF14 in adipose tissues than nonrisk allele carriers. To investigate how adipocyte KLF14 regulates metabolic traits in a sex-dependent manner, we characterized high-fat diet-fed male and female mice with adipocyte-specific Klf14 deletion or overexpression. Klf14 deletion resulted in increased fat mass in female mice and decreased fat mass in male mice. Female Klf14-deficient mice had overall smaller adipocytes in subcutaneous fat depots but larger adipocytes in parametrial depots, indicating a shift in lipid storage from subcutaneous to visceral fat depots. They had reduced metabolic rates and increased respiratory exchange ratios consistent with increased use of carbohydrates as an energy source. Fasting- and isoproterenol-induced adipocyte lipolysis was defective in female Klf14-deficient mice, and concomitantly, adipocyte triglycerides lipase mRNA levels were downregulated. Female Klf14-deficient mice cleared blood triglyceride and nonesterified fatty acid less efficiently than wild-type. Finally, adipocyte-specific overexpression of Klf14 resulted in lower total body fat in female but not male mice. Taken together, consistent with human studies, adipocyte KLF14 deficiency in female but not in male mice causes increased adiposity and redistribution of lipid storage from subcutaneous to visceral adipose tissues. Increasing KLF14 abundance in adipocytes of females with obesity and T2D may provide a novel treatment option to alleviate metabolic abnormalities.
Collapse
Affiliation(s)
- Qianyi Yang
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
- Corresponding authors: Qianyi Yang, , and Mete Civelek,
| | - Jameson Hinkle
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
| | - Jordan N. Reed
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA
| | - Redouane Aherrahrou
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
| | - Zhiwen Xu
- Department of Chemistry, College of Arts and Sciences, University of Virginia, Charlottesville, VA
| | - Thurl E. Harris
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA
| | - Erin J. Stephenson
- Department of Anatomy, College of Graduate Studies & Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL
| | - Kiran Musunuru
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Susanna R. Keller
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA
| | - Mete Civelek
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA
- Corresponding authors: Qianyi Yang, , and Mete Civelek,
| |
Collapse
|
16
|
Błachnio-Zabielska AU, Roszczyc-Owsiejczuk K, Imierska M, Pogodzińska K, Rogalski P, Daniluk J, Zabielski P. CerS1 but Not CerS5 Gene Silencing, Improves Insulin Sensitivity and Glucose Uptake in Skeletal Muscle. Cells 2022; 11:206. [PMID: 35053322 PMCID: PMC8773817 DOI: 10.3390/cells11020206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle is perceived as a major tissue in glucose and lipid metabolism. High fat diet (HFD) lead to the accumulation of intramuscular lipids, including: long chain acyl-CoA, diacylglycerols, and ceramides. Ceramides are considered to be one of the most important lipid groups in the generation of skeletal muscle insulin resistance. So far, it has not been clearly established whether all ceramides adversely affect the functioning of the insulin pathway, or whether there are certain ceramide species that play a pivotal role in the induction of insulin resistance. Therefore, we designed a study in which the expression of CerS1 and CerS5 genes responsible for the synthesis of C18:0-Cer and C16:0-Cer, respectively, was locally silenced in the gastrocnemius muscle of HFD-fed mice through in vivo electroporation-mediated shRNA plasmids. Our study indicates that HFD feeding induced both, the systemic and skeletal muscle insulin resistance, which was accompanied by an increase in the intramuscular lipid levels, decreased activation of the insulin pathway and, consequently, a decrease in the skeletal muscle glucose uptake. CerS1 silencing leads to a reduction in C18:0-Cer content, with a subsequent increase in the activity of the insulin pathway, and an improvement in skeletal muscle glucose uptake. Such effects were not visible in case of CerS5 silencing, which indicates that the accumulation of C18:0-Cer plays a decisive role in the induction of skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- Agnieszka U. Błachnio-Zabielska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland; (K.R.-O.); (M.I.); (K.P.)
| | - Kamila Roszczyc-Owsiejczuk
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland; (K.R.-O.); (M.I.); (K.P.)
| | - Monika Imierska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland; (K.R.-O.); (M.I.); (K.P.)
| | - Karolina Pogodzińska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Mickiewicza 2c, 15-089 Bialystok, Poland; (K.R.-O.); (M.I.); (K.P.)
| | - Paweł Rogalski
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland; (P.R.); (J.D.)
| | - Jarosław Daniluk
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland; (P.R.); (J.D.)
| | - Piotr Zabielski
- Department of Medical Biology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
17
|
Laskar P, Dufès C. Emergence of cationic polyamine dendrimersomes: design, stimuli sensitivity and potential biomedical applications. NANOSCALE ADVANCES 2021; 3:6007-6026. [PMID: 34765868 PMCID: PMC8548884 DOI: 10.1039/d1na00536g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 06/01/2023]
Abstract
For decades, self-assembled lipid vesicles have been widely used in clinics as nanoscale delivery systems for various biomedical applications, including treatment of various diseases. Due to their core-shell architecture and versatile nature, they have been successfully used as carriers for the delivery of a wide range of therapeutic cargos, including drugs and nucleic acids, in cancer treatment. Recently, surface-modified polyamine dendrimer-based vesicles, or dendrimersomes, have emerged as promising alternatives to lipid vesicles for various biomedical applications, due to their ease of synthesis, non-immunogenicity, stability in circulation and lower size polydispersity. This mini-review provides an overview of the recent advances resulting from the use of biomimetic hydrophobically-modified polyamine-based dendrimersomes towards biomedical applications, focusing mainly on the two most widely used polyamine dendrimers, namely polyamidoamine (PAMAM) and poly(propylene imine) (PPI) dendrimers.
Collapse
Affiliation(s)
- Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley McAllen TX 78504 USA
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde 161 Cathedral Street Glasgow G4 0RE UK
| |
Collapse
|
18
|
Auzmendi J, Akyuz E, Lazarowski A. The role of P-glycoprotein (P-gp) and inwardly rectifying potassium (Kir) channels in sudden unexpected death in epilepsy (SUDEP). Epilepsy Behav 2021; 121:106590. [PMID: 31706919 DOI: 10.1016/j.yebeh.2019.106590] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the major cause of death that affects patients with epilepsy. The risk of SUDEP increases according to the frequency and severity of uncontrolled seizures; therefore, SUDEP risk is higher in patients with refractory epilepsy (RE), in whom most antiepileptic drugs (AEDs) are ineffective for both seizure control and SUDEP prevention. Consequently, RE and SUDEP share a multidrug resistance (MDR) phenotype, which is mainly associated with brain overexpression of ABC-transporters such as P-glycoprotein (P-gp). The activity of P-gp can also contribute to membrane depolarization and affect the normal function of neurons and cardiomyocytes. Other molecular regulators of membrane potential are the inwardly rectifying potassium channels (Kir), whose genetic variants have been related to both epilepsy and heart dysfunctions. Although it has been suggested that dysfunctions of the cardiac, respiratory, and brainstem arousal systems are the causes of SUDEP, the molecular basis for explaining its dysfunctions remain unknown. In rats, repetitive seizures or status epilepticus induced high expression of P-gp and loss Kir expression in the brain and heart, and promoted membrane depolarization, malignant bradycardia, and the high rate of mortality. Here we reviewed clinical and experimental evidences suggesting that abnormal expression of depolarizing/repolarizing factors as P-gp and Kir could favor persistent depolarization of membranes without any rapid functional recovery capacity. This condition induced by convulsive stress could be the molecular mechanism leading to acquired severe bradycardia, as an ineffective heart response generating the appropriate scenario for SUDEP development. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Jerónimo Auzmendi
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina; INFIBIOC, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica (FFyB), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Enes Akyuz
- Yozgat Bozok University, Medical Faculty, Department of Biophysics, Erdoğan Akdağ Yerleşkesi, 66100 Yozgat, Turkey
| | - Alberto Lazarowski
- INFIBIOC, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica (FFyB), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.
| |
Collapse
|
19
|
Zingg JM, Vlad A, Ricciarelli R. Oxidized LDLs as Signaling Molecules. Antioxidants (Basel) 2021; 10:antiox10081184. [PMID: 34439432 PMCID: PMC8389018 DOI: 10.3390/antiox10081184] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Levels of oxidized low-density lipoproteins (oxLDLs) are usually low in vivo but can increase whenever the balance between formation and scavenging of free radicals is impaired. Under normal conditions, uptake and degradation represent the physiological cellular response to oxLDL exposure. The uptake of oxLDLs is mediated by cell surface scavenger receptors that may also act as signaling molecules. Under conditions of atherosclerosis, monocytes/macrophages and vascular smooth muscle cells highly exposed to oxLDLs tend to convert to foam cells due to the intracellular accumulation of lipids. Moreover, the atherogenic process is accelerated by the increased expression of the scavenger receptors CD36, SR-BI, LOX-1, and SRA in response to high levels of oxLDL and oxidized lipids. In some respects, the effects of oxLDLs, involving cell proliferation, inflammation, apoptosis, adhesion, migration, senescence, and gene expression, can be seen as an adaptive response to the rise of free radicals in the vascular system. Unlike highly reactive radicals, circulating oxLDLs may signal to cells at more distant sites and possibly trigger a systemic antioxidant defense, thus elevating the role of oxLDLs to that of signaling molecules with physiological relevance.
Collapse
Affiliation(s)
- Jean-Marc Zingg
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Correspondence: (J.-M.Z.); (R.R.); Tel.: +1-(305)-2433531 (J.-M.Z.); +39-010-3538831 (R.R.)
| | - Adelina Vlad
- Physiology Department, “Carol Davila” UMPh, 020021 Bucharest, Romania;
| | - Roberta Ricciarelli
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence: (J.-M.Z.); (R.R.); Tel.: +1-(305)-2433531 (J.-M.Z.); +39-010-3538831 (R.R.)
| |
Collapse
|
20
|
Xu S, Chaudhary O, Rodríguez-Morales P, Sun X, Chen D, Zappasodi R, Xu Z, Pinto AFM, Williams A, Schulze I, Farsakoglu Y, Varanasi SK, Low JS, Tang W, Wang H, McDonald B, Tripple V, Downes M, Evans RM, Abumrad NA, Merghoub T, Wolchok JD, Shokhirev MN, Ho PC, Witztum JL, Emu B, Cui G, Kaech SM. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8 + T cells in tumors. Immunity 2021; 54:1561-1577.e7. [PMID: 34102100 PMCID: PMC9273026 DOI: 10.1016/j.immuni.2021.05.003] [Citation(s) in RCA: 427] [Impact Index Per Article: 106.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/03/2021] [Accepted: 05/04/2021] [Indexed: 01/21/2023]
Abstract
A common metabolic alteration in the tumor microenvironment (TME) is lipid accumulation, a feature associated with immune dysfunction. Here, we examined how CD8+ tumor infiltrating lymphocytes (TILs) respond to lipids within the TME. We found elevated concentrations of several classes of lipids in the TME and accumulation of these in CD8+ TILs. Lipid accumulation was associated with increased expression of CD36, a scavenger receptor for oxidized lipids, on CD8+ TILs, which also correlated with progressive T cell dysfunction. Cd36-/- T cells retained effector functions in the TME, as compared to WT counterparts. Mechanistically, CD36 promoted uptake of oxidized low-density lipoproteins (OxLDL) into T cells, and this induced lipid peroxidation and downstream activation of p38 kinase. Inhibition of p38 restored effector T cell functions in vitro, and resolution of lipid peroxidation by overexpression of glutathione peroxidase 4 restored functionalities in CD8+ TILs in vivo. Thus, an oxidized lipid-CD36 axis promotes intratumoral CD8+ T cell dysfunction and serves as a therapeutic avenue for immunotherapies.
Collapse
Affiliation(s)
- Shihao Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Omkar Chaudhary
- Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Patricia Rodríguez-Morales
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Xiaoli Sun
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dan Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ziyan Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Antonio F M Pinto
- Mass Spectrometry Core for Proteomics and Metabolomics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - April Williams
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Isabell Schulze
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yagmur Farsakoglu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Siva Karthik Varanasi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jun Siong Low
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA; Fondazione per l'istituto di ricerca in biomedicina, Bellinzona, Switzerland
| | - Wenxi Tang
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Haiping Wang
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Bryan McDonald
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Victoria Tripple
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nada A Abumrad
- Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maxim N Shokhirev
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Joseph L Witztum
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brinda Emu
- Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Guoliang Cui
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
21
|
Goldberg IJ, Cabodevilla AG, Samovski D, Cifarelli V, Basu D, Abumrad NA. Lipolytic enzymes and free fatty acids at the endothelial interface. Atherosclerosis 2021; 329:1-8. [PMID: 34130222 DOI: 10.1016/j.atherosclerosis.2021.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 01/17/2023]
Abstract
Lipids released from circulating lipoproteins by intravascular action of lipoprotein lipase (LpL) reach parenchymal cells in tissues with a non-fenestrated endothelium by transfer through or around endothelial cells. The actions of LpL are controlled at multiple sites, its synthesis and release by myocytes and adipocytes, its transit and association with the endothelial cell luminal surface, and finally its activation and inhibition by a number of proteins and by its product non-esterified fatty acids. Multiple pathways mediate endothelial transit of lipids into muscle and adipose tissues. These include movement of fatty acids via the endothelial cell fatty acid transporter CD36 and movement of whole or partially LpL-hydrolyzed lipoproteins via other apical endothelial cell receptors such as SR-B1and Alk1. Lipids also likely change the barrier function of the endothelium and operation of the paracellular pathway around endothelial cells. This review summarizes in vitro and in vivo support for the key role of endothelial cells in delivery of lipids and highlights incompletely understood processes that are the focus of active investigation.
Collapse
Affiliation(s)
- Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Dmitri Samovski
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO, USA
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Nada A Abumrad
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
22
|
Sesorova IS, Dimov ID, Kashin AD, Sesorov VV, Karelina NR, Zdorikova MA, Beznoussenko GV, Mirоnоv AA. Cellular and sub-cellular mechanisms of lipid transport from gut to lymph. Tissue Cell 2021; 72:101529. [PMID: 33915359 DOI: 10.1016/j.tice.2021.101529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 02/26/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022]
Abstract
Although the general structure of the barrier between the gut and the blood is well known, many details are still missing. Here, we analyse the literature and our own data related to lipid transcytosis through adult mammalian enterocytes, and their absorption into lymph at the tissue level of the intestine. After starvation, the Golgi complex (GC) of enterocytes is in a resting state. The addition of lipids in the form of chyme leads to the initial appearance of pre-chylomicrons (ChMs) in the tubules of the smooth endoplasmic reticulum, which are attached at the basolateral plasma membrane, immediately below the 'belt' of the adhesive junctions. Then pre-ChMs move into the cisternae of the rough endoplasmic reticulum and then into the expansion of the perforated Golgi cisternae. Next, they pass through the GC, and are concentrated in the distensions of the perforated cisternae on the trans-side of the GC. The arrival of pre-ChMs at the GC leads to the transition of the GC to a state of active transport, with formation of intercisternal connections, attachment of cis-most and trans-most perforated cisternae to the medial Golgi cisternae, and disappearance of COPI vesicles. Post-Golgi carriers then deliver ChMs to the basolateral plasma membrane, fuse with it, and secret ChMs into the intercellular space between enterocytes at the level of their interdigitating contacts. Finally, ChMs are squeezed out into the interstitium through pores in the basal membrane, most likely due to the function of the actin-myosin 'cuff' around the interdigitating contacts. These pores appear to be formed by protrusions of the dendritic cells and the enterocytes per se. ChMs are absorbed from the interstitium into the lymphatic capillaries through the special oblique contacts between endothelial cells, which function as valves through the contraction-relaxation of bundles of smooth muscle cells in the interstitium. Lipid overloading of enterocytes results in accumulation of cytoplasmic lipid droplets, an increase in diameter of ChMs, inhibition of intra-Golgi transport, and fusion of ChMs in the interstitium. Here, we summarise and analyse recent findings, and discuss their functional implications.
Collapse
Affiliation(s)
- Irina S Sesorova
- Department of Anatomy, Saint Petersburg State Paediatric Medical University, S. Petersburg, Russia
| | - Ivan D Dimov
- Department of Anatomy, Ivanovo State Medical Academy, Ivanovo, Russia
| | - Alexandre D Kashin
- Department of Anatomy, Saint Petersburg State Paediatric Medical University, S. Petersburg, Russia
| | - Vitaly V Sesorov
- Department of Anatomy, Saint Petersburg State Paediatric Medical University, S. Petersburg, Russia
| | | | - Maria A Zdorikova
- Department of Anatomy, Saint Petersburg State Paediatric Medical University, S. Petersburg, Russia
| | | | | |
Collapse
|
23
|
Laskar P, Somani S, Mullin M, Tate RJ, Warzecha M, Bowering D, Keating P, Irving C, Leung HY, Dufès C. Octadecyl chain-bearing PEGylated poly(propyleneimine)-based dendrimersomes: physicochemical studies, redox-responsiveness, DNA condensation, cytotoxicity and gene delivery to cancer cells. Biomater Sci 2021; 9:1431-1448. [PMID: 33404026 DOI: 10.1039/d0bm01441a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
Stimuli-responsive nanocarriers have become increasingly important for nucleic acid and drug delivery in cancer therapy. Here, we report the synthesis, characterization and evaluation of disulphide-linked, octadecyl (C18 alkyl) chain-bearing PEGylated generation 3-diaminobutyric polypropylenimine dendrimer-based vesicles (or dendrimersomes) for gene delivery. The lipid-bearing PEGylated dendrimer was successfully synthesized through in situ two-step reaction. It was able to spontaneously self-assemble into stable, cationic, nanosized vesicles, with low critical aggregation concentration value, and also showed redox-responsiveness in presence of a glutathione concentration similar to that of the cytosolic reducing environment. In addition, it was able to condense more than 70% of DNA at dendrimer: DNA weight ratios of 5 : 1 and higher. This dendriplex resulted in an enhanced cellular uptake of DNA at dendrimer: DNA weight ratios of 10 : 1 and 20 : 1, by up to 16-fold and by up to 28-fold compared with naked DNA in PC-3 and DU145 prostate cancer cell lines respectively. At a dendrimer: DNA weight ratio of 20 : 1, it led to an increase in gene expression in PC-3 and DU145 cells, compared with DAB dendriplex. These octadecyl chain-bearing, PEGylated dendrimer-based vesicles are therefore promising redox-sensitive drug and gene delivery systems for potential applications in combination cancer therapy.
Collapse
Affiliation(s)
- Partha Laskar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abumrad NA, Cabodevilla AG, Samovski D, Pietka T, Basu D, Goldberg IJ. Endothelial Cell Receptors in Tissue Lipid Uptake and Metabolism. Circ Res 2021; 128:433-450. [PMID: 33539224 DOI: 10.1161/circresaha.120.318003] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipid uptake and metabolism are central to the function of organs such as heart, skeletal muscle, and adipose tissue. Although most heart energy derives from fatty acids (FAs), excess lipid accumulation can cause cardiomyopathy. Similarly, high delivery of cholesterol can initiate coronary artery atherosclerosis. Hearts and arteries-unlike liver and adrenals-have nonfenestrated capillaries and lipid accumulation in both health and disease requires lipid movement from the circulation across the endothelial barrier. This review summarizes recent in vitro and in vivo findings on the importance of endothelial cell receptors and uptake pathways in regulating FAs and cholesterol uptake in normal physiology and cardiovascular disease. We highlight clinical and experimental data on the roles of ECs in lipid supply to tissues, heart, and arterial wall in particular, and how this affects organ metabolism and function. Models of FA uptake into ECs suggest that receptor-mediated uptake predominates at low FA concentrations, such as during fasting, whereas FA uptake during lipolysis of chylomicrons may involve paracellular movement. Similarly, in the setting of an intact arterial endothelial layer, recent and historic data support a role for receptor-mediated processes in the movement of lipoproteins into the subarterial space. We conclude with thoughts on the need to better understand endothelial lipid transfer for fuller comprehension of the pathophysiology of hyperlipidemia, and lipotoxic diseases such as some forms of cardiomyopathy and atherosclerosis.
Collapse
Affiliation(s)
- Nada A Abumrad
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Dmitri Samovski
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Terri Pietka
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| |
Collapse
|
25
|
Kim J, Moon J, Park CH, Lee J, Cheng H, Floyd ZE, Chang JS. NT-PGC-1α deficiency attenuates high-fat diet-induced obesity by modulating food intake, fecal fat excretion and intestinal fat absorption. Sci Rep 2021; 11:1323. [PMID: 33446719 PMCID: PMC7809341 DOI: 10.1038/s41598-020-79823-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Transcriptional coactivator PGC-1α and its splice variant NT-PGC-1α regulate metabolic adaptation by modulating many gene programs. Selective ablation of PGC-1α attenuates diet-induced obesity through enhancing fatty acid oxidation and thermogenesis by upregulation of NT-PGC-1α in brown adipose tissue (BAT). Recently, we have shown that selective ablation of NT-PGC-1α reduces fatty acid oxidation in BAT. Thus, the objective of this study was to test our hypothesis that NT-PGC-1α−/− mice would be more prone to diet-induced obesity. Male and female NT-PGC-1α+/+ (WT) and NT-PGC-1α−/− mice were fed a regular chow or 60% high-fat (HF) diet for 16 weeks. Contrary to our expectations, both male and female NT-PGC-1α−/− mice fed HFD were protected from diet-induced obesity, with more pronounced effects in females. This lean phenotype was primarily driven by reduced dietary fat intake. Intriguingly, HFD-fed female, but not male, NT-PGC-1α−/− mice further exhibited decreased feed efficiency, which was closely associated with increased fecal fat excretion and decreased uptake of fatty acids by the intestinal enterocytes and adipocytes with a concomitant decrease in fatty acid transporter gene expression. Collectively, our results highlight the role for NT-PGC-1α in regulating whole body lipid homeostasis under HFD conditions.
Collapse
Affiliation(s)
- Jihyun Kim
- Laboratory of Gene Regulation and Metabolism, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Jiyoung Moon
- Laboratory of Gene Regulation and Metabolism, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Chul-Hong Park
- Laboratory of Gene Regulation and Metabolism, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Jisu Lee
- Laboratory of Gene Regulation and Metabolism, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Helia Cheng
- Laboratory of Gene Regulation and Metabolism, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Z Elizabeth Floyd
- Laboratory of Ubiquitin Biology, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Ji Suk Chang
- Laboratory of Gene Regulation and Metabolism, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
26
|
Cheng Y, Gan-Schreier H, Seeßle J, Staffer S, Tuma-Kellner S, Khnykin D, Stremmel W, Merle U, Herrmann T, Chamulitrat W. Methionine- and Choline-Deficient Diet Enhances Adipose Lipolysis and Leptin Release in aP2-Cre Fatp4-Knockout Mice. Mol Nutr Food Res 2020; 64:e2000361. [PMID: 32991778 DOI: 10.1002/mnfr.202000361] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/22/2020] [Indexed: 12/11/2022]
Abstract
SCOPE Inadequate intake of choline commonly leads to liver diseases. Methionine- and choline-deficient diets (MCDD) induce fatty liver in mice which is partly mediated by triglyceride (TG) lipolysis in white adipose tissues (WATs). Because Fatp4 knockdown has been shown to increase adipocyte lipolysis in vitro, here, the effects of MCDD on WAT lipolysis in aP2-Cre Fatp4-knockout (Fatp4A-/- ) mice are determined. METHODS AND RESULTS Isolated WATs of Fatp4A-/- mice exposed to MCD medium show an increase in lipolysis, and the strongest effect is noted on glycerol release from subcutaneous fat. Fatp4A-/- mice fed with MCDD for 4 weeks show an increase in serum glycerol, TG, and leptin levels associated with the activation of hormone-sensitive lipase in subcutaneous fat. Chow-fed Fatp4A-/- mice also show an increase in serum leptin and very-low-density lipoproteins as well as liver phosphatidylcholine and sphingomyelin levels. Both chow- and MCDD-fed Fatp4A-/- mice show a decrease in serum ketone and WAT sphingomyelin levels which supports a metabolic shift to TG for subsequent WAT lipolysis CONCLUSIONS: Adipose Fatp4 deficiency leads to TG lipolysis and leptin release, which are exaggerated by MCDD. The data imply hyperlipidemia risk by a low dietary choline intake and gene mutations that increase adipose TG levels.
Collapse
Affiliation(s)
- Yuting Cheng
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Hongying Gan-Schreier
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Jessica Seeßle
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Simone Staffer
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Denis Khnykin
- Department of Pathology and Center for Immune Regulation, Rikshospitalet University Hospital, 0424, Oslo, Norway
| | - Wolfgang Stremmel
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Thomas Herrmann
- Westkuesten Hospital, Esmarchstraße 50, 25746, Heide, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, University of Heidelberg Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| |
Collapse
|
27
|
GPAT Gene Silencing in Muscle Reduces Diacylglycerols Content and Improves Insulin Action in Diet-Induced Insulin Resistance. Int J Mol Sci 2020; 21:ijms21197369. [PMID: 33036203 PMCID: PMC7583033 DOI: 10.3390/ijms21197369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/23/2020] [Accepted: 10/02/2020] [Indexed: 02/03/2023] Open
Abstract
Skeletal muscle is an important tissue responsible for glucose and lipid metabolism. High-fat diet (HFD) consumption is associated with the accumulation of bioactive lipids: long chain acyl-CoA, diacylglycerols (DAG) and ceramides. This leads to impaired insulin signaling in skeletal muscle. There is little data on the involvement of DAG in the development of these disorders. Therefore, to clarify this enigma, the gene encoding glycerol-3-phosphate acyltransferase enzyme (GPAT, responsible for DAG synthesis) was silenced through shRNA interference in the gastrocnemius muscle of animals with diet-induced insulin resistance. This work shows that HFD induces insulin resistance, which is accompanied by an increase in the concentration of plasma fatty acids and the level of bioactive lipids in muscle. The increase in these lipids inhibits the insulin pathway and reduces muscle glucose uptake. GPAT silencing through electroporation with shRNA plasmid leads to a reduction in DAG and triacylglycerol (TAG) content, an increase in the activity of the insulin pathway and glucose uptake without a significant effect on ceramide content. This work clearly shows that DAG accumulation has a significant effect on the induction of muscle insulin resistance and that inhibition of DAG synthesis through GPAT modulation may be a potential target in the treatment of insulin resistance.
Collapse
|
28
|
FcRn overexpression in human cancer drives albumin recycling and cell growth; a mechanistic basis for exploitation in targeted albumin-drug designs. J Control Release 2020; 322:53-63. [PMID: 32145268 DOI: 10.1016/j.jconrel.2020.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/11/2020] [Accepted: 03/03/2020] [Indexed: 11/21/2022]
Abstract
Albumin accumulation in tumours could reflect a role of albumin in transport of endogenous nutrient cargos required for cellular growth and not just a suggested source of amino acids; a role driven by albumin engagement with its cognate cellular recycling neonatal Fc receptor. We investigate the hypothesis that albumin cellular recruitment is increased by higher human FcRn (hFcRn) expression in human cancer tissue that provides the mechanistic basis for exploitation in albumin-based drug designs engineered to optimise this process. Eight out of ten different human cancer tissue types screened for hFcRn expression by immunohistochemistry (310 samples) exhibited significantly higher hFcRn expression compared to healthy tissues. Accelerated tumour growth over 28 days in mice inoculated with hFcRn-expressing HT-29 human colorectal cancer cell xenografts, compared to CRISPR/Cas9 hFcRn-knockout HT-29, suggests a hFcRn-mediated tumour growth effect. Direct correlation between hFcRn expression and albumin recycling supports hFcRn-mediated diversion of albumin from lysosomal degradation. Two-fold increase in accumulation of fluorescent labelled high-binding hFcRn albumin, compared to wild type albumin, in luciferase MDA-MB-231-Luc-D3H2LN breast cancer xenografts was shown. This work identifies overexpression of hFcRn in several human cancer types with mechanistic data suggesting hFcRn-driven albumin recruitment for increased cellular growth that has the potential to be exploited with high hFcRn-binding albumin variants for targeted therapies.
Collapse
|
29
|
Yakah W, Singh P, Perides G, Brown J, Freedman SD, Martin CR. Developmental Accretion of Docosahexaenoic Acid Is Independent of Fatty Acid Transporter Expression in Brain and Lung Tissues of C57BL/6 and Fat1 Mice. J Nutr 2019; 149:1724-1731. [PMID: 31179494 PMCID: PMC6768804 DOI: 10.1093/jn/nxz074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/21/2018] [Accepted: 03/26/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Developmental expression of fatty acid transporters and their role in polyunsaturated fatty acid concentrations in the postnatal period have not been evaluated. OBJECTIVE We hypothesized that transporter expression is developmentally regulated, tissue-specific, and that expression can modulate fatty acid accretion independently of diet. METHODS Brain and lung transporter expression were quantified in C57BL/6 wild-type (WT) and Fat1 mice. Pups were dam-fed until day 21. Dams were fed AIN-76A 10% corn oil to represent a typical North American/European diet. After weaning, mice were fed the same diet as dams. Gene expression of Fatp1, Fatp4, Fabp5, and Fat/cd36 was quantified by quantitative reverse transcriptase-polymerase chain reaction. Fatty acid concentrations were measured by GC-MS. RESULTS Brain docosahexaenoic acid (DHA) concentrations increased from day 3 to day 28 in both genotypes, with higher concentrations at days 3 and 14 in Fat1 than in WT mice [median (IQR)]: 10.7 (10.6-11.2) mol% compared with 6.6 (6.4-7.2) mol% and 12.5 (12.4-12.9) mol% compared with 8.9 (8.7-9.1) mol%, respectively; P < 0.05). During DHA accrual, transporter expression decreased. Fold changes in brain Fatp4, Fabp5, and Fat/cd36 were inversely correlated with fold changes in brain DHA concentrations in Fat1 relative to WT mice (ρ = -0.85, -0.75, and -0.78, respectively; P ≤ 0.001). Lung DHA concentrations were unchanged across the 3 time points for both genotypes. Despite unchanging DHA concentrations, there was increased expression of Fatp1 at days 14 and 28 (5-fold), Fatp4 at day 14 (2.3-fold), and Fabp5 at day 14 (3.8-fold) relative to day 3 in Fat1 mice. In WT mice, Fatp1 increased almost 5-fold at day 28 relative to day 3. There was no correlation between lung transporters and DHA concentrations in Fat1 relative to WT mice. CONCLUSIONS Development of fatty acid transporter expression in C57BL/6 WT and Fat1 mice is genotype and tissue specific. Further, postnatal accretion of brain DHA appears independent of transporter status, with tissue concentrations representing dietary contributions.
Collapse
Affiliation(s)
- William Yakah
- Department of Neuroscience, Michigan State University, East Lansing, MI
| | | | | | | | - Steven D Freedman
- Division of Gastroenterology, Boston, MA,Division of Translational Research, Boston, MA
| | - Camilia R Martin
- Division of Translational Research, Boston, MA,Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA,Address correspondence to CRM (e-mail: )
| |
Collapse
|
30
|
Cheng V, Kimball DR, Conboy DJC. Determination of the Rate-Limiting Step in Fatty Acid Transport. J Phys Chem B 2019; 123:7157-7168. [DOI: 10.1021/acs.jpcb.9b05162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Victoria Cheng
- Department of Chemistry, University of Utah, 315 South 1400 East, RM 2020, Salt Lake City, Utah 84112, United States
| | - Dylan R. Kimball
- Department of Chemistry, University of Utah, 315 South 1400 East, RM 2020, Salt Lake City, Utah 84112, United States
| | - Dr. John C. Conboy
- Department of Chemistry, University of Utah, 315 South 1400 East, RM 2020, Salt Lake City, Utah 84112, United States
| |
Collapse
|
31
|
Zong X, Cao X, Wang H, Xiao X, Wang Y, Lu Z. Cathelicidin-WA Facilitated Intestinal Fatty Acid Absorption Through Enhancing PPAR-γ Dependent Barrier Function. Front Immunol 2019; 10:1674. [PMID: 31379865 PMCID: PMC6650583 DOI: 10.3389/fimmu.2019.01674] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/04/2019] [Indexed: 01/28/2023] Open
Abstract
The molecular mechanisms underlying the cellular uptake of long-chain fatty acids and the regulation of this process have been debated in recent decades. Here, we established an intestinal barrier dysfunction model in mice and Caco2 cell line by Lipopolysaccharide (LPS), and evaluated the fatty acid uptake capacity of the intestine. We found that LPS stimulation restricted the absorption of long chain fatty acid (LCFA), while Cathelicidin-WA (CWA) pretreatment facilitated this physiological process. At the molecular level, our results demonstrated that the stimulatory effects of CWA on intestinal lipid absorption were dependent on cluster determinant 36 and fatty acid transport protein 4, but not fatty acid–binding protein. Further, an enhanced intestinal barrier was observed in vivo and in vitro when CWA alleviated the fatty acid absorption disorder induced by LPS stimulation. Mechanistically, peroxisome proliferator-activated receptor (PPAR-γ) signaling was considered as a key pathway for CWA to enhance LCFA absorption and barrier function. Treatment with a PPAR-γ inhibitor led to impaired intestinal barrier function and suppressed LCFA uptake. Moreover, once PPAR-γ signaling was blocked, CWA pretreatment could not maintain the stability of the intestinal epithelial cell barrier or LCFA uptake after LPS stimulation. Collectively, these findings suggested that PPAR-γ may serve as a target for specific therapies aimed at alleviating fatty acid uptake disorder, and CWA showed considerable potential as a new PPAR-γ agonist to strengthen intestinal barrier function against fatty acid malabsorption.
Collapse
Affiliation(s)
- Xin Zong
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoxuan Cao
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Hong Wang
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiao Xiao
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yizhen Wang
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Zeqing Lu
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
32
|
Claus S, Jezierska S, Van Bogaert INA. Protein‐facilitated transport of hydrophobic molecules across the yeast plasma membrane. FEBS Lett 2019; 593:1508-1527. [DOI: 10.1002/1873-3468.13469] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Silke Claus
- Biochemical and Microbial Technology Universiteit Gent Belgium
| | | | - Inge N. A. Van Bogaert
- Lab. of Industrial Microbiology and Biocatalysis Faculty of Bioscience Engineering Ghent University Belgium
| |
Collapse
|
33
|
Shokry E, Raab R, Kirchberg FF, Hellmuth C, Klingler M, Demmelmair H, Koletzko B, Uhl O. Prolonged monitoring of postprandial lipid metabolism after a western meal rich in linoleic acid and carbohydrates. Appl Physiol Nutr Metab 2019; 44:1189-1198. [PMID: 30893569 DOI: 10.1139/apnm-2018-0798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Today, awareness has been raised regarding high consumption of n-6 polyunsaturated fatty acids (PUFA) in western diets. A comprehensive analysis of total and individual postprandial fatty acids profiles would provide insights into metabolic turnover and related health effects. After an overnight fast, 9 healthy adults consumed a mixed meal comprising 97 g carbohydrate and 45 g fat, of which 26.4 g was linoleic acid (LA). Nonesterified fatty acids (NEFA), phospholipid fatty acids (PL-FA) and triacylglycerol fatty acids (TG-FA) were monitored in plasma samples, at baseline and hourly over a 7-h postprandial period. Total TG-FA concentration peaked at 2 h after the meal and steadily decreased thereafter. LA from TG18:2n-6 and behenic acid from TG22:0 showed the highest response among TG-FA, with a biphasic response detected for the former. PL-FA exhibited no change. Total NEFA initially decreased to nadir at 1 h, then increased to peak at 7 h. The individual NEFA showed the same response curve except LA and some very-long-chain saturated fatty acids (VLCSFA, ≥20 carbon chain length) that markedly increased shortly after the meal intake. The similarities and dissimilarities in lipid profiles between study subjects at different time points were visualized using nonmetric multi-dimensional scaling. Overall, the results indicate that postprandial levels of LA and VLCSFA, either as NEFA or TG, were most affected by the test meal, which might provide an explanation for the health effects of this dietary lifestyle characterized by high intake of mixed meals rich in n-6 PUFA.
Collapse
Affiliation(s)
- Engy Shokry
- Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany.,Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany
| | - Roxana Raab
- Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany.,Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany
| | - Franca F Kirchberg
- Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany.,Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany
| | - Christian Hellmuth
- Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany.,Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany
| | - Mario Klingler
- Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany.,Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany
| | - Hans Demmelmair
- Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany.,Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany
| | - Berthold Koletzko
- Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany.,Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany
| | - Olaf Uhl
- Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany.,Ludwig-Maximilians-Universität (LMU) München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Centre, Munich, Germany
| |
Collapse
|
34
|
Tian L, Wen A, Dong S, Yan P. Molecular Characterization of Microtubule Affinity-Regulating Kinase4 from Sus scrofa and Promotion of Lipogenesis in Primary Porcine Placental Trophoblasts. Int J Mol Sci 2019; 20:ijms20051206. [PMID: 30857324 PMCID: PMC6429113 DOI: 10.3390/ijms20051206] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/02/2019] [Accepted: 03/05/2019] [Indexed: 11/28/2022] Open
Abstract
This study aimed to characterize the full-length cDNA of MARK4 in Sus scrofa, and evaluated its potential role in the regulation of lipid accumulation in pig placental trophoblasts and analyzed signaling pathways involved, thereby providing insights into mechanisms for placental lipotoxicity induced by excessive back-fat during pregnancy of sows. The cDNA obtained with 5′ and 3′ RACE amplification covered 3216 bp with an open reading frame of 2259 bp encoding 752 amino acids. Multiple alignments and phylogenetic analysis revealed MARK4 protein of Sus scrofa had a high homology (95%–99%) to that of other higher vertebrates. After transfection, enhanced MARK4 significantly promoted lipogenesis in pig trophoblasts, as evidenced by accelerated lipid accumulation and consistently increased mRNA expressions of lipogenic genes DGAT1, LPIN1, LPIN3, LPL, PPARδ and SREBP-1c. Meanwhile, PPARγ remarkably inhibited the stimulating effect of MARK4 on non-receptor-mediated lipid accumulation in trophoblasts. Further analyses revealed WNT signaling enhanced lipid accumulation and activation of MARK4 in pig trophoblast cells. Finally, we demonstrated that WNT/β-catenin signal pathway is involved in MARK4 activated lipogenesis. These results suggest that MARK4 promotes lipid accumulation in porcine placental trophoblasts and can be considered as a potential regulator of lipotoxicity associated with maternal obesity in the pig placenta.
Collapse
Affiliation(s)
- Liang Tian
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Aiyou Wen
- College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China.
| | - Shusheng Dong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Peishi Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
35
|
Zong X, Zhao J, Wang H, Lu Z, Wang F, Du H, Wang Y. Mettl3 Deficiency Sustains Long-Chain Fatty Acid Absorption through Suppressing Traf6-Dependent Inflammation Response. THE JOURNAL OF IMMUNOLOGY 2018; 202:567-578. [PMID: 30567729 DOI: 10.4049/jimmunol.1801151] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/13/2018] [Indexed: 01/24/2023]
Abstract
A better understanding of the molecular mechanism of intestinal fatty acid absorption could lead to novel approaches to treatment and prevention of fatty acid-related metabolic diseases. Although it is confirmed that absorption of long-chain fatty acids (LCFAs) decreases during the pathological processes, the genetic basis and molecular mechanisms remain largely unknown. N 6-methyladenosine (m6A) is the most prevalent internal modification on eukaryotic mRNA. Recently, m6A has been found to play important roles in inflammation and antiviral responses. In this study, we show that deficiency of Mettl3, the core methyltransferase of m6A, exerts antimalabsorption of LCFA activity in vitro through inhibiting the inflammation response mediated by LPS. To substantiate this finding further, we found the levels of triglycerides were also sustained in cells with depleted Mettl3, which were cultured in Transwell to polarize with villus formation to simulate the situation in vivo. Mechanistically, depletion of Mettl3 decreases the m6A level of Traf6 mRNA, thereby its transcripts are entrapped in the nucleus, followed by the decreased expression of Traf6, leading to the suppression of NF-κB and MAPK signaling pathway. Thus, the inflammation response was suppressed, resulting in the sustained absorption of LCFA. Moreover, we found that ectopic expression of Traf6 largely abolishes the sustained absorption LCFA in Mettl3 depletion cells. Collectively, silencing Mettl3 could sustain LCFA absorption through blocking the TRAF6-dependent inflammation response. Our work uncovers a critical function of m6A methylation and provides insight into critical roles of Mettl3 in LCFA absorption and inflammatory disease.
Collapse
Affiliation(s)
- Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and
| | - Jing Zhao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and
| | - Hong Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and
| | - Zeqing Lu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province 310058, People's Republic of China
| | - Fengqin Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province 310058, People's Republic of China
| | - Huahua Du
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province 310058, People's Republic of China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and .,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province 310058, People's Republic of China
| |
Collapse
|
36
|
Afrin S, Khan MRI, Zhang W, Wang Y, Zhang W, He L, Ma G. Membrane-Located Expression of Thioesterase From Acinetobacter baylyi Enhances Free Fatty Acid Production With Decreased Toxicity in Synechocystis sp. PCC6803. Front Microbiol 2018; 9:2842. [PMID: 30538684 PMCID: PMC6277518 DOI: 10.3389/fmicb.2018.02842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
It has been previously reported that photosynthetic production of extracellular free fatty acids (FFAs) in cyanobacteria was realized by thioesterases (TesA) mediated hydrolysis of fatty acyl-ACP in cytosol and excretion of the FFA outside of the cell. However, two major issues related to the genetically modified strains need to be addressed before the scale-up commercial application becomes possible: namely, the toxicity of FFAs, and the diversity of carbon lengths of fatty acids that could mimic the fossil fuel. To address those issues, we hypothesized that generating FFAs near membrane could facilitate rapid excretion of the FFA outside of the cell and thus decrease toxicity caused by intracellular FFAs in the cytosolic expression of thioesterase. To realize this, we localized a leaderless thioesterase (AcTesA) from Acinetobacter baylyi on the cytosolic side of the inner membrane of Synechocystis sp. PCC6803 using a membrane scaffolding system. The engineered strain with AcTesA on its membrane (mAcT) produced extracellular FFAs up to 171.9 ± 13.22 mg⋅L-1 compared with 40.24 ± 10.94 and 1.904 ± 0.158 mg⋅L-1 in the cytosol-expressed AcTesA (AcT) and wild-type (WT) strains, respectively. Moreover, the mAcT strain generated around 1.5 and 1.9 times less reactive oxygen species than AcT and WT, respectively. Approximately 78% of total FFAs were secreted with an average rate of 1 mg⋅L-1⋅h-1, which was higher than 0.44 mg⋅L-1⋅h-1 reported previously. In the case of mAcT strain, 60% of total secreted FFAs was monounsaturated (C18:1) which is the preferable biodiesel component. Therefore, the engineered mAcT strain shows enhanced FFAs production with less toxicity which is highly desirable for biodiesel production.
Collapse
Affiliation(s)
- Shajia Afrin
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Md Rezaul Islam Khan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Weiyi Zhang
- Shanghai Animal Disease Control Center, Shanghai, China
| | - Yushu Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Weiwen Zhang
- Laboratory of Synthetic Microbiology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Gang Ma
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
37
|
Drews B, Milojevic V, Giller K, Ulbrich S. Fatty acid profile of blood plasma and oviduct and uterine fluid during early and late luteal phase in the horse. Theriogenology 2018; 114:258-265. [DOI: 10.1016/j.theriogenology.2018.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/28/2018] [Accepted: 04/04/2018] [Indexed: 12/21/2022]
|
38
|
The effect of high fat diet and metformin treatment on liver lipids accumulation and their impact on insulin action. Sci Rep 2018; 8:7249. [PMID: 29739997 PMCID: PMC5940807 DOI: 10.1038/s41598-018-25397-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 04/18/2018] [Indexed: 12/27/2022] Open
Abstract
We sought to determine whether metformin treatment reverses a high-fat diet (HFD)-induced hepatic insulin resistance (IRes) and to identify lipid intermediates involved in induction of liver IRes. The experiments were conducted on male Wistar rats divided into three groups: 1. Control, 2. fed HFD and 3. fed HFD and treated with metformin. The animals were infused with a [U-13C]palmitate to measure fractional lipid synthesis rate. This allowed for the calculation of fractional synthesis rate of signaling lipids (FSR) through the estimation of their isotopic enrichment. Liver ceramide (Cer), diacylglycerol (DAG) and acyl-carnitine concentration and enrichment were analyzed by LC/MS/MS. The content of proteins involved in lipid metabolism and insulin signaling were analyzed by Western Blot. HFD treatment increased the content and FSR of DAG and Cer in the liver which was accompanied by systemic insulin resistance and inhibition of hepatic insulin signaling pathway under insulin stimulation. Metformin treatment ameliorated systemic insulin resistance and augmented the hepatic insulin signaling cascade. It reduced both the concentration and FSR of Cer, DAG, and increased acyl-carnitine content and the expression of mitochondrial markers. We postulate, that in liver, the insulin sensitizing effect of metformin depends on augmentation of mitochondrial β-oxidation, which protects from hepatic accumulation of both the Cer and DAG and preserves insulin sensitivity under HFD consumption. Moreover, we showed that hepatic content of Cer and DAG corresponds with their respective FSR.
Collapse
|
39
|
Howell GE, McDevitt E, Henein L, Mulligan C, Young D. "Trans-nonachlor increases extracellular free fatty acid accumulation and de novo lipogenesis to produce hepatic steatosis in McArdle-RH7777 cells". Toxicol In Vitro 2018; 50:285-292. [PMID: 29654899 DOI: 10.1016/j.tiv.2018.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/28/2018] [Accepted: 04/05/2018] [Indexed: 12/19/2022]
Abstract
Recent studies suggest there may be an environmental exposure component to the development and progression of non-alcoholic fatty liver disease (NAFLD) involving the organochlorine (OC) pesticides or their metabolites. However, the roles of OC compounds in the development of NAFLD has not been fully elucidated. Therefore, the current study was designed to determine if exposure to trans-nonachlor, a prevalent OC compound, could promote hepatocyte lipid accumulation and determine potential pro-steatotic mechanisms. McArdle-RH7777 (McA) hepatoma cells were incubated with trans-nonachlor for 24 h then neutral lipid accumulation was determined by Oil Red O staining. Exposure to trans-nonachlor produced a concentration dependent increase in neutral lipid accumulation. Trans-nonachlor also increased extracellular free fatty acid-induced neutral lipid accumulation which appears to be due at least in part to increased free fatty acid accumulation as evident by increased accumulation of Bodipy labeled dodecanoic acid. Additionally, 14C-acetate incorporation into total cellular lipids was increased by trans-nonachlor implicating increased de novo lipogenesis (DNL) as a potential mediator of trans-nonachlor-induced neutral lipid accumulation. Taken together, the present data indicate exposure to trans-nonachlor has a direct, pro-steatotic effect on hepatocytes to increase lipid accumulation through the combinatorial actions of extracellular free fatty acid accumulation and increased DNL.
Collapse
Affiliation(s)
- George Eli Howell
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762, United States.
| | - Erin McDevitt
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762, United States
| | - Lucie Henein
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762, United States
| | - Charlee Mulligan
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762, United States
| | - Darian Young
- Mississippi State University College of Veterinary Medicine, Department of Basic Sciences, Mississippi State, MS 39762, United States
| |
Collapse
|
40
|
Liu P, Peng L, Zhang H, Tang PMK, Zhao T, Yan M, Zhao H, Huang X, Lan H, Li P. Tangshen Formula Attenuates Diabetic Nephropathy by Promoting ABCA1-Mediated Renal Cholesterol Efflux in db/db Mice. Front Physiol 2018; 9:343. [PMID: 29681863 PMCID: PMC5897509 DOI: 10.3389/fphys.2018.00343] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/20/2018] [Indexed: 01/01/2023] Open
Abstract
The commonly prescribed Tangshen Formula (TSF) is a traditional Chinese formulation that has been shown to reduce plasma lipid metabolism and proteinuria and improve the estimated glomerular filtration rate (eGFR) in patients with diabetic kidney disease. This study investigated the underlying mechanism whereby TSF regulates renal lipid accumulation and ameliorates diabetic renal injuries in spontaneous diabetic db/db mice and in vitro in sodium palmitate (PA)-stimulated and Abca1-SiRNA-transfected mouse tubular epithelial cells (mTECs). The results revealed that TSF treatment significantly ameliorated the renal injuries by lowering urinary albumin excretion and improving renal tissue injuries in diabetic (db/db) mice. Interestingly, the treatment with TSF also resulted in decreased cholesterol levels in the renal tissues of db/db mice, which was associated with increased expression of the peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), the Liver X receptors (LXR), and ATP-binding cassette subfamily A member 1 (ABCA1), suggesting that TSF might attenuate diabetic kidney injury via a mechanism associated with improving cholesterol efflux in the diabetic kidney. This was investigated in vitro in mTECs, and the results showed that TSF reduced the PA-stimulated cholesterol accumulation in mTECs. Mechanistically, the addition of TSF was capable of reversing PA-induced downregulation of PGC-1α, LXR, and ABCA1 expression and cholesterol accumulation in mTECs, suggesting that TSF might act the protection via the PGC-1α-LXR-ABCA1 pathway to improve the cholesterol efflux in the renal tissues of db/db mice. This was further confirmed by silencing ABCA1 to block the promotive effect of TSF on cholesterol efflux in vitro. In conclusion, TSF might ameliorate diabetic kidney injuries by promoting ABCA1-mediated renal cholesterol efflux.
Collapse
Affiliation(s)
- Peng Liu
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.,Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Liang Peng
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Haojun Zhang
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tingting Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Meihua Yan
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Hailing Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoru Huang
- Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Huiyao Lan
- Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ping Li
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
41
|
Cifarelli V, Abumrad NA. Intestinal CD36 and Other Key Proteins of Lipid Utilization: Role in Absorption and Gut Homeostasis. Compr Physiol 2018; 8:493-507. [PMID: 29687890 PMCID: PMC6247794 DOI: 10.1002/cphy.c170026] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Several proteins have been implicated in fatty acid (FA) transport by enterocytes including the scavenger receptor CD36 (SR-B2), the scavenger receptor B1 (SR-B1) a member of the CD36 family and the FA transport protein 4 (FATP4). Here, we review the regulation of enterocyte FA uptake and its function in lipid absorption including prechylomicron formation, assembly and transport. Emphasis is given to CD36, which is abundantly expressed along the digestive tract of rodents and humans and has been the most studied. We also address the pleiotropic functions of CD36 that go beyond lipid absorption and metabolism to include recent evidence of its impact on intestinal homeostasis and barrier maintenance. Areas of progress involving contribution of membrane phospholipid remodeling and of cytosolic FA-binding proteins, FABP1 and FABP2 to fat absorption will be covered. © 2018 American Physiological Society. Compr Physiol 8:493-507, 2018.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Internal Medicine, Center for Human Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| | - Nada A. Abumrad
- Department of Internal Medicine, Center for Human Nutrition, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
42
|
Khan S, Cabral PD, Schilling WP, Schmidt ZW, Uddin AN, Gingras A, Madhavan SM, Garvin JL, Schelling JR. Kidney Proximal Tubule Lipoapoptosis Is Regulated by Fatty Acid Transporter-2 (FATP2). J Am Soc Nephrol 2017; 29:81-91. [PMID: 28993506 DOI: 10.1681/asn.2017030314] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/08/2017] [Indexed: 11/03/2022] Open
Abstract
Albuminuria and tubular atrophy are among the highest risks for CKD progression to ESRD. A parsimonious mechanism involves leakage of albumin-bound nonesterified fatty acids (NEFAs) across the damaged glomerular filtration barrier and subsequent reabsorption by the downstream proximal tubule, causing lipoapoptosis. We sought to identify the apical proximal tubule transporter that mediates NEFA uptake and cytotoxicity. We observed transporter-mediated uptake of fluorescently labeled NEFA in cultured proximal tubule cells and microperfused rat proximal tubules, with greater uptake from the apical surface than from the basolateral surface. Protein and mRNA expression analyses revealed that kidney proximal tubules express transmembrane fatty acid transporter-2 (FATP2), encoded by Slc27a2, but not the other candidate transporters CD36 and free fatty acid receptor 1. Kidney FATP2 localized exclusively to proximal tubule epithelial cells along the apical but not the basolateral membrane. Treatment of mice with lipidated albumin to induce proteinuria caused a decrease in the proportion of tubular epithelial cells and an increase in the proportion of interstitial space in kidneys from wild-type but not Slc27a2-/- mice. Ex vivo microperfusion and in vitro experiments with NEFA-bound albumin at concentrations that mimic apical proximal tubule exposure during glomerular injury revealed significantly reduced NEFA uptake and palmitate-induced apoptosis in microperfused Slc27a2-/- proximal tubules and Slc27a2-/- or FATP2 shRNA-treated proximal tubule cell lines compared with wild-type or scrambled oligonucleotide-treated cells, respectively. We conclude that FATP2 is a major apical proximal tubule NEFA transporter that regulates lipoapoptosis and may be an amenable target for the prevention of CKD progression.
Collapse
Affiliation(s)
- Shenaz Khan
- Department of Medicine, The MetroHealth System and
| | - Pablo D Cabral
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - William P Schilling
- Department of Medicine, The MetroHealth System and.,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| | | | - Asif N Uddin
- Department of Medicine, The MetroHealth System and
| | | | | | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| | | |
Collapse
|
43
|
Lee E, Choi J, Lee HS. Palmitate induces mitochondrial superoxide generation and activates AMPK in podocytes. J Cell Physiol 2017; 232:3209-3217. [PMID: 28214337 DOI: 10.1002/jcp.25867] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/17/2017] [Indexed: 12/21/2022]
Abstract
Studies have shown that high levels of serum free fatty acids (FFAs) are associated with lipotoxicity and type 2 diabetes. Palmitic acid (PA) is the predominant circulating saturated FFA, yet its role in the pathogenesis of diabetic nephropathy (DN) is not clear. Recently, one study suggested that mitochondrial superoxide production is related to AMP-activated protein kinase (AMPK) activity in diabetic mice kidneys. To elucidate the link between PA and oxidative stress and AMPK activity in DN, we compared the cultured murine podocytes exposed to PA and oleic acid (OA). Incubation of cells with 250 μM PA or OA induced a translocation of CD36, a fatty acid transport protein, with intracellular lipid accumulation. PA, but not OA, induced mitochondrial superoxide and hydrogen peroxide (H2 O2 ) generation in podocytes, as shown by enhanced fluorescence of MitoSOX Red and dichlorofluorescein (DCF), respectively. Costimulation of PA-treated cells with the H2 O2 scavenger catalase abolished the PA-induced DCF fluorescence. Only PA induced mitochondrial damage as shown by electron microscopy. The AMPK activity was determined by immunoblotting, measuring the ratio of phosphorylated AMPK (p-AMPK) to total AMPK. Only PA significantly increased the p-AMPK levels compared with controls. Addition of catalase to PA-treated cells did not affect the PA-stimulated p-AMPK levels. Collectively, our results indicate that PA induces mitochondrial superoxide and H2 O2 generation in cultured podocytes, which may not be directly linked to AMPK activation. Given that, PA seems to play an important role in the pathogenesis of DN through lipotoxicity initiated by mitochondrial superoxide overproduction.
Collapse
Affiliation(s)
- Eugene Lee
- Renal Pathology Lab, Hankook Kidney and Diabetes Institute, Seoul, Korea
| | - Jin Choi
- Renal Pathology Lab, Hankook Kidney and Diabetes Institute, Seoul, Korea
| | - Hyun Soon Lee
- Renal Pathology Lab, Hankook Kidney and Diabetes Institute, Seoul, Korea
| |
Collapse
|
44
|
Rezhdo O, Speciner L, Carrier R. Lipid-associated oral delivery: Mechanisms and analysis of oral absorption enhancement. J Control Release 2016; 240:544-560. [PMID: 27520734 PMCID: PMC5082615 DOI: 10.1016/j.jconrel.2016.07.050] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 01/29/2023]
Abstract
The majority of newly discovered oral drugs are poorly water soluble, and co-administration with lipids has proven effective in significantly enhancing bioavailability of some compounds with low aqueous solubility. Yet, lipid-based delivery technologies have not been widely employed in commercial oral products. Lipids can impact drug transport and fate in the gastrointestinal (GI) tract through multiple mechanisms including enhancement of solubility and dissolution kinetics, enhancement of permeation through the intestinal mucosa, and triggering drug precipitation upon lipid emulsion depletion (e.g., by digestion). The effect of lipids on drug absorption is currently not quantitatively predictable, in part due to the multiple complex dynamic processes that can be impacted by lipids. Quantitative mechanistic analysis of the processes significant to lipid system function and overall impact on drug absorption can aid in the understanding of drug-lipid interactions in the GI tract and exploitation of such interactions to achieve optimal lipid-based drug delivery. In this review, we discuss the impact of co-delivered lipids and lipid digestion on drug dissolution, partitioning, and absorption in the context of the experimental tools and associated kinetic expressions used to study and model these processes. The potential benefit of a systems-based consideration of the concurrent multiple dynamic processes occurring upon co-dosing lipids and drugs to predict the impact of lipids on drug absorption and enable rational design of lipid-based delivery systems is presented.
Collapse
Affiliation(s)
- Oljora Rezhdo
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, United States
| | - Lauren Speciner
- Department of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, United States
| | - Rebecca Carrier
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, United States.
| |
Collapse
|
45
|
The role of CD36 in the regulation of myocardial lipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1450-60. [DOI: 10.1016/j.bbalip.2016.03.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 12/29/2022]
|
46
|
Effects of Chain Length and Saturability of Fatty Acids on Phospholipids and Proteins in Plasma Membranes of Bovine Mammary Gland. J Membr Biol 2016; 249:743-756. [DOI: 10.1007/s00232-016-9919-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 07/16/2016] [Indexed: 10/21/2022]
|
47
|
Howell III GE, Mulligan C, Young D, Kondakala S. Exposure to chlorpyrifos increases neutral lipid accumulation with accompanying increased de novo lipogenesis and decreased triglyceride secretion in McArdle-RH7777 hepatoma cells. Toxicol In Vitro 2016; 32:181-9. [DOI: 10.1016/j.tiv.2016.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 12/09/2015] [Accepted: 01/06/2016] [Indexed: 01/14/2023]
|
48
|
Pascual F, Coleman RA. Fuel availability and fate in cardiac metabolism: A tale of two substrates. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1425-33. [PMID: 26993579 DOI: 10.1016/j.bbalip.2016.03.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/12/2022]
Abstract
The heart's extraordinary metabolic flexibility allows it to adapt to normal changes in physiology in order to preserve its function. Alterations in the metabolic profile of the heart have also been attributed to pathological conditions such as ischemia and hypertrophy; however, research during the past decade has established that cardiac metabolic adaptations can precede the onset of pathologies. It is therefore critical to understand how changes in cardiac substrate availability and use trigger events that ultimately result in heart dysfunction. This review examines the mechanisms by which the heart obtains fuels from the circulation or from mobilization of intracellular stores. We next describe experimental models that exhibit either an increase in glucose use or a decrease in FA oxidation, and how these aberrant conditions affect cardiac metabolism and function. Finally, we highlight the importance of alternative, relatively under-investigated strategies for the treatment of heart failure. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.
Collapse
Affiliation(s)
- Florencia Pascual
- Department of Nutrition, University of North Carolina at Chapel Hill, 27599, USA.
| | - Rosalind A Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, 27599, USA.
| |
Collapse
|
49
|
Abstract
Various intestinal functions exhibit circadian rhythmicity. Disruptions in these rhythms as in shift workers and transcontinental travelers are associated with intestinal discomfort. Circadian rhythms are controlled at the molecular level by core clock and clock-controlled genes. These clock genes are expressed in intestinal cells, suggesting that they might participate in the circadian regulation of intestinal functions. A major function of the intestine is nutrient absorption. Here, we will review absorption of proteins, carbohydrates, and lipids and circadian regulation of various transporters involved in their absorption. A better understanding of circadian regulation of intestinal absorption might help control several metabolic disorders and attenuate intestinal discomfort associated with disruptions in sleep-wake cycles.
Collapse
Affiliation(s)
- M Mahmood Hussain
- Department of Cell Biology and Pediatrics, SUNY Downstate Medical Center, Brooklyn, New York, USA, and VA New York Harbor Healthcare System, Brooklyn, New York, USA
| | - Xiaoyue Pan
- Department of Cell Biology and Pediatrics, SUNY Downstate Medical Center, Brooklyn, New York, USA, and VA New York Harbor Healthcare System, Brooklyn, New York, USA
| |
Collapse
|
50
|
Dose apolipoprotein AV has influence on the regulation of fatty acids and triglyceride metabolism in cardiomyocyte in case of obesity. Int J Cardiol 2015; 185:109-11. [PMID: 25791104 DOI: 10.1016/j.ijcard.2015.03.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/07/2015] [Indexed: 11/23/2022]
|