101
|
Wu D, Zhang K, Khan FA, Wu Q, Pandupuspitasari NS, Tang Y, Guan K, Sun F, Huang C. The emerging era of lactate: A rising star in cellular signaling and its regulatory mechanisms. J Cell Biochem 2023; 124:1067-1081. [PMID: 37566665 DOI: 10.1002/jcb.30458] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/19/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Cellular metabolites are ancient molecules with pleiotropic implications in health and disease. Beyond their cognate roles, they have signaling functions as the ligands for specific receptors and the precursors for epigenetic or posttranslational modifications. Lactate has long been recognized as a metabolic waste and fatigue product mainly produced from glycolytic metabolism. Recent evidence however suggests lactate is an unique molecule with diverse signaling attributes in orchestration of numerous biological processes, including tumor immunity and neuronal survival. The copious metabolic and non-metabolic functions of lactate mediated by its bidirectional shuttle between cells or intracellular organelles lead to a phenotype called "lactormone." Importantly, the mechanisms of lactate signaling, via acting as a molecular sensor and a regulator of NAD+ metabolism and AMP-activated protein kinase signaling, and via the newly identified lactate-driven lactylation, have been discovered. Further, we include a brief discussion about the autocrine regulation of efferocytosis by lactate in Sertoli cells which favoraerobic glycolysis. By emphasizing a repertoire of the most recent discovered mechanisms of lactate signaling, this review will open tantalizing avenues for future investigations cracking the regulatory topology of lactate signaling covered in the veil of mystery.
Collapse
Affiliation(s)
- Di Wu
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Kejia Zhang
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, Ministry of Research and Technology National Research and Innovation Agency, Jakarta, Indonesia
| | - Qin Wu
- Jinan Second People's Hospital & The Ophthalmologic Hospital of Jinan, Jinan, China
| | | | - Yuan Tang
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, China
| | - Fei Sun
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Chunjie Huang
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| |
Collapse
|
102
|
Yang LG, March ZM, Stephenson RA, Narayan PS. Apolipoprotein E in lipid metabolism and neurodegenerative disease. Trends Endocrinol Metab 2023; 34:430-445. [PMID: 37357100 PMCID: PMC10365028 DOI: 10.1016/j.tem.2023.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/27/2023]
Abstract
Dysregulation of lipid metabolism has emerged as a central component of many neurodegenerative diseases. Variants of the lipid transport protein, apolipoprotein E (APOE), modulate risk and resilience in several neurodegenerative diseases including late-onset Alzheimer's disease (LOAD). Allelic variants of the gene, APOE, alter the lipid metabolism of cells and tissues and have been broadly associated with several other cellular and systemic phenotypes. Targeting APOE-associated metabolic pathways may offer opportunities to alter disease-related phenotypes and consequently, attenuate disease risk and impart resilience to multiple neurodegenerative diseases. We review the molecular, cellular, and tissue-level alterations to lipid metabolism that arise from different APOE isoforms. These changes in lipid metabolism could help to elucidate disease mechanisms and tune neurodegenerative disease risk and resilience.
Collapse
Affiliation(s)
- Linda G Yang
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Zachary M March
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Roxan A Stephenson
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Priyanka S Narayan
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.; National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA; Center for Alzheimer's and Related Dementias (CARD), National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
103
|
Lopez-Lopez A, Valenzuela R, Rodriguez-Perez AI, Guerra MJ, Labandeira-Garcia JL, Muñoz A. Interactions between Angiotensin Type-1 Antagonists, Statins, and ROCK Inhibitors in a Rat Model of L-DOPA-Induced Dyskinesia. Antioxidants (Basel) 2023; 12:1454. [PMID: 37507992 PMCID: PMC10376833 DOI: 10.3390/antiox12071454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/06/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Statins have been proposed for L-DOPA-induced dyskinesia (LID) treatment. Statin anti-dyskinetic effects were related to the inhibition of the Ras-ERK pathway. However, the mechanisms responsible for the anti-LID effect are unclear. Changes in cholesterol homeostasis and oxidative stress- and inflammation-related mechanisms such as angiotensin II and Rho-kinase (ROCK) inhibition may be involved. The nigra and striatum of dyskinetic rats showed increased levels of cholesterol, ROCK, and the inflammatory marker IL-1β, which were reduced by the angiotensin type-1 receptor (AT1) antagonist candesartan, simvastatin, and the ROCK inhibitor fasudil. As observed for LID, angiotensin II-induced, via AT1, increased levels of cholesterol and ROCK in the rat nigra and striatum. In cultured dopaminergic neurons, angiotensin II increased cholesterol biosynthesis and cholesterol efflux without changes in cholesterol uptake. In astrocytes, angiotensin induced an increase in cholesterol uptake, decrease in biosynthesis, and no change in cholesterol efflux, suggesting a neuronal accumulation of cholesterol that is reduced via transfer to astrocytes. Our data suggest mutual interactions between angiotensin/AT1, cholesterol, and ROCK pathways in LID, which are attenuated by the corresponding inhibitors. Interestingly, these three drugs have also been suggested as neuroprotective treatments against Parkinson's disease. Therefore, they may reduce dyskinesia and the progression of the disease using common mechanisms.
Collapse
Affiliation(s)
- Andrea Lopez-Lopez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Rita Valenzuela
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Ana Isabel Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - María J Guerra
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Jose Luis Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Ana Muñoz
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
104
|
Lan ZQ, Ge ZY, Lv SK, Zhao B, Li CX. The regulatory role of lipophagy in central nervous system diseases. Cell Death Discov 2023; 9:229. [PMID: 37414782 DOI: 10.1038/s41420-023-01504-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023] Open
Abstract
Lipid droplets (LDs) are the organelles for storing neutral lipids, which are broken down when energy is insufficient. It has been suggested that excessive accumulation of LDs can affect cellular function, which is important to coordinate homeostasis of lipids in vivo. Lysosomes play an important role in the degradation of lipids, and the process of selective autophagy of LDs through lysosomes is known as lipophagy. Dysregulation of lipid metabolism has recently been associated with a variety of central nervous system (CNS) diseases, but the specific regulatory mechanisms of lipophagy in these diseases remain to be elucidated. This review summarizes various forms of lipophagy and discusses the role that lipophagy plays in the development of CNS diseases in order to reveal the related mechanisms and potential therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Zhuo-Qing Lan
- Department of General practice medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, P.R. China
| | - Zi-Yi Ge
- Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Shu-Kai Lv
- Department of General practice medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, P.R. China
| | - Bing Zhao
- Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R. China.
| | - Cai-Xia Li
- Department of General practice medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, P.R. China.
- Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R. China.
| |
Collapse
|
105
|
Rai M, Carter SM, Shefali SA, Chawla G, Tennessen JM. Characterization of genetic and molecular tools for studying the endogenous expression of Lactate dehydrogenase in Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545165. [PMID: 37398276 PMCID: PMC10312709 DOI: 10.1101/2023.06.15.545165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Drosophila melanogaster larval development relies on a specialized metabolic state that utilizes carbohydrates and other dietary nutrients to promote rapid growth. One unique feature of the larval metabolic program is that Lactate Dehydrogenase (Ldh) activity is highly elevated during this growth phase when compared to other stages of the fly life cycle, indicating that Ldh serves a key role in promoting juvenile development. Previous studies of larval Ldh activity have largely focused on the function of this enzyme at the whole animal level, however, Ldh expression varies significantly among larval tissues, raising the question of how this enzyme promotes tissue-specific growth programs. Here we characterize two transgene reporters and an antibody that can be used to study Ldh expression in vivo . We find that all three tools produce similar Ldh expression patterns. Moreover, these reagents demonstrate that the larval Ldh expression pattern is complex, suggesting the purpose of this enzyme varies across cell types. Overall, our studies validate a series of genetic and molecular reagents that can be used to study glycolytic metabolism in the fly.
Collapse
Affiliation(s)
- Madhulika Rai
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Sarah M. Carter
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Geetanjali Chawla
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institute of Eminence (SNIoE), Dadri, Uttar Pradesh 201314, India
| | | |
Collapse
|
106
|
Gerstner JR, Flores CC, Lefton M, Rogers B, Davis CJ. FABP7: a glial integrator of sleep, circadian rhythms, plasticity, and metabolic function. Front Syst Neurosci 2023; 17:1212213. [PMID: 37404868 PMCID: PMC10315501 DOI: 10.3389/fnsys.2023.1212213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/02/2023] [Indexed: 07/06/2023] Open
Abstract
Sleep and circadian rhythms are observed broadly throughout animal phyla and influence neural plasticity and cognitive function. However, the few phylogenetically conserved cellular and molecular pathways that are implicated in these processes are largely focused on neuronal cells. Research on these topics has traditionally segregated sleep homeostatic behavior from circadian rest-activity rhythms. Here we posit an alternative perspective, whereby mechanisms underlying the integration of sleep and circadian rhythms that affect behavioral state, plasticity, and cognition reside within glial cells. The brain-type fatty acid binding protein, FABP7, is part of a larger family of lipid chaperone proteins that regulate the subcellular trafficking of fatty acids for a wide range of cellular functions, including gene expression, growth, survival, inflammation, and metabolism. FABP7 is enriched in glial cells of the central nervous system and has been shown to be a clock-controlled gene implicated in sleep/wake regulation and cognitive processing. FABP7 is known to affect gene transcription, cellular outgrowth, and its subcellular localization in the fine perisynaptic astrocytic processes (PAPs) varies based on time-of-day. Future studies determining the effects of FABP7 on behavioral state- and circadian-dependent plasticity and cognitive processes, in addition to functional consequences on cellular and molecular mechanisms related to neural-glial interactions, lipid storage, and blood brain barrier integrity will be important for our knowledge of basic sleep function. Given the comorbidity of sleep disturbance with neurological disorders, these studies will also be important for our understanding of the etiology and pathophysiology of how these diseases affect or are affected by sleep.
Collapse
Affiliation(s)
- Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Micah Lefton
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Brooke Rogers
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
107
|
Ralhan I, Chang J, Moulton MJ, Goodman LD, Lee NY, Plummer G, Pasolli HA, Matthies D, Bellen HJ, Ioannou MS. Autolysosomal exocytosis of lipids protect neurons from ferroptosis. J Cell Biol 2023; 222:e202207130. [PMID: 37036445 PMCID: PMC10098143 DOI: 10.1083/jcb.202207130] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/25/2022] [Accepted: 03/20/2023] [Indexed: 04/11/2023] Open
Abstract
During oxidative stress neurons release lipids that are internalized by glia. Defects in this coordinated process play an important role in several neurodegenerative diseases. Yet, the mechanisms of lipid release and its consequences on neuronal health are unclear. Here, we demonstrate that lipid-protein particle release by autolysosome exocytosis protects neurons from ferroptosis, a form of cell death driven by lipid peroxidation. We show that during oxidative stress, peroxidated lipids and iron are released from neurons by autolysosomal exocytosis which requires the exocytic machinery VAMP7 and syntaxin 4. We observe membrane-bound lipid-protein particles by TEM and demonstrate that these particles are released from neurons using cryoEM. Failure to release these lipid-protein particles causes lipid hydroperoxide and iron accumulation and sensitizes neurons to ferroptosis. Our results reveal how neurons protect themselves from peroxidated lipids. Given the number of brain pathologies that involve ferroptosis, defects in this pathway likely play a key role in the pathophysiology of neurodegenerative disease.
Collapse
Affiliation(s)
- Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Canada
| | - Jinlan Chang
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Matthew J. Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
| | - Lindsey D. Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
| | - Nathanael Y.J. Lee
- Department of Physiology, University of Alberta, Edmonton, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Canada
| | - Greg Plummer
- Faculty of Medicine & Dentistry Cell Imaging Core, University of Alberta, Edmonton, Canada
| | - H. Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Doreen Matthies
- 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, USA
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Maria S. Ioannou
- Department of Physiology, University of Alberta, Edmonton, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
108
|
Li Y, Chang P, Sankaran S, Jang H, Nie Y, Zeng A, Hussain S, Wu JY, Chen X, Shi L. Bioorthogonal Stimulated Raman Scattering Imaging Uncovers Lipid Metabolic Dynamics in Drosophila Brain During Aging. GEN BIOTECHNOLOGY 2023; 2:247-261. [PMID: 37363411 PMCID: PMC10286263 DOI: 10.1089/genbio.2023.0017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
Studies have shown that brain lipid metabolism is associated with biological aging and influenced by dietary and genetic manipulations; however, the underlying mechanisms are elusive. High-resolution imaging techniques propose a novel and potent approach to understanding lipid metabolic dynamics in situ. Applying deuterium water (D2O) probing with stimulated Raman scattering (DO-SRS) microscopy, we revealed that lipid metabolic activity in Drosophila brain decreased with aging in a sex-dependent manner. Female flies showed an earlier occurrence of lipid turnover decrease than males. Dietary restriction (DR) and downregulation of insulin/IGF-1 signaling (IIS) pathway, two scenarios for lifespan extension, led to significant enhancements of brain lipid turnover in old flies. Combining SRS imaging with deuterated bioorthogonal probes (deuterated glucose and deuterated acetate), we discovered that, under DR treatment and downregulation of IIS pathway, brain metabolism shifted to use acetate as a major carbon source for lipid synthesis. For the first time, our study directly visualizes and quantifies spatiotemporal alterations of lipid turnover in Drosophila brain at the single organelle (lipid droplet) level. Our study not only demonstrates a new approach for studying brain lipid metabolic activity in situ but also illuminates the interconnection of aging, dietary, and genetic manipulations on brain lipid metabolic regulation.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Phyllis Chang
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Shriya Sankaran
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Hongje Jang
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Yuhang Nie
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Audrey Zeng
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Sahran Hussain
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Jane Y. Wu
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Xu Chen
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Lingyan Shi
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
109
|
Bresgen N, Kovacs M, Lahnsteiner A, Felder TK, Rinnerthaler M. The Janus-Faced Role of Lipid Droplets in Aging: Insights from the Cellular Perspective. Biomolecules 2023; 13:912. [PMID: 37371492 PMCID: PMC10301655 DOI: 10.3390/biom13060912] [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: 03/13/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
It is widely accepted that nine hallmarks-including mitochondrial dysfunction, epigenetic alterations, and loss of proteostasis-exist that describe the cellular aging process. Adding to this, a well-described cell organelle in the metabolic context, namely, lipid droplets, also accumulates with increasing age, which can be regarded as a further aging-associated process. Independently of their essential role as fat stores, lipid droplets are also able to control cell integrity by mitigating lipotoxic and proteotoxic insults. As we will show in this review, numerous longevity interventions (such as mTOR inhibition) also lead to strong accumulation of lipid droplets in Saccharomyces cerevisiae, Caenorhabditis elegans, Drosophila melanogaster, and mammalian cells, just to name a few examples. In mammals, due to the variety of different cell types and tissues, the role of lipid droplets during the aging process is much more complex. Using selected diseases associated with aging, such as Alzheimer's disease, Parkinson's disease, type II diabetes, and cardiovascular disease, we show that lipid droplets are "Janus"-faced. In an early phase of the disease, lipid droplets mitigate the toxicity of lipid peroxidation and protein aggregates, but in a later phase of the disease, a strong accumulation of lipid droplets can cause problems for cells and tissues.
Collapse
Affiliation(s)
- Nikolaus Bresgen
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Melanie Kovacs
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Angelika Lahnsteiner
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Thomas Klaus Felder
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mark Rinnerthaler
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| |
Collapse
|
110
|
Zadoorian A, Du X, Yang H. Lipid droplet biogenesis and functions in health and disease. Nat Rev Endocrinol 2023:10.1038/s41574-023-00845-0. [PMID: 37221402 DOI: 10.1038/s41574-023-00845-0] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/25/2023]
Abstract
Ubiquitous yet unique, lipid droplets are intracellular organelles that are increasingly being recognized for their versatility beyond energy storage. Advances uncovering the intricacies of their biogenesis and the diversity of their physiological and pathological roles have yielded new insights into lipid droplet biology. Despite these insights, the mechanisms governing the biogenesis and functions of lipid droplets remain incompletely understood. Moreover, the causal relationship between the biogenesis and function of lipid droplets and human diseases is poorly resolved. Here, we provide an update on the current understanding of the biogenesis and functions of lipid droplets in health and disease, highlighting a key role for lipid droplet biogenesis in alleviating cellular stresses. We also discuss therapeutic strategies of targeting lipid droplet biogenesis, growth or degradation that could be applied in the future to common diseases, such as cancer, hepatic steatosis and viral infection.
Collapse
Affiliation(s)
- Armella Zadoorian
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Ximing Du
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
111
|
Chaudhari S, Ware AP, Jasti DB, Gorthi SP, Acharya LP, Bhat M, Mallya S, Satyamoorthy K. Exome sequencing of choreoacanthocytosis reveals novel mutations in VPS13A and co-mutation in modifier gene(s). Mol Genet Genomics 2023; 298:965-976. [PMID: 37209156 DOI: 10.1007/s00438-023-02032-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/04/2023] [Indexed: 05/22/2023]
Abstract
Choreoacanthocytosis, one of the forms of neuroacanthocytosis, is caused by mutations in vacuolar protein sorting-associated protein A (VPS13A), and is often misdiagnosed with other form of neuroacanthocytosis with discrete genetic defects. The phenotypic variations among the patients with VPS13A mutations significantly obfuscates the understanding of the disease and treatment strategies. In this study, two unrelated cases were identified, exhibiting the core phenotype of neuroacanthocytosis but with considerable clinical heterogeneity. Case 1 presented with an additional Parkinsonism phenotype, whereas seizures were evident in case 2. To decipher the genetic basis, whole exome sequencing followed by validation with Sanger sequencing was performed. A known homozygous pathogenic nonsense mutation (c.799C > T; p.R267X) in exon 11 of the VPS13A gene was identified in case 1 that resulted in a truncated protein. A novel missense mutation (c.9263T > G; p.M3088R) in exon 69 of VPS13A identified in case 2 was predicted as pathogenic. In silico analysis of the p.M3088R mutation at the C-terminus of VPS13A suggests a loss of interaction with TOMM40 and may disrupt mitochondrial localization. We also observed an increase in mitochondrial DNA copy numbers in case 2. Mutation analysis revealed benign heterozygous variants in interacting partners of VPS13A such as VAPA in case 1. Our study confirmed the cases as ChAc and identified the novel homozygous variant of VPS13A (c.9263T > G; p.M3088R) within the mutation spectrum of VPS13A-associated ChAc. Furthermore, mutations in VPS13A and co-mutations in its potential interacting partner(s) might contribute to the diverse clinical manifestations of ChAc, which requires further study.
Collapse
Affiliation(s)
- Sima Chaudhari
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Akshay Pramod Ware
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Dushyanth Babu Jasti
- Department of Neurology, Kasturba Medical College, Manipal, Karnataka, 576104, India
| | - Sankar Prasad Gorthi
- Department of Neurology, Kasturba Medical College, Manipal, Karnataka, 576104, India
- Department of Neurology, Bharati Hospital and Research Center, Bharati Vidyapeeth (Deemed to be University) Medical College and Hospital, Dhankawadi, Pune, Maharashtra, 411043, India
| | - Lavanya Prakash Acharya
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Manoj Bhat
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
112
|
Khan MA, Khan ZA, Shoeb F, Fatima G, Khan RH, Khan MM. Role of de novo lipogenesis in inflammation and insulin resistance in alzheimer's disease. Int J Biol Macromol 2023; 242:124859. [PMID: 37187418 DOI: 10.1016/j.ijbiomac.2023.124859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023]
Abstract
Patients with Alzheimer's disease (AD) display both peripheral tissue and brain insulin resistance, the later could be a potential risk factor for cognitive dysfunction. While certain degree of inflammation is required for inducing insulin resistance, underlying mechanism(s) remains unclear. Evidence from diverse research domains suggest that elevated intracellular fatty acids of de novo pathway can induce insulin resistance even without triggering inflammation; however, the effect of saturated fatty acids (SFAs) could be detrimental due the development of proinflammatory cues. In this context, evidence suggest that while lipid/fatty acid accumulation is a characteristic feature of brain pathology in AD, dysregulated de novo lipogenesis could be a potential source for lipid/fatty acid accumulation. Therefore, therapies aimed at regulating de novo lipogenesis could be effective in improving insulin sensitivity and cognitive function in patients with AD.
Collapse
Affiliation(s)
- Mohsin Ali Khan
- Research and Development Unit, Era's Lucknow Medical College and Hospital, Aligarh, UP, India
| | - Zaw Ali Khan
- Research and Development Unit, Era's Lucknow Medical College and Hospital, Aligarh, UP, India
| | - Fouzia Shoeb
- Department of Personalized and Molecular Medicine, Aligarh, UP, India
| | - Ghizal Fatima
- Laboratory of Chronobiology, Department of Biotechnology, Aligarh, UP, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Faculty of Life sciences, Aligarh Muslim University, Aligarh, UP, India
| | - Mohammad M Khan
- Laboratory of Chronobiology, Department of Biotechnology, Aligarh, UP, India; Laboratory of Translational Neurology and Molecular Psychiatry, Era's Lucknow Medical College and Hospital, Faculty of Science, Era University, Sarfarazganj, Lucknow, UP, India.
| |
Collapse
|
113
|
Wynne ME, Ogunbona O, Lane AR, Gokhale A, Zlatic SA, Xu C, Wen Z, Duong DM, Rayaprolu S, Ivanova A, Ortlund EA, Dammer EB, Seyfried NT, Roberts BR, Crocker A, Shanbhag V, Petris M, Senoo N, Kandasamy S, Claypool SM, Barrientos A, Wingo A, Wingo TS, Rangaraju S, Levey AI, Werner E, Faundez V. APOE expression and secretion are modulated by mitochondrial dysfunction. eLife 2023; 12:e85779. [PMID: 37171075 PMCID: PMC10231934 DOI: 10.7554/elife.85779] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/11/2023] [Indexed: 05/13/2023] Open
Abstract
Mitochondria influence cellular function through both cell-autonomous and non-cell autonomous mechanisms, such as production of paracrine and endocrine factors. Here, we demonstrate that mitochondrial regulation of the secretome is more extensive than previously appreciated, as both genetic and pharmacological disruption of the electron transport chain caused upregulation of the Alzheimer's disease risk factor apolipoprotein E (APOE) and other secretome components. Indirect disruption of the electron transport chain by gene editing of SLC25A mitochondrial membrane transporters as well as direct genetic and pharmacological disruption of either complexes I, III, or the copper-containing complex IV of the electron transport chain elicited upregulation of APOE transcript, protein, and secretion, up to 49-fold. These APOE phenotypes were robustly expressed in diverse cell types and iPSC-derived human astrocytes as part of an inflammatory gene expression program. Moreover, age- and genotype-dependent decline in brain levels of respiratory complex I preceded an increase in APOE in the 5xFAD mouse model. We propose that mitochondria act as novel upstream regulators of APOE-dependent cellular processes in health and disease.
Collapse
Affiliation(s)
- Meghan E Wynne
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Oluwaseun Ogunbona
- Department of Cell Biology, Emory UniversityAtlantaUnited States
- Department of Pathology and Laboratory Medicine, Emory UniversityAtlantaUnited States
| | - Alicia R Lane
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Avanti Gokhale
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | | | - Chongchong Xu
- Department of Psychiatry and Behavioral Sciences, Emory UniversityAtlantaUnited States
| | - Zhexing Wen
- Department of Cell Biology, Emory UniversityAtlantaUnited States
- Department of Psychiatry and Behavioral Sciences, Emory UniversityAtlantaUnited States
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Duc M Duong
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Sruti Rayaprolu
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Anna Ivanova
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Eric A Ortlund
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Eric B Dammer
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | | | - Blaine R Roberts
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Amanda Crocker
- Program in Neuroscience, Middlebury CollegeMiddleburyUnited States
| | - Vinit Shanbhag
- Department of Biochemistry, University of MissouriColumbiaUnited States
| | - Michael Petris
- Department of Biochemistry, University of MissouriColumbiaUnited States
| | - Nanami Senoo
- Department of Physiology, Johns Hopkins UniversityBaltimoreUnited States
| | | | | | - Antoni Barrientos
- Department of Neurology and Biochemistry & Molecular Biology, University of MiamiMiamiUnited States
| | - Aliza Wingo
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Thomas S Wingo
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Srikant Rangaraju
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Allan I Levey
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Erica Werner
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Victor Faundez
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| |
Collapse
|
114
|
Chung HL, Ye Q, Park YJ, Zuo Z, Mok JW, Kanca O, Tattikota SG, Lu S, Perrimon N, Lee HK, Bellen HJ. Very-long-chain fatty acids induce glial-derived sphingosine-1-phosphate synthesis, secretion, and neuroinflammation. Cell Metab 2023; 35:855-874.e5. [PMID: 37084732 PMCID: PMC10160010 DOI: 10.1016/j.cmet.2023.03.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/10/2023] [Accepted: 03/29/2023] [Indexed: 04/23/2023]
Abstract
VLCFAs (very-long-chain fatty acids) are the most abundant fatty acids in myelin. Hence, during demyelination or aging, glia are exposed to higher levels of VLCFA than normal. We report that glia convert these VLCFA into sphingosine-1-phosphate (S1P) via a glial-specific S1P pathway. Excess S1P causes neuroinflammation, NF-κB activation, and macrophage infiltration into the CNS. Suppressing the function of S1P in fly glia or neurons, or administration of Fingolimod, an S1P receptor antagonist, strongly attenuates the phenotypes caused by excess VLCFAs. In contrast, elevating the VLCFA levels in glia and immune cells exacerbates these phenotypes. Elevated VLCFA and S1P are also toxic in vertebrates based on a mouse model of multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE). Indeed, reducing VLCFA with bezafibrate ameliorates the phenotypes. Moreover, simultaneous use of bezafibrate and fingolimod synergizes to improve EAE, suggesting that lowering VLCFA and S1P is a treatment avenue for MS.
Collapse
Affiliation(s)
- Hyung-Lok Chung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Qi Ye
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ye-Jin Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jung-Wan Mok
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | | | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nobert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Hyun Kyoung Lee
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
115
|
Sheng L, Shields EJ, Gospocic J, Sorida M, Ju L, Byrns CN, Carranza F, Berger SL, Bonini N, Bonasio R. Ensheathing glia promote increased lifespan and healthy brain aging. Aging Cell 2023; 22:e13803. [PMID: 36840361 PMCID: PMC10186613 DOI: 10.1111/acel.13803] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/26/2023] Open
Abstract
Glia have an emergent role in brain aging and disease. In the Drosophila melanogaster brain, ensheathing glia function as phagocytic cells and respond to acute neuronal damage, analogous to mammalian microglia. We previously reported changes in glia composition over the life of ants and fruit flies, including a decline in the relative proportion of ensheathing glia with time. How these changes influence brain health and life expectancy is unknown. Here, we show that ensheathing glia but not astrocytes decrease in number during Drosophila melanogaster brain aging. The remaining ensheathing glia display dysregulated expression of genes involved in lipid metabolism and apoptosis, which may lead to lipid droplet accumulation, cellular dysfunction, and death. Inhibition of apoptosis rescued the decline of ensheathing glia with age, improved the neuromotor performance of aged flies, and extended lifespan. Furthermore, an expanded ensheathing glia population prevented amyloid-beta accumulation in a fly model of Alzheimer's disease and delayed the premature death of the diseased animals. These findings suggest that ensheathing glia play a vital role in regulating brain health and animal longevity.
Collapse
Affiliation(s)
- Lihong Sheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Emily J. Shields
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Urology and Institute of NeuropathologyMedical Center–University of FreiburgFreiburgGermany
| | - Janko Gospocic
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Urology and Institute of NeuropathologyMedical Center–University of FreiburgFreiburgGermany
| | - Masato Sorida
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Linyang Ju
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - China N. Byrns
- Medical Scientist Training ProgramUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Neuroscience Graduate GroupUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Faith Carranza
- Department of BiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Shelley L. Berger
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of BiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of GeneticsUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Nancy Bonini
- Neuroscience Graduate GroupUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of BiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Roberto Bonasio
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Urology and Institute of NeuropathologyMedical Center–University of FreiburgFreiburgGermany
| |
Collapse
|
116
|
Windham IA, Ragusa JV, Wallace ED, Wagner CH, White KK, Cohen S. APOE traffics to astrocyte lipid droplets and modulates triglyceride saturation and droplet size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538740. [PMID: 37162939 PMCID: PMC10168303 DOI: 10.1101/2023.04.28.538740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The E4 variant of APOE strongly predisposes individuals to late-onset Alzheimer's disease. We demonstrate that in response to neutral lipid synthesis, apolipoprotein E (APOE) in astrocytes can avoid translocation into the ER lumen and traffic to lipid droplets (LDs) via membrane bridges at ER-LD contacts. APOE knockdown promotes fewer, larger LDs containing more unsaturated triglyceride. This LD size distribution phenotype was rescued by chimeric APOE that targets only LDs. APOE4 - expressing astrocytes also form a small number of large LDs enriched in unsaturated triglyceride. Additionally, the larger LDs in APOE4 cells exhibit impaired turnover and increased sensitivity to lipid peroxidation. Our data indicate that APOE plays a previously unrecognized role as an LD surface protein that regulates LD size and composition. APOE4 is a toxic gain of function variant that causes aberrant LD composition and morphology. We propose that APOE4 astrocytes with large, unsaturated LDs are sensitized to lipid peroxidation or lipotoxicity, which could contribute to Alzheimer's disease risk. Summary Windham et al . discover that APOE in astrocytes can traffic to lipid droplets (LDs), where it modulates LD composition and size. Astrocytes expressing the Alzheimer's risk variant APOE4 form large LDs with impaired turnover and increased peroxidation sensitivity.
Collapse
|
117
|
Zorec R, Vardjan N. Adrenergic regulation of astroglial aerobic glycolysis and lipid metabolism: Towards a noradrenergic hypothesis of neurodegeneration. Neurobiol Dis 2023; 182:106132. [PMID: 37094775 DOI: 10.1016/j.nbd.2023.106132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023] Open
Abstract
Ageing is a key factor in the development of cognitive decline and dementia, an increasing and challenging problem of the modern world. The most commonly diagnosed cognitive decline is related to Alzheimer's disease (AD), the pathophysiology of which is poorly understood. Several hypotheses have been proposed. The cholinergic hypothesis is the oldest, however, recently the noradrenergic system has been considered to have a role as well. The aim of this review is to provide evidence that supports the view that an impaired noradrenergic system is causally linked to AD. Although dementia is associated with neurodegeneration and loss of neurons, this likely develops due to a primary failure of homeostatic cells, astrocytes, abundant and heterogeneous neuroglial cells in the central nervous system (CNS). The many functions that astrocytes provide to maintain the viability of neural networks include the control of ionic balance, neurotransmitter turnover, synaptic connectivity and energy balance. This latter function is regulated by noradrenaline, released from the axon varicosities of neurons arising from the locus coeruleus (LC), the primary site of noradrenaline release in the CNS. The demise of the LC is linked to AD, whereby a hypometabolic CNS state is observed clinically. This is likely due to impaired release of noradrenaline in the AD brain during states of arousal, attention and awareness. These functions controlled by the LC are needed for learning and memory formation and require activation of the energy metabolism. In this review, we address first the process of neurodegeneration and cognitive decline, highlighting the function of astrocytes. Cholinergic and/or noradrenergic deficits lead to impaired astroglial function. Then, we focus on adrenergic control of astroglial aerobic glycolysis and lipid droplet metabolism, which play a protective role but also promote neurodegeneration under some circumstances, supporting the noradrenergic hypothesis of cognitive decline. We conclude that targeting astroglial metabolism, glycolysis and/or mitochondrial processes may lead to important new developments in the future when searching for medicines to prevent or even halt cognitive decline.
Collapse
Affiliation(s)
- Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia.
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia.
| |
Collapse
|
118
|
Vaughen JP, Theisen E, Clandinin TR. From seconds to days: Neural plasticity viewed through a lipid lens. Curr Opin Neurobiol 2023; 80:102702. [PMID: 36965206 DOI: 10.1016/j.conb.2023.102702] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/31/2023] [Accepted: 02/16/2023] [Indexed: 03/27/2023]
Abstract
Many adult neurons are dynamically remodeled across timescales ranging from the rapid addition and removal of specific synaptic connections, to largescale structural plasticity events that reconfigure circuits over hours, days, and months. Membrane lipids, including brain-enriched sphingolipids, play crucial roles in these processes. In this review, we summarize progress at the intersection of neuronal activity, lipids, and structural remodeling. We highlight how brain activity modulates lipid metabolism to enable adaptive structural plasticity, and showcase glia as key players in membrane remodeling. These studies reveal that lipids act as critical signaling molecules that instruct the dynamic architecture of the brain.
Collapse
Affiliation(s)
- John P Vaughen
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, United States; Department of Developmental Biology, Stanford University, Stanford, CA, 94305, United States. https://twitter.com/gliaful
| | - Emma Theisen
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, United States. https://twitter.com/emmaktheisen
| | - Thomas R Clandinin
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, United States.
| |
Collapse
|
119
|
Scott H, Novikov B, Ugur B, Allen B, Mertsalov I, Monagas-Valentin P, Koff M, Baas Robinson S, Aoki K, Veizaj R, Lefeber DJ, Tiemeyer M, Bellen H, Panin V. Glia-neuron coupling via a bipartite sialylation pathway promotes neural transmission and stress tolerance in Drosophila. eLife 2023; 12:e78280. [PMID: 36946697 PMCID: PMC10110239 DOI: 10.7554/elife.78280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
Modification by sialylated glycans can affect protein functions, underlying mechanisms that control animal development and physiology. Sialylation relies on a dedicated pathway involving evolutionarily conserved enzymes, including CMP-sialic acid synthetase (CSAS) and sialyltransferase (SiaT) that mediate the activation of sialic acid and its transfer onto glycan termini, respectively. In Drosophila, CSAS and DSiaT genes function in the nervous system, affecting neural transmission and excitability. We found that these genes function in different cells: the function of CSAS is restricted to glia, while DSiaT functions in neurons. This partition of the sialylation pathway allows for regulation of neural functions via a glia-mediated control of neural sialylation. The sialylation genes were shown to be required for tolerance to heat and oxidative stress and for maintenance of the normal level of voltage-gated sodium channels. Our results uncovered a unique bipartite sialylation pathway that mediates glia-neuron coupling and regulates neural excitability and stress tolerance.
Collapse
Affiliation(s)
- Hilary Scott
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Boris Novikov
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Berrak Ugur
- Departments of Molecular and Human Genetics and Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Brooke Allen
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Ilya Mertsalov
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Pedro Monagas-Valentin
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Melissa Koff
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Sarah Baas Robinson
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Raisa Veizaj
- Translational Metabolic Laboratory, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical CenterNijmegenNetherlands
| | - Dirk J Lefeber
- Translational Metabolic Laboratory, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical CenterNijmegenNetherlands
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Hugo Bellen
- Departments of Molecular and Human Genetics and Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Vladislav Panin
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| |
Collapse
|
120
|
Chen ZP, Wang S, Zhao X, Fang W, Wang Z, Ye H, Wang MJ, Ke L, Huang T, Lv P, Jiang X, Zhang Q, Li L, Xie ST, Zhu JN, Hang C, Chen D, Liu X, Yan C. Lipid-accumulated reactive astrocytes promote disease progression in epilepsy. Nat Neurosci 2023; 26:542-554. [PMID: 36941428 DOI: 10.1038/s41593-023-01288-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 02/20/2023] [Indexed: 03/23/2023]
Abstract
Reactive astrocytes play an important role in neurological diseases, but their molecular and functional phenotypes in epilepsy are unclear. Here, we show that in patients with temporal lobe epilepsy (TLE) and mouse models of epilepsy, excessive lipid accumulation in astrocytes leads to the formation of lipid-accumulated reactive astrocytes (LARAs), a new reactive astrocyte subtype characterized by elevated APOE expression. Genetic knockout of APOE inhibited LARA formation and seizure activities in epileptic mice. Single-nucleus RNA sequencing in TLE patients confirmed the existence of a LARA subpopulation with a distinct molecular signature. Functional studies in epilepsy mouse models and human brain slices showed that LARAs promote neuronal hyperactivity and disease progression. Targeting LARAs by intervention with lipid transport and metabolism could thus provide new therapeutic options for drug-resistant TLE.
Collapse
Affiliation(s)
- Zhang-Peng Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China.
- Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China.
| | - Suji Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xiansen Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wen Fang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zhengge Wang
- Department of Radiology, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Epilepsy Center, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Haojie Ye
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Meng-Ju Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ling Ke
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Tengfei Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Pin Lv
- Department of Radiology, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xiaohong Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China
| | - Qipeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Liang Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Shu-Tao Xie
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Chunhua Hang
- Department of Neurosurgery, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Dijun Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Xiangyu Liu
- Epilepsy Center, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
- Department of Neurosurgery, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China.
- Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China.
- Epilepsy Center, the Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
- Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, Nanjing, China.
| |
Collapse
|
121
|
Abnormal lipid metabolism shapes astrocyte reactivity in patients with epilepsy. Nat Neurosci 2023; 26:535-536. [PMID: 36941430 DOI: 10.1038/s41593-023-01298-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
|
122
|
St-Pierre MK, Carrier M, González Ibáñez F, Khakpour M, Wallman MJ, Parent M, Tremblay MÈ. Astrocytes display ultrastructural alterations and heterogeneity in the hippocampus of aged APP-PS1 mice and human post-mortem brain samples. J Neuroinflammation 2023; 20:73. [PMID: 36918925 PMCID: PMC10015698 DOI: 10.1186/s12974-023-02752-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
The past decade has witnessed increasing evidence for a crucial role played by glial cells, notably astrocytes, in Alzheimer's disease (AD). To provide novel insights into the roles of astrocytes in the pathophysiology of AD, we performed a quantitative ultrastructural characterization of their intracellular contents and parenchymal interactions in an aged mouse model of AD pathology, as aging is considered the main risk factor for developing AD. We compared 20-month-old APP-PS1 and age-matched C57BL/6J male mice, among the ventral hippocampus CA1 strata lacunosum-moleculare and radiatum, two hippocampal layers severely affected by AD pathology. Astrocytes in both layers interacted more with synaptic elements and displayed more ultrastructural markers of increased phagolysosomal activity in APP-PS1 versus C57BL6/J mice. In addition, we investigated the ultrastructural heterogeneity of astrocytes, describing in the two examined layers a dark astrocytic state that we characterized in terms of distribution, interactions with AD hallmarks, and intracellular contents. This electron-dense astrocytic state, termed dark astrocytes, was observed throughout the hippocampal parenchyma, closely associated with the vasculature, and possessed several ultrastructural markers of cellular stress. A case study exploring the hippocampal head of an aged human post-mortem brain sample also revealed the presence of a similar electron-dense, dark astrocytic state. Overall, our study provides the first ultrastructural quantitative analysis of astrocytes among the hippocampus in aged AD pathology, as well as a thorough characterization of a dark astrocytic state conserved from mouse to human.
Collapse
Affiliation(s)
- Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada.,Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Fernando González Ibáñez
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada.,Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Mohammadparsa Khakpour
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Marie-Josée Wallman
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Center, Quebec City, QC, Canada
| | - Martin Parent
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Center, Quebec City, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada. .,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada. .,Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada. .,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada. .,Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada. .,Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
123
|
Cerasuolo M, Papa M, Colangelo AM, Rizzo MR. Alzheimer’s Disease from the Amyloidogenic Theory to the Puzzling Crossroads between Vascular, Metabolic and Energetic Maladaptive Plasticity. Biomedicines 2023; 11:biomedicines11030861. [PMID: 36979840 PMCID: PMC10045635 DOI: 10.3390/biomedicines11030861] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive and degenerative disease producing the most common type of dementia worldwide. The main pathogenetic hypothesis in recent decades has been the well-known amyloidogenic hypothesis based on the involvement of two proteins in AD pathogenesis: amyloid β (Aβ) and tau. Amyloid deposition reported in all AD patients is nowadays considered an independent risk factor for cognitive decline. Vascular damage and blood–brain barrier (BBB) failure in AD is considered a pivotal mechanism for brain injury, with increased deposition of both immunoglobulins and fibrin. Furthermore, BBB dysfunction could be an early sign of cognitive decline and the early stages of clinical AD. Vascular damage generates hypoperfusion and relative hypoxia in areas with high energy demand. Long-term hypoxia and the accumulation within the brain parenchyma of neurotoxic molecules could be seeds of a self-sustaining pathological progression. Cellular dysfunction comprises all the elements of the neurovascular unit (NVU) and neuronal loss, which could be the result of energy failure and mitochondrial impairment. Brain glucose metabolism is compromised, showing a specific region distribution. This energy deficit worsens throughout aging. Mild cognitive impairment has been reported to be associated with a glucose deficit in the entorhinal cortex and in the parietal lobes. The current aim is to understand the complex interactions between amyloid β (Aβ) and tau and elements of the BBB and NVU in the brain. This new approach aimed at the study of metabolic mechanisms and energy insufficiency due to mitochondrial impairment would allow us to define therapies aimed at predicting and slowing down the progression of AD.
Collapse
Affiliation(s)
- Michele Cerasuolo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- SYSBIO Centre of Systems Biology ISBE-IT, 20126 Milan, Italy
- Correspondence:
| | - Anna Maria Colangelo
- SYSBIO Centre of Systems Biology ISBE-IT, 20126 Milan, Italy
- Laboratory of Neuroscience “R. Levi-Montalcini”, Department of Biotechnology and Biosciences, NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| |
Collapse
|
124
|
A Perspective on the Link between Mitochondria-Associated Membranes (MAMs) and Lipid Droplets Metabolism in Neurodegenerative Diseases. BIOLOGY 2023; 12:biology12030414. [PMID: 36979106 PMCID: PMC10045954 DOI: 10.3390/biology12030414] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Mitochondria interact with the endoplasmic reticulum (ER) through contacts called mitochondria-associated membranes (MAMs), which control several processes, such as the ER stress response, mitochondrial and ER dynamics, inflammation, apoptosis, and autophagy. MAMs represent an important platform for transport of non-vesicular phospholipids and cholesterol. Therefore, this region is highly enriched in proteins involved in lipid metabolism, including the enzymes that catalyze esterification of cholesterol into cholesteryl esters (CE) and synthesis of triacylglycerols (TAG) from fatty acids (FAs), which are then stored in lipid droplets (LDs). LDs, through contact with other organelles, prevent the toxic consequences of accumulation of unesterified (free) lipids, including lipotoxicity and oxidative stress, and serve as lipid reservoirs that can be used under multiple metabolic and physiological conditions. The LDs break down by autophagy releases of stored lipids for energy production and synthesis of membrane components and other macromolecules. Pathological lipid deposition and autophagy disruption have both been reported to occur in several neurodegenerative diseases, supporting that lipid metabolism alterations are major players in neurodegeneration. In this review, we discuss the current understanding of MAMs structure and function, focusing on their roles in lipid metabolism and the importance of autophagy in LDs metabolism, as well as the changes that occur in neurogenerative diseases.
Collapse
|
125
|
Galkina OV, Vetrovoy OV, Krasovskaya IE, Eschenko ND. Role of Lipids in Regulation of Neuroglial Interactions. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:337-352. [PMID: 37076281 DOI: 10.1134/s0006297923030045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 03/28/2023]
Abstract
Lipids comprise an extremely heterogeneous group of compounds that perform a wide variety of biological functions. Traditional view of lipids as important structural components of the cell and compounds playing a trophic role is currently being supplemented by information on the possible participation of lipids in signaling, not only intracellular, but also intercellular. The review article discusses current data on the role of lipids and their metabolites formed in glial cells (astrocytes, oligodendrocytes, microglia) in communication of these cells with neurons. In addition to metabolic transformations of lipids in each type of glial cells, special attention is paid to the lipid signal molecules (phosphatidic acid, arachidonic acid and its metabolites, cholesterol, etc.) and the possibility of their participation in realization of synaptic plasticity, as well as in other possible mechanisms associated with neuroplasticity. All these new data can significantly expand our knowledge about the regulatory functions of lipids in neuroglial relationships.
Collapse
Affiliation(s)
- Olga V Galkina
- Biochemistry Department, Faculty of Biology, Saint-Petersburg State University, St. Petersburg, 199034, Russia.
| | - Oleg V Vetrovoy
- Biochemistry Department, Faculty of Biology, Saint-Petersburg State University, St. Petersburg, 199034, Russia
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, 199034, Russia
| | - Irina E Krasovskaya
- Biochemistry Department, Faculty of Biology, Saint-Petersburg State University, St. Petersburg, 199034, Russia
| | - Nataliya D Eschenko
- Biochemistry Department, Faculty of Biology, Saint-Petersburg State University, St. Petersburg, 199034, Russia
| |
Collapse
|
126
|
Affiliation(s)
- Luis F Rubio-Atonal
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Maria S Ioannou
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada.
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
127
|
Yin F. Lipid metabolism and Alzheimer's disease: clinical evidence, mechanistic link and therapeutic promise. FEBS J 2023; 290:1420-1453. [PMID: 34997690 PMCID: PMC9259766 DOI: 10.1111/febs.16344] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/14/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative disorder with multifactorial etiology, intersecting genetic and environmental risk factors, and a lack of disease-modifying therapeutics. While the abnormal accumulation of lipids was described in the very first report of AD neuropathology, it was not until recent decades that lipid dyshomeostasis became a focus of AD research. Clinically, lipidomic and metabolomic studies have consistently shown alterations in the levels of various lipid classes emerging in early stages of AD brains. Mechanistically, decades of discovery research have revealed multifaceted interactions between lipid metabolism and key AD pathogenic mechanisms including amyloidogenesis, bioenergetic deficit, oxidative stress, neuroinflammation, and myelin degeneration. In the present review, converging evidence defining lipid dyshomeostasis in AD is summarized, followed by discussions on mechanisms by which lipid metabolism contributes to pathogenesis and modifies disease risk. Furthermore, lipid-targeting therapeutic strategies, and the modification of their efficacy by disease stage, ApoE status, and metabolic and vascular profiles, are reviewed.
Collapse
Affiliation(s)
- Fei Yin
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA.,Department of Pharmacology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA.,Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
128
|
Accumulation of amyloid-β in the brain of mouse models of Alzheimer's disease is modified by altered gene expression in the presence of human apoE isoforms during aging. Neurobiol Aging 2023; 123:63-74. [PMID: 36638682 DOI: 10.1016/j.neurobiolaging.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
Apolipoprotein E4 (apoE4) is a risk factor for Alzheimer's disease (AD). Here, we investigated brain amyloid-β (Aβ) accumulation throughout the aging process in an amyloid precursor protein (APP) knock-in (KI) mouse model of AD that expresses human APPNL-G-F with or without human apoE4 or apoE3. Brain Aβ42 levels were significantly lower in 9-month-old mice that express human isoforms of apoE than in age-matched APP-KI control mice. Linear accumulation of Aβ42 began in 5-month-old apoE4 mice, and a strong increase in Aβ42 levels was observed in 21-month-old apoE3 mice. Aβ42 levels in cerebroventricular fluid were higher in apoE3 than in apoE4 mice at 6-7 months of age, suggesting that apoE3 is more efficient at clearing Aβ42 than apoE4 at these ages. However, apoE3 protein levels were lower than apoE4 protein levels in the brains of 21-month-old apoE3 and apoE4 mice, respectively, which may explain the rapid increase in brain Aβ42 burden in apoE3 mice. We identified genes that were downregulated in a human apoE-dependent (apoE4 > apoE3) and age-dependent (apoE3 = apoE4) manner, which may regulate brain Aβ burden and/or AD progression. Analysis of gene expression in AD mouse models helps identify molecular mechanisms of pleiotropy by the human APOE gene during aging.
Collapse
|
129
|
Nemtsova Y, Steinert BL, Wharton KA. Compartment specific mitochondrial dysfunction in Drosophila knock-in model of ALS reversed by altered gene expression of OXPHOS subunits and pro-fission factor Drp1. Mol Cell Neurosci 2023; 125:103834. [PMID: 36868541 DOI: 10.1016/j.mcn.2023.103834] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal multisystem neurodegenerative disease, characterized by a loss in motor function. ALS is genetically diverse, with mutations in genes ranging from those regulating RNA metabolism, like TAR DNA-binding protein (TDP-43) and Fused in sarcoma (FUS), to those that act to maintain cellular redox homeostasis, like superoxide dismutase 1 (SOD1). Although varied in genetic origin, pathogenic and clinical commonalities are clearly evident between cases of ALS. Defects in mitochondria is one such common pathology, thought to occur prior to, rather than as a consequence of symptom onset, making these organelles a promising therapeutic target for ALS, as well as other neurodegenerative diseases. Depending on the homeostatic needs of neurons throughout life, mitochondria are normally shuttled to different subcellular compartments to regulate metabolite and energy production, lipid metabolism, and buffer calcium. While originally considered a motor neuron disease due to the dramatic loss in motor function accompanied by motor neuron cell death in ALS patients, many studies have now implicated non-motor neurons and glial cells alike. Defects in non-motor neuron cell types often preceed motor neuron death suggesting their dysfunction may initiate and/or facilitate the decline in motor neuron health. Here, we investigate mitochondria in a Drosophila Sod1 knock-in model of ALS. In depth, in vivo, examination reveals mitochondrial dysfunction evident prior to onset of motor neuron degeneration. Genetically encoded redox biosensors identify a general disruption in the electron transport chain (ETC). Compartment specific abnormalities in mitochondrial morphology is observed in diseased sensory neurons, accompanied by no apparent defects in the axonal transport machinery, but instead an increase in mitophagy in synaptic regions. The decrease in networked mitochondria at the synapse is reversed upon downregulation of the pro-fission factor Drp1. Furthermore, altered expression of specific OXPHOS subunits reverses ALS-associated defects in mitochondrial morphology and function.
Collapse
Affiliation(s)
- Y Nemtsova
- Molecular Biology, Cell Biology, and Biochemistry Department, Brown University, Providence, RI 02912, United States.
| | - B L Steinert
- Molecular Biology, Cell Biology, and Biochemistry Department, Brown University, Providence, RI 02912, United States.
| | - K A Wharton
- Molecular Biology, Cell Biology, and Biochemistry Department, Brown University, Providence, RI 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI 02912, United States.
| |
Collapse
|
130
|
Mi Y, Qi G, Vitali F, Shang Y, Raikes AC, Wang T, Jin Y, Brinton RD, Gu H, Yin F. Loss of fatty acid degradation by astrocytic mitochondria triggers neuroinflammation and neurodegeneration. Nat Metab 2023; 5:445-465. [PMID: 36959514 PMCID: PMC10202034 DOI: 10.1038/s42255-023-00756-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 02/03/2023] [Indexed: 03/25/2023]
Abstract
Astrocytes provide key neuronal support, and their phenotypic transformation is implicated in neurodegenerative diseases. Metabolically, astrocytes possess low mitochondrial oxidative phosphorylation (OxPhos) activity, but its pathophysiological role in neurodegeneration remains unclear. Here, we show that the brain critically depends on astrocytic OxPhos to degrade fatty acids (FAs) and maintain lipid homeostasis. Aberrant astrocytic OxPhos induces lipid droplet (LD) accumulation followed by neurodegeneration that recapitulates key features of Alzheimer's disease (AD), including synaptic loss, neuroinflammation, demyelination and cognitive impairment. Mechanistically, when FA load overwhelms astrocytic OxPhos capacity, elevated acetyl-CoA levels induce astrocyte reactivity by enhancing STAT3 acetylation and activation. Intercellularly, lipid-laden reactive astrocytes stimulate neuronal FA oxidation and oxidative stress, activate microglia through IL-3 signalling, and inhibit the biosynthesis of FAs and phospholipids required for myelin replenishment. Along with LD accumulation and impaired FA degradation manifested in an AD mouse model, we reveal a lipid-centric, AD-resembling mechanism by which astrocytic mitochondrial dysfunction progressively induces neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Yashi Mi
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Guoyuan Qi
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Francesca Vitali
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA
- Department of Neurology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA
| | - Yuan Shang
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Adam C Raikes
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Tian Wang
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA
- Department of Neurology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA
| | - Yan Jin
- Center of Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Roberta D Brinton
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA
- Department of Neurology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA
- Department of Pharmacology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Haiwei Gu
- Center of Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA.
- Department of Pharmacology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA.
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
131
|
Aging and memory are altered by genetically manipulating lactate dehydrogenase in the neurons or glia of flies. Aging (Albany NY) 2023; 15:947-981. [PMID: 36849157 PMCID: PMC10008500 DOI: 10.18632/aging.204565] [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: 11/04/2022] [Accepted: 02/20/2023] [Indexed: 03/01/2023]
Abstract
The astrocyte-neuron lactate shuttle hypothesis posits that glial-generated lactate is transported to neurons to fuel metabolic processes required for long-term memory. Although studies in vertebrates have revealed that lactate shuttling is important for cognitive function, it is uncertain if this form of metabolic coupling is conserved in invertebrates or is influenced by age. Lactate dehydrogenase (Ldh) is a rate limiting enzyme that interconverts lactate and pyruvate. Here we genetically manipulated expression of Drosophila melanogaster lactate dehydrogenase (dLdh) in neurons or glia to assess the impact of altered lactate metabolism on invertebrate aging and long-term courtship memory at different ages. We also assessed survival, negative geotaxis, brain neutral lipids (the core component of lipid droplets) and brain metabolites. Both upregulation and downregulation of dLdh in neurons resulted in decreased survival and memory impairment with age. Glial downregulation of dLdh expression caused age-related memory impairment without altering survival, while upregulated glial dLdh expression lowered survival without disrupting memory. Both neuronal and glial dLdh upregulation increased neutral lipid accumulation. We provide evidence that altered lactate metabolism with age affects the tricarboxylic acid (TCA) cycle, 2-hydroxyglutarate (2HG), and neutral lipid accumulation. Collectively, our findings indicate that the direct alteration of lactate metabolism in either glia or neurons affects memory and survival but only in an age-dependent manner.
Collapse
|
132
|
Tanaka Y, Minami Y, Endo M. Ror1 promotes PPARα-mediated fatty acid metabolism in astrocytes. Genes Cells 2023; 28:307-318. [PMID: 36811220 DOI: 10.1111/gtc.13013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023]
Abstract
Ror1 signaling regulates cell polarity, migration, proliferation, and differentiation during developmental morphogenesis, and plays an important role in regulating neurogenesis in the embryonic neocortices. However, the role of Ror1 signaling in the brains after birth remains largely unknown. Here, we found that expression levels of Ror1 in the mouse neocortices increase during the postnatal period, when astrocytes mature and start expressing GFAP. Indeed, Ror1 is highly expressed in cultured postmitotic mature astrocytes. RNA-Seq analysis revealed that Ror1 expressed in cultured astrocytes mediates upregulated expression of genes related to fatty acid (FA) metabolism, including the gene encoding carnitine palmitoyl-transferase 1a (Cpt1a), the rate-limiting enzyme of mitochondrial fatty acid β-oxidation (FAO). We also found that Ror1 promotes the degradation of lipid droplets (LDs) accumulated in the cytoplasm of cultured astrocytes after oleic acid loading, and that suppressed expression of Ror1 decreases the amount of FAs localized at mitochondria, intracellular ATP levels, and expression levels of peroxisome proliferator-activated receptor α (PPARα) target genes, including Cpt1a. Collectively, these findings indicate that Ror1 signaling promotes PPARα-mediated transcription of FA metabolism-related genes, thereby facilitating the availability of FAs derived from LDs for mitochondrial FAO in the mature astrocytes.
Collapse
Affiliation(s)
- Yuki Tanaka
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Mitsuharu Endo
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| |
Collapse
|
133
|
Pan X, Dutta D, Lu S, Bellen HJ. Sphingolipids in neurodegenerative diseases. Front Neurosci 2023; 17:1137893. [PMID: 36875645 PMCID: PMC9978793 DOI: 10.3389/fnins.2023.1137893] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/27/2023] [Indexed: 02/18/2023] Open
Abstract
Neurodegenerative Diseases (NDDs) are a group of disorders that cause progressive deficits of neuronal function. Recent evidence argues that sphingolipid metabolism is affected in a surprisingly broad set of NDDs. These include some lysosomal storage diseases (LSDs), hereditary sensory and autonomous neuropathy (HSAN), hereditary spastic paraplegia (HSP), infantile neuroaxonal dystrophy (INAD), Friedreich's ataxia (FRDA), as well as some forms of amyotrophic lateral sclerosis (ALS) and Parkinson's disease (PD). Many of these diseases have been modeled in Drosophila melanogaster and are associated with elevated levels of ceramides. Similar changes have also been reported in vertebrate cells and mouse models. Here, we summarize studies using fly models and/or patient samples which demonstrate the nature of the defects in sphingolipid metabolism, the organelles that are implicated, the cell types that are initially affected, and potential therapeutics for these diseases.
Collapse
Affiliation(s)
- Xueyang Pan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
134
|
Kaplan L, Drexler C, Pfaller AM, Brenna S, Wunderlich KA, Dimitracopoulos A, Merl-Pham J, Perez MT, Schlötzer-Schrehardt U, Enzmann V, Samardzija M, Puig B, Fuchs P, Franze K, Hauck SM, Grosche A. Retinal regions shape human and murine Müller cell proteome profile and functionality. Glia 2023; 71:391-414. [PMID: 36334068 DOI: 10.1002/glia.24283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/08/2022]
Abstract
The human macula is a highly specialized retinal region with pit-like morphology and rich in cones. How Müller cells, the principal glial cell type in the retina, are adapted to this environment is still poorly understood. We compared proteomic data from cone- and rod-rich retinae from human and mice and identified different expression profiles of cone- and rod-associated Müller cells that converged on pathways representing extracellular matrix and cell adhesion. In particular, epiplakin (EPPK1), which is thought to play a role in intermediate filament organization, was highly expressed in macular Müller cells. Furthermore, EPPK1 knockout in a human Müller cell-derived cell line led to a decrease in traction forces as well as to changes in cell size, shape, and filopodia characteristics. We here identified EPPK1 as a central molecular player in the region-specific architecture of the human retina, which likely enables specific functions under the immense mechanical loads in vivo.
Collapse
Affiliation(s)
- Lew Kaplan
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Corinne Drexler
- Max Perutz Labs, Department of Biochemistry and Cell Biology, University of Vienna, Vienna Biocenter Campus (VBC), Vienna, Austria
- Vienna Biocenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Anna M Pfaller
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kirsten A Wunderlich
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Andrea Dimitracopoulos
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Juliane Merl-Pham
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Maria-Theresa Perez
- Department of Clinical Sciences, Division of Ophthalmology, Lund University, Lund, Sweden
- NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden
| | | | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Marijana Samardzija
- Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Fuchs
- Max Perutz Labs, Department of Biochemistry and Cell Biology, University of Vienna, Vienna Biocenter Campus (VBC), Vienna, Austria
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Institute of Medical Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
135
|
Danielli M, Perne L, Jarc Jovičić E, Petan T. Lipid droplets and polyunsaturated fatty acid trafficking: Balancing life and death. Front Cell Dev Biol 2023; 11:1104725. [PMID: 36776554 PMCID: PMC9911892 DOI: 10.3389/fcell.2023.1104725] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Lipid droplets are fat storage organelles ubiquitously distributed across the eukaryotic kingdom. They have a central role in regulating lipid metabolism and undergo a dynamic turnover of biogenesis and breakdown to meet cellular requirements for fatty acids, including polyunsaturated fatty acids. Polyunsaturated fatty acids esterified in membrane phospholipids define membrane fluidity and can be released by the activity of phospholipases A2 to act as ligands for nuclear receptors or to be metabolized into a wide spectrum of lipid signaling mediators. Polyunsaturated fatty acids in membrane phospholipids are also highly susceptible to lipid peroxidation, which if left uncontrolled leads to ferroptotic cell death. On the one hand, lipid droplets act as antioxidant organelles that control polyunsaturated fatty acid storage in triglycerides in order to reduce membrane lipid peroxidation, preserve organelle function and prevent cell death, including ferroptosis. On the other hand, lipid droplet breakdown fine-tunes the delivery of polyunsaturated fatty acids into metabolic and signaling pathways, but unrestricted lipid droplet breakdown may also lead to the release of lethal levels of polyunsaturated fatty acids. Precise regulation of lipid droplet turnover is thus essential for polyunsaturated fatty acid distribution and cellular homeostasis. In this review, we focus on emerging aspects of lipid droplet-mediated regulation of polyunsaturated fatty acid trafficking, including the management of membrane lipid peroxidation, ferroptosis and lipid mediator signaling.
Collapse
Affiliation(s)
| | | | | | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
136
|
Singh L, Nair L, Kumar D, Arora MK, Bajaj S, Gadewar M, Mishra SS, Rath SK, Dubey AK, Kaithwas G, Choudhary M, Singh M. Hypoxia induced lactate acidosis modulates tumor microenvironment and lipid reprogramming to sustain the cancer cell survival. Front Oncol 2023; 13:1034205. [PMID: 36761981 PMCID: PMC9906992 DOI: 10.3389/fonc.2023.1034205] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023] Open
Abstract
It is well known that solid hypoxic tumour cells oxidise glucose through glycolysis, and the end product of this pathway is fermented into lactate which accumulates in the tumour microenvironment (TME). Initially, it was proclaimed that cancer cells cannot use lactate; therefore, they dump it into the TME and subsequently augment the acidity of the tumour milieu. Furthermore, the TME acts as a lactate sink with stope variable amount of lactate in different pathophysiological condition. Regardless of the amount of lactate pumped out within TME, it disappears immediately which still remains an unresolved puzzle. Recent findings have paved pathway in exploring the main role of lactate acidosis in TME. Cancer cells utilise lactate in the de novo fatty acid synthesis pathway to initiate angiogenesis and invasiveness, and lactate also plays a crucial role in the suppression of immunity. Furthermore, lactate re-programme the lipid biosynthetic pathway to develop a metabolic symbiosis in normoxic, moderately hypoxic and severely hypoxic cancer cells. For instance: severely hypoxic cancer cells enable to synthesizing poly unsaturated fatty acids (PUFA) in oxygen scarcity secretes excess of lactate in TME. Lactate from TME is taken up by the normoxic cancer cells whereas it is converted back to PUFAs after a sequence of reactions and then liberated in the TME to be utilized in the severely hypoxic cancer cells. Although much is known about the role of lactate in these biological processes, the exact molecular pathways that are involved remain unclear. This review attempts to understand the molecular pathways exploited by lactate to initiate angiogenesis, invasiveness, suppression of immunity and cause re-programming of lipid synthesis. This review will help the researchers to develop proper understanding of lactate associated bimodal regulations of TME.
Collapse
Affiliation(s)
- Lakhveer Singh
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Lakshmi Nair
- Department of Pharmaceutical Science, Assam University (A Central University), Silchar, Assam, India
| | - Dinesh Kumar
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Mandeep Kumar Arora
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Sakshi Bajaj
- Chaudhary Devi Lal College of Pharmacy, Yamuna Nagar, India
| | - Manoj Gadewar
- School of Medical and Allied Sciences, KR Mangalam University, Gurgaon, India
| | | | - Santosh Kumar Rath
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Amit Kumar Dubey
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Gaurav Kaithwas
- Department of Pharmaceutical Science, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Manjusha Choudhary
- University Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, India,*Correspondence: Manjusha Choudhary, ; Manjari Singh,
| | - Manjari Singh
- Department of Pharmaceutical Science, Assam University (A Central University), Silchar, Assam, India,*Correspondence: Manjusha Choudhary, ; Manjari Singh,
| |
Collapse
|
137
|
Cyto-Genotoxic and Behavioral Effects of Flubendiamide in Allium cepa Root Cells, Drosophila melanogaster and Molecular Docking Studies. Int J Mol Sci 2023; 24:ijms24021565. [PMID: 36675079 PMCID: PMC9861014 DOI: 10.3390/ijms24021565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/28/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Flubendiamide (FLB) is an insecticide that is commonly employed to control pests on a variety of vegetables and fruits, with low toxicity for non-target organisms. However, due to its widespread use, the environmental risks and food safety have become major concerns. In this study, the toxicity potential of FLB was studied in the model organisms, Allium cepa and Drosophila melanogaster. The cyto-genotoxic effects of FLB on the root growth, mitotic index (MI), chromosomal aberrations (CAs) and deoxyribonucleic acid (DNA) damage in A. cepa root meristematic cells were investigated using the root growth inhibition Allium test and Comet assays. FLB caused CAs in the form of disturbed ana-telophase, chromosome laggards, stickiness, anaphase-bridge and polyploidy depending on the concentration and the exposure time. The toxicity and genotoxicity of FLB at various doses (0.001, 0.01, 0.1 and 1 mM) on D. melanogaster were investigated from the point of view of larval weight and movement, pupal formation success, pupal position, emergence success and DNA damage, respectively. FLB exposure led to a significant reduction of the locomotor activity at the highest concentration. While DNA damage increased significantly in the FLB-treated onions depending on the concentration and time, DNA damage in the FLB-treated D. melanogaster significantly increased only at the highest dose compared to that which occurred in the control group. Moreover, to provide a mechanistic insight into the genotoxic and locomotion-disrupting effects of FLB, molecular docking simulations of this pesticide were performed against the DNA and diamondback moth (DBM) ryanodine receptor (RyR) Repeat34 domain. The docking studies revealed that FLB binds strongly to a DNA region that is rich in cytosine-guanine-adenine bases (C-G-A) in the minor groove, and it displayed a remarkable binding affinity against the DBM RyR Repeat34 domain.
Collapse
|
138
|
Satouh Y, Sato K. Reorganization, specialization, and degradation of oocyte maternal components for early development. Reprod Med Biol 2023; 22:e12505. [PMID: 36726596 PMCID: PMC9884333 DOI: 10.1002/rmb2.12505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 01/30/2023] Open
Abstract
Background Oocyte components are maternally provided, solely determine oocyte quality, and coordinately determine embryo quality with zygotic gene expression. During oocyte maturation, maternal organelles are drastically reorganized and specialized to support oocyte characteristics. A large number of maternal components are actively degraded after fertilization and gradually replaced by zygotic gene products. The molecular basis and the significance of these processes on oocyte/embryo quality are not fully understood. Methods Firstly, recent findings in organelle characteristics of other cells or oocytes from model organisms are introduced for further understanding of oocyte organelle reorganization/specialization. Secondly, recent progress in studies on maternal components degradation and their molecular mechanisms are introduced. Finally, future applications of these advancements for predicting mammalian oocyte/embryo quality are discussed. Main findings The significance of cellular surface protein degradation via endocytosis for embryonic development, and involvement of biogenesis of lipid droplets in embryonic quality, were recently reported using mammalian model organisms. Conclusion Identifying key oocyte component characteristics and understanding their dynamics may lead to new applications in oocyte/embryo quality prediction and improvement. To implement these multidimensional concepts, development of new technical approaches that allow us to address the complexity and efficient studies using model organisms are required.
Collapse
Affiliation(s)
- Yuhkoh Satouh
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular RegulationGunma UniversityMaebashiJapan
| | - Ken Sato
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular RegulationGunma UniversityMaebashiJapan
| |
Collapse
|
139
|
Koppel SJ, Wilkins HM, Weidling IW, Wang X, Menta BW, Swerdlow RH. β-Hydroxybutyrate preferentially enhances neuron over astrocyte respiration while signaling cellular quiescence. Mitochondrion 2023; 68:125-137. [PMID: 36516926 PMCID: PMC9805520 DOI: 10.1016/j.mito.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/13/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
While ketone bodies support overall brain energy metabolism, it is increasingly clear specific brain cell types respond differently to ketone body availability. Here, we characterized how SH-SY5Y neuroblastoma cell, primary neuron, and primary astrocyte bioenergetics and nutrient sensing pathways respond to β-hydroxybutyrate (βOHB). SH-SY5Y cells and primary neurons, but not astrocytes, exposed to βOHB increased respiration and decreased PI3K-Akt-mTOR signaling. Despite increased carbon availability and respiration, SH-SY5Y cells treated with βOHB reduced their overall metabolic activity and cell cycling rate. Levels of the quiescence-regulating Yamanaka factors increased to a broader extent in SH-SY5Y cells and primary neurons. We propose a βOHB-induced increase in neuron respiration, accompanied by activation of quiescence associated pathways, could alleviate bioenergetic stress and limit cell senescence. This in turn could potentially benefit conditions, including brain aging and neurodegenerative diseases, that feature bioenergetic decline and cell senescence.
Collapse
Affiliation(s)
- Scott J Koppel
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA; Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA; Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ian W Weidling
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA; Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Xiaowan Wang
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Blaise W Menta
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA; Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA; Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA; Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
140
|
Fang X, Zhou Y, Chen S, Xu X, Ke J, Zhou Y, Wei H, Fu B. Natural killer cells promote intra-cellular-infected trophoblasts survival via APOD-LRP1 axis. Immunology 2022. [PMID: 36562137 DOI: 10.1111/imm.13621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are known for their potent ability to kill stressed cells, whereas host cells infected with intra-cellular bacteria may also be benefit from the selective killing function of NK cells and survive. The mechanism of how NK cells protect host cells infected with intra-cellular bacteria is still unclear. Here, we discovered that decidual NK (dNK) cells cannot only eliminate intra-cellular bacteria which infected trophoblasts, but can also synthesize more lipids and transport lipids to trophoblasts to avoid their apoptosis. Mechanically, NK cells synthesize more lipids accompanied by increasing expression of apolipoprotein APOD. Lipids in NK cells can be delivered to trophoblast cells through APOD, maintaining adequate lipid droplet content and lipid metabolism homeostasis in trophoblasts. Blocking the APOD receptor LRP1 abolished lipid transport from NK cells to trophoblasts, and the reduction of lipid droplets caused by bacterial infection in trophoblast cells could not be restored, culminating in cell apoptosis. Our study provides new evidence for the immune surveillance and protective effect of NK cells on embryos during early pregnancy.
Collapse
Affiliation(s)
- Xi Fang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yonggang Zhou
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Siao Chen
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiuxiu Xu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Jieqi Ke
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Binqing Fu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
141
|
Bayazid AB, Lim BO. Quercetin Is An Active Agent in Berries against Neurodegenerative Diseases Progression through Modulation of Nrf2/HO1. Nutrients 2022; 14:5132. [PMID: 36501161 PMCID: PMC9737775 DOI: 10.3390/nu14235132] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
Berries are well-known fruits for their antioxidant effects due to their high content of flavonoids, and quercetin is one of the potent bioactive flavonoids. Although oxidative stress is an inevitable outcome in cells due to energy uptake and metabolism and other factors, excessive oxidative stress is considered a pivotal mediator for the cell death and leads to the progression of neurodegenerative diseases (NDDs). Furthermore, oxidative stress triggers inflammation that leads to neuronal cell loss. Alzheimer's, Parkinson's, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and so on are the main neurodegenerative diseases. Hence, AD and PD are the most affected NDDs and cause the most lethality without any effective cure. Since AD and PD are the most common NDDs, therefore, in this study, we will describe the effect of oxidative stress on AD and PD. Targeting oxidative stress could be a very effective way to prevent and cure NDDs. Thus, the nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO1) are potent endogenous antioxidant modulatory pathways, which also show cytoprotective activities. Modulation of Nrf2/HO1 signaling pathways through a biological approach could be an effective way to treat with NDDs. Quercetin is a natural polyphenol, which protects neurodegeneration, remarkably by suppressing oxidative stress and inflammation. Thus, quercetin could be a very effective agent against NDDs. We will discuss the benefits and challenges of quercetin to treat against NDDs, focusing on molecular biology.
Collapse
Affiliation(s)
- Al Borhan Bayazid
- Medicinal Biosciences, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Beong Ou Lim
- Medicinal Biosciences, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
- Human Bioscience Corporate R&D Center, Human Bioscience Corp. 268 Chungwondaero, Chungju 27478, Republic of Korea
| |
Collapse
|
142
|
Gao L, Zang X, Qiao H, Moussian B, Wang Y. Xenobiotic responses of Drosophila melanogaster to insecticides with different modes of action and entry. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 111:e21958. [PMID: 35942563 DOI: 10.1002/arch.21958] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 06/15/2023]
Abstract
Depending on their chemical structure, insecticides enter the insect body either through the cuticle or by ingestion (mode of entry [MoE]), and, naturally, harm or even kill insects through different mechanisms (modes of action). In parallel, they trigger a systemic detoxification response, especially by activation of detoxification gene expression. We monitored the acute genetic alterations of known xenobiotic response target genes against five different insecticides with two most common MoEs (contact toxicity and stomach toxicity), found that: 1. only a few genes were detected responding to acute exposure to insecticides (LD90 ); 2. The expression of cyp12d1 was upregulated in all experiments, except for dichlorodiphenyltrichloroethane exposure, suggesting that cyp12d1 is a general first response gene of the xenobiotic response; 3. The contact and stomach entries did not show any notable difference, both MoEs induced the response of JNK signaling pathway, possibly serving as the driver of the response of cyp12d1 and a few other genes. In conclusion, the changes in gene expression levels were relatively modest and no significant differences were found between the two MoEs, so the insecticide entry route does not seem to have an impact on the detoxification response. However, the two MoEs of the same insecticide showed different efficiencies in our test. Thus, the study of these two MoEs will help to develop more efficient release and management methods for the use of such insecticides.
Collapse
Affiliation(s)
- Lujuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xiya Zang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Huanhuan Qiao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Bernard Moussian
- Animal Genetics, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany
- Université Côte d'Azur, INRAE, CNRS, Institut Sophia Agrobiotech, Sophia Antipolis, Parc Valrose, France
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
143
|
Chen S, Huang X. Cytosolic lipolysis in non-adipose tissues: energy provision and beyond. FEBS J 2022; 289:7385-7398. [PMID: 34407292 DOI: 10.1111/febs.16161] [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: 04/09/2021] [Revised: 07/18/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022]
Abstract
Cytosolic lipolysis is a well-defined biochemical process that plays important roles in the mobilization of stored neutral lipids. Lipid turnover, regulated by cytosolic lipolysis, has been extensively studied in adipose tissue, liver, and muscle. The storage and utilization of neutral lipids is a basic function of most, if not all, tissues and cells. In this review, we focus on the functions of cytosolic lipolysis mainly in non-adipose tissues and in several physiological processes, including cancer, longevity, and pathogen infection. The mechanisms underlying the impact of cytosolic lipolysis on these events will be discussed. Detailed understanding of cytosolic lipolysis in both adipose and non-adipose tissues will have implications for future clinical translation.
Collapse
Affiliation(s)
- Siyu Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
144
|
Nitta Y, Sugie A. Studies of neurodegenerative diseases using Drosophila and the development of novel approaches for their analysis. Fly (Austin) 2022; 16:275-298. [PMID: 35765969 PMCID: PMC9336468 DOI: 10.1080/19336934.2022.2087484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 02/09/2023] Open
Abstract
The use of Drosophila in neurodegenerative disease research has contributed to the identification of modifier genes for the pathology. The basis for neurodegenerative disease occurrence in Drosophila is the conservation of genes across species and the ability to perform rapid genetic analysis using a compact brain. Genetic findings previously discovered in Drosophila can reveal molecular pathologies involved in human neurological diseases in later years. Disease models using Drosophila began to be generated during the development of genetic engineering. In recent years, results of reverse translational research using Drosophila have been reported. In this review, we discuss research on neurodegenerative diseases; moreover, we introduce various methods for quantifying neurodegeneration in Drosophila.
Collapse
Affiliation(s)
- Yohei Nitta
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Atsushi Sugie
- Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
145
|
Natarajaseenivasan K, Garcia A, Velusamy P, Shanmughapriya S, Langford D. Citrate shuttling in astrocytes is required for processing cocaine-induced neuron-derived excess peroxidated fatty acids. iScience 2022; 25:105407. [PMID: 36389000 PMCID: PMC9646946 DOI: 10.1016/j.isci.2022.105407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 08/25/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Disturbances in lipid metabolism in the CNS contribute to neurodegeneration and cognitive impairments. Through tight metabolic coupling, astrocytes provide energy to neurons by delivering lactate and cholesterol and by taking up and processing neuron-derived peroxidated fatty acids (pFA). Disruption of CNS lipid homeostasis is observed in people who use cocaine and in several neurodegenerative disorders, including HIV. The brain's main source of energy is aerobic glycolysis, but numerous studies report a switch to β-oxidation of FAs in response to cocaine. Unlike astrocytes, in response to cocaine, neurons cannot efficiently consume excess pFAs for energy. Accumulation of pFA in neurons induces autophagy and release of pFA. Astrocytes endocytose the pFA for oxidation as an energy source. Our data show that blocking mitochondrial/cytosolic citrate transport reduces the neurotrophic capacity of astrocytes, leading to decreased neuronal fitness.
Collapse
Affiliation(s)
- Kalimuthusamy Natarajaseenivasan
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Department of Microbiology, Bharathidasan University, Tiruchirapalli, India
| | - Alvaro Garcia
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Prema Velusamy
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Santhanam Shanmughapriya
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Dianne Langford
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
146
|
Ignatenko O, Malinen S, Rybas S, Vihinen H, Nikkanen J, Kononov A, Jokitalo ES, Ince-Dunn G, Suomalainen A. Mitochondrial dysfunction compromises ciliary homeostasis in astrocytes. J Biophys Biochem Cytol 2022; 222:213692. [PMID: 36383135 PMCID: PMC9674092 DOI: 10.1083/jcb.202203019] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/19/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
Astrocytes, often considered as secondary responders to neurodegeneration, are emerging as primary drivers of brain disease. Here we show that mitochondrial DNA depletion in astrocytes affects their primary cilium, the signaling organelle of a cell. The progressive oxidative phosphorylation deficiency in astrocytes induces FOXJ1 and RFX transcription factors, known as master regulators of motile ciliogenesis. Consequently, a robust gene expression program involving motile cilia components and multiciliated cell differentiation factors are induced. While the affected astrocytes still retain a single cilium, these organelles elongate and become remarkably distorted. The data suggest that chronic activation of the mitochondrial integrated stress response (ISRmt) in astrocytes drives anabolic metabolism and promotes ciliary elongation. Collectively, our evidence indicates that an active signaling axis involving mitochondria and primary cilia exists and that ciliary signaling is part of ISRmt in astrocytes. We propose that metabolic ciliopathy is a novel pathomechanism for mitochondria-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Olesia Ignatenko
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Satu Malinen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sofiia Rybas
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Helena Vihinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Joni Nikkanen
- Cardiovascular Research Institute, University of California, San Francisco, CA
| | | | - Eija S. Jokitalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Gulayse Ince-Dunn
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu Suomalainen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland,HUS Diagnostics, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
147
|
Agrawal I, Lim YS, Ng SY, Ling SC. Deciphering lipid dysregulation in ALS: from mechanisms to translational medicine. Transl Neurodegener 2022; 11:48. [DOI: 10.1186/s40035-022-00322-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractLipids, defined by low solubility in water and high solubility in nonpolar solvents, can be classified into fatty acids, glycerolipids, glycerophospholipids, sphingolipids, and sterols. Lipids not only regulate integrity and fluidity of biological membranes, but also serve as energy storage and bioactive molecules for signaling. Causal mutations in SPTLC1 (serine palmitoyltransferase long chain subunit 1) gene within the lipogenic pathway have been identified in amyotrophic lateral sclerosis (ALS), a paralytic and fatal motor neuron disease. Furthermore, lipid dysmetabolism within the central nervous system and circulation is associated with ALS. Here, we aim to delineate the diverse roles of different lipid classes and understand how lipid dysmetabolism may contribute to ALS pathogenesis. Among the different lipids, accumulation of ceramides, arachidonic acid, and lysophosphatidylcholine is commonly emerging as detrimental to motor neurons. We end with exploring the potential ALS therapeutics by reducing these toxic lipids.
Collapse
|
148
|
Derepression of the USP22-FASN axis by p53 loss under oxidative stress drives lipogenesis and tumorigenesis. Cell Death Dis 2022; 8:445. [PMID: 36333288 PMCID: PMC9636132 DOI: 10.1038/s41420-022-01241-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Overproduction of reactive oxygen species (ROS) and aberrant lipid metabolism are established hallmarks of cancer; however, the role of ROS in lipid synthesis during tumorigenesis is almost unknown. Herein, we show that ROS regulates lipid synthesis and thus controls colorectal tumorigenesis through a p53-dependent mechanism. In p53 wild-type colorectal cancer (CRC) cells, hydrogen peroxide (H2O2)-induced p53 expression represses the transcription of deubiquitinase USP22, which otherwise deubiquitinates and stabilizes Fatty Acid Synthase (FASN), and thus inhibits fatty acid synthesis. Whereas, in p53-deficient CRC cells, ROS-mediated inhibition of USP22 is relieved, leading to FASN stabilization, which thus promotes lipid synthesis and tumor growth. In human CRC specimens, USP22 expression is positively correlated with FASN expression. Our study demonstrates that ROS critically regulates lipid synthesis and tumorigenesis through the USP22-FASN axis in a p53-dependent manner, and targeting the USP22-FASN axis may represent a potential strategy for the treatment of colorectal cancer.
Collapse
|
149
|
Yang R, Wei Y, Zhao M, Shi M, Zhao Y, Sun P. PBA functionalized single-atom Fe for efficient therapy of multidrug-resistant bacterial infections. Colloids Surf B Biointerfaces 2022; 219:112811. [PMID: 36067683 DOI: 10.1016/j.colsurfb.2022.112811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 10/31/2022]
Abstract
The abuse of antibiotics has led to the emergence of multidrug-resistant bacterial strains worldwide, which greatly threatens human health. In the present work, we developed single-atom catalysts (SACs) with atomically dispersed Fe as catalytic sites (Fe-SACs) to combat multidrug-resistant bacteria by elevating cellular reactive oxygen species (ROS). Our intensive studies confirmed that Fe-SACs were successfully prepared and exhibited excellent catalase (CAT)-, oxidase (OXD)-, and peroxidase (POD)-like activities. To enhance water dispersibility, biosafety and the interactions between the nanodrugs and gram-positive bacteria, phenylboronic acid group-functionalized carboxylated chitosan (CCS-PBA) was coated on the surface of Fe-SACs to yield Fe-SACs@CCS-PBA for in vitro and in vivo studies. The synergistic catalytic activity and photothermal activity of Fe-SACs@CCS-PBA effectively overcame multidrug-resistant bacterial strains (MRSA) in vitro and significantly accelerated wound healing in vivo, suggesting the great potential of SACs to overcome infectious disease caused by multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Ruigeng Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yueru Wei
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Mengyang Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Mengxiao Shi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, Henan, PR China; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Pengchao Sun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, Henan, PR China; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
150
|
Wu Y, Chen K, Li L, Hao Z, Wang T, Liu Y, Xing G, Liu Z, Li H, Yuan H, Lu J, Zhang C, Zhang J, Zhao D, Wang J, Nie J, Ye D, Pan G, Chan WY, Liu X. Plin2-mediated lipid droplet mobilization accelerates exit from pluripotency by lipidomic remodeling and histone acetylation. Cell Death Differ 2022; 29:2316-2331. [PMID: 35614132 PMCID: PMC9613632 DOI: 10.1038/s41418-022-01018-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/07/2022] [Accepted: 05/08/2022] [Indexed: 12/29/2022] Open
Abstract
Metabolic switch is critical for cell fate determination through metabolic functions, epigenetic modifications, and gene expression. However, the mechanisms underlying these alterations and their functional roles remain unclear. Here, we show that Plin2-mediated moderate lipid hydrolysis is critical for pluripotency of embryonic stem cells (ESCs). Upon exit from pluripotency, lipid droplet (LD)-associated protein Plin2 is recognized by Hsc70 and degraded via chaperone-mediated autophagy to facilitate LD mobilization. Enhancing lipid hydrolysis by Plin2 knockout promotes pluripotency exit, which is recovered by ATGL inhibition. Mechanistically, excessive lipid hydrolysis induces a dramatic lipidomic remodeling characterized by decreased cardiolipin and phosphatidylethanolamine, which triggers defects in mitochondrial cristae and fatty acid oxidation, resulting in reduced acetyl-CoA and histone acetylation. Our results reveal how LD mobilization is regulated and its critical role in ESC pluripotency, and indicate the mechanism linking LD homeostasis to mitochondrial remodeling and epigenetic regulation, which might shed light on development and diseases.
Collapse
Affiliation(s)
- Yi Wu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Keshi Chen
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Linpeng Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zhihong Hao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianyu Wang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yang Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangsuo Xing
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zichao Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Heying Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Hao Yuan
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jianghuan Lu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | | | | | - Danyun Zhao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Junwei Wang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jinfu Nie
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Dan Ye
- Fudan University, Shanghai, 200433, China
| | - Guangjin Pan
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wai-Yee Chan
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| |
Collapse
|