1
|
Ma Y, Dong T, Luan F, Yang J, Miao F, Wei P. Interaction of major facilitator superfamily domain containing 2A with the blood-brain barrier. Neural Regen Res 2025; 20:2133-2152. [PMID: 39248155 DOI: 10.4103/nrr.nrr-d-24-00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/08/2024] [Indexed: 09/10/2024] Open
Abstract
The functional and structural integrity of the blood-brain barrier is crucial in maintaining homeostasis in the brain microenvironment; however, the molecular mechanisms underlying the formation and function of the blood-brain barrier remain poorly understood. The major facilitator superfamily domain containing 2A has been identified as a key regulator of blood-brain barrier function. It plays a critical role in promoting and maintaining the formation and functional stability of the blood-brain barrier, in addition to the transport of lipids, such as docosahexaenoic acid, across the blood-brain barrier. Furthermore, an increasing number of studies have suggested that major facilitator superfamily domain containing 2A is involved in the molecular mechanisms of blood-brain barrier dysfunction in a variety of neurological diseases; however, little is known regarding the mechanisms by which major facilitator superfamily domain containing 2A affects the blood-brain barrier. This paper provides a comprehensive and systematic review of the close relationship between major facilitator superfamily domain containing 2A proteins and the blood-brain barrier, including their basic structures and functions, cross-linking between major facilitator superfamily domain containing 2A and the blood-brain barrier, and the in-depth studies on lipid transport and the regulation of blood-brain barrier permeability. This comprehensive systematic review contributes to an in-depth understanding of the important role of major facilitator superfamily domain containing 2A proteins in maintaining the structure and function of the blood-brain barrier and the research progress to date. This will not only help to elucidate the pathogenesis of neurological diseases, improve the accuracy of laboratory diagnosis, and optimize clinical treatment strategies, but it may also play an important role in prognostic monitoring. In addition, the effects of major facilitator superfamily domain containing 2A on blood-brain barrier leakage in various diseases and the research progress on cross-blood-brain barrier drug delivery are summarized. This review may contribute to the development of new approaches for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yilun Ma
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Taiwei Dong
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Fei Luan
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Juanjuan Yang
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi'an, Shaanxi Province, China
| | - Feng Miao
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Peifeng Wei
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi'an, Shaanxi Province, China
| |
Collapse
|
2
|
Hachem M, Ali AH, Yildiz I, Landry C, Gosselet F. Investigation of Lysophospholipids-DHA transport across an in vitro human model of blood brain barrier. Heliyon 2024; 10:e38871. [PMID: 39421371 PMCID: PMC11483317 DOI: 10.1016/j.heliyon.2024.e38871] [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: 05/10/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Several studies emphasized on the preventive and therapeutic potential of Docosahexaenoic Acid (DHA, 22:6n-3) supplementation in chronic and age-related disorders including neurodegenerative diseases. Researchers principally studied the cerebral accretion of Lysophosphatidylcholine (LysoPC-DHA), the furthermost vital Lysophospholipid-DHA (LysoPL-DHA) in blood plasma. Nevertheless, the cerebral bioavailability of other LysoPL-DHA forms including Lysophosphatidylethanolamine (LysoPE-DHA), and Lysophosphatidylserine (LysoPS-DHA) were not extensively examined even though their vital biological functions in the brain. Hence, the aim of the present study was to evaluate the toxicity and transport of DHA in comparison to several LysoPL-DHA including LysoPC-DHA, LysoPE-DHA and LysoPS-DHA across a human model of blood-brain barrier (BBB). The human brain-like endothelial cells (hBLECs) monolayer tightness was evaluated by the parallel assessment of the permeability of fluorescent marker Lucifer yellow (LY) and revealed the absence of toxicity of non-esterified DHA and all LysoPL-DHA towards hBLECs. LysoPC-DHA, LysoPE-DHA and LysoPS-DHA displayed a higher recovery in the abluminal medium in comparison to non-esterified DHA at 30, 60 and 120 min post-incubation. Among all, LysoPS-DHA revealed the highest apparent coefficient permeability (Papp) 85.87 ± 4.24 x 10-6 cm s-1 and was significantly different than DHA, LysoPC-DHA and LysoPE-DHA. More interestingly, when studying the time course of Papp of DHA, LysoPC-DHA and LysoPE-DHA, at different post-incubation time, this permeability decreases with time especially for LysoPC-DHA and LysoPE-DHA, not for DHA. Furthermore, LysoPS-DHA exhibited the highest intracellular accumulation (10.39 ± 0.49 %) in hBLECs in comparison to all other tested lipids. Finally, differences in 3D structures and molecular electrostatic potential maps calculation of LysoPL-DHA could explain the dissimilar cerebral uptake of LysoPL-DHA. Altogether, our findings raise the novel hypothesis that LysoPS-DHA may represent a preferred physiological carrier of DHA to the brain.
Collapse
Affiliation(s)
- Mayssa Hachem
- Department of Chemistry, College of Engineering and Physical Sciences, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Abdelmoneim H. Ali
- Department of Chemical & Petroleum Engineering, College of Engineering and Physical Sciences, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Ibrahim Yildiz
- Department of Chemistry, Khalifa University of Sciences and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Christophe Landry
- Artois University, Blood-Brain Barrier Laboratory, UR 2465, Lens, France
| | - Fabien Gosselet
- Artois University, Blood-Brain Barrier Laboratory, UR 2465, Lens, France
| |
Collapse
|
3
|
Powell TL, Barentsen K, Vaughan O, Uhlson C, Zemski Berry K, Erickson K, Faer K, Chassen SS, Jansson T. Knockdown of Placental Major Facilitator Superfamily Domain Containing 2a in Pregnant Mice Reduces Fetal Brain Growth and Phospholipid Docosahexaenoic Acid Content. Nutrients 2023; 15:4956. [PMID: 38068814 PMCID: PMC10708493 DOI: 10.3390/nu15234956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
INTRODUCTION Docosahexaenoic acid (DHA) is an n-3 long chain polyunsaturated fatty acid critical for fetal brain development that is transported to the fetus from the mother by the placenta. The lysophosphatidylcholine (LPC) transporter, Major Facilitator Superfamily Domain Containing 2a (MFSD2a), is localized in the basal plasma membrane of the syncytiotrophoblast of the human placenta, and MFSD2a expression correlates with umbilical cord blood LPC-DHA levels in human pregnancy. We hypothesized that placenta-specific knockdown of MFSD2a in pregnant mice reduces phospholipid DHA accumulation in the fetal brain. METHODS Mouse blastocysts (E3.5) were transduced with an EGFP-expressing lentivirus containing either an shRNA targeting MFSD2a or a non-coding sequence (SCR), then transferred to pseudopregnant females. At E18.5, fetuses were weighed and their placenta, brain, liver and plasma were collected. MFSD2a mRNA expression was determined by qPCR in the brain, liver and placenta and phospholipid DHA was quantified by LC-MS/MS. RESULTS MFSD2a-targeting shRNA reduced placental mRNA MFSD2a expression by 38% at E18.5 (n = 45, p < 0.008) compared with SCR controls. MFSD2a expression in the fetal brain and liver were unchanged. Fetal brain weight was reduced by 13% (p = 0.006). Body weight, placenta and liver weights were unaffected. Fetal brain phosphatidyl choline and phosphatidyl ethanolamine DHA content was lower in fetuses with placenta-specific MFSD2a knockdown. CONCLUSIONS Placenta-specific reduction in expression of the LPC-DHA transporter MFSD2a resulted in reduced fetal brain weight and lower phospholipid DHA content in the fetal brain. These data provide mechanistic evidence that placental MFSD2a mediates maternal-fetal transfer of LPC-DHA, which is critical for brain growth.
Collapse
Affiliation(s)
- Theresa L. Powell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Kenneth Barentsen
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Owen Vaughan
- Department of Maternal and Fetal Medicine, EGA Institute for Women’s Heath, University College London, 86-96 Chenies Mews, London WC1E 6HX, UK
| | - Charis Uhlson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Karin Zemski Berry
- Department of Medicine, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Kathryn Erickson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Kelsey Faer
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Stephanie S. Chassen
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Chin CF, Galam DL, Gao L, Tan BC, Wong BH, Chua GL, Loke RY, Lim YC, Wenk MR, Lim MS, Leow WQ, Goh GB, Torta F, Silver DL. Blood-derived lysophospholipid sustains hepatic phospholipids and fat storage necessary for hepatoprotection in overnutrition. J Clin Invest 2023; 133:e171267. [PMID: 37463052 PMCID: PMC10471173 DOI: 10.1172/jci171267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/12/2023] [Indexed: 09/02/2023] Open
Abstract
The liver has a high demand for phosphatidylcholine (PC), particularly in overnutrition, where reduced phospholipid levels have been implicated in the development of nonalcoholic fatty liver disease (NAFLD). Whether other pathways exist in addition to de novo PC synthesis that contribute to hepatic PC pools remains unknown. Here, we identified the lysophosphatidylcholine (LPC) transporter major facilitator superfamily domain containing 2A (Mfsd2a) as critical for maintaining hepatic phospholipid pools. Hepatic Mfsd2a expression was induced in patients having NAFLD and in mice in response to dietary fat via glucocorticoid receptor action. Mfsd2a liver-specific deficiency in mice (L2aKO) led to a robust nonalcoholic steatohepatitis-like (NASH-like) phenotype within just 2 weeks of dietary fat challenge associated with reduced hepatic phospholipids containing linoleic acid. Reducing dietary choline intake in L2aKO mice exacerbated liver pathology and deficiency of liver phospholipids containing polyunsaturated fatty acids (PUFAs). Treating hepatocytes with LPCs containing oleate and linoleate, two abundant blood-derived LPCs, specifically induced lipid droplet biogenesis and contributed to phospholipid pools, while LPC containing the omega-3 fatty acid docosahexaenoic acid (DHA) promoted lipid droplet formation and suppressed lipogenesis. This study revealed that PUFA-containing LPCs drive hepatic lipid droplet formation, suppress lipogenesis, and sustain hepatic phospholipid pools - processes that are critical for protecting the liver from excess dietary fat.
Collapse
Affiliation(s)
- Cheen Fei Chin
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Dwight L.A. Galam
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Liang Gao
- Singapore Lipidomics Incubator, Life Sciences Institute and
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Bryan C. Tan
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Bernice H. Wong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Geok-Lin Chua
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Randy Y.J. Loke
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Yen Ching Lim
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Markus R. Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute and
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Miao-Shan Lim
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Wei-Qiang Leow
- Department of Anatomical Pathology, Singapore General Hospital, and
| | - George B.B. Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
- Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute and
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - David L. Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| |
Collapse
|
5
|
Cheng YJ, Fan F, Zhang Z, Zhang HJ. Lipid metabolism in malignant tumor brain metastasis: reprogramming and therapeutic potential. Expert Opin Ther Targets 2023; 27:861-878. [PMID: 37668244 DOI: 10.1080/14728222.2023.2255377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 07/19/2023] [Accepted: 08/31/2023] [Indexed: 09/06/2023]
Abstract
INTRODUCTION Brain metastasis is a highly traumatic event in the progression of malignant tumors, often symbolizing higher mortality. Metabolic alterations are hallmarks of cancer, and the mask of lipid metabolic program rearrangement in cancer progression is gradually being unraveled. AREAS COVERED In this work, we reviewed clinical and fundamental studies related to lipid expression and activity changes in brain metastases originating from lung, breast, and cutaneous melanomas, respectively. Novel roles of lipid metabolic reprogramming in the development of brain metastasis from malignant tumors were identified and its potential as a therapeutic target was evaluated. Published literature and clinical studies in databases consisting of PubMed, Embase, Scopus and www.ClinicalTrials.gov from 1990 to 2022 were searched. EXPERT OPINION Lipid metabolic reprogramming in brain metastasis is involved in de novo lipid synthesis within low lipid availability environments, regulation of lipid uptake and storage, metabolic interactions between brain tumors and the brain microenvironment, and membrane lipid remodeling, in addition to being a second messenger for signal transduction. Although some lipid metabolism modulators work efficiently in preclinical models, there is still a long way to go from laboratory to clinic. This area of research holds assurance for the organ-targeted treatment of brain metastases through drug-regulated metabolic targets and dietary interventions.
Collapse
Affiliation(s)
- Yan-Jie Cheng
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, People's Republic of China
- Department of Oncology, Shanghai Fengxian District Central Hospital, Shanghai, China
| | - Fan Fan
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Zhong Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Hai-Jun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
6
|
Chen H, Ahmed S, Zhao H, Elghobashi-Meinhardt N, Dai Y, Kim JH, McDonald JG, Li X, Lee CH. Structural and functional insights into Spns2-mediated transport of sphingosine-1-phosphate. Cell 2023; 186:2644-2655.e16. [PMID: 37224812 PMCID: PMC10330195 DOI: 10.1016/j.cell.2023.04.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/23/2023] [Accepted: 04/19/2023] [Indexed: 05/26/2023]
Abstract
Sphingosine-1-phosphate (S1P) is an important signaling sphingolipid that regulates the immune system, angiogenesis, auditory function, and epithelial and endothelial barrier integrity. Spinster homolog 2 (Spns2) is an S1P transporter that exports S1P to initiate lipid signaling cascades. Modulating Spns2 activity can be beneficial in treatments of cancer, inflammation, and immune diseases. However, the transport mechanism of Spns2 and its inhibition remain unclear. Here, we present six cryo-EM structures of human Spns2 in lipid nanodiscs, including two functionally relevant intermediate conformations that link the inward- and outward-facing states, to reveal the structural basis of the S1P transport cycle. Functional analyses suggest that Spns2 exports S1P via facilitated diffusion, a mechanism distinct from other MFS lipid transporters. Finally, we show that the Spns2 inhibitor 16d attenuates the transport activity by locking Spns2 in the inward-facing state. Our work sheds light on Spns2-mediated S1P transport and aids the development of advanced Spns2 inhibitors.
Collapse
Affiliation(s)
- Hongwen Chen
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shahbaz Ahmed
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hongtu Zhao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Yaxin Dai
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jae Hun Kim
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaochun Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Chia-Hsueh Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
7
|
Nguyen C, Lei HT, Lai LTF, Gallenito MJ, Mu X, Matthies D, Gonen T. Lipid flipping in the omega-3 fatty-acid transporter. Nat Commun 2023; 14:2571. [PMID: 37156797 PMCID: PMC10167227 DOI: 10.1038/s41467-023-37702-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/28/2023] [Indexed: 05/10/2023] Open
Abstract
Mfsd2a is the transporter for docosahexaenoic acid (DHA), an omega-3 fatty acid, across the blood brain barrier (BBB). Defects in Mfsd2a are linked to ailments from behavioral and motor dysfunctions to microcephaly. Mfsd2a transports long-chain unsaturated fatty-acids, including DHA and α-linolenic acid (ALA), that are attached to the zwitterionic lysophosphatidylcholine (LPC) headgroup. Even with the recently determined structures of Mfsd2a, the molecular details of how this transporter performs the energetically unfavorable task of translocating and flipping lysolipids across the lipid bilayer remains unclear. Here, we report five single-particle cryo-EM structures of Danio rerio Mfsd2a (drMfsd2a): in the inward-open conformation in the ligand-free state and displaying lipid-like densities modeled as ALA-LPC at four distinct positions. These Mfsd2a snapshots detail the flipping mechanism for lipid-LPC from outer to inner membrane leaflet and release for membrane integration on the cytoplasmic side. These results also map Mfsd2a mutants that disrupt lipid-LPC transport and are associated with disease.
Collapse
Affiliation(s)
- Chi Nguyen
- Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsiang-Ting Lei
- Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA, 20147, USA
| | - Louis Tung Faat Lai
- Unit on Structural Biology, Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marc J Gallenito
- Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xuelang Mu
- Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Doreen Matthies
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA, 20147, USA.
- Unit on Structural Biology, Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Tamir Gonen
- Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Departments of Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
8
|
Chua GL, Tan BC, Loke RYJ, He M, Chin CF, Wong BH, Kuk ACY, Ding M, Wenk MR, Guan L, Torta F, Silver DL. Mfsd2a utilizes a flippase mechanism to mediate omega-3 fatty acid lysolipid transport. Proc Natl Acad Sci U S A 2023; 120:e2215290120. [PMID: 36848557 PMCID: PMC10013850 DOI: 10.1073/pnas.2215290120] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/27/2023] [Indexed: 03/01/2023] Open
Abstract
Major Facilitator Superfamily Domain containing 2a (Mfsd2a) is a sodium-dependent lysophosphatidylcholine (LPC) transporter expressed at the blood-brain barrier that constitutes the main pathway by which the brain obtains omega-3 fatty acids, such as docosahexanoic acid. Mfsd2a deficiency in humans results in severe microcephaly, underscoring the importance of LPC transport by Mfsd2a for brain development. Biochemical studies and recent cryo-electron microscopy (cryo-EM) structures of Mfsd2a bound to LPC suggest that Mfsd2a transports LPC via an alternating access mechanism between outward-facing and inward-facing conformational states in which the LPC inverts during transport between the outer and inner leaflet of a membrane. However, direct biochemical evidence of flippase activity by Mfsd2a has not been demonstrated and it is not understood how Mfsd2a could invert LPC between the outer and inner leaflet of the membrane in a sodium-dependent manner. Here, we established a unique in vitro assay using recombinant Mfsd2a reconstituted in liposomes that exploits the ability of Mfsd2a to transport lysophosphatidylserine (LPS) coupled with a small molecule LPS binding fluorophore that allowed for monitoring of directional flipping of the LPS headgroup from the outer to the inner liposome membrane. Using this assay, we demonstrate that Mfsd2a flips LPS from the outer to the inner leaflet of a membrane bilayer in a sodium-dependent manner. Furthermore, using cryo-EM structures as guides together with mutagenesis and a cell-based transport assay, we identify amino acid residues important for Mfsd2a activity that likely constitute substrate interaction domains. These studies provide direct biochemical evidence that Mfsd2a functions as a lysolipid flippase.
Collapse
Affiliation(s)
- Geok-Lin Chua
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Bryan C. Tan
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Randy Y. J. Loke
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Menglan He
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Cheen-Fei Chin
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Bernice H. Wong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Alvin C. Y. Kuk
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| | - Mei Ding
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore117456, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117596, Singapore
| | - Markus R. Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore117456, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117596, Singapore
| | - Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX79430
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore117456, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117596, Singapore
| | - David L. Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore169857, Singapore
| |
Collapse
|
9
|
Hasannejad-Asl B, Pooresmaeil F, Choupani E, Dabiri M, Behmardi A, Fadaie M, Fathi M, Moosavi SA, Takamoli S, Hemati E, Naei VY, Kazemi-Lomedasht F. Nanoparticles as Powerful Tools for Crossing the Blood-brain Barrier. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:18-26. [PMID: 35196974 DOI: 10.2174/1871527321666220222092655] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/06/2022] [Accepted: 01/16/2022] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier (BBB) is considered an important protective barrier in the central nervous system (CNS). The barrier is mainly formed by endothelial cells (ECs) interconnected by various junctions such as tight junctions (TJs), gap junctions, and adherent junctions. They collectively constitute an intensive barrier to the transit of different substances into the brain, selectively permitting small molecules to pass through by passive movement but holding off large ones such as peptides and proteins to cross the brain. Hence some molecules selectively transfer across the BBB by active routes via transcytosis. The BBB also forms a barrier against neurotoxins as well as pathogenic agents. Although various CNS disorders like Alzheimer's disease (AD) and Parkinson's disease (PD) could hamper the integrity of the border. Nevertheless, the BBB acts as a barrier for CNS disorders treatment because it prevents the drugs from reaching their target in the CNS. In recent years, different strategies, including osmotic disruption of BBB or chemical modification of drugs, have been used to transfer the chemotherapeutic agents into brain substances. Nowadays, nanoparticles (NPs) have been used as an effective and non-invasive tool for drug delivery and diagnosis of CNS disorders. In this review, we discuss the structural characteristic of BBB, safe passageways to cross the BBB, and the relation of barrier lesions with different CNS disorders. In the end, we explore the progress in drug delivery, diagnosis, imaging, and treatment of CNS disorders using nanoparticles.
Collapse
Affiliation(s)
- Behnam Hasannejad-Asl
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farkhondeh Pooresmaeil
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Edris Choupani
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mehran Dabiri
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Abtin Behmardi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mahmood Fadaie
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Majid Fathi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Seyed Akbar Moosavi
- Department of Medical Laboratory Sciences, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Neuroscience Research Center (NRC) The Iran University of Medical Science, Allied Health Department, Tehran, Iran
- Tehran Women Hospital, AST Genetic Lab, Tehran, Iran
| | - Shahla Takamoli
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Ehsan Hemati
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Yaghoubi Naei
- Immunology Research Center, Mashhad University of Medical Science, Mashhad, Iran
| | - Fatemeh Kazemi-Lomedasht
- Department of Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
10
|
Lo Van A, Bernoud-Hubac N, Lagarde M. Esterification of Docosahexaenoic Acid Enhances Its Transport to the Brain and Its Potential Therapeutic Use in Brain Diseases. Nutrients 2022; 14:4550. [PMID: 36364810 PMCID: PMC9656701 DOI: 10.3390/nu14214550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 10/22/2023] Open
Abstract
Docosahexaenoic acid-containing lysophosphatidylcholine (DHA-LysoPC) is presented as the main transporter of DHA from blood plasma to the brain. This is related to the major facilitator superfamily domain-containing protein 2A (Mfsd2a) symporter expression in the blood-brain barrier that recognizes the various lyso-phospholipids that have choline in their polar head. In order to stabilize the DHA moiety at the sn-2 position of LysoPC, the sn-1 position was esterified by the shortest acetyl chain, creating the structural phospholipid 1-acetyl,2-docosahexaenoyl-glycerophosphocholine (AceDoPC). This small structure modification allows the maintaining of the preferential brain uptake of DHA over non-esterified DHA. Additional properties were found for AceDoPC, such as antioxidant properties, especially due to the aspirin-like acetyl moiety, as well as the capacity to generate acetylcholine in response to the phospholipase D cleavage of the polar head. Esterification of DHA within DHA-LysoPC or AceDoPC could elicit more potent neuroprotective effects against neurological diseases.
Collapse
Affiliation(s)
- Amanda Lo Van
- Univ Lyon, INSA Lyon, CNRS, LaMCoS, UMR5259, 69621 Villeurbanne, France
| | | | | |
Collapse
|
11
|
Cui XY, Jiang S, Wang CC, Yang JY, Zhao YC, Xue CH, Wang YM, Zhang TT. Comparative Analyses of EPA-Phosphatidylcholine, EPA-Lysophosphatidylcholine, and DHA-Lysophosphatidylcholine on DHA and EPA Repletion in n-3 PUFA-Deficient Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:13327-13339. [PMID: 36197792 DOI: 10.1021/acs.jafc.2c06462] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) play an important role in maintaining the physiological functions of tissues, and the beneficial effects of DHA/EPA in phospholipid forms have been widely reported. Although lysophosphatidylcholine (LPC) is considered to be the preferred form of DHA supplementation for the brain, the kinetics of DHA and EPA recovery and corresponding changes of n-6 docosapentaenoic acid (DPA) and arachidonic acid (AA) levels in different phospholipid molecules and different tissues after administration of EPA in phosphatidylcholine (PC) and LPC forms and DHA in the LPC form are not clear. Here, we measured the total fatty acids in tissues and fatty acid composition of different phospholipid molecules after gavage administration of equal molar amounts of EPA/DHA in mice with n-3 polyunsaturated fatty acid (PUFA) deficiency induced by maternal dietary deprivation of n-3 PUFA during pregnancy and lactation. The results showed that dietary supplementation with EPA-PC, EPA-LPC, and DHA-LPC exhibited different priorities for EPA or DHA accretion and supplementation efficiency curves in different tissues during the developing period. EPA-PC exhibited a more optimal efficacy in DHA and EPA repletion in serum and hepatic total fatty acids. In terms of DHA recovery in the brain, EPA-LPC and DHA-LPC showed great effects. Meanwhile, the DHA level in total fatty acids and major fractions of phospholipids (PC, PE, and PI + PS) in the heart and bone marrow with the supplementation of DHA-LPC displayed a relatively considerable increase compared with that of EPA supplementation groups. The study provides a reference for the time course of DHA or EPA recovery in phospholipid molecular species in different tissues after the supplementation of EPA-PC, EPA-LPC, and DHA-LPC.
Collapse
Affiliation(s)
- Xiao-Yu Cui
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| | - Shan Jiang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| | - Jin-Yue Yang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| | - Ying-Cai Zhao
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
- Laboratory of Marine Drugs & Biological Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, Shandong, P. R. China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
- Laboratory of Marine Drugs & Biological Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, Shandong, P. R. China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| |
Collapse
|
12
|
Spns1 is a lysophospholipid transporter mediating lysosomal phospholipid salvage. Proc Natl Acad Sci U S A 2022; 119:e2210353119. [PMID: 36161949 DOI: 10.1073/pnas.2210353119] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The lysosome is central to the degradation of proteins, carbohydrates, and lipids and their salvage back to the cytosol for reutilization. Lysosomal transporters for amino acids, sugars, and cholesterol have been identified, and the metabolic fates of these molecules in the cytoplasm have been elucidated. Remarkably, it is not known whether lysosomal salvage exists for glycerophospholipids, the major constituents of cellular membranes. By using a transport assay screen against orphan lysosomal transporters, we identified the major facilitator superfamily protein Spns1 that is ubiquitously expressed in all tissues as a proton-dependent lysophosphatidylcholine (LPC) and lysophosphatidylethanolamine (LPE) transporter, with LPC and LPE being the lysosomal breakdown products of the most abundant eukaryotic phospholipids, phosphatidylcholine and phosphatidylethanolamine, respectively. Spns1 deficiency in cells, zebrafish embryos, and mouse liver resulted in lysosomal accumulation of LPC and LPE species with pathological consequences on lysosomal function. Flux analysis using stable isotope-labeled phospholipid apolipoprotein E nanodiscs targeted to lysosomes showed that LPC was transported out of lysosomes in an Spns1-dependent manner and re-esterified back into the cytoplasmic pools of phosphatidylcholine. Our findings identify a phospholipid salvage pathway from lysosomes to the cytosol that is dependent on Spns1 and critical for maintaining normal lysosomal function.
Collapse
|
13
|
Popov LD. Deciphering the relationship between caveolae-mediated intracellular transport and signalling events. Cell Signal 2022; 97:110399. [PMID: 35820545 DOI: 10.1016/j.cellsig.2022.110399] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
Abstract
The caveolae-mediated transport across polarized epithelial cell barriers has been largely deciphered in the last decades and is considered the second essential intracellular transfer mechanism, after the clathrin-dependent endocytosis. The basic cell biology knowledge was supplemented recently, with the molecular mechanisms beyond caveolae generation implying the key contribution of the lipid-binding proteins (the structural protein Caveolin and the adapter protein Cavin), along with the bulb coat stabilizing molecules PACSIN-2 and Eps15 homology domain protein-2. The current attention is focused also on caveolae architecture (such as the bulb coat, the neck, the membrane funnel inside the bulb, and the associated receptors), and their specific tasks during the intracellular transport of various cargoes. Here, we resume the present understanding of the assembly, detachment, and internalization of caveolae from the plasma membrane lipid raft domains, and give an updated view on transcytosis and endocytosis, the two itineraries of cargoes transport via caveolae. The review adds novel data on the signalling molecules regulating caveolae intracellular routes and on the transport dysregulation in diseases. The therapeutic possibilities offered by exploitation of Caveolin-1 expression and caveolae trafficking, and the urgent issues to be uncovered conclude the review.
Collapse
Affiliation(s)
- Lucia-Doina Popov
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania.
| |
Collapse
|
14
|
Walter JD, Remm S, Seeger MA. Fatty acid transporter MFSD2A is a multifunctional gatekeeper in brain and placenta. Nat Struct Mol Biol 2022; 29:504-506. [PMID: 35710837 PMCID: PMC7615496 DOI: 10.1038/s41594-022-00788-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
MFSD2A mediates uptake of the essential fatty acid DHA across the blood–brain barrier. Separately, via interactions with syncytin-2, MFSD2A contributes to the formation of the mother–fetus placental boundary. Cryo-EM analysis of a human MFSD2A–syncytin-2 complex provides new insights into how MFSD2A performs these dual roles.
Collapse
Affiliation(s)
- Justin D Walter
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland.
| | - Sille Remm
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Markus A Seeger
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
15
|
Structural insights into the lysophospholipid brain uptake mechanism and its inhibition by syncytin-2. Nat Struct Mol Biol 2022; 29:604-612. [PMID: 35710838 DOI: 10.1038/s41594-022-00786-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 05/01/2022] [Indexed: 12/29/2022]
Abstract
Brain development and function require uptake of essential omega-3 fatty acids in the form of lysophosphatidylcholine via major-facilitator superfamily transporter MFSD2A, a potential pharmaceutical target to modulate blood-brain barrier (BBB) permeability. MFSD2A is also the receptor of endogenous retroviral envelope syncytin-2 (SYNC2) in human placenta, where it mediates cell-cell fusion and formation of the maternal-fetal interface. Here, we report a cryo-electron microscopy structure of the human MFSD2A-SYNC2 complex that reveals a large hydrophobic cavity in the transporter C-terminal domain to occlude long aliphatic chains. The transporter architecture suggests an alternating-access transport mechanism for lipid substrates in mammalian MFS transporters. SYNC2 establishes an extensive binding interface with MFSD2A, and a SYNC2-soluble fragment acts as a long-sought-after inhibitor of MFSD2A transport. Our work uncovers molecular mechanisms important to brain and placenta development and function, and SYNC2-mediated inhibition of MFSD2A transport suggests strategies to aid delivery of therapeutic macromolecules across the BBB.
Collapse
|
16
|
Wong BH, Mei D, Chua GL, Galam DL, Wenk MR, Torta F, Silver DL. The lipid transporter Mfsd2a maintains pulmonary surfactant homeostasis. J Biol Chem 2022; 298:101709. [PMID: 35150739 PMCID: PMC8914330 DOI: 10.1016/j.jbc.2022.101709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/18/2022] Open
Abstract
Pulmonary surfactant is a lipoprotein complex essential for lung function, and insufficiency or altered surfactant composition is associated with major lung diseases, such as acute respiratory distress syndromes, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. Pulmonary surfactant is primarily composed of phosphatidylcholine (PC) in complex with specialized surfactant proteins and secreted by alveolar type 2 (AT2) cells. Surfactant homeostasis on the alveolar surface is balanced by the rates of synthesis and secretion with reuptake and recycling by AT2 cells, with some degradation by pulmonary macrophages and loss up the bronchial tree. However, whether phospholipid (PL) transporters exist in AT2 cells to mediate reuptake of surfactant PL remains to be identified. Here, we demonstrate that major facilitator superfamily domain containing 2a (Mfsd2a), a sodium-dependent lysophosphatidylcholine (LPC) transporter, is expressed at the apical surface of AT2 cells. A mouse model with inducible AT2 cell–specific deficiency of Mfsd2a exhibited AT2 cell hypertrophy with reduced total surfactant PL levels because of reductions in the most abundant surfactants, PC containing dipalmitic acid, and PC species containing the omega-3 fatty acid docosahexaenoic acid. These changes in surfactant levels and composition were mirrored by similar changes in the AT2 cell lipidome. Mechanistically, direct tracheal instillation of fluorescent LPC and PC probes indicated that Mfsd2a mediates the uptake of LPC generated by pulmonary phospholipase activity in the alveolar space. These studies reveal that Mfsd2a-mediated LPC uptake is quantitatively important in maintaining surfactant homeostasis and identify this lipid transporter as a physiological component of surfactant recycling.
Collapse
Affiliation(s)
- Bernice H Wong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Ding Mei
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Geok Lin Chua
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Dwight L Galam
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David L Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
17
|
Lambert E, Mehdipour AR, Schmidt A, Hummer G, Perez C. Evidence for a trap-and-flip mechanism in a proton-dependent lipid transporter. Nat Commun 2022; 13:1022. [PMID: 35197476 PMCID: PMC8866510 DOI: 10.1038/s41467-022-28361-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/23/2022] [Indexed: 02/08/2023] Open
Abstract
Transport of lipids across membranes is fundamental for diverse biological pathways in cells. Multiple ion-coupled transporters take part in lipid translocation, but their mechanisms remain largely unknown. Major facilitator superfamily (MFS) lipid transporters play central roles in cell wall synthesis, brain development and function, lipids recycling, and cell signaling. Recent structures of MFS lipid transporters revealed overlapping architectural features pointing towards a common mechanism. Here we used cysteine disulfide trapping, molecular dynamics simulations, mutagenesis analysis, and transport assays in vitro and in vivo, to investigate the mechanism of LtaA, a proton-dependent MFS lipid transporter essential for lipoteichoic acid synthesis in the pathogen Staphylococcus aureus. We reveal that LtaA displays asymmetric lateral openings with distinct functional relevance and that cycling through outward- and inward-facing conformations is essential for transport activity. We demonstrate that while the entire amphipathic central cavity of LtaA contributes to lipid binding, its hydrophilic pocket dictates substrate specificity. We propose that LtaA catalyzes lipid translocation by a ‘trap-and-flip’ mechanism that might be shared among MFS lipid transporters. LtaA catalyzes glycolipid translocation by a ‘trap-and-flip’ mechanism, pointing to a shared mechanistic model among MFS lipid transporters. Asymmetric lateral openings allow access of the entire lipid substrate to the amphipathic central cavity.
Collapse
Affiliation(s)
| | | | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Gerhard Hummer
- Institute of Biophysics, Goethe University Frankfurt, Frankfurt am Main, Germany.,Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Camilo Perez
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
18
|
Lin ZZ, Li ZQ, Li JJ, Yu CL, Yang CW, Ran JS, Yin LQ, Zhang DH, Zhang GF, Liu YP. Mfsd2a Promotes the Proliferation, Migration, Differentiation and Adipogenesis of Chicken Intramuscular Preadipocytes. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2022. [DOI: 10.1590/1806-9061-2021-1547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- ZZ Lin
- Sichuan Agricultural University, China
| | - ZQ Li
- Sichuan Agricultural University, China
| | - JJ Li
- Sichuan Agricultural University, China
| | - CL Yu
- Sichuan Animal Science Academy, China
| | - CW Yang
- Sichuan Animal Science Academy, China
| | - JS Ran
- Sichuan Agricultural University, China
| | - LQ Yin
- Sichuan Agricultural University, China
| | - DH Zhang
- Sichuan Agricultural University, China
| | - GF Zhang
- Sichuan Agricultural University, China
| | - YP Liu
- Sichuan Agricultural University, China
| |
Collapse
|
19
|
Nguyen YTK, Ha HTT, Nguyen TH, Nguyen LN. The role of SLC transporters for brain health and disease. Cell Mol Life Sci 2021; 79:20. [PMID: 34971415 PMCID: PMC11071821 DOI: 10.1007/s00018-021-04074-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/05/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
The brain exchanges nutrients and small molecules with blood via the blood-brain barrier (BBB). Approximately 20% energy intake for the body is consumed by the brain. Glucose is known for its critical roles for energy production and provides substrates for biogenesis in neurons. The brain takes up glucose via glucose transporters GLUT1 and 3, which are expressed in several neural cell types. The brain is also equipped with various transport systems for acquiring amino acids, lactate, ketone bodies, lipids, and cofactors for neuronal functions. Unraveling the mechanisms by which the brain takes up and metabolizes these nutrients will be key in understanding the nutritional requirements in the brain. This could also offer opportunities for therapeutic interventions in several neurological disorders. For instance, emerging evidence suggests a critical role of lactate as an alternative energy source for neurons. Neuronal cells express monocarboxylic transporters to acquire lactate. As such, treatment of GLUT1-deficient patients with ketogenic diets to provide the brain with alternative sources of energy has been shown to improve the health of the patients. Many transporters are present in the brain, but only a small number has been characterized. In this review, we will discuss about the roles of solute carrier (SLC) transporters at the blood brain barrier (BBB) and neural cells, in transport of nutrients and metabolites in the brain.
Collapse
Affiliation(s)
- Yen T K Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Hoa T T Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Tra H Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
- SLING/Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational and Cardiovascular Disease Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
| |
Collapse
|
20
|
Spieth L, Berghoff SA, Stumpf SK, Winchenbach J, Michaelis T, Watanabe T, Gerndt N, Düking T, Hofer S, Ruhwedel T, Shaib AH, Willig K, Kronenberg K, Karst U, Frahm J, Rhee JS, Minguet S, Möbius W, Kruse N, von der Brelie C, Michels P, Stadelmann C, Hülper P, Saher G. Anesthesia triggers drug delivery to experimental glioma in mice by hijacking caveolar transport. Neurooncol Adv 2021; 3:vdab140. [PMID: 34647026 PMCID: PMC8500692 DOI: 10.1093/noajnl/vdab140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background Pharmaceutical intervention in the CNS is hampered by the shielding function of the blood–brain barrier (BBB). To induce clinical anesthesia, general anesthetics such as isoflurane readily penetrate the BBB. Here, we investigated whether isoflurane can be utilized for therapeutic drug delivery. Methods Barrier function in primary endothelial cells was evaluated by transepithelial/transendothelial electrical resistance, and nanoscale STED and SRRF microscopy. In mice, BBB permeability was quantified by extravasation of several fluorescent tracers. Mouse models including the GL261 glioma model were evaluated by MRI, immunohistochemistry, electron microscopy, western blot, and expression analysis. Results Isoflurane enhances BBB permeability in a time- and concentration-dependent manner. We demonstrate that, mechanistically, isoflurane disturbs the organization of membrane lipid nanodomains and triggers caveolar transport in brain endothelial cells. BBB tightness re-establishes directly after termination of anesthesia, providing a defined window for drug delivery. In a therapeutic glioblastoma trial in mice, simultaneous exposure to isoflurane and cytotoxic agent improves efficacy of chemotherapy. Conclusions Combination therapy, involving isoflurane-mediated BBB permeation with drug administration has far-reaching therapeutic implications for CNS malignancies.
Collapse
Affiliation(s)
- Lena Spieth
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Stefan A Berghoff
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Sina K Stumpf
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Jan Winchenbach
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Thomas Michaelis
- Max-Planck-Institut für biophysikalische Chemie, Biomedizinische NMR, Göttingen, Germany
| | - Takashi Watanabe
- Max-Planck-Institut für biophysikalische Chemie, Biomedizinische NMR, Göttingen, Germany
| | - Nina Gerndt
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Tim Düking
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Sabine Hofer
- Max-Planck-Institut für biophysikalische Chemie, Biomedizinische NMR, Göttingen, Germany
| | - Torben Ruhwedel
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany.,Max-Planck-Institute of Experimental Medicine, Electron Microscopy Core Unit, Göttingen, Germany
| | - Ali H Shaib
- Max-Planck-Institute of Experimental Medicine, Department of Molecular Neurobiology, Göttingen, Germany
| | - Katrin Willig
- Max-Planck-Institute of Experimental Medicine, Group of Optical Nanoscopy in Neuroscience, Göttingen, Germany.,University Medical Center, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Katharina Kronenberg
- Westfälische Wilhelms-Universität Münster, Institute of Inorganic and Analytical Chemistry, Münster, Germany
| | - Uwe Karst
- Westfälische Wilhelms-Universität Münster, Institute of Inorganic and Analytical Chemistry, Münster, Germany
| | - Jens Frahm
- Max-Planck-Institut für biophysikalische Chemie, Biomedizinische NMR, Göttingen, Germany
| | - Jeong Seop Rhee
- Max-Planck-Institute of Experimental Medicine, Department of Molecular Neurobiology, Göttingen, Germany
| | - Susana Minguet
- Albert-Ludwigs-University of Freiburg, Faculty of Biology, Freiburg, Germany. Signalling Research Centres BIOSS and CIBSS, Freiburg, Germany. Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Wiebke Möbius
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany.,Max-Planck-Institute of Experimental Medicine, Electron Microscopy Core Unit, Göttingen, Germany.,University Medical Center, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Niels Kruse
- University Medical Center Göttingen, Institute for Neuropathology, Göttingen, Germany
| | | | - Peter Michels
- University Medical Center Göttingen, Institute for Anesthesiology, Göttingen, Germany
| | - Christine Stadelmann
- University Medical Center Göttingen, Institute for Neuropathology, Göttingen, Germany
| | - Petra Hülper
- Klinikum Oldenburg, Oldenburg, University Hospital, Germany
| | - Gesine Saher
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| |
Collapse
|
21
|
Huang B, Li X. The Role of Mfsd2a in Nervous System Diseases. Front Neurosci 2021; 15:730534. [PMID: 34566571 PMCID: PMC8461068 DOI: 10.3389/fnins.2021.730534] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022] Open
Abstract
Major facilitator superfamily (MFS) is the maximum and most diversified membrane transporter, acting as uniporters, symporters and antiporters. MFS is considered to have a good development potential in the transport of drugs for the treatment of brain diseases. The major facilitator superfamily domain containing protein 2a (Mfsd2a) is a member of MFS. Mfsd2a-knockout mice have shown a marked decrease of docosahexaenoic acid (DHA) level in brain, exhibiting neuron loss, microcephaly and cognitive deficits, as DHA acts essentially in brain growth and integrity. Mfsd2a has attracted more and more attention in the study of nervous system diseases because of its critical role in maintaining the integrity of the blood-brain barrier (BBB) and transporting DHA, including inhibiting cell transport in central nervous system endothelial cells, alleviating BBB injury, avoiding BBB injury in cerebral hemorrhage model, acting as a carrier etc. Up to now, the clinical research of Mfsd2a in nervous system diseases is rare. This article reviewed the current research progress of Mfsd2a in nervous system diseases. It summarized the physiological functions of Mfsd2a in the occurrence and development of intracranial hemorrhage (ICH), Alzheimer's disease (AD), sepsis-associated encephalopathy (SAE), autosomal recessive primary microcephaly (MCPH) and intracranial tumor, aiming to provide ideas for the basic research and clinical application of Mfsd2a.
Collapse
Affiliation(s)
- Bei Huang
- Operational Management Office, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Khuller K, Yigit G, Martínez Grijalva C, Altmüller J, Thiele H, Nürnberg P, Elcioglu NH, Yeter B, Hehr U, Stein A, Della Marina A, Köninger A, Depienne C, Kaiser FJ, Wollnik B, Kuechler A. MFSD2A-associated primary microcephaly - Expanding the clinical and mutational spectrum of this ultra-rare disease. Eur J Med Genet 2021; 64:104310. [PMID: 34400370 DOI: 10.1016/j.ejmg.2021.104310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/25/2021] [Accepted: 08/12/2021] [Indexed: 01/07/2023]
Abstract
MFSD2A, a member of the major facilitator superfamily (MFS), is a transmembrane transporter responsible for the uptake of specific essential fatty acids through the blood-brain barrier (BBB) to the brain. The transporter is crucial for early embryonic brain development and a major factor in the formation and maintenance of the BBB. Mfsd2a-knockout mice show a leakage of the BBB in early embryonic stages and develop a phenotype characterized by microcephaly, cognitive impairment, and anxiety. So far, homozygous or compound heterozygous MFSD2A mutations in humans have only been reported in 13 different families with a total of 28 affected individuals. The phenotypical spectrum of patients with MFSD2A variants is rather broad but all patients present with microcephaly and severe intellectual disability, absent or limited speech, and walking difficulties. Severely affected patients develop seizures and show brain malformations and have, above all, a profound developmental delay hardly reaching any developmental motor milestones. Here, we report on two unrelated individuals with novel homozygous variants in the MFSD2A gene, presenting with severe primary microcephaly, brain malformations, profound developmental delay, and epilepsy, including hypsarrhythmia. Our findings extend the mutational spectrum of the bi-allelic MFSD2A variants causing autosomal recessive primary microcephaly type 15 and broaden the phenotypic spectrum associated with these pathogenic variants emphasizing the role of MFSD2A in early brain development.
Collapse
Affiliation(s)
| | - Gökhan Yigit
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany.
| | | | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.
| | - Nursel H Elcioglu
- Department of Pediatric Genetics, Marmara University Medical School, Istanbul, Turkey; Eastern Mediterranean University School of Medicine, Cyprus, Mersin, 10, Turkey.
| | - Burcu Yeter
- Department of Pediatric Genetics, Marmara University Medical School, Istanbul, Turkey.
| | - Ute Hehr
- Center for Human Genetics, and Department of Human Genetics, University of Regensburg, Regensburg, Germany.
| | - Anja Stein
- Department of Pediatrics, Neonatology, University Hospital Essen, Germany.
| | - Adela Della Marina
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University of Essen, Germany, University Hospital Essen, Germany.
| | - Angela Köninger
- Department of Obstetrics and Gynaecology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | | | - Frank J Kaiser
- Institute for Human Genetics, University Hospital Essen, Germany.
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| | - Alma Kuechler
- Institute for Human Genetics, University Hospital Essen, Germany.
| |
Collapse
|
23
|
Guan L, Hariharan P. X-ray crystallography reveals molecular recognition mechanism for sugar binding in a melibiose transporter MelB. Commun Biol 2021; 4:931. [PMID: 34341464 PMCID: PMC8329300 DOI: 10.1038/s42003-021-02462-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022] Open
Abstract
Major facilitator superfamily_2 transporters are widely found from bacteria to mammals. The melibiose transporter MelB, which catalyzes melibiose symport with either Na+, Li+, or H+, is a prototype of the Na+-coupled MFS transporters, but its sugar recognition mechanism has been a long-unsolved puzzle. Two high-resolution X-ray crystal structures of a Salmonella typhimurium MelB mutant with a bound ligand, either nitrophenyl-α-d-galactoside or dodecyl-β-d-melibioside, were refined to a resolution of 3.05 or 3.15 Å, respectively. In the substrate-binding site, the interaction of both galactosyl moieties on the two ligands with MelBSt are virturally same, so the sugar specificity determinant pocket can be recognized, and hence the molecular recognition mechanism for sugar binding in MelB has been deciphered. The conserved cation-binding pocket is also proposed, which directly connects to the sugar specificity pocket. These key structural findings have laid a solid foundation for our understanding of the cooperative binding and symport mechanisms in Na+-coupled MFS transporters, including eukaryotic transporters such as MFSD2A. Guan and Hariharan report two crystal structures of melibiose transporter MelB in complex with substrate analogs, nitrophenyl-galactoside, and dodecyl-melibioside. Both structures revealed similar specific site for sugar recognition and resolved the cation-binding pocket, advancing the understanding of MelB and related transporters.
Collapse
Affiliation(s)
- Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Parameswaran Hariharan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
24
|
Wood CAP, Zhang J, Aydin D, Xu Y, Andreone BJ, Langen UH, Dror RO, Gu C, Feng L. Structure and mechanism of blood-brain-barrier lipid transporter MFSD2A. Nature 2021; 596:444-448. [PMID: 34349262 PMCID: PMC8884080 DOI: 10.1038/s41586-021-03782-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
MFSD2A is a sodium-dependent lysophosphatidylcholine symporter that is responsible for the uptake of docosahexaenoic acid into the brain1,2, which is crucial for the development and performance of the brain3. Mutations that affect MFSD2A cause microcephaly syndromes4,5. The ability of MFSD2A to transport lipid is also a key mechanism that underlies its function as an inhibitor of transcytosis to regulate the blood-brain barrier6,7. Thus, MFSD2A represents an attractive target for modulating the permeability of the blood-brain barrier for drug delivery. Here we report the cryo-electron microscopy structure of mouse MFSD2A. Our structure defines the architecture of this important transporter, reveals its unique extracellular domain and uncovers its substrate-binding cavity. The structure-together with our functional studies and molecular dynamics simulations-identifies a conserved sodium-binding site, reveals a potential lipid entry pathway and helps to rationalize MFSD2A mutations that underlie microcephaly syndromes. These results shed light on the critical lipid transport function of MFSD2A and provide a framework to aid in the design of specific modulators for therapeutic purposes.
Collapse
Affiliation(s)
- Chase A P Wood
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jinru Zhang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Deniz Aydin
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Computer Science, Stanford University, Stanford, CA, USA.,Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Yan Xu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Urs H Langen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ron O Dror
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Computer Science, Stanford University, Stanford, CA, USA.,Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Liang Feng
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
25
|
Cater RJ, Chua GL, Erramilli SK, Keener JE, Choy BC, Tokarz P, Chin CF, Quek DQY, Kloss B, Pepe JG, Parisi G, Wong BH, Clarke OB, Marty MT, Kossiakoff AA, Khelashvili G, Silver DL, Mancia F. Structural basis of omega-3 fatty acid transport across the blood-brain barrier. Nature 2021; 595:315-319. [PMID: 34135507 PMCID: PMC8266758 DOI: 10.1038/s41586-021-03650-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023]
Abstract
Docosahexaenoic acid is an omega-3 fatty acid that is essential for neurological development and function, and it is supplied to the brain and eyes predominantly from dietary sources1-6. This nutrient is transported across the blood-brain and blood-retina barriers in the form of lysophosphatidylcholine by major facilitator superfamily domain containing 2A (MFSD2A) in a Na+-dependent manner7,8. Here we present the structure of MFSD2A determined using single-particle cryo-electron microscopy, which reveals twelve transmembrane helices that are separated into two pseudosymmetric domains. The transporter is in an inward-facing conformation and features a large amphipathic cavity that contains the Na+-binding site and a bound lysolipid substrate, which we confirmed using native mass spectrometry. Together with our functional analyses and molecular dynamics simulations, this structure reveals details of how MFSD2A interacts with substrates and how Na+-dependent conformational changes allow for the release of these substrates into the membrane through a lateral gate. Our work provides insights into the molecular mechanism by which this atypical major facility superfamily transporter mediates the uptake of lysolipids into the brain, and has the potential to aid in the delivery of neurotherapeutic agents.
Collapse
Affiliation(s)
- Rosemary J Cater
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Geok Lin Chua
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Satchal K Erramilli
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - James E Keener
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Brendon C Choy
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Piotr Tokarz
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Cheen Fei Chin
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Debra Q Y Quek
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Brian Kloss
- Center on Membrane Protein Production and Analysis, New York Structural Biology Center, New York, NY, USA
| | - Joseph G Pepe
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Giacomo Parisi
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Bernice H Wong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael T Marty
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, NY, USA.
- Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| | - David L Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
26
|
Nguyen TQ, Vu TM, Tukijan F, Muralidharan S, Foo JC, Li Chin JF, Hasan Z, Torta F, Nguyen LN. Erythrocytes efficiently utilize exogenous sphingosines for S1P synthesis and export via Mfsd2b. J Biol Chem 2020; 296:100201. [PMID: 33334894 PMCID: PMC7948482 DOI: 10.1074/jbc.ra120.012941] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 11/24/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a potent lipid mediator that exerts its activity via activation of five different G protein–coupled receptors, designated as S1P1–5. This potent lipid mediator is synthesized from the sphingosine precursor by two sphingosine kinases (SphK1 and 2) and must be exported to exert extracellular signaling functions. We recently identified Mfsd2b as the S1P transporter in the hematopoietic system. However, the sources of sphingosine for S1P synthesis and the transport mechanism of Mfsd2b in erythrocytes remain to be determined. Here, we show that erythrocytes efficiently take up exogenous sphingosine and that a de novo synthesis pathway in part provides sphingosines to erythrocytes. The uptake of sphingosine in erythrocytes is facilitated by the activity of SphK1. By converting sphingosine into S1P, SphK1 indirectly increases the influx of sphingosine, a process that is irreversible in erythrocytes. Our results explain for the abnormally high amount of sphingosine accumulation in Mfsd2b knockout erythrocytes. Furthermore, we show that Mfsd2b utilizes a proton gradient to facilitate the release of S1P. The negatively charged residues D95 and T157 are essential for Mfsd2b transport activity. Of interest, we also discovered an S1P analog that inhibits S1P export from erythrocytes, providing evidence that sphingosine analogs can be used to inhibit S1P export by Mfsd2b. Collectively, our results highlight that erythrocytes are efficient in sphingosine uptake for S1P production and the release of S1P is dependent on Mfsd2b functions.
Collapse
Affiliation(s)
- Toan Q Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Thiet Minh Vu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Farhana Tukijan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sneha Muralidharan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Juat Chin Foo
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Zafrul Hasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Federico Torta
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; SLING/Immunology Program, Life Sciences Institute, National University of Singapore, Singapore; Immunology Translational and Cardiovascular Disease Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
27
|
Emerging roles of lysophospholipids in health and disease. Prog Lipid Res 2020; 80:101068. [PMID: 33068601 DOI: 10.1016/j.plipres.2020.101068] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 12/22/2022]
Abstract
Lipids are abundant and play essential roles in human health and disease. The main functions of lipids are building blocks for membrane biogenesis. However, lipids are also metabolized to produce signaling molecules. Here, we discuss the emerging roles of circulating lysophospholipids. These lysophospholipids consist of lysoglycerophospholipids and lysosphingolipids. They are both present in cells at low concentration, but their concentrations in extracellular fluids are significantly higher. The biological functions of some of these lysophospholipids have been recently revealed. Remarkably, some of the lysophospholipids play pivotal signaling roles as well as being precursors for membrane biogenesis. Revealing how circulating lysophospholipids are produced, released, transported, and utilized in multi-organ systems is critical to understand their functions. The discovery of enzymes, carriers, transporters, and membrane receptors for these lysophospholipids has shed light on their physiological significance. In this review, we summarize the biological roles of these lysophospholipids via discussing about the proteins regulating their functions. We also discuss about their potential impacts to human health and diseases.
Collapse
|
28
|
Semba RD. Perspective: The Potential Role of Circulating Lysophosphatidylcholine in Neuroprotection against Alzheimer Disease. Adv Nutr 2020; 11:760-772. [PMID: 32190891 PMCID: PMC7360459 DOI: 10.1093/advances/nmaa024] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/02/2020] [Accepted: 02/19/2020] [Indexed: 12/28/2022] Open
Abstract
Alzheimer disease (AD), the most common cause of dementia, is a progressive disorder involving cognitive impairment, loss of learning and memory, and neurodegeneration affecting wide areas of the cerebral cortex and hippocampus. AD is characterized by altered lipid metabolism in the brain. Lower concentrations of long-chain PUFAs have been described in the frontal cortex, entorhinal cortex, and hippocampus in the brain in AD. The brain can synthesize only a few fatty acids; thus, most fatty acids must enter the brain from the blood. Recent studies show that PUFAs such as DHA (22:6) are transported across the blood-brain barrier (BBB) in the form of lysophosphatidylcholine (LPC) via a specific LPC receptor at the BBB known as the sodium-dependent LPC symporter 1 (MFSD2A). Higher dietary PUFA intake is associated with decreased risk of cognitive decline and dementia in observational studies; however, PUFA supplementation, with fatty acids esterified in triacylglycerols did not prevent cognitive decline in clinical trials. Recent studies show that LPC is the preferred carrier of PUFAs across the BBB into the brain. An insufficient pool of circulating LPC containing long-chain fatty acids could potentially limit the supply of long-chain fatty acids to the brain, including PUFAs such as DHA, and play a role in the pathobiology of AD. Whether adults with low serum LPC concentrations are at greater risk of developing cognitive decline and AD remains a major gap in knowledge. Preventing and treating cognitive decline and the development of AD remain a major challenge. The LPC pathway is a promising area for future investigators to identify modifiable risk factors for AD.
Collapse
Affiliation(s)
- Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Biallelic MFSD2A variants associated with congenital microcephaly, developmental delay, and recognizable neuroimaging features. Eur J Hum Genet 2020; 28:1509-1519. [PMID: 32572202 DOI: 10.1038/s41431-020-0669-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/30/2020] [Accepted: 06/04/2020] [Indexed: 12/15/2022] Open
Abstract
Major Facilitator Superfamily Domain containing 2a (MFSD2A) is an essential endothelial lipid transporter at the blood-brain barrier. Biallelic variants affecting function in MFSD2A cause autosomal recessive primary microcephaly 15 (MCPH15, OMIM# 616486). We sought to expand our knowledge of the phenotypic spectrum of MCPH15 and demonstrate the underlying mechanism of inactivation of the MFSD2A transporter. We carried out detailed analysis of the clinical and neuroradiological features of a series of 27 MCPH15 cases, including eight new individuals from seven unrelated families. Genetic investigation was performed through exome sequencing (ES). Structural insights on the human Mfsd2a model and in-vitro biochemical assays were used to investigate the functional impact of the identified variants. All patients had primary microcephaly and severe developmental delay. Brain MRI showed variable degrees of white matter reduction, ventricular enlargement, callosal hypodysgenesis, and pontine and vermian hypoplasia. ES led to the identification of six novel biallelic MFSD2A variants (NG_053084.1, NM_032793.5: c.556+1G>A, c.748G>T; p.(Val250Phe), c.750_753del; p.(Cys251SerfsTer3), c.977G>A; p.(Arg326His), c.1386_1435del; p.(Gln462HisfsTer17), and c.1478C>T; p.(Pro493Leu)) and two recurrent variants (NM_032793.5: c.593C>T; p.(Thr198Met) and c.476C>T; p.(Thr159Met)). All these variants and the previously reported NM_032793.5: c.490C>A; p.(Pro164Thr) resulted in either reduced MFSD2A expression and/or transport activity. Our study further delineates the phenotypic spectrum of MCPH15, refining its clinical and neuroradiological characterization and supporting that MFSD2A deficiency causes early prenatal brain developmental disruption. We also show that poor MFSD2A expression despite normal transporter activity is a relevant pathomechanism in MCPH15.
Collapse
|
30
|
Liu P, Zhu W, Chen C, Yan B, Zhu L, Chen X, Peng C. The mechanisms of lysophosphatidylcholine in the development of diseases. Life Sci 2020; 247:117443. [DOI: 10.1016/j.lfs.2020.117443] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023]
|
31
|
Shao Y, Tian HY, Zhang JJ, Kharrati-Koopaee H, Guo X, Zhuang XL, Li ML, Nanaie HA, Dehghani Tafti E, Shojaei B, Reza Namavar M, Sotoudeh N, Oluwakemi Ayoola A, Li JL, Liang B, Esmailizadeh A, Wang S, Wu DD. Genomic and Phenotypic Analyses Reveal Mechanisms Underlying Homing Ability in Pigeon. Mol Biol Evol 2020; 37:134-148. [PMID: 31501895 DOI: 10.1093/molbev/msz208] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The homing pigeon was selectively bred from the domestic pigeon for a homing ability over long distances, a very fascinating but complex behavioral trait. Here, we generate a total of 95 whole genomes from diverse pigeon breeds. Comparing the genomes from the homing pigeon population with those from other breeds identifies candidate positively selected genes, including many genes involved in the central nervous system, particularly spatial learning and memory such as LRP8. Expression profiling reveals many neuronal genes displaying differential expression in the hippocampus, which is the key organ for memory and navigation and exhibits significantly larger size in the homing pigeon. In addition, we uncover a candidate gene GSR (encoding glutathione-disulfide reductase) experiencing positive selection in the homing pigeon. Expression profiling finds that GSR is highly expressed in the wattle and visual pigment cell layer, and displays increased expression levels in the homing pigeon. In vitro, a magnetic field stimulates increases in calcium ion concentration in cells expressing pigeon GSR. These findings support the importance of the hippocampus (functioning in spatial memory and navigation) for homing ability, and the potential involvement of GSR in pigeon magnetoreception.
Collapse
Affiliation(s)
- Yong Shao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Hang-Yu Tian
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Jing-Jing Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hamed Kharrati-Koopaee
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran.,Institute of Biotechnology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Xing Guo
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xiao-Lin Zhuang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Ming-Li Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | | | - Elahe Dehghani Tafti
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Bahador Shojaei
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohammad Reza Namavar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Histomorphometry and Stereology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Narges Sotoudeh
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Adeola Oluwakemi Ayoola
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Jia-Li Li
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Bin Liang
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Ali Esmailizadeh
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Shu Wang
- School of Basic Medical Sciences, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Dong-Dong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
32
|
Wong BH, Silver DL. Mfsd2a: A Physiologically Important Lysolipid Transporter in the Brain and Eye. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:223-234. [PMID: 32705603 DOI: 10.1007/978-981-15-6082-8_14] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lipids and essential fatty acids are required for normal brain development and continued photoreceptor membrane biogenesis for the maintenance of vision. The blood-brain barrier and blood-eye barriers prohibit the free diffusion of solutes into the brain and eye so that transporter-mediated uptake predominates at these barriers. The major facilitator superfamily of transporters constitutes one of the largest families of facilitative transporters across all domains of life. A unique family member, major facilitator superfamily domain containing 2a (Mfsd2a) is a lysophosphatidylcholine (LPC) transporter expressed at the blood-brain and blood-retinal barriers and demonstrated to be the major pathway for brain and eye accretion of docosahexaenoic acid (DHA) as an LPC. In addition to LPC-DHA, Mfsd2a can transport other LPCs containing mono- and polyunsaturated fatty acids. Mfsd2a deficiency in mouse and humans results in severe microcephaly, underscoring the importance of LPC transport in brain development. Beyond its role in brain development, LPC-DHA uptake in the brain and eye negatively regulates de novo lipogenesis. This review focuses on the current understanding of the physiological roles of Mfsd2a in the brain and eye and the proposed transport mechanism of Mfsd2a.
Collapse
Affiliation(s)
- Bernice H Wong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - David L Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
33
|
Sánchez-Campillo M, Ruiz-Palacios M, Ruiz-Alcaraz AJ, Prieto-Sánchez MT, Blanco-Carnero JE, Zornoza M, Ruiz-Pastor MJ, Demmelmair H, Sánchez-Solís M, Koletzko B, Larqué E. Child Head Circumference and Placental MFSD2a Expression Are Associated to the Level of MFSD2a in Maternal Blood During Pregnancy. Front Endocrinol (Lausanne) 2020; 11:38. [PMID: 32117064 PMCID: PMC7012934 DOI: 10.3389/fendo.2020.00038] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/20/2020] [Indexed: 12/14/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a world-wide health challenge, which prevalence is expected to increase in parallel to the epidemic of obesity. Children born from GDM mothers have lower levels of docosahexaenoic acid (DHA) in cord blood, which might influence their neurodevelopment. Recently, the membrane transporter Major Family Super Domain 2a (MFSD2a) was associated with the selective transportation of DHA as lysophospholipids. The expression of the DHA membrane transporter MFSD2a is lower in GDM placentas, which could affect materno-fetal DHA transport. Humans with homozygous inactivating mutations in the MFSD2a gene present severe microcephaly and intellectual impairments. Herein, we intended to identify early blood biomarkers that may be of use during pregnancy to monitor the offspring development and the adequate nutritional interventions, such as nutritional supplementation, that may be selected to improve it. We evaluated MFSD2a expression in maternal blood at the third trimester of pregnancy, and its potential relationship with the expression of placental MFSD2a at delivery and child outcomes. Three groups of pregnant women were recruited: 25 controls, 23 GDM with dietary treatment, and 20 GDM with insulin treatment. Maternal and neonatal anthropometric and biochemical parameters were evaluated. MFSD2a was analyzed in placenta, blood and serum. MFSD2a protein expression in maternal blood was significantly lower in GDM groups and correlated with placental MFSD2a and Z-score neonatal head circumference during the first 6 months of life. The cord/maternal serum ratio of DHA, a solid indicator of materno-fetal DHA transport, was reduced in GDM groups and correlated with MFSD2a in maternal blood at the third trimester and in placenta at delivery. This indicates that altered MFSD2a levels in maternal blood during pregnancy might influence placental nutrient transport and fetal neurodevelopment. Furthermore, MFSD2a levels in maternal blood on the third trimester were inversely correlated to DHA in maternal serum lyso-PL. Thus, the level of MFSD2a in maternal blood could be used as a potential biomarker for the early detection of disturbances of MFSD2a expression during pregnancy and the subsequent consequences for the neurodevelopment of the child, as well as it may help to choose the optimal treatment approach for the affected subjects.
Collapse
Affiliation(s)
- María Sánchez-Campillo
- Department of Physiology, Faculty of Biology, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - María Ruiz-Palacios
- Department of Physiology, Faculty of Biology, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Antonio J. Ruiz-Alcaraz
- Department of Biochemistry, Molecular Biology B and Immunology, Faculty of Medicine, Regional Campus of International Excellence Campus Mare Nostrum and Biomedical Research Institute of Murcia (IMIB), University of Murcia, Murcia, Spain
| | | | | | - Matilde Zornoza
- Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - María José Ruiz-Pastor
- Department of Physiology, Faculty of Biology, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Hans Demmelmair
- Ludwig-Maximilians-University Munich, Dr. von Hauner Children's Hospital, München, Germany
| | | | - Berthold Koletzko
- Ludwig-Maximilians-University Munich, Dr. von Hauner Children's Hospital, München, Germany
| | - Elvira Larqué
- Department of Physiology, Faculty of Biology, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, Murcia, Spain
- *Correspondence: Elvira Larqué
| |
Collapse
|
34
|
Decreased Blood Level of MFSD2a as a Potential Biomarker of Alzheimer's Disease. Int J Mol Sci 2019; 21:ijms21010070. [PMID: 31861865 PMCID: PMC6981746 DOI: 10.3390/ijms21010070] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/29/2022] Open
Abstract
The protein Major Facilitator Superfamily Domain containing 2A (MFSD2a) was recently described as the primary carrier for docosahexaenoic acid (DHA) into the brain. Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by lower DHA levels in blood lipids. The aim of this study was to investigate the expression of MFSD2a in the whole blood and brain as a potential biomarker of AD. Three groups were established: 38 healthy controls, 48 subjects with moderate AD (GDS4), and 47 with severe AD (GDS6). We analyzed postmortem brain samples from the hippocampus of 11 healthy controls and 11 severe AD patients. Fatty acid (FA) was determined in serum and brain by gas chromatography. Blood and brain MFSD2a protein expression was analyzed by Western blotting. We found a significant and progressive decline of MFSD2a levels in blood of AD patients (Control 0.83 ± 0.13, GDS4 0.72 ± 0.09, GDS6 0.48 ± 0.05*, p ˂ 0.01). We also corroborated a significant reduction of DHA and other n-3 long-chain polyunsaturated FA in serum of AD. No differences were found in MFSD2a expression or FA levels in brain of controls and AD subjects. MFSD2A carrier was analyzed in AD patients for the first time and the level of MFSD2a in the whole blood could be a potential biomarker of this disease.
Collapse
|
35
|
Omega-3 Docosahexaenoic Acid Is a Mediator of Fate-Decision of Adult Neural Stem Cells. Int J Mol Sci 2019; 20:ijms20174240. [PMID: 31480215 PMCID: PMC6747551 DOI: 10.3390/ijms20174240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
The mammalian brain is enriched with lipids that serve as energy catalyzers or secondary messengers of essential signaling pathways. Docosahexaenoic acid (DHA) is an omega-3 fatty acid synthesized de novo at low levels in humans, an endogenous supply from its precursors, and is mainly incorporated from nutrition, an exogeneous supply. Decreased levels of DHA have been reported in the brains of patients with neurodegenerative diseases. Preventing this decrease or supplementing the brain with DHA has been considered as a therapy for the DHA brain deficiency that could be linked with neuronal death or neurodegeneration. The mammalian brain has, however, a mechanism of compensation for loss of neurons in the brain: neurogenesis, the birth of neurons from neural stem cells. In adulthood, neurogenesis is still present, although at a slower rate and with low efficiency, where most of the newly born neurons die. Neural stem/progenitor cells (NSPCs) have been shown to require lipids for proper metabolism for proliferation maintenance and neurogenesis induction. Recent studies have focused on the effects of these essential lipids on the neurobiology of NSPCs. This review aimed to introduce the possible use of DHA to impact NSPC fate-decision as a therapy for neurodegenerative diseases.
Collapse
|
36
|
Bazinet RP, Bernoud-Hubac N, Lagarde M. How the plasma lysophospholipid and unesterified fatty acid pools supply the brain with docosahexaenoic acid. Prostaglandins Leukot Essent Fatty Acids 2019; 142:1-3. [PMID: 30773208 DOI: 10.1016/j.plefa.2018.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022]
Abstract
The brain requires a constant supply of docosahexaenoic acid (DHA) from blood to maintain DHA levels within the brain. Several plasma pools have been proposed to supply the brain with DHA, including plasma lipoproteins, lysophosphatidylcholine and unesterified fatty acids. Here we briefly review the evidence for each plasma pool supplying the brain highlighting controversies and remaining questions. We conclude that circulating lysophosphatidylcholine has a higher brain/body partition coefficient than unesterified DHA while unesterified DHA entry into the brain is more rapid.
Collapse
Affiliation(s)
- Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| | - Nathalie Bernoud-Hubac
- Univ-Lyon, Inserm UMR 1060, Inra UMR 1397, INSA-Lyon, IMBL, 20 Ave A., Einstein, 69100 Villeurbanne, France
| | - Michel Lagarde
- Univ-Lyon, Inserm UMR 1060, Inra UMR 1397, INSA-Lyon, IMBL, 20 Ave A., Einstein, 69100 Villeurbanne, France
| |
Collapse
|
37
|
Yalagala PCR, Sugasini D, Dasarathi S, Pahan K, Subbaiah PV. Dietary lysophosphatidylcholine-EPA enriches both EPA and DHA in the brain: potential treatment for depression. J Lipid Res 2019; 60:566-578. [PMID: 30530735 PMCID: PMC6399499 DOI: 10.1194/jlr.m090464] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/07/2018] [Indexed: 12/21/2022] Open
Abstract
EPA and DHA protect against multiple metabolic and neurologic disorders. Although DHA appears more effective for neuroinflammatory conditions, EPA is more beneficial for depression. However, the brain contains negligible amounts of EPA, and dietary supplements fail to increase it appreciably. We tested the hypothesis that this failure is due to absorption of EPA as triacylglycerol, whereas the transporter at the blood-brain barrier requires EPA as lysophosphatidylcholine (LPC). We compared tissue uptake in normal mice gavaged with equal amounts (3.3 μmol/day) of either LPC-EPA or free EPA (surrogate for current supplements) for 15 days and also measured target gene expression. Compared with the no-EPA control, LPC-EPA increased brain EPA >100-fold (from 0.03 to 4 μmol/g); free EPA had little effect. Furthermore, LPC-EPA, but not free EPA, increased brain DHA 2-fold. Free EPA increased EPA in adipose tissue, and both supplements increased EPA and DHA in the liver and heart. Only LPC-EPA increased EPA and DHA in the retina, and expression of brain-derived neurotrophic factor, cyclic AMP response element binding protein, and 5-hydroxy tryptamine (serotonin) receptor 1A in the brain. These novel results show that brain EPA can be increased through diet. Because LPC-EPA increased both EPA and DHA in the brain, it may help in the treatment of depression as well as neuroinflammatory diseases, such as Alzheimer's disease.
Collapse
|
38
|
Ferchaud-Roucher V, Kramer A, Silva E, Pantham P, Weintraub ST, Jansson T, Powell TL. A potential role for lysophosphatidylcholine in the delivery of long chain polyunsaturated fatty acids to the fetal circulation. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:394-402. [PMID: 30572119 DOI: 10.1016/j.bbalip.2018.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/10/2018] [Accepted: 12/15/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Véronique Ferchaud-Roucher
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Anita Kramer
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elena Silva
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Priyadarshini Pantham
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, IL, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center San Antonio, TX, USA
| | - Thomas Jansson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Theresa L Powell
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
39
|
Lin Y, Deepak RNVK, Zheng JZ, Fan H, Zheng L. A dual substrate-accessing mechanism of a major facilitator superfamily protein facilitates lysophospholipid flipping across the cell membrane. J Biol Chem 2018; 293:19919-19931. [PMID: 30373772 DOI: 10.1074/jbc.ra118.005548] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/23/2018] [Indexed: 11/06/2022] Open
Abstract
Lysophospholipid transporter (LplT) is a member of the major facilitator superfamily present in many Gram-negative bacteria. LplT catalyzes flipping of lysophospholipids (LPLs) across the bacterial inner membrane, playing an important role in bacterial membrane homeostasis. We previously reported that LplT promotes both uptake of exogenous LPLs and intramembranous LPL flipping across the bilayer. To gain mechanistic insight into this dual LPL-flipping activity, here we implemented a combination of computational approaches and LPL transport analyses to study LPL binding of and translocation by LplT. Our results suggest that LplT translocates LPLs through an elongated cavity exhibiting an extremely asymmetric polarity. We found that two D(E)N motifs form a head group-binding site, in which the carboxylate group of Asp-30 is important for LPL head group recognition. Substitutions of residues in the head group-binding site disrupted both LPL uptake and flipping activities. However, alteration of hydrophobic residues on the interface between the N- and C-terminal domains impaired LPL flipping specifically, resulting in LPLs accumulation in the membrane, but LPL uptake remained active. These results suggest a dual substrate-accessing mechanism, in which LplT recruits LPLs to its substrate-binding site via two routes, either from its extracellular entry or through a membrane-embedded groove between transmembrane helices, and then moves them toward the inner membrane leaflet. This LPL-flipping mechanism is likely conserved in many bacterial species, and our findings illustrate how LplT adjusts the major facilitator superfamily translocation pathway to perform its versatile lipid homeostatic functions.
Collapse
Affiliation(s)
- Yibin Lin
- From the Department of Biochemistry and Molecular Biology, Center for Membrane Biology, the University of Texas Health Science Center at Houston McGovern Medical School, Houston Texas 77030
| | - R N V Krishna Deepak
- the Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 138671 Singapore, and
| | - Jonathan Zixiang Zheng
- From the Department of Biochemistry and Molecular Biology, Center for Membrane Biology, the University of Texas Health Science Center at Houston McGovern Medical School, Houston Texas 77030
| | - Hao Fan
- the Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 138671 Singapore, and .,the Department of Biological Sciences (DBS), National University of Singapore, 117558 Singapore, and Center for Computational Biology, DUKE-NUS Medical School, 169857 Singapore
| | - Lei Zheng
- From the Department of Biochemistry and Molecular Biology, Center for Membrane Biology, the University of Texas Health Science Center at Houston McGovern Medical School, Houston Texas 77030,
| |
Collapse
|
40
|
Eser Ocak P, Ocak U, Sherchan P, Zhang JH, Tang J. Insights into major facilitator superfamily domain-containing protein-2a (Mfsd2a) in physiology and pathophysiology. What do we know so far? J Neurosci Res 2018; 98:29-41. [PMID: 30345547 DOI: 10.1002/jnr.24327] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/20/2018] [Accepted: 08/28/2018] [Indexed: 01/02/2023]
Abstract
Major facilitator superfamily domain-containing protein-2a (Mfsd2a) which was considered as an orphan transporter has recently gained attention for its regulatory role in the maintenance of proper functioning of the blood-brain barrier. Besides the major role of Mfsd2a in maintaining the barrier function, increasing evidence has emerged with regard to the contributions of Mfsd2a to various biological processes such as transport, cell fusion, cell cycle, inflammation and regeneration, managing tumor growth, functioning of other organs with barrier functions or responses to injury. The purpose of this article is to review the different roles of Mfsd2a and its involvement in the physiological and pathophysiological processes primarily in the central nervous system and throughout the mammalian body under the lights of the current literature.
Collapse
Affiliation(s)
- Pinar Eser Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Umut Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
41
|
Abstract
Dietary and supplemental intake of the ω-3 fatty acid docosahexaenoic acid (DHA) reduces risk of Alzheimer’s disease (AD) and ameliorates symptoms. The apolipoprotein E (APOE)4 allele is the strongest risk factor for sporadic AD, exclusive of age. APOE4 carriers respond well to the DHA present in fish but do not respond as well to dietary supplements. The mechanisms behind this varied response remain unknown. I posit that the difference is that fish contain DHA in phospholipid form, whereas fish oil supplements do not. This influences whether DHA is metabolized to nonesterified DHA (free DHA) or a phospholipid form called lysophosphatidylcholine DHA (DHA-lysoPC). Free DHA is transported across the outer membrane leaflet of the blood–brain barrier (BBB) via passive diffusion, and DHA-lysoPC is transported across the inner membrane leaflet of the BBB via the major facilitator superfamily domain-containing protein 2A. I propose that APOE4 carriers have impaired brain transport of free DHA but not of DHA-lysoPC, as a consequence of a breakdown in the outer membrane leaflet of the BBB, putting them at increased risk for AD. Dietary sources of DHA in phospholipid form may provide a means to increase plasma levels of DHA-lysoPC, thereby decreasing the risk of AD.—Patrick, R. P. Role of phosphatidylcholine-DHA in preventing APOE4-associated Alzheimer’s disease.
Collapse
Affiliation(s)
- Rhonda P Patrick
- University of California San Francisco Benioff, Children's Hospital Oakland Research Institute, Oakland, California, USA
| |
Collapse
|
42
|
Comprehensive review on the molecular genetics of autosomal recessive primary microcephaly (MCPH). Genet Res (Camb) 2018; 100:e7. [PMID: 30086807 DOI: 10.1017/s0016672318000046] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Primary microcephaly (MCPH) is an autosomal recessive sporadic neurodevelopmental ailment with a trivial head size characteristic that is below 3-4 standard deviations. MCPH is the smaller upshot of an architecturally normal brain; a significant decrease in size is seen in the cerebral cortex. At birth MCPH presents with non-progressive mental retardation, while secondary microcephaly (onset after birth) presents with and without other syndromic features. MCPH is a neurogenic mitotic syndrome nevertheless pretentious patients demonstrate normal neuronal migration, neuronal apoptosis and neural function. Eighteen MCPH loci (MCPH1-MCPH18) have been mapped to date from various populations around the world and contain the following genes: Microcephalin, WDR62, CDK5RAP2, CASC5, ASPM, CENPJ, STIL, CEP135, CEP152, ZNF335, PHC1, CDK6, CENPE, SASS6, MFSD2A, ANKLE2, CIT and WDFY3, clarifying our understanding about the molecular basis of microcephaly genetic disorder. It has previously been reported that phenotype disease is caused by MCB gene mutations and the causes of this phenotype are disarrangement of positions and organization of chromosomes during the cell cycle as a result of mutated DNA, centriole duplication, neurogenesis, neuronal migration, microtubule dynamics, transcriptional control and the cell cycle checkpoint having some invisible centrosomal process that can manage the number of neurons that are produced by neuronal precursor cells. Furthermore, researchers inform us about the clinical management of families that are suffering from MCPH. Establishment of both molecular understanding and genetic advocating may help to decrease the rate of this ailment. This current review study examines newly identified genes along with previously identified genes involved in autosomal recessive MCPH.
Collapse
|
43
|
FREE-RADICAL OXIDATION AND OXYGENATION OF HEMOGLOBIN AT EXACERBATION OF CYTOMEGALOVIRUS INFECTION IN THE SECOND TRIMESTER OF PREGNANCY. ACTA BIOMEDICA SCIENTIFICA 2018. [DOI: 10.29413/abs.2018-3.4.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The aim of the study was to assess free radical processes and their effect on oxygenation of hemoglobin in the blood of 18-21 weeks pregnant women with exacerbation of cytomegalovirus infection. We examined 40 pregnant women with exacerbation of cytomegalovirus infection and various levels of specific antibodies (IgM) at 18-21 weeks and 30 pregnant women without cytomegalovirus. The spectrophotometric method was used to determine the content of oxyhemoglobin, methemoglobin, thiobarbituric acid (TBA)-active products, superoxide dismutase in blood erythrocytes; thin layer chromatography – phosphotidylcholine, lysophosphatidylcholine; gas-liquid chromatography – arachidonic acid. Exacerbation of cytomegalovirus infection changes the activity of free radical oxidation processes, the severity of which is determined by the level of IgM antibodies and is manifested by a decrease in superoxide dismutase (p = 0.000) and phosphatidylcholine (p = 0.000), an increase in phospholipase A2 (p = 0.000), lysophosphatidylcholine (p = 0.000), arachidonic acid (p = 0.000) and TBA-active products (p = 0.000). Accumulation of superoxide anion radical and products of lipoperoxidation in erythrocytes decreases oxyhemoglobin (p = 0.000) and increases methemoglobin (p = 0.000). Exacerbation of cytomegalovirus infection at 18-21 weeks is associated with the enhancement of free radical lipid oxidation and a deficiency in the antioxidant activity of superoxide dismutase. It decreases oxygen transport properties and increases phagocytosis by red blood monocytes; and pregnant women develop further hemic hypoxia.
Collapse
|
44
|
Harel T, Quek DQY, Wong BH, Cazenave-Gassiot A, Wenk MR, Fan H, Berger I, Shmueli D, Shaag A, Silver DL, Elpeleg O, Edvardson S. Homozygous mutation in MFSD2A, encoding a lysolipid transporter for docosahexanoic acid, is associated with microcephaly and hypomyelination. Neurogenetics 2018; 19:227-235. [DOI: 10.1007/s10048-018-0556-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/09/2018] [Indexed: 01/05/2023]
|
45
|
Lo Van A, Sakayori N, Hachem M, Belkouch M, Picq M, Fourmaux B, Lagarde M, Osumi N, Bernoud-Hubac N. Targeting the Brain with a Neuroprotective Omega-3 Fatty Acid to Enhance Neurogenesis in Hypoxic Condition in Culture. Mol Neurobiol 2018; 56:986-999. [DOI: 10.1007/s12035-018-1139-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/21/2018] [Indexed: 12/01/2022]
|
46
|
Perland E, Bagchi S, Klaesson A, Fredriksson R. Characteristics of 29 novel atypical solute carriers of major facilitator superfamily type: evolutionary conservation, predicted structure and neuronal co-expression. Open Biol 2018; 7:rsob.170142. [PMID: 28878041 PMCID: PMC5627054 DOI: 10.1098/rsob.170142] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/31/2017] [Indexed: 12/21/2022] Open
Abstract
Solute carriers (SLCs) are vital as they are responsible for a major part of the molecular transport over lipid bilayers. At present, there are 430 identified SLCs, of which 28 are called atypical SLCs of major facilitator superfamily (MFS) type. These are MFSD1, 2A, 2B, 3, 4A, 4B, 5, 6, 6 L, 7, 8, 9, 10, 11, 12, 13A, 14A and 14B; SV2A, SV2B and SV2C; SVOP and SVOPL; SPNS1, SPNS2 and SPNS3; and UNC93A and UNC93B1. We studied their fundamental properties, and we also included CLN3, an atypical SLC not yet belonging to any protein family (Pfam) clan, because its involvement in the same neuronal degenerative disorders as MFSD8. With phylogenetic analyses and bioinformatic sequence comparisons, the proteins were divided into 15 families, denoted atypical MFS transporter families (AMTF1-15). Hidden Markov models were used to identify orthologues from human to Drosophila melanogaster and Caenorhabditis elegans Topology predictions revealed 12 transmembrane segments (for all except CLN3), corresponding to the common MFS structure. With single-cell RNA sequencing and in situ proximity ligation assay on brain cells, co-expressions of several atypical SLCs were identified. Finally, the transcription levels of all genes were analysed in the hypothalamic N25/2 cell line after complete amino acid starvation, showing altered expression levels for several atypical SLCs.
Collapse
Affiliation(s)
- Emelie Perland
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Sonchita Bagchi
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Axel Klaesson
- Pharmaceutical Cell Biology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
47
|
Dragoni S, Turowski P. Polarised VEGFA Signalling at Vascular Blood–Neural Barriers. Int J Mol Sci 2018; 19:ijms19051378. [PMID: 29734754 PMCID: PMC5983809 DOI: 10.3390/ijms19051378] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
At blood–neural barriers, endothelial VEGFA signalling is highly polarised, with entirely different responses being triggered by luminal or abluminal stimulation. These recent findings were made in a field which is still in its mechanistic infancy. For a long time, endothelial polarity has intuitively been presumed, and likened to that of epithelial cells, but rarely demonstrated. In the cerebral and the retinal microvasculature, the uneven distribution of VEGF receptors 1 and 2, with the former predominant on the luminal and the latter on the abluminal face of the endothelium, leads to a completely polarised signalling response to VEGFA. Luminal VEGFA activates VEGFR1 homodimers and AKT, leading to a cytoprotective response, whilst abluminal VEGFA induces vascular leakage via VEGFR2 homodimers and p38. Whilst these findings do not provide a complete picture of VEGFA signalling in the microvasculature—there are still unclear roles for heterodimeric receptor complexes as well as co-receptors—they provide essential insight into the adaptation of vascular systems to environmental cues that are naturally different, depending on whether they are present on the blood or tissue side. Importantly, sided responses are not only restricted to VEGFA, but exist for other important vasoactive agents.
Collapse
Affiliation(s)
- Silvia Dragoni
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| | - Patric Turowski
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
48
|
MFSD2B is a sphingosine 1-phosphate transporter in erythroid cells. Sci Rep 2018; 8:4969. [PMID: 29563527 PMCID: PMC5862976 DOI: 10.1038/s41598-018-23300-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/02/2018] [Indexed: 12/22/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is an intercellular signaling molecule present in blood. Erythrocytes have a central role in maintaining the S1P concentration in the blood stream. We previously demonstrated that S1P is exported from erythrocytes by a glyburide-sensitive S1P transporter. However, the gene encoding the S1P transporter in erythrocytes is unknown. In this study, we found that the mouse erythroid cell line, MEDEP-E14, has S1P export activity and exhibits properties that are consistent with those of erythrocytes. Using microarray analysis of MEDEP-E14 cells and its parental cell line, E14TG2a, we identified several candidate genes for S1P export activity. Of those genes, only one gene, Mfsd2b, showed S1P transport activity. The properties of S1P release by MFSD2B were similar to those in erythrocytes. Moreover, knockout of MFSD2B in MEDEP-E14 cells decreased S1P export from the cells. These results strongly suggest that MFSD2B is a novel S1P transporter in erythroid cells.
Collapse
|
49
|
Chouinard-Watkins R, Lacombe RJS, Bazinet RP. Mechanisms regulating brain docosahexaenoic acid uptake: what is the recent evidence? Curr Opin Clin Nutr Metab Care 2018; 21:71-77. [PMID: 29206690 DOI: 10.1097/mco.0000000000000440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW To summarize recent advances pertaining to the mechanisms regulating brain docosahexaenoic acid (DHA) uptake. DHA is an omega-3 polyunsaturated fatty acid highly enriched in neuronal membranes and it is implicated in several important neurological processes. However, DHA synthesis is extremely limited within the brain. RECENT FINDINGS There are two main plasma pools that supply the brain with DHA: the nonesterified pool and the lysophosphatidylcholine (lysoPtdCho) pool. Quantitatively, plasma nonesterified-DHA (NE-DHA) is the main contributor to brain DHA. Fatty acid transport protein 1 (FATP1) in addition to fatty acid-binding protein 5 (FABP5) are key players that regulate brain uptake of NE-DHA. However, the plasma half-life of lysoPtdCho-DHA and its brain partition coefficient are higher than those of NE-DHA after intravenous administration. SUMMARY The mechanisms regulating brain DHA uptake are more complicated than once believed, but recent advances provide some clarity notably by suggesting that FATP1 and FABP5 are key contributors to cellular uptake of DHA at the blood-brain barrier. Elucidating how DHA enters the brain is important as we might be able to identify methods to better deliver DHA to the brain as a potential therapeutic.
Collapse
Affiliation(s)
- Raphaël Chouinard-Watkins
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
50
|
Brain docosahexaenoic acid uptake and metabolism. Mol Aspects Med 2018; 64:109-134. [PMID: 29305120 DOI: 10.1016/j.mam.2017.12.004] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/21/2017] [Accepted: 12/28/2017] [Indexed: 12/22/2022]
Abstract
Docosahexaenoic acid (DHA) is the most abundant n-3 polyunsaturated fatty acid in the brain where it serves to regulate several important processes and, in addition, serves as a precursor to bioactive mediators. Given that the capacity of the brain to synthesize DHA locally is appreciably low, the uptake of DHA from circulating lipid pools is essential to maintaining homeostatic levels. Although, several plasma pools have been proposed to supply the brain with DHA, recent evidence suggests non-esterified-DHA and lysophosphatidylcholine-DHA are the primary sources. The uptake of DHA into the brain appears to be regulated by a number of complementary pathways associated with the activation and metabolism of DHA, and may provide mechanisms for enrichment of DHA within the brain. Following entry into the brain, DHA is esterified into and recycled amongst membrane phospholipids contributing the distribution of DHA in brain phospholipids. During neurotransmission and following brain injury, DHA is released from membrane phospholipids and converted to bioactive mediators which regulate signaling pathways important to synaptogenesis, cell survival, and neuroinflammation, and may be relevant to treating neurological diseases. In the present review, we provide a comprehensive overview of brain DHA metabolism, encompassing many of the pathways and key enzymatic regulators governing brain DHA uptake and metabolism. In addition, we focus on the release of non-esterified DHA and subsequent production of bioactive mediators and the evidence of their proposed activity within the brain. We also provide a brief review of the evidence from post-mortem brain analyses investigating DHA levels in the context of neurological disease and mood disorder, highlighting the current disparities within the field.
Collapse
|