1
|
Xing C, Zhang C, Xu Z, Wang Y, Lu W, Liu X, Zhang Y, Ma J, Yang S, Du Y, Xu G, Liu Y. Genome-wide CRISPR screening identifies LRP1 as an entry factor for SFTSV. Nat Commun 2025; 16:4036. [PMID: 40301361 PMCID: PMC12041282 DOI: 10.1038/s41467-025-59305-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne disease caused by the SFTS virus (SFTSV), which has high mortality rates and poses a significant threat to public health. To identify potential therapeutic targets against SFTSV, we conduct genome-wide knockout screening, which identifies the previously known host factor CCR2, and reveals prolow-density lipoprotein receptor-related protein 1 (LRP1) as an entry factor for SFTSV. Knockdown or knockout of LRP1 significantly attenuate SFTSV infection in mouse embryonic fibroblasts (MEFs). Additionally, inhibition of LRP1 suppresses SFTSV pseudovirus infection in MEFs, suggesting its role in viral entry. The interaction between the SFTSV glycoprotein Gn and LRP1 via the CLI and CLII domains is revealed by co-IP and surface plasmon resonance (SPR). Moreover, LRP1 antagonists and neutralizing antibodies effectively attenuate SFTSV infection in MEFs. Administration of an LRP1-neutralizing antibody in a lethal male mouse model reduces the viral load, mitigates tissue damage, and improves survival. This study identifies LRP1 as a host entry receptor for SFTSV, providing a target for therapeutic strategy development.
Collapse
Affiliation(s)
- Chen Xing
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Cong Zhang
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Hefei, China
| | - Zhihao Xu
- The First Clinical Medical School, Anhui Medical University, Hefei, China
| | - Yajie Wang
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wanqing Lu
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiaohan Liu
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yingying Zhang
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jingyuan Ma
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Shuqi Yang
- Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yinan Du
- Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Hefei, China
| | - Gang Xu
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
- Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Yan Liu
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
- Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Hefei, China.
- State Key Laboratory for Safe Mining of Deep Coal Resources and Environment Protection, School of Public Health, Anhui University of Science and Technology, Huainan, China.
| |
Collapse
|
2
|
Ferhat L, Soussi R, Masse M, Kyriatzis G, Girard S, Gassiot F, Gaudin N, Laurencin M, Bernard A, Bôle A, Ferracci G, Smirnova M, Roman F, Dive V, Cisternino S, Temsamani J, David M, Lécorché P, Jacquot G, Khrestchatisky M. A peptide-neurotensin conjugate that crosses the blood-brain barrier induces pharmacological hypothermia associated with anticonvulsant, neuroprotective, and anti-inflammatory properties following status epilepticus in mice. eLife 2025; 13:RP100527. [PMID: 40152901 PMCID: PMC11952754 DOI: 10.7554/elife.100527] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
Preclinical and clinical studies show that mild to moderate hypothermia is neuroprotective in sudden cardiac arrest, ischemic stroke, perinatal hypoxia/ischemia, traumatic brain injury, and seizures. Induction of hypothermia largely involves physical cooling therapies, which induce several clinical complications, while some molecules have shown to be efficient in pharmacologically induced hypothermia (PIH). Neurotensin (NT), a 13 amino acid neuropeptide that regulates body temperature, interacts with various receptors to mediate its peripheral and central effects. NT induces PIH when administered intracerebrally. However, these effects are not observed if NT is administered peripherally, due to its rapid degradation and poor passage of the blood-brain barrier (BBB). We conjugated NT to peptides that bind the low-density lipoprotein receptor (LDLR) to generate 'vectorized' forms of NT with enhanced BBB permeability. We evaluated their effects in epileptic conditions following peripheral administration. One of these conjugates, VH-N412, displayed improved stability, binding potential to both the LDLR and NTSR-1, rodent/human cross-reactivity and improved brain distribution. In a mouse model of kainate (KA)-induced status epilepticus (SE), VH-N412 elicited rapid hypothermia associated with anticonvulsant effects, potent neuroprotection, and reduced hippocampal inflammation. VH-N412 also reduced sprouting of the dentate gyrus mossy fibers and preserved learning and memory skills in the treated mice. In cultured hippocampal neurons, VH-N412 displayed temperature-independent neuroprotective properties. To the best of our knowledge, this is the first report describing the successful treatment of SE with PIH. In all, our results show that vectorized NT may elicit different neuroprotection mechanisms mediated by hypothermia and/or by intrinsic neuroprotective properties.
Collapse
Affiliation(s)
- Lotfi Ferhat
- Aix-Marseille Univ, CNRS, INP, Inst NeurophysiopatholMarseilleFrance
| | - Rabia Soussi
- Aix-Marseille Univ, CNRS, INP, Inst NeurophysiopatholMarseilleFrance
| | - Maxime Masse
- VECT-HORUS SAS, Faculté de MédecineMarseilleFrance
| | | | - Stéphane Girard
- Aix-Marseille Univ, CNRS, INP, Inst NeurophysiopatholMarseilleFrance
- VECT-HORUS SAS, Faculté de MédecineMarseilleFrance
| | | | | | | | - Anne Bernard
- Aix-Marseille Univ, CNRS, INP, Inst NeurophysiopatholMarseilleFrance
| | - Angélique Bôle
- Aix-Marseille Univ, CNRS, INP, Inst NeurophysiopatholMarseilleFrance
| | | | - Maria Smirnova
- Université Paris Cité, INSERM UMRS 1144, Optimisation Thérapeutique en NeuropsychopharmacologieParisFrance
| | - François Roman
- Aix-Marseille Univ, CNRS, INP, Inst NeurophysiopatholMarseilleFrance
| | | | - Salvatore Cisternino
- Université Paris Cité, INSERM UMRS 1144, Optimisation Thérapeutique en NeuropsychopharmacologieParisFrance
- Pharmacie, Hôpital Universitaire Necker – Enfants Malades, AP-HPParisFrance
| | | | - Marion David
- VECT-HORUS SAS, Faculté de MédecineMarseilleFrance
| | | | | | | |
Collapse
|
3
|
Ramírez-Melo LM, Estrada-Luna D, Rubio-Ruiz ME, Castañeda-Ovando A, Fernández-Martínez E, Jiménez-Osorio AS, Pérez-Méndez Ó, Carreón-Torres E. Relevance of Lipoprotein Composition in Endothelial Dysfunction and the Development of Hypertension. Int J Mol Sci 2025; 26:1125. [PMID: 39940892 PMCID: PMC11817739 DOI: 10.3390/ijms26031125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Endothelial dysfunction and chronic inflammation are determining factors in the development and progression of chronic degenerative diseases, such as hypertension and atherosclerosis. Among the shared pathophysiological characteristics of these two diseases is a metabolic disorder of lipids and lipoproteins. Therefore, the contents and quality of the lipids and proteins of lipoproteins become the targets of therapeutic objective. One of the stages of lipoprotein formation occurs through the incorporation of dietary lipids by enterocytes into the chylomicrons. Consequently, the composition, structure, and especially the properties of lipoproteins could be modified through the intake of bioactive compounds. The objective of this review is to describe the roles of the different lipid and protein components of lipoproteins and their receptors in endothelial dysfunction and the development of hypertension. In addition, we review the use of some non-pharmacological treatments that could improve endothelial function and/or prevent endothelial damage. The reviewed information contributes to the understanding of lipoproteins as vehicles of regulatory factors involved in the modulation of inflammatory and hemostatic processes, the attenuation of oxidative stress, and the neutralization of toxins, rather than only cholesterol and phospholipid transporters. For this review, a bibliographic search was carried out in different online metabases.
Collapse
Affiliation(s)
- Lisette Monsibaez Ramírez-Melo
- Nutrition Academic Area Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico;
| | - Diego Estrada-Luna
- Nursing Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (D.E.-L.); (A.S.J.-O.)
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Tlalpan, Mexico City 14080, Mexico;
| | - Araceli Castañeda-Ovando
- Chemistry Academic Area, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Hidalgo, Mexico;
| | - Eduardo Fernández-Martínez
- Medicine Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Hidalgo, Mexico;
| | - Angélica Saraí Jiménez-Osorio
- Nursing Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (D.E.-L.); (A.S.J.-O.)
| | - Óscar Pérez-Méndez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Mexico City 14080, Mexico;
- Tecnológico de Monterrey, Campus Ciudad de México, Mexico City 14380, Mexico
| | - Elizabeth Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Mexico City 14080, Mexico;
| |
Collapse
|
4
|
Li W, Chen C, Xu B, Chen J, Yang M, Gao L, Zhou J. The LDL Receptor-Related Protein 1: Mechanisms and roles in promoting Aβ efflux transporter in Alzheimer's disease. Biochem Pharmacol 2025; 231:116643. [PMID: 39577706 DOI: 10.1016/j.bcp.2024.116643] [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: 08/21/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
The LDL Receptor-Related Protein 1(LRP1), a member of the Low-density Lipoprotein (LDL) receptor family, is a multifunctional cellular transporter and signaling receptor, this includes regulation of lipid metabolism, cell migration and signaling. Abnormal accumulation of amyloid beta (Aβ) in the brain is thought to be the main pathological change in Alzheimer's disease. By binding to a variety of ligands, LRP1 is involved in the internalization and degradation of Aβ, thereby affecting the course of Alzheimer's disease (AD). Here, we discuss the main mechanisms by which LRP1 mediates Aβ degradation and clearance and several current therapeutic approaches targeting LRP1. Finally, we concluded that modulating the expression level of LRP1 is an effective way to attenuate Aβ deposition and ameliorate AD. Abbreviations: LRP1, LDL Receptor-Related Protein 1;LDL, Low Density Lipoprotein; Aβ, amyloid beta; AD, Alzheimer's disease; APP, amyloid precursor protein; ApoE, apolipoprotein E; TGF, growth factor; MMP, matrix metalloproteinase;TAT, thrombin-antithrombin complex; BBB, blood-brain barrier; MMP-9,cyclophilin A (CypA)-matrix metalloproteinase-9; VMC, Vascular Mural Cell; IDE,insulin degrading enzyme; EVs, extracellular vesicles; sLRP1,shed LRP1; BDNF, brain-derived neurotrophin; IGF-1,insulin-like growth factor 1; NGF, nerve growth factor; MAPK,mitogen-activated protein kinase; ERK1/2,exogenous signal-regulated kinase1/2;JNK, c-Jun amino-terminal kinase; TLR4, toll-like receptor 4; NF-κB,nuclear factor-κB; GCAP,guanylate cyclase-activating protein; KD, ketogenic diet;KB, ketone body; BLECs,Brain-like endothelial cell; BYHWD, Buyang Huanwu decoction; LGZG, Linguizhugan decoction;P- gp, P-glycoprotein;PPARγ, Peroxisome proliferator-activated receptor γ;SP16,SERPIN peptide 16; Asx, Astaxanthin; Bex, Bexarotene.
Collapse
Affiliation(s)
- Weiyi Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, China
| | - Canyu Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Bo Xu
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Jixiang Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Mingxia Yang
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, China
| | - Lili Gao
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Jiecan Zhou
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China; The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
5
|
Itrat N, Nisa MU, Al‐Asmari F, Ramadan MF, Zongo E. A double-blind, randomized control trial to investigate the therapeutic potential of garlic scapes for high apoprotein E levels in a high-Fat diet-induced hypercholesteremic rat model. Food Sci Nutr 2024; 12:7607-7619. [PMID: 39479679 PMCID: PMC11521695 DOI: 10.1002/fsn3.4331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 11/02/2024] Open
Abstract
Hypercholesteremia is the main contributor to metabolic diseases, including obesity, hypertension, and diabetes, which are the primary global sources of morbidity and death rates. Garlic scapes, a member of the Allium sativum family and a rich source of antioxidants, are utilized in various cuisine preparations due to their unique flavors and tastes. The current study examined garlic scape powder's effect on apoprotein E and its ability to decrease cholesterol. In an in vivo experiment, normal, healthy Wistar albino rats (weeks) were divided into a negative control group (NC, n = 10) and a high-fat diet-raised group (n = 50) until they achieved cholesterol ≥250 mg/dL. Hypercholesteremic rats were further divided randomly into five groups: positive control (PC), standard group (fenofibrate 20 mg/kg bwt), and treatment groups G1, G2, and G3 that were administered with garlic scape powder 400 mg, 800 mg, and 1200 mg/kg bwt orally, respectively, for 3 months. The blood samples were examined for cholesterol, triglycerides, high-density lipoproteins (HDLs), low-density lipoproteins, apoprotein E, albumin levels, alanine aminotransferase (ALT), and aspartate aminotransferase (AST). The liver tissues of the rats were subjected to histopathology. The lipid profile was assessed using serum kit techniques, whereas an ELISA kit was used to evaluate apoprotein E, and a serum kit was used to estimate ALT and AST. In comparison to all other groups except NC, the highest dose of 1200 mg/kg bwt of garlic scapes significantly (p ≤ .05) increased serum insulin (13.66 ± 0.72 μU/mL), apoprotein E levels (6.08 ± 0.10 mg/dL), HDL (42.1 ± 1.81 mg/dL), and reduce TG (88.7 ± 1.64 mg/dL) and decreased overall cholesterol levels (67.9 ± 1.17 mg/dL). Except for NC, all treatment groups had significantly (p ≤ .05) lower ALT and AST values than PC. To sum up, powdered garlic scapes may be a great way to avoid hyperlipidemia, which raises the risk of cardiovascular illnesses. ALT and AST levels were significantly (p ≤ .05) reduced in all treatment groups compared to PC, except for NC. In conclusion, garlic scape powder may be an excellent source to prevent hyperlipidemia, a risk factor for cardiovascular diseases. In addition, powdered garlic scapes supplementation at high doses may be used as an alternative natural source in functional foods to halt hyperlipidemia without liver toxicity in the long term.
Collapse
Affiliation(s)
- Nizwa Itrat
- Department of Nutritional Sciences, Faculty of Medical SciencesGovernment College UniversityFaisalabadPunjabPakistan
| | - Mahr un Nisa
- Department of Nutritional Sciences, Faculty of Medical SciencesGovernment College UniversityFaisalabadPunjabPakistan
| | - Fahad Al‐Asmari
- Department of Food and Nutrition Sciences, College of Agricultural and Food SciencesKing Faisal UniversityAl‐AhsaSaudi Arabia
| | - Mohamed Fawzy Ramadan
- Department of Clinical Nutrition, Faculty of Applied Medical SciencesUmm Al‐Qura UniversityMakkahSaudi Arabia
| | - Eliasse Zongo
- Laboratoire de Recherche et d'Enseignement en Santé et Biotechnologies AnimalesUniversité Nazi BONIBobo Dioulasso 01Burkina Faso
| |
Collapse
|
6
|
Guo A, Wu Q, Yan X, Chen K, Liu Y, Liang D, Yang Y, Luo Q, Xiong M, Yu Y, Fei E, Chen F. Differential roles of lysosomal cholesterol transporters in the development of C. elegans NMJs. Life Sci Alliance 2024; 7:e202402584. [PMID: 39084875 PMCID: PMC11291935 DOI: 10.26508/lsa.202402584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Cholesterol homeostasis in neurons is critical for synapse formation and maintenance. Neurons with impaired cholesterol uptake undergo progressive synapse loss and eventual degeneration. To investigate the molecular mechanisms of neuronal cholesterol homeostasis and its role during synapse development, we studied motor neurons of Caenorhabditis elegans because these neurons rely on dietary cholesterol. Combining lipidomic analysis, we discovered that NCR-1, a lysosomal cholesterol transporter, promotes cholesterol absorption and synapse development. Loss of ncr-1 causes smaller synapses, and low cholesterol exacerbates the deficits. Moreover, NCR-1 deficiency hinders the increase in synapses under high cholesterol. Unexpectedly, NCR-2, the NCR-1 homolog, increases the use of cholesterol and sphingomyelins and impedes synapse formation. NCR-2 deficiency causes an increase in synapses regardless of cholesterol concentration. Inhibiting the degradation or synthesis of sphingomyelins can induce or suppress the synaptic phenotypes in ncr-2 mutants. Our findings indicate that neuronal cholesterol homeostasis is differentially controlled by two lysosomal cholesterol transporters and highlight the importance of neuronal cholesterol homeostasis in synapse development.
Collapse
Affiliation(s)
- Amin Guo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qi Wu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xin Yan
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Kanghua Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuxiang Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dingfa Liang
- Queen Mary School of Nanchang University, Jiangxi Medical College, Nanchang, China
| | - Yuxiao Yang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qunfeng Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Mingtao Xiong
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Erkang Fei
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fei Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Alassaf M, Rajan A. Adipocyte metabolic state regulates glial phagocytic function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614765. [PMID: 39386724 PMCID: PMC11463506 DOI: 10.1101/2024.09.24.614765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Obesity and type 2 diabetes are well-established risk factors for neurodegenerative disorders1-4, yet the underlying mechanisms remain poorly understood. The adipocyte-brain axis is crucial for brain function, as adipocytes secrete signaling molecules, including lipids and adipokines, that impinge on neural circuits to regulate feeding and energy expenditure5. Disruptions in the adipocyte-brain axis are associated with neurodegenerative conditions6, but the causal links are not fully understood. Neural debris accumulates with age and injury, and glial phagocytic function is crucial for clearing this debris and maintaining a healthy brain microenvironment7-9. Using adult Drosophila, we investigate how adipocyte metabolism influences glial phagocytic activity in the brain. We demonstrate that a prolonged obesogenic diet increases adipocyte fatty acid oxidation and ketogenesis. Genetic manipulations that mimic obesogenic diet-induced changes in adipocyte lipid and mitochondrial metabolism unexpectedly reduce the expression of the phagocytic receptor Draper in Drosophila microglia-like cells in the brain. We identify Apolpp-the Drosophila equivalent of human apolipoprotein B (ApoB)-as a critical adipocyte-derived signal that regulates glial phagocytosis. Additionally, we show that Lipoprotein Receptor 1 (LpR1), the LDL receptor on phagocytic glia, is required for glial capacity to clear injury-induced neuronal debris. Our findings establish that adipocyte-brain lipoprotein signaling regulates glial phagocytic function, revealing a novel pathway that links adipocyte metabolic disorders with neurodegeneration.
Collapse
Affiliation(s)
- Mroj Alassaf
- Basic Sciences Division, Fred Hutch, Seattle, WA-98109. The USA
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, WA-98109. The USA
| |
Collapse
|
8
|
Liu E, Zhang Y, Wang JZ. Updates in Alzheimer's disease: from basic research to diagnosis and therapies. Transl Neurodegener 2024; 13:45. [PMID: 39232848 PMCID: PMC11373277 DOI: 10.1186/s40035-024-00432-x] [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: 03/12/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, characterized pathologically by extracellular deposition of β-amyloid (Aβ) into senile plaques and intracellular accumulation of hyperphosphorylated tau (pTau) as neurofibrillary tangles. Clinically, AD patients show memory deterioration with varying cognitive dysfunctions. The exact molecular mechanisms underlying AD are still not fully understood, and there are no efficient drugs to stop or reverse the disease progression. In this review, we first provide an update on how the risk factors, including APOE variants, infections and inflammation, contribute to AD; how Aβ and tau become abnormally accumulated and how this accumulation plays a role in AD neurodegeneration. Then we summarize the commonly used experimental models, diagnostic and prediction strategies, and advances in periphery biomarkers from high-risk populations for AD. Finally, we introduce current status of development of disease-modifying drugs, including the newly officially approved Aβ vaccines, as well as novel and promising strategies to target the abnormal pTau. Together, this paper was aimed to update AD research progress from fundamental mechanisms to the clinical diagnosis and therapies.
Collapse
Affiliation(s)
- Enjie Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yao Zhang
- Department of Endocrine, Liyuan Hospital, Key Laboratory of Ministry of Education for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Jian-Zhi Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
9
|
Zhou J, Zhang L, Peng J, Zhang X, Zhang F, Wu Y, Huang A, Du F, Liao Y, He Y, Xie Y, Gu L, Kuang C, Ou W, Xie M, Tu T, Pang J, Zhang D, Guo K, Feng Y, Yin S, Cao Y, Li T, Jiang Y. Astrocytic LRP1 enables mitochondria transfer to neurons and mitigates brain ischemic stroke by suppressing ARF1 lactylation. Cell Metab 2024; 36:2054-2068.e14. [PMID: 38906140 DOI: 10.1016/j.cmet.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 09/11/2023] [Accepted: 05/23/2024] [Indexed: 06/23/2024]
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is an endocytic/signaling cell-surface receptor that regulates diverse cellular functions, including cell survival, differentiation, and proliferation. LRP1 has been previously implicated in the pathogenesis of neurodegenerative disorders, but there are inconsistencies in its functions. Therefore, whether and how LRP1 maintains brain homeostasis remains to be clarified. Here, we report that astrocytic LRP1 promotes astrocyte-to-neuron mitochondria transfer by reducing lactate production and ADP-ribosylation factor 1 (ARF1) lactylation. In astrocytes, LRP1 suppressed glucose uptake, glycolysis, and lactate production, leading to reduced lactylation of ARF1. Suppression of astrocytic LRP1 reduced mitochondria transfer into damaged neurons and worsened ischemia-reperfusion injury in a mouse model of ischemic stroke. Furthermore, we examined lactate levels in human patients with stroke. Cerebrospinal fluid (CSF) lactate was elevated in stroke patients and inversely correlated with astrocytic mitochondria. These findings reveal a protective role of LRP1 in brain ischemic stroke by enabling mitochondria-mediated astrocyte-neuron crosstalk.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Lifang Zhang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Jianhua Peng
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xianhui Zhang
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Fan Zhang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yuanyuan Wu
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - An Huang
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Fengling Du
- Department of Neonatology, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yuyan Liao
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yijing He
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yuke Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Long Gu
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Chenghao Kuang
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Wei Ou
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Maodi Xie
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tianqi Tu
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Jinwei Pang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Dingkun Zhang
- Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kecheng Guo
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yue Feng
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Shigang Yin
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yang Cao
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yong Jiang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
10
|
Lin H, Fu L, Zhou X, Yu A, Chen Y, Liao W, Shu G, Zhang L, Tan L, Liang H, Wang Z, Deng Q, Wang J, Jin M, Chen Z, Wei J, Cao J, Chen W, Li X, Li P, Lu J, Luo J. LRP1 induces anti-PD-1 resistance by modulating the DLL4-NOTCH2-CCL2 axis and redirecting M2-like macrophage polarisation in bladder cancer. Cancer Lett 2024; 593:216807. [PMID: 38462037 DOI: 10.1016/j.canlet.2024.216807] [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: 10/31/2023] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
The tumour microenvironment (TME) drives bladder cancer (BLCA) progression. Targeting the TME has emerged as a promising strategy for BLCA treatment in recent years. Furthermore, checkpoint blockade therapies are only beneficial for a minority of patients with BLCA, and drug resistance is a barrier to achieving significant clinical effects of anti-programmed cell death protein-1 (PD-1)/programmed death protein ligand-1 (PD-L1) therapy. In this study, higher low-density lipoprotein receptor-related protein 1 (LRP1) levels were related to a poorer prognosis for patients with various cancers, including those with higher grades and later stages of BLCA. Enrichment analysis demonstrated that LRP1 plays a role in the epithelial-mesenchymal transition (EMT), NOTCH signalling pathway, and ubiquitination. LRP1 knockdown in BLCA cells delayed BLCA progression both in vivo and in vitro. Furthermore, LRP1 knockdown suppressed EMT, reduced DLL4-NOTCH2 signalling activity, and downregulated M2-like macrophage polarisation. Patients with BLCA and higher LRP1 levels responded weakly to anti-PD-1 therapy in the IMvigor210 cohort. Moreover, LRP1 knockdown enhanced the therapeutic effects of anti-PD-1 in mice. Taken together, our findings suggest that LRP1 is a potential target for improving the efficacy of anti-PD-1/PD-L1 therapy by preventing EMT and M2-like macrophage polarisation by blocking the DLL4-NOTCH2 axis.
Collapse
Affiliation(s)
- Hansen Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liangmin Fu
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinwei Zhou
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Anze Yu
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuhang Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wuyuan Liao
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guannan Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Lizhen Zhang
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lei Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hui Liang
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhu Wang
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Qiong Deng
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Jieyan Wang
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Meiyu Jin
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhenhua Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinhuan Wei
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiazheng Cao
- Department of Urology, Jiangmen Central Hospital, Haibang Street 23, Pengjiang District, Jiangmen, 529030, Guangdong Province, China
| | - Wei Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaofei Li
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Pengju Li
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Jun Lu
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Junhang Luo
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
11
|
Ozdilek B, Agirbasli M. Soluble LRP-1 in Parkinson's disease: clues for paradoxical effects. Int J Neurosci 2024; 134:620-627. [PMID: 36184975 DOI: 10.1080/00207454.2022.2131552] [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: 06/17/2020] [Revised: 12/29/2021] [Accepted: 09/21/2022] [Indexed: 10/17/2022]
Abstract
Background: Low density lipoprotein receptor-related protein-1 (LRP-1) is highly expressed in the central nervous system and plays a role in neurodegenerative disorders. The available data on this subject-matter seem to support the presence of a correlation between LRP-1 levels and abnormal aggregation of a plurality of proteins, including tau, amyloid, and α‑synuclein. Understanding the molecular mechanisms underlying Parkinson's disease (PD) is critical for development of new therapies. Aim: To investigate serum soluble LRP-1 (sLRP-1) concentrations in patients with PD and explored their potential role as a biomarker in diagnosis and prognosis of disease. Methods: Based on well-defined inclusion and exclusion criteria, we have included 133 PD patients and 45 healthy controls. The clinical severity was assessed using Hoehn Yahr and Unified PD Rating Scale (UPDRS). Following a fasting period, venous blood samples were taken, and centrifuged. Serum samples were stored until analysis. sLRP-1 was measured by ELISA assay. Results: The median of serum sLRP-1 levels was higher in PD patients compared to that in healthy controls, but without reaching a statistical significance. There was a positive, but statistically insignificant, correlation between sLRP-1 levels and duration of disease. sLRP-1 levels had a significant correlation with UPDRS Parts I and IV. Patients with hypertension showed lower levels of sLRP-1. Conclusion: The present study suggests that serum sLRP-1 concentrations are associated with the factors influencing prognosis of PD and disease severity. Further studies are needed to definitively determine whether or not sLRP-1 can be utilized as a diagnostic and prognostic biomarker for PD.
Collapse
Affiliation(s)
- Betul Ozdilek
- Department of Neurology, Istanbul Medeniyet University Faculty of Medicine, Istanbul, Turkey
- Clinic of Neurology, Ministry of Health Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Mehmet Agirbasli
- Department of Cardiology, Istanbul Medeniyet University Faculty of Medicine, Istanbul, Turkey
- Clinic of Cardiology, Ministry of Health Goztepe Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
12
|
Yin J, Chen HL, Grigsby-Brown A, He Y, Cotten ML, Short J, Dermady A, Lei J, Gibbs M, Cheng ES, Zhang D, Long C, Xu L, Zhong T, Abzalimov R, Haider M, Sun R, He Y, Zhou Q, Tjandra N, Yuan Q. Glia-derived secretory fatty acid binding protein Obp44a regulates lipid storage and efflux in the developing Drosophila brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588417. [PMID: 38645138 PMCID: PMC11030299 DOI: 10.1101/2024.04.10.588417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Glia derived secretory factors play diverse roles in supporting the development, physiology, and stress responses of the central nervous system (CNS). Through transcriptomics and imaging analyses, we have identified Obp44a as one of the most abundantly produced secretory proteins from Drosophila CNS glia. Protein structure homology modeling and Nuclear Magnetic Resonance (NMR) experiments reveal Obp44a as a fatty acid binding protein (FABP) with a high affinity towards long-chain fatty acids in both native and oxidized forms. Further analyses demonstrate that Obp44a effectively infiltrates the neuropil, traffics between neuron and glia, and is secreted into hemolymph, acting as a lipid chaperone and scavenger to regulate lipid and redox homeostasis in the developing brain. In agreement with this essential role, deficiency of Obp44a leads to anatomical and behavioral deficits in adult animals and elevated oxidized lipid levels. Collectively, our findings unveil the crucial involvement of a noncanonical lipid chaperone to shuttle fatty acids within and outside the brain, as needed to maintain a healthy brain lipid environment. These findings could inspire the design of novel approaches to restore lipid homeostasis that is dysregulated in CNS diseases.
Collapse
Affiliation(s)
- Jun Yin
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Hsueh-Ling Chen
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Anna Grigsby-Brown
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Yi He
- Fermentation Facility, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Myriam L Cotten
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR
| | - Jacob Short
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Aidan Dermady
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Jingce Lei
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Mary Gibbs
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Ethan S Cheng
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Dean Zhang
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Caixia Long
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Lele Xu
- Advanced Science Research Center, The City University of New York, New York, NY
- Ph.D. Program in Biology, The Graduate Center of the City University of New York, New York, NY
| | - Tiffany Zhong
- Neuroscience Program, Princeton University, Princeton, NJ
| | - Rinat Abzalimov
- Advanced Science Research Center, The City University of New York, New York, NY
| | - Mariam Haider
- Department of Cell and Developmental Biology, Vanderbilt Brain Institute, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN
| | - Rong Sun
- Department of Cell and Developmental Biology, Vanderbilt Brain Institute, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN
| | - Ye He
- Advanced Science Research Center, The City University of New York, New York, NY
- Ph.D. Program in Biology, The Graduate Center of the City University of New York, New York, NY
| | - Qiangjun Zhou
- Department of Cell and Developmental Biology, Vanderbilt Brain Institute, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN
| | - Nico Tjandra
- Laboratory of Molecular Biophysics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Quan Yuan
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| |
Collapse
|
13
|
Windham IA, Cohen S. The cell biology of APOE in the brain. Trends Cell Biol 2024; 34:338-348. [PMID: 37805344 PMCID: PMC10995109 DOI: 10.1016/j.tcb.2023.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/23/2023] [Accepted: 09/08/2023] [Indexed: 10/09/2023]
Abstract
Apolipoprotein E (APOE) traffics lipids in the central nervous system. The E4 variant of APOE is a major genetic risk factor for Alzheimer's disease (AD) and a multitude of other neurodegenerative diseases, yet the molecular mechanisms by which APOE4 drives disease are still unclear. A growing collection of studies in iPSC models, knock-in mice, and human postmortem brain tissue have demonstrated that APOE4 expression in astrocytes and microglia is associated with the accumulation of cytoplasmic lipid droplets, defects in endolysosomal trafficking, impaired mitochondrial metabolism, upregulation of innate immune pathways, and a transition into a reactive state. In this review, we collate these developments and suggest testable mechanistic hypotheses that could explain common APOE4 phenotypes.
Collapse
Affiliation(s)
- Ian A Windham
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC, USA
| | - Sarah Cohen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
14
|
Wang W, Chen X, Chen J, Xu M, Liu Y, Yang S, Zhao W, Tan S. Engineering lentivirus envelope VSV-G for liver targeted delivery of IDOL-shRNA to ameliorate hypercholesterolemia and atherosclerosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102115. [PMID: 38314097 PMCID: PMC10835450 DOI: 10.1016/j.omtn.2024.102115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 01/05/2024] [Indexed: 02/06/2024]
Abstract
Lentiviral vectors (LVs) have been widely used as a tool for gene therapies. However, tissue-selective transduction after systemic delivery remains a challenge. Inducible degrader of low-density lipoprotein receptor is an attractive target for treating hypercholesterolemia. Here, a liver-targeted LV, CS8-LV-shIDOL, is developed by incorporating a hepatocyte-targeted peptide derived from circumsporozoite protein (CSP) into the lentivirus envelope for liver-targeted delivery of IDOL-shRNA (short hairpin RNA) to alleviate hypercholesterolemia. Tail-vein injection of CS8-LV-shIDOL results in extremely high accumulation in liver and nearly undetectable levels in other organs in mice. In addition, it shows superior therapeutic efficacy in lowering serum low-density lipoprotein cholesterol (LDL-C) and reducing atherosclerotic lesions over unmodified LV-shIDOL in hyperlipidemic mice. Mechanically, the envelope-engineered CS8-LV-shIDOL can enter liver cells via low-density lipoprotein receptor-related protein (LRP). Thus, this study provides a novel approach for liver-targeted delivery of IDOL-shRNA to treat hypercholesterolemia by using an envelope-engineered LV, and this delivery system has great potential for liver-targeted transgene therapy.
Collapse
Affiliation(s)
- Wei Wang
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Xuemei Chen
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Jiali Chen
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Menglong Xu
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Liu
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Shijie Yang
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Wenfeng Zhao
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Shuhua Tan
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
15
|
Wei W, Sun H, Yang B, Song E, Song Y. Coronal ApoE Protein Combines with LRP1 to Inactivate GSK3β That Mitigates Silica Nanoparticle-Induced Brain Lesion. ACS Chem Neurosci 2024; 15:808-815. [PMID: 38315060 DOI: 10.1021/acschemneuro.3c00728] [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] [Indexed: 02/07/2024] Open
Abstract
Silica nanoparticles (SiO2 NPs) are widely used engineered materials that warrant their obvious environmental exposure risk. Our previous study has shown that different routes of SiO2 NP exposure on the glycogen synthase kinase 3 beta (GSK3β) activity were related to the serum proteins enriched on the surface of SiO2 NPs, which implied that a particular protein in the serum changed the inherent toxic behavior of SiO2 NPs and inhibited the activation of GSK3β by SiO2 NPs. Here, we identified that the SiO2 NP surface enriched a large amount of apolipoprotein E (ApoE), and the ApoE protein corona bound to the lipoprotein receptor-related protein 1 (LRP1) to inactivate GSK3β, thereby reducing the damage of SiO2 NPs to the brain. This work presented the first evidence that specific biocorona reduced the toxicity of SiO2 NPs at the molecular level, which helped to elucidate the role of specific corona components on nanotoxicity.
Collapse
Affiliation(s)
- Wei Wei
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Hang Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Bingwei Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| |
Collapse
|
16
|
Kang J, Zhang C, Wang Y, Peng J, Berger B, Perrimon N, Shen J. Lipophorin receptors genetically modulate neurodegeneration caused by reduction of Psn expression in the aging Drosophila brain. Genetics 2024; 226:iyad202. [PMID: 37996068 PMCID: PMC10763532 DOI: 10.1093/genetics/iyad202] [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: 10/11/2023] [Revised: 11/01/2023] [Accepted: 11/12/2023] [Indexed: 11/25/2023] Open
Abstract
Mutations in the Presenilin (PSEN) genes are the most common cause of early-onset familial Alzheimer's disease (FAD). Studies in cell culture, in vitro biochemical systems, and knockin mice showed that PSEN mutations are loss-of-function mutations, impairing γ-secretase activity. Mouse genetic analysis highlighted the importance of Presenilin (PS) in learning and memory, synaptic plasticity and neurotransmitter release, and neuronal survival, and Drosophila studies further demonstrated an evolutionarily conserved role of PS in neuronal survival during aging. However, molecular pathways that interact with PS in neuronal survival remain unclear. To identify genetic modifiers that modulate PS-dependent neuronal survival, we developed a new DrosophilaPsn model that exhibits age-dependent neurodegeneration and increases of apoptosis. Following a bioinformatic analysis, we tested top ranked candidate genes by selective knockdown (KD) of each gene in neurons using two independent RNAi lines in Psn KD models. Interestingly, 4 of the 9 genes enhancing neurodegeneration in Psn KD flies are involved in lipid transport and metabolism. Specifically, neuron-specific KD of lipophorin receptors, lpr1 and lpr2, dramatically worsens neurodegeneration in Psn KD flies, and overexpression of lpr1 or lpr2 does not alleviate Psn KD-induced neurodegeneration. Furthermore, lpr1 or lpr2 KD alone also leads to neurodegeneration, increased apoptosis, climbing defects, and shortened lifespan. Lastly, heterozygotic deletions of lpr1 and lpr2 or homozygotic deletions of lpr1 or lpr2 similarly lead to age-dependent neurodegeneration and further exacerbate neurodegeneration in Psn KD flies. These findings show that LpRs modulate Psn-dependent neuronal survival and are critically important for neuronal integrity in the aging brain.
Collapse
Affiliation(s)
- Jongkyun Kang
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chen Zhang
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yuhao Wang
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jian Peng
- Department of Computer Science, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Jie Shen
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Takano-Kawabe K, Matoba K, Nakamura Y, Moriyama M. Low Density Lipoprotein Receptor-related Protein 2 Expression and Function in Cultured Astrocytes and Microglia. Neurochem Res 2024; 49:199-211. [PMID: 37702891 DOI: 10.1007/s11064-023-04022-7] [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: 07/26/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023]
Abstract
Activation of glial cells, astrocytes and microglia, has been observed in neurodegenerative diseases including Alzheimer's disease (AD). Amyloid β (Aβ), which is aggregated and the aggregation is detected as characteristic pathology in AD brain, is known to be produced by neurons and to activate glial cells. Clearance of Aβ from the brain via active transport system is important to prevent the accumulation and aggregation. Low density lipoprotein receptor-related protein 2 (LRP2/megalin) is an Aβ transporter. However, expression and contribution of LRP2 in astrocytes and microglia remain to be clarified. In the present study, we examined the expression of LRP2 and its roles in cultured astrocytes prepared from rat embryonic brain cortex and mouse microglial cell line BV-2. Both cultured rat astrocytes and BV-2 cells expressed LRP2 mRNA detected by RT-PCR. When lipopolysaccharide (LPS) or all-trans retinoic acid (ATRA) were added to BV-2 cells, LRP2 mRNA expression and uptake of microbeads, Aβ and insulin were increased. On the other hand, LPS decreased LRP2 expression and uptake of Aβ and insulin in cultured astrocytes. Knockdown of LRP2 using siRNA attenuated the LPS- or ATRA-increased uptake of microbeads, Aβ and insulin in BV-2 cells. These results suggest that LRP2 was expressed in both astrocytes and microglia and might be involved in endocytosis activities. Adequate control of LRP2 expression and function in astrocytes and microglia might regulate Aβ and insulin levels in brain and would be a potential target in AD pathology.
Collapse
Affiliation(s)
- Katsura Takano-Kawabe
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan.
| | - Kazuyuki Matoba
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| | - Yoichi Nakamura
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| | - Mitsuaki Moriyama
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| |
Collapse
|
18
|
Gao Y, Ye S, Tang Y, Tong W, Sun S. Brain cholesterol homeostasis and its association with neurodegenerative diseases. Neurochem Int 2023; 171:105635. [PMID: 37949118 DOI: 10.1016/j.neuint.2023.105635] [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: 09/07/2023] [Revised: 10/17/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
The brain is the most cholesterol-rich organ in mammals. However, cholesterol metabolism in the brain is completely independent of other tissues due to the presence of the blood-brain barrier (BBB). Neurons, astrocytes and oligodendrocytes are the main cells responsible for cholesterol synthesis in the brain. The cholesterol content in the brain is maintained at a relatively constant level under strict regulation of synthesis, transport, and turnover, that is, brain cholesterol homeostasis. Once this balance is disrupted, neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD) ensue. This review summarizes the processes controlling cholesterol homeostasis with respect to the synthesis, transport and turnover of cholesterol in the brain. We further focus on how cholesterol imbalance contributes to neurodegenerative diseases to explore the possibilities to modulate the key steps involved, which will provide clues for the development of therapies for the treatment of central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Yi Gao
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shiying Ye
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuehong Tang
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China.
| | - Shaowei Sun
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
19
|
Wasser CR, Werthmann GC, Hall EM, Kuhbandner K, Wong CH, Durakoglugil MS, Herz J. Regulation of the hippocampal translatome by Apoer2-ICD release. Mol Neurodegener 2023; 18:62. [PMID: 37726747 PMCID: PMC10510282 DOI: 10.1186/s13024-023-00652-1] [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: 06/13/2023] [Accepted: 08/24/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND ApoE4, the most significant genetic risk factor for late-onset Alzheimer's disease (AD), sequesters a pro-synaptogenic Reelin receptor, Apoer2, in the endosomal compartment and prevents its normal recycling. In the adult brain, Reelin potentiates excitatory synapses and thereby protects against amyloid-β toxicity. Recently, a gain-of-function mutation in Reelin that is protective against early-onset AD has been described. Alternative splicing of the Apoer2 intracellular domain (Apoer2-ICD) regulates Apoer2 signaling. Splicing of juxtamembraneous exon 16 alters the γ-secretase mediated release of the Apoer2-ICD as well as synapse number and LTP, and inclusion of exon 19 ameliorates behavioral deficits in an AD mouse model. The Apoer2-ICD has also been shown to alter transcription of synaptic genes. However, the role of Apoer2-ICD release upon transcriptional regulation and its role in AD pathogenesis is unknown. METHODS To assess in vivo mRNA-primed ribosomes specifically in hippocampi transduced with Apoer2-ICD splice variants, we crossed wild-type, cKO, and Apoer2 cleavage-resistant mice to a Cre-inducible translating ribosome affinity purification (TRAP) model. This allowed us to perform RNA-Seq on ribosome-loaded mRNA harvested specifically from hippocampal cells transduced with Apoer2-ICDs. RESULTS Across all conditions, we observed ~4,700 altered translating transcripts, several of which comprise key synaptic components such as extracellular matrix and focal adhesions with concomitant perturbation of critical signaling cascades, energy metabolism, translation, and apoptosis. We further demonstrated the ability of the Apoer2-ICD to rescue many of these altered transcripts, underscoring the importance of Apoer2 splicing in synaptic homeostasis. A variety of these altered genes have been implicated in AD, demonstrating how dysregulated Apoer2 splicing may contribute to neurodegeneration. CONCLUSIONS Our findings demonstrate how alternative splicing of the APOE and Reelin receptor Apoer2 and release of the Apoer2-ICD regulates numerous translating transcripts in mouse hippocampi in vivo. These transcripts comprise a wide range of functions, and alterations in these transcripts suggest a mechanistic basis for the synaptic deficits seen in Apoer2 mutant mice and AD patients. Our findings, together with the recently reported AD-protective effects of a Reelin gain-of-function mutation in the presence of an early-onset AD mutation in Presenilin-1, implicate the Reelin/Apoer2 pathway as a target for AD therapeutics.
Collapse
Affiliation(s)
- Catherine R Wasser
- Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-9046, USA
- Center for Translational Neurodegeneration Research, Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, USA
| | - Gordon C Werthmann
- Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-9046, USA
- Center for Translational Neurodegeneration Research, Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, USA
| | - Eric M Hall
- Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-9046, USA
- Center for Translational Neurodegeneration Research, Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, USA
| | - Kristina Kuhbandner
- Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-9046, USA
- Center for Translational Neurodegeneration Research, Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, USA
| | - Connie H Wong
- Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-9046, USA
- Center for Translational Neurodegeneration Research, Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, USA
| | - Murat S Durakoglugil
- Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-9046, USA
- Center for Translational Neurodegeneration Research, Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-9046, USA.
- Center for Translational Neurodegeneration Research, Department of Molecular Genetics, UT Southwestern, 5323 Harry Hines Blvd, Dallas, TX, USA.
- Department of Neuroscience, UT Southwestern, Dallas, TX, USA.
- Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
20
|
Ullah R, Lee EJ. Advances in Amyloid-β Clearance in the Brain and Periphery: Implications for Neurodegenerative Diseases. Exp Neurobiol 2023; 32:216-246. [PMID: 37749925 PMCID: PMC10569141 DOI: 10.5607/en23014] [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: 04/20/2023] [Revised: 07/25/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
This review examines the role of impaired amyloid-β clearance in the accumulation of amyloid-β in the brain and the periphery, which is closely associated with Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). The molecular mechanism underlying amyloid-β accumulation is largely unknown, but recent evidence suggests that impaired amyloid-β clearance plays a critical role in its accumulation. The review provides an overview of recent research and proposes strategies for efficient amyloid-β clearance in both the brain and periphery. The clearance of amyloid-β can occur through enzymatic or non-enzymatic pathways in the brain, including neuronal and glial cells, blood-brain barrier, interstitial fluid bulk flow, perivascular drainage, and cerebrospinal fluid absorption-mediated pathways. In the periphery, various mechanisms, including peripheral organs, immunomodulation/immune cells, enzymes, amyloid-β-binding proteins, and amyloid-β-binding cells, are involved in amyloid-β clearance. Although recent findings have shed light on amyloid-β clearance in both regions, opportunities remain in areas where limited data is available. Therefore, future strategies that enhance amyloid-β clearance in the brain and/or periphery, either through central or peripheral clearance approaches or in combination, are highly encouraged. These strategies will provide new insight into the disease pathogenesis at the molecular level and explore new targets for inhibiting amyloid-β deposition, which is central to the pathogenesis of sporadic AD (amyloid-β in parenchyma) and CAA (amyloid-β in blood vessels).
Collapse
Affiliation(s)
- Rahat Ullah
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Neurology, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
21
|
Cao Y, Wang H, Jin P, Ma F, Zhou X. Identification and Characterization of the Very-Low-Density Lipoprotein Receptor Gene from Branchiostoma belcheri: Insights into the Origin and Evolution of the Low-Density Lipoprotein Receptor Gene Family. Animals (Basel) 2023; 13:2193. [PMID: 37443991 DOI: 10.3390/ani13132193] [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: 03/11/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Low-density lipoprotein receptors (LDLRs) are a class of cell-surface endocytosis receptors that are mainly involved in cholesterol homeostasis and cellular signal transduction. Very-low-density lipoprotein receptors (VLDLRs), which are members of the LDLR family, have been regarded as multi-function receptors that fulfill diverse physiological functions. However, no VLDLR gene has been identified in protochordates to date. As a representative protochordate species, amphioxi are the best available example of vertebrate ancestors. Identifying and characterizing the VLDLR gene in amphioxi has high importance for exploring the evolutionary process of the LDLR family. With this study, a new amphioxus VLDLR gene (designated AmphiVLDLR) was cloned and characterized using RACE-PCR. The 3217 nt transcript of the AmphiVLDLR had a 2577 nt ORF, and the deduced 858 amino acids were highly conserved within vertebrate VLDLRs according to their primary structure and three-dimensional structure, both of which contained five characteristic domains. In contrast to other vertebrate VLDLRs, which had a conserved genomic structure organization with 19 exons and 18 introns, the AmphiVLDLR had 13 exons and 12 introns. The results of a selective pressure analysis showed that the AmphiVLDLR had numerous positive selection sites. Furthermore, the tissue expression of AmphiVLDLR using RT-qPCR showed that AmphiVLDLR RNA expression levels were highest in the gills and muscles, moderate in the hepatic cecum and gonads, and lowest in the intestines. The results of the evolutionary analysis demonstrated that the AmphiVLDLR gene is a new member of the VLDLR family whose family members have experienced duplications and deletions over the evolutionary process. These results imply that the functions of LDLR family members have also undergone differentiation. In summary, we found a new VLDLR gene homolog (AmphiVLDLR) in amphioxi. Our results provide insight into the function and evolution of the LDLR gene family.
Collapse
Affiliation(s)
- Yunpeng Cao
- School of Chemistry and Biological Engineering, Nanjing Normal University Taizhou College, Taizhou 225300, China
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Haili Wang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Xue Zhou
- School of Chemistry and Biological Engineering, Nanjing Normal University Taizhou College, Taizhou 225300, China
| |
Collapse
|
22
|
Wasser C, Werthmann GC, Hall EM, Kuhbandner K, Wong CH, Durakoglugil MS, Herz J. Apoer2-ICD-dependent regulation of hippocampal ribosome mRNA loading. RESEARCH SQUARE 2023:rs.3.rs-3040567. [PMID: 37461529 PMCID: PMC10350194 DOI: 10.21203/rs.3.rs-3040567/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Background ApoE4, the most significant genetic risk factor for late-onset Alzheimer's disease (AD), sequesters a pro-synaptogenic Reelin receptor, Apoer2, in the endosomal compartment and prevents its normal recycling. In the adult brain, Reelin potentiates excitatory synapses and thereby protects against amyloid-β toxicity. Recently, a gain-of-function mutation in Reelin that is protective against early-onset AD has been described. Alternative splicing of the Apoer2 intracellular domain (Apoer2-ICD) regulates Apoer2 signaling. Splicing of juxtamembraneous exon 16 alters the g-secretase mediated release of the Apoer2-ICD as well as synapse number and LTP, and inclusion of exon 19 ameliorates behavioral deficits in an AD mouse model. The Apoer2-ICD has also been shown to alter transcription of synaptic genes. However, the role of Apoer2 splicing for transcriptional regulation and its role in AD pathogenesis is unknown. Methods To assess in vivo mRNA-primed ribosomes specifically in hippocampi transduced with Apoer2-ICD splice variants, we crossed wild-type, cKO, and Apoer2 cleavage-resistant mice to a Cre-inducible translating ribosome affinity purification (TRAP) model. This allowed us to perform RNA-Seq on ribosome-loaded mRNA harvested specifically from hippocampal cells transduced with Apoer2-ICDs. Results Across all conditions, we observed ~ 4,700 altered ribosome-associated transcripts, several of which comprise key synaptic components such as extracellular matrix and focal adhesions with concomitant perturbation of critical signaling cascades, energy metabolism, translation, and apoptosis. We further demonstrated the ability of the Apoer2-ICD to rescue many of these altered transcripts, underscoring the importance of Apoer2 splicing in synaptic homeostasis. A variety of these altered genes have been implicated in AD, demonstrating how dysregulated Apoer2 splicing may contribute to neurodegeneration. Conclusions Our findings demonstrate how alternative splicing of the APOE and Reelin receptor Apoer2 and release of the Apoer2-ICD regulates numerous ribosome-associated transcripts in mouse hippocampi in vivo . These transcripts comprise a wide range of functions, and alterations in these transcripts suggest a mechanistic basis for the synaptic deficits seen in Apoer2 mutant mice and AD patients. Our findings, together with the recently reported AD-protective effects of a Reelin gain-of-function mutation in the presence of an early-onset AD mutation in Presenilin-1, implicate the Reelin/Apoer2 pathway as a target for AD therapeutics.
Collapse
Affiliation(s)
- Catherine Wasser
- UT Southwestern: The University of Texas Southwestern Medical Center
| | | | - Eric M Hall
- UT Southwestern: The University of Texas Southwestern Medical Center
| | | | - Connie H Wong
- UT Southwestern: The University of Texas Southwestern Medical Center
| | | | - Joachim Herz
- UT Southwestern: The University of Texas Southwestern Medical Center
| |
Collapse
|
23
|
Alexander A, Herz J, Calvier L. Reelin through the years: From brain development to inflammation. Cell Rep 2023; 42:112669. [PMID: 37339050 PMCID: PMC10592530 DOI: 10.1016/j.celrep.2023.112669] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/09/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Reelin was originally identified as a regulator of neuronal migration and synaptic function, but its non-neuronal functions have received far less attention. Reelin participates in organ development and physiological functions in various tissues, but it is also dysregulated in some diseases. In the cardiovascular system, Reelin is abundant in the blood, where it contributes to platelet adhesion and coagulation, as well as vascular adhesion and permeability of leukocytes. It is a pro-inflammatory and pro-thrombotic factor with important implications for autoinflammatory and autoimmune diseases such as multiple sclerosis, Alzheimer's disease, arthritis, atherosclerosis, or cancer. Mechanistically, Reelin is a large secreted glycoprotein that binds to several membrane receptors, including ApoER2, VLDLR, integrins, and ephrins. Reelin signaling depends on the cell type but mostly involves phosphorylation of NF-κB, PI3K, AKT, or JAK/STAT. This review focuses on non-neuronal functions and the therapeutic potential of Reelin, while highlighting secretion, signaling, and functional similarities between cell types.
Collapse
Affiliation(s)
- Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
24
|
Faissner A. Low-density lipoprotein receptor-related protein-1 (LRP1) in the glial lineage modulates neuronal excitability. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1190240. [PMID: 37383546 PMCID: PMC10293750 DOI: 10.3389/fnetp.2023.1190240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023]
Abstract
The low-density lipoprotein related protein receptor 1 (LRP1), also known as CD91 or α-Macroglobulin-receptor, is a transmembrane receptor that interacts with more than 40 known ligands. It plays an important biological role as receptor of morphogens, extracellular matrix molecules, cytokines, proteases, protease inhibitors and pathogens. In the CNS, it has primarily been studied as a receptor and clearance agent of pathogenic factors such as Aβ-peptide and, lately, Tau protein that is relevant for tissue homeostasis and protection against neurodegenerative processes. Recently, it was found that LRP1 expresses the Lewis-X (Lex) carbohydrate motif and is expressed in the neural stem cell compartment. The removal of Lrp1 from the cortical radial glia compartment generates a strong phenotype with severe motor deficits, seizures and a reduced life span. The present review discusses approaches that have been taken to address the neurodevelopmental significance of LRP1 by creating novel, lineage-specific constitutive or conditional knockout mouse lines. Deficits in the stem cell compartment may be at the root of severe CNS pathologies.
Collapse
|
25
|
Zhang L, Xia Y, Gui Y. Neuronal ApoE4 in Alzheimer's disease and potential therapeutic targets. Front Aging Neurosci 2023; 15:1199434. [PMID: 37333457 PMCID: PMC10272394 DOI: 10.3389/fnagi.2023.1199434] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
The most prevalent genetic risk factor for Alzheimer's disease (AD) is Apolipoprotein E (ApoE), a gene located on chromosome 19 that encodes three alleles (e2, e3, and e4) that give rise to the ApoE subtypes E2, E3, and E4, respectively. E2 and E4 have been linked to increased plasma triglyceride concentrations and are known to play a critical role in lipoprotein metabolism. The prominent pathological features of AD mainly include senile plaques formed by amyloid β (Aβ42) aggregation and neuronal fibrous tangles (NFTs), and the deposited plaques are mainly composed of Aβ hyperphosphorylation and truncated head. In the central nervous system, the ApoE protein is primarily derived from astrocytes, but ApoE is also produced when neurons are stressed or affected by certain stress, injury, and aging conditions. ApoE4 in neurons induces Aβ and tau protein pathologies, leading to neuroinflammation and neuronal damage, impairing learning and memory functions. However, how neuronal ApoE4 mediates AD pathology remains unclear. Recent studies have shown that neuronal ApoE4 may lead to greater neurotoxicity, which increases the risk of AD development. This review focuses on the pathophysiology of neuronal ApoE4 and explains how neuronal ApoE4 mediates Aβ deposition, pathological mechanisms of tau protein hyperphosphorylation, and potential therapeutic targets.
Collapse
|
26
|
Joseph BB, Naslavsky N, Binti S, Conquest S, Robison L, Bai G, Homer RO, Grant BD, Caplan S, Fay DS. Conserved NIMA kinases regulate multiple steps of endocytic trafficking. PLoS Genet 2023; 19:e1010741. [PMID: 37099601 PMCID: PMC10166553 DOI: 10.1371/journal.pgen.1010741] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/08/2023] [Accepted: 04/11/2023] [Indexed: 04/27/2023] Open
Abstract
Human NIMA-related kinases have primarily been studied for their roles in cell cycle progression (NEK1/2/6/7/9), checkpoint-DNA-damage control (NEK1/2/4/5/10/11), and ciliogenesis (NEK1/4/8). We previously showed that Caenorhabditis elegans NEKL-2 (NEK8/9 homolog) and NEKL-3 (NEK6/7 homolog) regulate apical clathrin-mediated endocytosis (CME) in the worm epidermis and are essential for molting. Here we show that NEKL-2 and NEKL-3 also have distinct roles in controlling endosome function and morphology. Specifically, loss of NEKL-2 led to enlarged early endosomes with long tubular extensions but showed minimal effects on other compartments. In contrast, NEKL-3 depletion caused pronounced defects in early, late, and recycling endosomes. Consistently, NEKL-2 was strongly localized to early endosomes, whereas NEKL-3 was localized to multiple endosomal compartments. Loss of NEKLs also led to variable defects in the recycling of two resident cargoes of the trans-Golgi network (TGN), MIG-14/Wntless and TGN-38/TGN38, which were missorted to lysosomes after NEKL depletion. In addition, defects were observed in the uptake of clathrin-dependent (SMA-6/Type I BMP receptor) and independent cargoes (DAF-4/Type II BMP receptor) from the basolateral surface of epidermal cells after NEKL-2 or NEKL-3 depletion. Complementary studies in human cell lines further showed that siRNA knockdown of the NEKL-3 orthologs NEK6 and NEK7 led to missorting of the mannose 6-phosphate receptor from endosomes. Moreover, in multiple human cell types, depletion of NEK6 or NEK7 disrupted both early and recycling endosomal compartments, including the presence of excess tubulation within recycling endosomes, a defect also observed after NEKL-3 depletion in worms. Thus, NIMA family kinases carry out multiple functions during endocytosis in both worms and humans, consistent with our previous observation that human NEKL-3 orthologs can rescue molting and trafficking defects in C. elegans nekl-3 mutants. Our findings suggest that trafficking defects could underlie some of the proposed roles for NEK kinases in human disease.
Collapse
Affiliation(s)
- Braveen B. Joseph
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Naava Naslavsky
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shaonil Binti
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Sylvia Conquest
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Lexi Robison
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Ge Bai
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
| | - Rafael O. Homer
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Barth D. Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, United States of America
| | - Steve Caplan
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - David S. Fay
- Department of Molecular Biology, College of Agriculture Life Sciences, and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| |
Collapse
|
27
|
Cao R, Chen P, Wang H, Jing H, Zhang H, Xing G, Luo B, Pan J, Yu Z, Xiong WC, Mei L. Intrafusal-fiber LRP4 for muscle spindle formation and maintenance in adult and aged animals. Nat Commun 2023; 14:744. [PMID: 36765071 PMCID: PMC9918736 DOI: 10.1038/s41467-023-36454-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Proprioception is sensed by muscle spindles for precise locomotion and body posture. Unlike the neuromuscular junction (NMJ) for muscle contraction which has been well studied, mechanisms of spindle formation are not well understood. Here we show that sensory nerve terminals are disrupted by the mutation of Lrp4, a gene required for NMJ formation; inducible knockout of Lrp4 in adult mice impairs sensory synapses and movement coordination, suggesting that LRP4 is required for spindle formation and maintenance. LRP4 is critical to the expression of Egr3 during development; in adult mice, it interacts in trans with APP and APLP2 on sensory terminals. Finally, spindle sensory endings and function are impaired in aged mice, deficits that could be diminished by LRP4 expression. These observations uncovered LRP4 as an unexpected regulator of muscle spindle formation and maintenance in adult and aged animals and shed light on potential pathological mechanisms of abnormal muscle proprioception.
Collapse
Affiliation(s)
- Rangjuan Cao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peng Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongyang Jing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongsheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Guanglin Xing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Bin Luo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jinxiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Zheng Yu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
28
|
Dembélé J, Liao JH, Liu TP, Chen YP. Overcoming Cytosolic Delivery Barriers of Proteins Using Denatured Protein-Conjugated Mesoporous Silica Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2023; 15:432-451. [PMID: 36562665 PMCID: PMC9896485 DOI: 10.1021/acsami.2c17544] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Intracellular delivery of therapeutic proteins has increased advantages over current small-molecule drugs and gene therapies, especially in therapeutic efficacies for a broad spectrum of diseases. Hence, developing the protein therapeutics approach provides a needed alternative. Here, we designed a mesoporous silica nanoparticle (MSN)-mediated protein delivery approach and demonstrated effective intracellular delivery of the denatured superoxide dismutase (SOD) protein, overcoming the delivery challenges and achieving higher enzymatic activity than native SOD-conjugated MSNs. The denatured SOD-conjugated MSN delivery strategy provides benefits of reduced size and steric hindrance, increased protein flexibility without distorting its secondary structure, exposure of the cell-penetrating peptide transactivator of transcription for enhanced efficient delivery, and a change in the corona protein composition, enabling cytosolic delivery. After delivery, SOD displayed a specific activity around threefold higher than in our previous reports. Furthermore, the in vivo biosafety and therapeutic potential for neuron therapy were evaluated, demonstrating the biocompatibility and the effective antioxidant effect in Neuro-2a cells that protected neurite outgrowth from paraquat-induced reactive oxygen species attack. This study offers an opportunity to realize the druggable possibility of cytosolic proteins using MSNs.
Collapse
Affiliation(s)
- Julien Dembélé
- Graduate
Institute of Biomedical Materials & Tissue Engineering, College
of Biomedical Engineering, Taipei Medical
University, Taipei 11031, Taiwan
- Laboratory
of Toxicology, Environment and Health, Doctorate School of Health, University Joseph Ki-Zerbo, Ouaga 03 BP 7021, Burkina Faso
| | - Jou-Hsuan Liao
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Tsang-Pai Liu
- Department
of Surgery, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Yi-Ping Chen
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International
PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
29
|
Geng S, Paul F, Kowalczyk I, Raimundo S, Sporbert A, Mamo TM, Hammes A. Balancing WNT signalling in early forebrain development: The role of LRP4 as a modulator of LRP6 function. Front Cell Dev Biol 2023; 11:1173688. [PMID: 37091972 PMCID: PMC10119419 DOI: 10.3389/fcell.2023.1173688] [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: 02/24/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The specification of the forebrain relies on the precise regulation of WNT/ß-catenin signalling to support neuronal progenitor cell expansion, patterning, and morphogenesis. Imbalances in WNT signalling activity in the early neuroepithelium lead to congenital disorders, such as neural tube defects (NTDs). LDL receptor-related protein (LRP) family members, including the well-studied receptors LRP5 and LRP6, play critical roles in modulating WNT signalling capacity through tightly regulated interactions with their co-receptor Frizzled, WNT ligands, inhibitors and intracellular WNT pathway components. However, little is known about the function of LRP4 as a potential modulator of WNT signalling in the central nervous system. In this study, we investigated the role of LRP4 in the regulation of WNT signalling during early mouse forebrain development. Our results demonstrate that LRP4 can modulate LRP5- and LRP6-mediated WNT signalling in the developing forebrain prior to the onset of neurogenesis at embryonic stage 9.5 and is therefore essential for accurate neural tube morphogenesis. Specifically, LRP4 functions as a genetic modifier for impaired mitotic activity and forebrain hypoplasia, but not for NTDs in LRP6-deficient mutants. In vivo and in vitro data provide evidence that LRP4 is a key player in fine-tuning WNT signalling capacity and mitotic activity of mouse neuronal progenitors and of human retinal pigment epithelial (hTERT RPE-1) cells. Our data demonstrate the crucial roles of LRP4 and LRP6 in regulating WNT signalling and forebrain development and highlight the need to consider the interaction between different signalling pathways to understand the underlying mechanisms of disease. The findings have significant implications for our mechanistic understanding of how LRPs participate in controlling WNT signalling.
Collapse
Affiliation(s)
- Shuang Geng
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Fabian Paul
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Izabela Kowalczyk
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Sandra Raimundo
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tamrat Meshka Mamo
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| | - Annette Hammes
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| |
Collapse
|
30
|
Wang H, Wan HT, Wu B, Jian J, Ng AHM, Chung CYL, Chow EYC, Zhang J, Wong AOL, Lai KP, Chan TF, Zhang EL, Wong CKC. A Chromosome-level assembly of the Japanese eel genome, insights into gene duplication and chromosomal reorganization. Gigascience 2022; 11:giac120. [PMID: 36480030 PMCID: PMC9730501 DOI: 10.1093/gigascience/giac120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
Japanese eels (Anguilla japonica) are commercially important species, harvested extensively for food. Currently, this and related species (American and European eels) are challenging to breed on a commercial basis. As a result, the wild stock is used for aquaculture. Moreover, climate change, habitat loss, water pollution, and altered ocean currents affect eel populations negatively. Accordingly, the International Union for Conservation of Nature lists Japanese eels as endangered and on its red list. Here we presented a high-quality genome assembly for Japanese eels and demonstrated that large chromosome reorganizations occurred in the events of third-round whole-genome duplications (3R-WRDs). Several chromosomal fusions and fissions have reduced the ancestral protochromosomal number of 25 to 19 in the Anguilla lineage. A phylogenetic analysis of the expanded gene families showed that the olfactory receptors (group δ and ζ genes) and voltage-gated Ca2+ channels expanded significantly. Both gene families are crucial for olfaction and neurophysiology. Additional tandem and proximal duplications occurred following 3R-WGD to acquire immune-related genes for an adaptive advantage against various pathogens. The Japanese eel assembly presented here can be used to study other Anguilla species relating to evolution and conservation.
Collapse
Affiliation(s)
- Hongbo Wang
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), China
- Department of Computer Science, Hong Kong Baptist University, Hong Kong SAR
| | - Hin Ting Wan
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR
| | - Bin Wu
- BGI Genomics, BGI-Shenzhen, Shenzhen 518083, China
| | - Jianbo Jian
- BGI Genomics, BGI-Shenzhen, Shenzhen 518083, China
| | - Alice H M Ng
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR
| | - Claire Yik-Lok Chung
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong SAR
| | - Eugene Yui-Ching Chow
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong SAR
| | - Jizhou Zhang
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong SAR
| | - Anderson O L Wong
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), China
- School of Biological Sciences, the University of Hong Kong, Hong Kong SAR
| | - Keng Po Lai
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), China
- Key Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Ting Fung Chan
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), China
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Hong Kong SAR
| | - Eric Lu Zhang
- Department of Computer Science, Hong Kong Baptist University, Hong Kong SAR
| | - Chris Kong-Chu Wong
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), China
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong SAR
| |
Collapse
|
31
|
Lipophorin receptors regulate mushroom body development and complex behaviors in Drosophila. BMC Biol 2022; 20:198. [PMID: 36071487 PMCID: PMC9454125 DOI: 10.1186/s12915-022-01393-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 08/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Drosophila melanogaster lipophorin receptors (LpRs), LpR1 and LpR2, are members of the LDLR family known to mediate lipid uptake in a range of organisms from Drosophila to humans. The vertebrate orthologs of LpRs, ApoER2 and VLDL-R, function as receptors of a glycoprotein involved in development of the central nervous system, Reelin, which is not present in flies. ApoER2 and VLDL-R are associated with the development and function of the hippocampus and cerebral cortex, important association areas in the mammalian brain, as well as with neurodevelopmental and neurodegenerative disorders linked to those regions. It is currently unknown whether LpRs play similar roles in the Drosophila brain. RESULTS We report that LpR-deficient flies exhibit impaired olfactory memory and sleep patterns, which seem to reflect anatomical defects found in a critical brain association area, the mushroom bodies (MB). Moreover, cultured MB neurons respond to mammalian Reelin by increasing the complexity of their neurite arborization. This effect depends on LpRs and Dab, the Drosophila ortholog of the Reelin signaling adaptor protein Dab1. In vitro, two of the long isoforms of LpRs allow the internalization of Reelin, suggesting that Drosophila LpRs interact with human Reelin to induce downstream cellular events. CONCLUSIONS These findings demonstrate that LpRs contribute to MB development and function, supporting the existence of a LpR-dependent signaling in Drosophila, and advance our understanding of the molecular factors functioning in neural systems to generate complex behaviors in this model. Our results further emphasize the importance of Drosophila as a model to investigate the alterations in specific genes contributing to neural disorders.
Collapse
|
32
|
LRP1 is a neuronal receptor for α-synuclein uptake and spread. Mol Neurodegener 2022; 17:57. [PMID: 36056345 PMCID: PMC9438229 DOI: 10.1186/s13024-022-00560-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aggregation and spread of α-synuclein (α-Syn) protein and related neuronal toxicity are the key pathological features of Parkinson's disease (PD) and Lewy body dementia (LBD). Studies have shown that pathological species of α-Syn and tau can spread in a prion-like manner between neurons, although these two proteins have distinct pathological roles and contribute to different neurodegenerative diseases. It is reported that the low-density lipoprotein receptor-related protein 1 (LRP1) regulates the spread of tau proteins; however, the molecular regulatory mechanisms of α-Syn uptake and spread, and whether it is also regulated by LRP1, remain poorly understood. METHODS We established LRP1 knockout (LRP1-KO) human induced pluripotent stem cells (iPSCs) isogenic lines using a CRISPR/Cas9 strategy and generated iPSC-derived neurons (iPSNs) to test the role of LRP1 in α-Syn uptake. We treated the iPSNs with fluorescently labeled α-Syn protein and measured the internalization of α-Syn using flow cytometry. Three forms of α-Syn species were tested: monomers, oligomers, and pre-formed fibrils (PFFs). To examine whether the lysine residues of α-Syn are involved in LRP1-mediated uptake, we capped the amines of lysines on α-Syn with sulfo-NHS acetate and then measured the internalization. We also tested whether the N-terminus of α-Syn is critical for LRP1-mediated internalization. Lastly, we investigated the role of Lrp1 in regulating α-Syn spread with a neuronal Lrp1 conditional knockout (Lrp1-nKO) mouse model. We generated adeno-associated viruses (AAVs) that allowed for distinguishing the α-Syn expression versus spread and injected them into the hippocampus of six-month-old Lrp1-nKO mice and the littermate wild type (WT) controls. The spread of α-Syn was evaluated three months after the injection. RESULTS We found that the uptake of both monomeric and oligomeric α-Syn was significantly reduced in iPSNs with LRP1-KO compared with the WT controls. The uptake of α-Syn PFFs was also inhibited in LRP1-KO iPSNs, albeit to a much lesser extent compared to α-Syn monomers and oligomers. The blocking of lysine residues on α-Syn effectively decreased the uptake of α-Syn in iPSNs and the N-terminus of α-Syn was critical for LRP1-mediated α-Syn uptake. Finally, in the Lrp1-nKO mice, the spread of α-Syn was significantly reduced compared with the WT littermates. CONCLUSIONS We identified LRP1 as a key regulator of α-Syn neuronal uptake, as well as an important mediator of α-Syn spread in the brain. This study provides new knowledge on the physiological and pathological role of LRP1 in α-Syn trafficking and pathology, offering insight for the treatment of synucleinopathies.
Collapse
|
33
|
The Influence of Clusterin Glycosylation Variability on Selected Pathophysiological Processes in the Human Body. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7657876. [PMID: 36071866 PMCID: PMC9441386 DOI: 10.1155/2022/7657876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022]
Abstract
The present review gathers together the most important information about variability in clusterin molecular structure, its profile, and the degree of glycosylation occurring in human tissues and body fluids in the context of the utility of these characteristics as potential diagnostic biomarkers of selected pathophysiological conditions. The carbohydrate part of clusterin plays a crucial role in many biological processes such as endocytosis and apoptosis. Many pathologies associated with neurodegeneration, carcinogenesis, metabolic diseases, and civilizational diseases (e.g., cardiovascular incidents and male infertility) have been described as causes of homeostasis disturbance, in which the glycan part of clusterin plays a very important role. The results of the discussed studies suggest that glycoproteomic analysis of clusterin may help differentiate the severity of hippocampal atrophy, detect the causes of infertility with an immune background, and monitor the development of cancer. Understanding the mechanism of clusterin (CLU) action and its binding epitopes may enable to indicate new therapeutic goals. The carbohydrate part of clusterin is considered necessary to maintain its proper molecular conformation, structural stability, and proper systemic and/or local biological activity. Taking into account the wide spectrum of CLU action and its participation in many processes in the human body, further studies on clusterin glycosylation variability are needed to better understand the molecular mechanisms of many pathophysiological conditions. They can also provide the opportunity to find new biomarkers and enrich the panel of diagnostic parameters for diseases that still pose a challenge for modern medicine.
Collapse
|
34
|
Laser-responsive multi-functional nanoparticles for efficient combinational chemo-photodynamic therapy against breast cancer. Colloids Surf B Biointerfaces 2022; 216:112574. [PMID: 35623257 DOI: 10.1016/j.colsurfb.2022.112574] [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: 04/05/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023]
Abstract
Herein, novel laser-responsive multi-functional nanoparticles (NPs-Lip@PTX/CyA/Ce6) were fabricated with bovine serum albumins (BSA) based nanoparticles, which simultaneously carried chemotherapeutic drug paclitaxel (PTX) and P-gp inhibitor cyclosporin A (CyA), as core and photosensitizer agent Chlorin e6 (Ce6) loaded Tf-modified liposomal bilayer as shell. NPs-Lip@PTX/CyA/Ce6 exhibited apparent core-shell structure morphology with particle size of 160.9 ± 1.7 nm and zeta potential of - 26.7 ± 0.6 mV, indicating their excellent stability in aqueous solution. Besides, NPs-Lip@PTX/CyA/Ce6 possessed laser-responsive release profiles upon laser irradiation at specific wavelength, which was favor to exert efficient combinatorial chemo-photodynamic therapy and effectively reverse the multiple drug resistance (MDR). Under laser irradiation, as expected, NPs-Lip@PTX/CyA/Ce6 demonstrated superb intracellular ROS productivity and fantastic in vitro and in vivo anti-cancer therapy effect but absent of systemic toxicity. In conclusion, the nano-drug delivery system would be prospectively applied in clinic as resultful therapeutic tactic for investing compositional chemo-photodynamic therapy synergistically.
Collapse
|
35
|
Effects of Drosophila melanogaster regular exercise and apolipoprotein B knockdown on abnormal heart rhythm induced by a high-fat diet. PLoS One 2022; 17:e0262471. [PMID: 35657779 PMCID: PMC9165823 DOI: 10.1371/journal.pone.0262471] [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: 06/29/2021] [Accepted: 12/23/2021] [Indexed: 11/19/2022] Open
Abstract
Abnormal heart rhythm is a common cardiac dysfunction in obese patients, and its pathogenesis is related to systemic lipid accumulation. The cardiomyocyte-derived apoLpp (homologous gene in Drosophila of the human apolipoprotein B) plays an important role in whole-body lipid metabolism of Drosophila under a high-fat diet (HFD). Knockdown of apoLpp derived from cardiomyocytes can reduce HFD-induced weight gain and abdominal lipid accumulation. In addition, exercise can reduce the total amount of apoLpp in circulation. However, the relationship between regular exercise, cardiomyocyte-derived apoLpp and abnormal heart rhythm is unclear. We found that an HFD increased the level of triglyceride (TG) in the whole-body, lipid accumulation and obesity in Drosophila. Moreover, the expression of apoLpp in the heart increased sharply, the heart rate and arrhythmia index increased and fibrillation occurred. Conversely, regular exercise or cardiomyocyte-derived apoLpp knockdown reduced the TG level in the whole-body of Drosophila. This significantly reduced the arrhythmia induced by obesity, including the reduction of heart rate, arrhythmia index, and fibrillation. Under HFD conditions, flies with apoLpp knockdown in the heart could resist the abnormal cardiac rhythm caused by obesity after receiving regular exercise. HFD-induced obesity and abnormal cardiac rhythm may be related to the acute increase of cardiomyocyte-derived apoLpp. Regular exercise and inhibition of cardiomyocyte-derived apoLpp can reduce the HFD-induced abnormal cardiac rhythm.
Collapse
|
36
|
Zhao J, Zhang H, Fan X, Yu X, Huai J. Lipid Dyshomeostasis and Inherited Cerebellar Ataxia. Mol Neurobiol 2022; 59:3800-3828. [PMID: 35420383 PMCID: PMC9148275 DOI: 10.1007/s12035-022-02826-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/01/2022] [Indexed: 12/04/2022]
Abstract
Cerebellar ataxia is a form of ataxia that originates from dysfunction of the cerebellum, but may involve additional neurological tissues. Its clinical symptoms are mainly characterized by the absence of voluntary muscle coordination and loss of control of movement with varying manifestations due to differences in severity, in the site of cerebellar damage and in the involvement of extracerebellar tissues. Cerebellar ataxia may be sporadic, acquired, and hereditary. Hereditary ataxia accounts for the majority of cases. Hereditary ataxia has been tentatively divided into several subtypes by scientists in the field, and nearly all of them remain incurable. This is mainly because the detailed mechanisms of these cerebellar disorders are incompletely understood. To precisely diagnose and treat these diseases, studies on their molecular mechanisms have been conducted extensively in the past. Accumulating evidence has demonstrated that some common pathogenic mechanisms exist within each subtype of inherited ataxia. However, no reports have indicated whether there is a common mechanism among the different subtypes of inherited cerebellar ataxia. In this review, we summarize the available references and databases on neurological disorders characterized by cerebellar ataxia and show that a subset of genes involved in lipid homeostasis form a new group that may cause ataxic disorders through a common mechanism. This common signaling pathway can provide a valuable reference for future diagnosis and treatment of ataxic disorders.
Collapse
Affiliation(s)
- Jin Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Huan Zhang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xueyu Fan
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xue Yu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jisen Huai
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
37
|
Abe RJ, Abe JI, Nguyen MTH, Olmsted-Davis EA, Mamun A, Banerjee P, Cooke JP, Fang L, Pownall H, Le NT. Free Cholesterol Bioavailability and Atherosclerosis. Curr Atheroscler Rep 2022; 24:323-336. [PMID: 35332444 PMCID: PMC9050774 DOI: 10.1007/s11883-022-01011-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW As both a cholesterol acceptor and carrier in the reverse cholesterol transport (RCT) pathway, high-density lipoprotein (HDL) is putatively atheroprotective. However, current pharmacological therapies to increase plasma HDL cholesterol (HDL-c) concentration have paradoxically failed to prevent or reduce atherosclerosis and cardiovascular disease (CVD). Given that free cholesterol (FC) transfer between surfaces of lipoproteins and cells is reversible, excess plasma FC can be transferred to the cells of peripheral tissue sites resulting in atherosclerosis. Here, we summarize potential mechanisms contributing to this paradox and highlight the role of excess free cholesterol (FC) bioavailability in atherosclerosis vs. atheroprotection. RECENT FINDINGS Recent findings have established a complex relationship between HDL-c concentration and atherosclerosis. Systemic scavenger receptor class B type 1 (SR-B1) knock out (KO) mice exhibit with increased diet-induced atherosclerosis despite having an elevated plasma HDL-c concentration compared to wild type (WT) mice. The greater bioavailability of HDL-FC in SR-B1 vs. WT mice is associated with a higher FC content in multiple cell types and tissue sites. These results suggest that dysfunctional HDL with high FC bioavailability is atheroprone despite high HDL-c concentration. Past oversimplification of HDL-c involvement in cholesterol transport has led to the failures in HDL targeted therapy. Evidence suggests that FC-mediated functionality of HDL is of higher importance than its quantity; as a result, deciphering the regulatory mechanisms by which HDL-FC bioavailability can induce atherosclerosis can have far-reaching clinical implications.
Collapse
Affiliation(s)
- Rei J Abe
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Minh T H Nguyen
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | | | - Abrar Mamun
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Priyanka Banerjee
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - John P Cooke
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Longhou Fang
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Henry Pownall
- Weill Cornell Medicine, New York, NY, USA
- Center for Bioenergetics, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Nhat-Tu Le
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
- Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
38
|
Huang K, Liu Y, Perrimon N. Roles of Insect Oenocytes in Physiology and Their Relevance to Human Metabolic Diseases. FRONTIERS IN INSECT SCIENCE 2022; 2:859847. [PMID: 38468774 PMCID: PMC10926422 DOI: 10.3389/finsc.2022.859847] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/14/2022] [Indexed: 03/13/2024]
Abstract
Oenocytes are large secretory cells present in the abdomen of insects known to synthesize very-long-chain fatty acids to produce hydrocarbons and pheromones that mediate courtship behavior in adult flies. In recent years, oenocytes have been implicated in the regulation of energy metabolism. These hepatocyte-like cells accumulate lipid droplets under starvation and can non-autonomously regulate tracheal waterproofing and adipocyte lipid composition. Here, we summarize evidence, mostly from Drosophila, establishing that oenocytes perform liver-like functions. We also compare the functional differences in oenocytes and the fat body, another lipid storage tissue which also performs liver-like functions. Lastly, we examine signaling pathways that regulate oenocyte metabolism derived from other metabolic tissues, as well as oenocyte-derived signals that regulate energy homeostasis.
Collapse
Affiliation(s)
- Kerui Huang
- Department of Genetics, Harvard Medical School, Blavatnik Institute, Boston, MA, United States
| | - Ying Liu
- Department of Genetics, Harvard Medical School, Blavatnik Institute, Boston, MA, United States
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Blavatnik Institute, Boston, MA, United States
- Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, United States
| |
Collapse
|
39
|
Martens YA, Zhao N, Liu CC, Kanekiyo T, Yang AJ, Goate AM, Holtzman DM, Bu G. ApoE Cascade Hypothesis in the pathogenesis of Alzheimer's disease and related dementias. Neuron 2022; 110:1304-1317. [PMID: 35298921 PMCID: PMC9035117 DOI: 10.1016/j.neuron.2022.03.004] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/08/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022]
Abstract
The ε4 allele of the apolipoprotein E gene (APOE4) is a strong genetic risk factor for Alzheimer's disease (AD) and several other neurodegenerative conditions, including Lewy body dementia (LBD). The three APOE alleles encode protein isoforms that differ from one another only at amino acid positions 112 and 158: apoE2 (C112, C158), apoE3 (C112, R158), and apoE4 (R112, R158). Despite progress, it remains unclear how these small amino acid differences in apoE sequence among the three isoforms lead to profound effects on aging and disease-related pathways. Here, we propose a novel "ApoE Cascade Hypothesis" in AD and age-related cognitive decline, which states that the biochemical and biophysical properties of apoE impact a cascade of events at the cellular and systems levels, ultimately impacting aging-related pathogenic conditions including AD. As such, apoE-targeted therapeutic interventions are predicted to be more effective by addressing the biochemical phase of the cascade.
Collapse
Affiliation(s)
- Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Austin J Yang
- Division of Neuroscience, National Institute on Aging, Bethesda, MD, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
40
|
Bai Y, Dong X, Li M, Hei N, Ge C, Li M, Yin P, Zhang L. Pathological Characteristics of Malformations of Cortical Development in Golden Syrian Hamsters with LDLR Deficiency. J Neuropathol Exp Neurol 2021; 80:1143-1146. [PMID: 34718658 DOI: 10.1093/jnen/nlab103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yun Bai
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China.,Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei, China
| | - Xiaohui Dong
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Mingzhao Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Naiheng Hei
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei, China.,The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chen Ge
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China.,Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei, China
| | - Mei Li
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pengfei Yin
- Hebei INVIVO Biotechnology Co., Ltd., Shijiazhuang, Hebei, China
| | - Lianshan Zhang
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China.,Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei, China
| |
Collapse
|
41
|
Bian W, Tang M, Jiang H, Xu W, Hao W, Sui Y, Hou Y, Nie L, Zhang H, Wang C, Li N, Wang J, Qin J, Wu L, Ma X, Chen J, Wang W, Li X. Low-density-lipoprotein-receptor-related protein 1 mediates Notch pathway activation. Dev Cell 2021; 56:2902-2919.e8. [PMID: 34626540 DOI: 10.1016/j.devcel.2021.09.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 08/18/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022]
Abstract
The Notch signaling pathway controls cell growth, differentiation, and fate decisions, and its dysregulation has been linked to various human genetic disorders and cancers. To comprehensively understand the global organization of the Notch pathway and identify potential drug targets for Notch-related diseases, we established a protein interaction landscape for the human Notch pathway. By combining and analyzing genetic and phenotypic data with bioinformatics analysis, we greatly expanded this pathway and identified many key regulators, including low-density-lipoprotein-receptor-related protein 1 (LRP1). We demonstrated that LRP1 mediates the ubiquitination chain linkage switching of Delta ligands, which further affects ligand recycling, membrane localization, and stability. LRP1 inhibition led to Notch signaling inhibition and decreased tumorigenesis in leukemia models. Our study provides a glimpse into the Notch pathway interaction network and uncovers LRP1 as one critical regulator of the Notch pathway, as well as a possible therapeutic target for Notch-related cancers.
Collapse
Affiliation(s)
- Weixiang Bian
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Mengfan Tang
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Hua Jiang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Wenyan Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Wanyu Hao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Science, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Yue Sui
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yingnan Hou
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Litong Nie
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Huimin Zhang
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Chao Wang
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Nan Li
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiadong Wang
- Institute of Systems Biomedicine, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, People's Republic of China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Lianfeng Wu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Science, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| | - Junjie Chen
- Department of Experimental Radiation Oncology, the University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA.
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA.
| | - Xu Li
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China.
| |
Collapse
|
42
|
Van Valkenburgh J, Meuret C, Martinez AE, Kodancha V, Solomon V, Chen K, Yassine HN. Understanding the Exchange of Systemic HDL Particles Into the Brain and Vascular Cells Has Diagnostic and Therapeutic Implications for Neurodegenerative Diseases. Front Physiol 2021; 12:700847. [PMID: 34552500 PMCID: PMC8450374 DOI: 10.3389/fphys.2021.700847] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
High-density lipoproteins (HDLs) are complex, heterogenous lipoprotein particles, consisting of a large family of apolipoproteins, formed in subspecies of distinct shapes, sizes, and functions and are synthesized in both the brain and the periphery. HDL apolipoproteins are important determinants of Alzheimer’s disease (AD) pathology and vascular dementia, having both central and peripheral effects on brain amyloid-beta (Aβ) accumulation and vascular functions, however, the extent to which HDL particles (HLD-P) can exchange their protein and lipid components between the central nervous system (CNS) and the systemic circulation remains unclear. In this review, we delineate how HDL’s structure and composition enable exchange between the brain, cerebrospinal fluid (CSF) compartment, and vascular cells that ultimately affect brain amyloid metabolism and atherosclerosis. Accordingly, we then elucidate how modifications of HDL-P have diagnostic and therapeutic potential for brain vascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Juno Van Valkenburgh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Cristiana Meuret
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ashley E Martinez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Vibha Kodancha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Victoria Solomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
43
|
Kordes C, Bock HH, Reichert D, May P, Häussinger D. Hepatic stellate cells: current state and open questions. Biol Chem 2021; 402:1021-1032. [PMID: 34008380 DOI: 10.1515/hsz-2021-0180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/03/2021] [Indexed: 01/14/2023]
Abstract
This review article summarizes 20 years of our research on hepatic stellate cells within the framework of two collaborative research centers CRC575 and CRC974 at the Heinrich Heine University. Over this period, stellate cells were identified for the first time as mesenchymal stem cells of the liver, and important functions of these cells in the context of liver regeneration were discovered. Furthermore, it was determined that the space of Disse - bounded by the sinusoidal endothelium and hepatocytes - functions as a stem cell niche for stellate cells. Essential elements of this niche that control the maintenance of hepatic stellate cells have been identified alongside their impairment with age. This article aims to highlight previous studies on stellate cells and critically examine and identify open questions and future research directions.
Collapse
Affiliation(s)
- Claus Kordes
- Clinic of Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Hans H Bock
- Clinic of Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Doreen Reichert
- Clinic of Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Petra May
- Clinic of Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Dieter Häussinger
- Clinic of Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University, Moorenstraße 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
44
|
Wu Y, Johnson G, Zhao F, Wu Y, Zhao G, Brown A, You S, Zou MH, Song P. Features of Lipid Metabolism in Humanized ApoE Knockin Rat Models. Int J Mol Sci 2021; 22:ijms22158262. [PMID: 34361033 PMCID: PMC8347964 DOI: 10.3390/ijms22158262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022] Open
Abstract
Apolipoprotein E (ApoE), an essential plasma apolipoprotein, has three isoforms (E2, E3, and E4) in humans. E2 is associated with type III hyperlipoproteinemia. E4 is the major susceptibility gene to Alzheimer’s disease (AD) and coronary heart disease (CHD). We investigated lipid metabolism and atherosclerotic lesions of novel humanized ApoE knockin (hApoE KI) rats in comparison to wide-type (WT) and ApoE knockout (ApoE KO) rats. The hApoE2 rats showed the lowest bodyweight and white fat mass. hApoE2 rats developed higher serum total cholesterol (TC), total triglyceride (TG), and low- and very low density lipoprotein (LDL-C&VLDL-C). ApoE KO rats also exhibited elevated TC and LDL-C&VLDL-C. Only mild atherosclerotic lesions were detected in hApoE2 and ApoE KO aortic roots. Half of the hApoE2 rats developed hepatic nodular cirrhosis. A short period of the Paigen diet (PD) treatment led to the premature death of the hApoE2 and ApoE KO rats. Severe vascular wall thickening of the coronary and pulmonary arteries was observed in 4-month PD-treated hApoE4 rats. In conclusion, hApoE2 rats develop spontaneous hyperlipidemia and might be suitable for studies of lipid metabolism-related diseases. With the PD challenge, hApoE4 KI rats could be a novel model for the analysis of vascular remodeling.
Collapse
Affiliation(s)
- Yang Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA; (Y.W.); (G.J.); (F.Z.); (Y.W.); (S.Y.); (M.-H.Z.)
| | - Gem Johnson
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA; (Y.W.); (G.J.); (F.Z.); (Y.W.); (S.Y.); (M.-H.Z.)
| | - Fujie Zhao
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA; (Y.W.); (G.J.); (F.Z.); (Y.W.); (S.Y.); (M.-H.Z.)
| | - Yin Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA; (Y.W.); (G.J.); (F.Z.); (Y.W.); (S.Y.); (M.-H.Z.)
| | - Guojun Zhao
- Envigo RMS, Inc., St. Louis, MO 63146, USA; (G.Z.); (A.B.)
| | - Andrew Brown
- Envigo RMS, Inc., St. Louis, MO 63146, USA; (G.Z.); (A.B.)
| | - Shaojin You
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA; (Y.W.); (G.J.); (F.Z.); (Y.W.); (S.Y.); (M.-H.Z.)
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA; (Y.W.); (G.J.); (F.Z.); (Y.W.); (S.Y.); (M.-H.Z.)
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA; (Y.W.); (G.J.); (F.Z.); (Y.W.); (S.Y.); (M.-H.Z.)
- Correspondence: ; Tel.: +1-404-413-6636
| |
Collapse
|
45
|
Esteves P, Blanc L, Celle A, Dupin I, Maurat E, Amoedo N, Cardouat G, Ousova O, Gales L, Bellvert F, Begueret H, Thumerel M, Dupuy JW, Desbenoit N, Marthan R, Girodet PO, Rossignol R, Berger P, Trian T. Crucial role of fatty acid oxidation in asthmatic bronchial smooth muscle remodelling. Eur Respir J 2021; 58:13993003.04252-2020. [PMID: 33833033 DOI: 10.1183/13993003.04252-2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/26/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Bronchial smooth muscle (BSM) remodelling in asthma is related to an increased mitochondrial biogenesis and enhanced BSM cell proliferation in asthma. Since (i) mitochondria produce the highest levels of cellular energy and (ii) fatty acid beta-oxidation is the most powerful way to produce ATP, we hypothesized that, in asthmatic BSM cells, energetic metabolism is shifted towards the beta-oxidation of fatty acids. OBJECTIVES We aimed to characterize BSM cell metabolism in asthma both in vitro and ex vivo to identify a novel target for reducing BSM cell proliferation. METHODS Twenty-one asthmatic and 31 non-asthmatic patients were enrolled. We used metabolomic and proteomic approaches to study BSM cells. Oxidative stress, ATP synthesis, fatty acid endocytosis, metabolite production, metabolic capabilities, mitochondrial networks, cell proliferation and apoptosis were assessed on BSM cells. Fatty acid content was assessed in vivo using MALDI-spectrometry imaging. RESULTS Asthmatic BSM cells were characterized by an increased rate of mitochondrial respiration with a stimulated ATP production and mitochondrial β-oxidation. Fatty acid consumption was increased in asthmatic BSM both in vitro and ex vivo. Proteome remodelling of asthmatic BSM occurred via 2 canonical mitochondrial pathways. The levels of CPT2 and LDL-receptor, which internalize fatty acids through mitochondrial and cell membranes, respectively, were both increased in asthmatic BSM cells. Blocking CPT2 or LDL-receptor drastically and specifically reduced asthmatic BSM cell proliferation. CONCLUSION This study demonstrates a metabolic switch towards mitochondrial beta-oxidation in asthmatic BSM and identifies fatty acid metabolism as a new key target to reduce BSM remodelling in asthma.
Collapse
Affiliation(s)
- Pauline Esteves
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Landry Blanc
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,CNRS, UMR5248, Institute of Chemistry & Biology of Membranes & Nano objects, Functional Genomics Center (CGFB), Proteomics Facility, Université de Bordeaux, Bordeaux, France
| | - Alexis Celle
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Isabelle Dupin
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Elise Maurat
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Nivea Amoedo
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Guillaume Cardouat
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Olga Ousova
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Lara Gales
- CNRS 5504, INRA 792, INSA Toulouse, Toulouse Biotechnology Institute, Bio & Chemical Engineering, Université de Toulouse, MetaToul, Toulouse, France
| | - Florian Bellvert
- CNRS 5504, INRA 792, INSA Toulouse, Toulouse Biotechnology Institute, Bio & Chemical Engineering, Université de Toulouse, MetaToul, Toulouse, France
| | - Hugues Begueret
- Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France
| | - Matthieu Thumerel
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France
| | - Jean-William Dupuy
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,CNRS, UMR5248, Institute of Chemistry & Biology of Membranes & Nano objects, Functional Genomics Center (CGFB), Proteomics Facility, Université de Bordeaux, Bordeaux, France
| | - Nicolas Desbenoit
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,CNRS, UMR5248, Institute of Chemistry & Biology of Membranes & Nano objects, Functional Genomics Center (CGFB), Proteomics Facility, Université de Bordeaux, Bordeaux, France
| | - Roger Marthan
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France
| | - Pierre-Olivier Girodet
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France
| | - Rodrigue Rossignol
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Patrick Berger
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France.,Co-last author
| | - Thomas Trian
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France .,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Co-last author
| |
Collapse
|
46
|
Abstract
Cholesterol is a quantitatively and biologically significant constituent of all mammalian cell membrane, including those that comprise the retina. Retinal cholesterol homeostasis entails the interplay between de novo synthesis, uptake, intraretinal sterol transport, metabolism, and efflux. Defects in these complex processes are associated with several congenital and age-related disorders of the visual system. Herein, we provide an overview of the following topics: (a) cholesterol synthesis in the neural retina; (b) lipoprotein uptake and intraretinal sterol transport in the neural retina and the retinal pigment epithelium (RPE); (c) cholesterol efflux from the neural retina and the RPE; and (d) biology and pathobiology of defects in sterol synthesis and sterol oxidation in the neural retina and the RPE. We focus, in particular, on studies involving animal models of monogenic disorders pertinent to the above topics, as well as in vitro models using biochemical, metabolic, and omic approaches. We also identify current knowledge gaps and opportunities in the field that beg further research in this topic area.
Collapse
Affiliation(s)
- Sriganesh Ramachandra Rao
- Departments of Ophthalmology and Biochemistry and Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Steven J Fliesler
- Departments of Ophthalmology and Biochemistry and Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA.
| |
Collapse
|
47
|
LDL receptors and their role in targeted therapy for glioma: a review. Drug Discov Today 2021; 26:1212-1225. [PMID: 33609780 DOI: 10.1016/j.drudis.2021.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/15/2021] [Accepted: 02/06/2021] [Indexed: 11/22/2022]
Abstract
Gliomas are highly lethal forms of cancers occurring in the brain. Delivering the drugs into the brain is a major challenge to the treatment of gliomas because of the highly selectively permeable blood-brain barrier (BBB). Tapping the potential of receptor-mediated drug delivery systems using targeted nanoparticles (NPs) is a sought-after step forward toward successful glioma treatment. Several receptors are the focus of research for application in drug delivery. Low-density lipoprotein receptors (LDLR) are abundantly expressed in both healthy brains and diseased brains with a disrupted BBB. In this review, we discuss the LDLR and the types of NPs that have been used to target the brain via this receptor.
Collapse
|
48
|
Zhou X, Smith QR, Liu X. Brain penetrating peptides and peptide-drug conjugates to overcome the blood-brain barrier and target CNS diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1695. [PMID: 33470550 DOI: 10.1002/wnan.1695] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Nearly one in six people worldwide suffer from disorders of the central nervous system (CNS). There is an urgent need for effective strategies to improve the success rates in CNS drug discovery and development. The lack of effective technologies for delivering drugs and genes to the brain due to the blood-brain barrier (BBB), a structural barrier that effectively blocks most neurotherapeutic agents from reaching the brain, has posed a formidable hurdle for CNS drug development. Brain-homing and brain-penetrating molecular transport vectors, such as brain permeable peptides or BBB shuttle peptides, have shown promise in overcoming the BBB and ferrying the drug molecules to the brain. The BBB shuttle peptides are discovered by phage display technology or derived from natural neurotropic proteins or certain viruses and harness the receptor-mediated transcytosis molecular machinery for crossing the BBB. Brain permeable peptide-drug conjugates (PDCs), composed of BBB shuttle peptides, linkers, and drug molecules, have emerged as a promising CNS drug delivery system by taking advantage of the endogenous transcytosis mechanism and tricking the brain into allowing these bioactive molecules to pass the BBB. Here, we examine the latest development of brain-penetrating peptide shuttles and brain-permeable PDCs as molecular vectors to deliver small molecule drug payloads across the BBB to reach brain parenchyma. Emerging knowledge of the contribution of the peptides and their specific receptors expressed on the brain endothelial cells, choice of drug payloads, the design of PDCs, brain entry mechanisms, and delivery efficiency to the brain are highlighted. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Quentin R Smith
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| |
Collapse
|
49
|
Pan L, Tian Y, Sun H, Wang Y, Liu G. TMT-based proteomics analysis reveals the efficacy of jiangzhuo formula in improving the lipid profiles of dyslipidemia rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113390. [PMID: 32931881 DOI: 10.1016/j.jep.2020.113390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/17/2020] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiangzhuo Formula (JZF) is a traditional Chinese herbal prescription that is clinically applied to treat dyslipidemia. However, the mechanism underlying its efficacy remains unexplored. AIM OF THE STUDY This study aims to elucidate the underlying mechanisms, explore potential pathways, and identify the key proteins of JZF for the treatment of dyslipidemia. METHODS In this work, Q-Orbitrap high-resolution liquid chromatography mass spectrometry was used to identify the natural ingredients in JZF, rats with dyslipidemia were established via a high-fat diet for four weeks, then the dyslipidemia rats were treated with high-dose JZF (9 g/d) and low-dose JZF (4.5 g/d) for four weeks. After treatment, serum lipid detection and Oil-red-O staining were conducted to assess the efficacy of JZF in ameliorating dyslipidemia. Tandem mass tag (TMT) -based quantitative proteomics technology was then used to evaluate the roles and importance of proteins from the extracted hepatic tissue. The differentially expressed proteins were assessed by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, Gene Ontology (GO), and protein-protein interaction (PPI) networks. Western blot and PCR analysis were used to validate the potential targets regulated by JZF. RESULTS JZF could significantly improve the blood lipid profiles of serum and fat deposits of the liver. A total of 123 differentially expressed proteins were detected after JZF intervention, comprising 65 up-regulated proteins and 58 down-regulated proteins. The KEGG pathway analysis revealed that cholesterol metabolism, the PPAR signaling pathway, and bile secretion were the principal pathways involved in the disordered lipid metabolism, while GO analysis suggested that proteins that are located in the cell, regulate cellular processes, and show binding activity contribute to reductions in lipids. The combination of proteomics, Western blot, and PCR suggested that Apolipoprotein B (APOB), Apolipoprotein E (APOE), cholesterol 7 alpha-hydroxylase A1 (CYP7A1), and Hydroxymethylglutaryl-CoA synthase 1 (HMGCS1) might play critical roles in JZF's lipid-lowering network. CONCLUSION JZF can effectively improve lipid profiles via multiple pathways involved in cholesterol metabolism, the PPAR signaling pathway, and bile secretion. Generally, the proteomics techniques used in this research show that JZF could be a promising drug for the treatment of dyslipidemia.
Collapse
Affiliation(s)
- Linlin Pan
- Department of Chinese Medicine Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China.
| | - Yuan Tian
- Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China.
| | - Haiyang Sun
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China.
| | - Yao Wang
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China.
| | - Guirong Liu
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China.
| |
Collapse
|
50
|
Xie BS, Wang X, Pan YH, Jiang G, Feng JF, Lin Y. Apolipoprotein E, low-density lipoprotein receptor, and immune cells control blood-brain barrier penetration by AAV-PHP.eB in mice. Am J Cancer Res 2021; 11:1177-1191. [PMID: 33391529 PMCID: PMC7738887 DOI: 10.7150/thno.46992] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 10/25/2020] [Indexed: 12/27/2022] Open
Abstract
Rationale: The blood-brain barrier (BBB) prevents the effective delivery of therapeutic molecules to the central nervous system (CNS). A recently generated adeno-associated virus (AAV)-based vector, AAV-PHP.eB, has been found to penetrate the BBB more efficiently than other vectors including AAV-PHP.B. However, little is known about the mechanisms. In this study, we investigated how AAV-PHP.eB penetrates the BBB in mice. Methods: We injected AAV-PHP.eB into the bloodstream of wild-type C57BL/6 and BALB/c mice as well as mouse strains carrying genetic mutation in apolipoprotein E gene (Apoe) or low-density lipoprotein receptor gene (Ldlr), or lacking various components of the immune system. Then, we evaluated AAV-PHP.eB transduction to the brain and spinal cord in these mice. Results: We found that the transduction to the CNS of intravenous AAV-PHP.eB was more efficient in C57BL/6 than BALB/c mice, and significantly reduced in Apoe or Ldlr knockout C57BL/6 mice compared to wild-type C57BL/6 mice. Moreover, poor CNS transduction in BALB/c mice was dramatically increased by B-cell or natural killer-cell depletion. Conclusions: Our findings demonstrate that the ApoE-LDLR pathway underlies the CNS tropism of AAV-PHP.eB and that the immune system contributes to the strain specificity of AAV-PHP.eB.
Collapse
|