1
|
Yang H, Sun L, Bai X, Cai B, Tu Z, Fang C, Bian Y, Zhang X, Han X, Lv D, Zhang C, Li B, Luo S, Du B, Li L, Yao Y, Dong Z, Huang Z, Su G, Li H, Wang QK, Zhang M. Dysregulated RBM24 phosphorylation impairs APOE translation underlying psychological stress-induced cardiovascular disease. Nat Commun 2024; 15:10181. [PMID: 39580475 PMCID: PMC11585567 DOI: 10.1038/s41467-024-54519-0] [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: 04/26/2023] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
Psychological stress contributes to cardiovascular disease (CVD) and sudden cardiac death, yet its molecular basis remains obscure. RNA binding protein RBM24 plays a critical role in cardiac development, rhythm regulation, and cellular stress. Here, we show that psychological stress activates RBM24 S181 phosphorylation through eIF4E2-GSK3β signaling, which causally links psychological stress to CVD by promoting APOE translation (apolipoprotein E). Using an Rbm24 S181A KI mouse model, we show that impaired S181 phosphorylation leads to cardiac contractile dysfunction, atrial fibrillation, dyslipidemia, reduced muscle strength, behavioral abnormalities, and sudden death under acute and chronic psychological stressors. The impaired S181 phosphorylation of RBM24 inhibits cardiac translation, including APOE translation. Notably, cardiomyocyte-specific expression of APOE rescues cardiac electrophysiological abnormalities and contractile dysfunction, through preventing ROS stress and mitochondrial dysfunction. Moreover, RBM24-S181 phosphorylation acts as a serum marker for chronic stress in human. These results provide a functional link between RBM24 phosphorylation, eIF4E-regulated APOE translation, and psychological-stress-induced CVD.
Collapse
Affiliation(s)
- He Yang
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lei Sun
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xuemei Bai
- Center for Human Genome Research, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bingcheng Cai
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zepeng Tu
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chen Fang
- Center for Human Genome Research, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yusheng Bian
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaoyu Zhang
- Center for Human Genome Research, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xudong Han
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Dayin Lv
- Center for Human Genome Research, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chi Zhang
- Center for Human Genome Research, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bo Li
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | | | - Bingbing Du
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lan Li
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yufeng Yao
- Center for Human Genome Research, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhiqiang Dong
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhuowei Huang
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430010, China
| | - Guanhua Su
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Li
- Center for Human Genome Research, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China.
- School of Biotechnology of Shandong Polytechnic, Jinan, Shandong, 250101, China.
| | - Qing K Wang
- Center for Human Genome Research, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Min Zhang
- College of Biomedicine and Health, College of Life science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
2
|
Rueter J, Rimbach G, Bilke S, Tholey A, Huebbe P. Readdressing the Localization of Apolipoprotein E (APOE) in Mitochondria-Associated Endoplasmic Reticulum (ER) Membranes (MAMs): An Investigation of the Hepatic Protein-Protein Interactions of APOE with the Mitochondrial Proteins Lon Protease (LONP1), Mitochondrial Import Receptor Subunit TOM40 (TOMM40) and Voltage-Dependent Anion-Selective Channel 1 (VDAC1). Int J Mol Sci 2024; 25:10597. [PMID: 39408926 PMCID: PMC11476584 DOI: 10.3390/ijms251910597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
As a component of circulating lipoproteins, APOE binds to cell surface receptors mediating lipoprotein metabolism and cholesterol transport. A growing body of evidence, including the identification of a broad variety of cellular proteins interacting with APOE, suggests additional independent functions. Investigating cellular localization and protein-protein interactions in cultured human hepatocytes, we aimed to contribute to the elucidation of hitherto unnoted cellular functions of APOE. We observed a strong accumulation of APOE in MAMs, equally evident for the two major isoforms APOE3 and APOE4. Using mass spectrometry proteome analyses, novel and previously noted APOE interactors were identified, including the mitochondrial proteins TOMM40, LONP1 and VDAC1. All three interactors were present in MAM fractions, which we think initially facilitates interactions with APOE. LONP1 is a protease with chaperone activity, which migrated to MAMs in response to ER stress, displaying a reinforced interaction with APOE. We therefore hypothesize that APOE may help in the unfolded protein response (UPR) by acting as a co-chaperone in cooperation with LONP1 at the interface of mitochondria and ER membranes. The interaction of APOE with the integral proteins TOMM40 and VDAC1 may point to the formation of bridging complexes connecting mitochondria with other organelles.
Collapse
Affiliation(s)
- Johanna Rueter
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118 Kiel, Germany; (J.R.); (G.R.)
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118 Kiel, Germany; (J.R.); (G.R.)
| | - Stephanie Bilke
- Institute of Experimental Medicine, University of Kiel, Niemannsweg 11, 24105 Kiel, Germany
| | - Andreas Tholey
- Institute of Experimental Medicine, University of Kiel, Niemannsweg 11, 24105 Kiel, Germany
| | - Patricia Huebbe
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118 Kiel, Germany; (J.R.); (G.R.)
| |
Collapse
|
3
|
Wan S, He QY, Yang Y, Liu F, Zhang X, Guo X, Niu H, Wang Y, Liu YX, Ye WL, Li XM, ZhuanSun XM, Sun P, He XS, Hu G, Breuhahn K, Zhao H, Wu GQ, Wu H. SPARC Stabilizes ApoE to Induce Cholesterol-Dependent Invasion and Sorafenib Resistance in Hepatocellular Carcinoma. Cancer Res 2024; 84:1872-1888. [PMID: 38471084 DOI: 10.1158/0008-5472.can-23-2889] [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: 09/20/2023] [Revised: 01/11/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Dysregulation of cholesterol homeostasis is implicated in the development and progression of hepatocellular carcinoma (HCC) that is characterized by intrahepatic and early extrahepatic metastases. A better understanding of the underlying mechanisms regulating cholesterol metabolism in HCC could help identify strategies to circumvent the aggressive phenotype. Here, we found that high expression of intracellular SPARC (secreted protein acidic and rich in cysteine) was significantly associated with elevated cholesterol levels and an enhanced invasive phenotype in HCC. SPARC potentiated cholesterol accumulation in HCC cells during tumor progression by stabilizing the ApoE protein. Mechanistically, SPARC competitively bound to ApoE, impairing its interaction with the E3 ligase tripartite motif containing 21 (TRIM21) and preventing its ubiquitylation and subsequent degradation. ApoE accumulation led to cholesterol enrichment in HCC cells, stimulating PI3K-AKT signaling and inducing epithelial-mesenchymal transition (EMT). Importantly, sorafenib-resistant HCC cells were characterized by increased expression of intracellular SPARC, elevated cholesterol levels, and enhanced invasive capacity. Inhibiting SPARC expression or reducing cholesterol levels enhanced the sensitivity of HCC cells to sorafenib treatment. Together, these findings unveil interplay between SPARC and cholesterol homeostasis. Targeting SPARC-triggered cholesterol-dependent oncogenic signaling is a potential therapeutic strategy for advanced HCC. SIGNIFICANCE Intracellular SPARC boosts cholesterol availability to fuel invasion and drug resistance in hepatocellular carcinoma, providing a rational approach to improve the treatment of advanced liver cancer.
Collapse
Affiliation(s)
- Shan Wan
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Quan-Yao He
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Yun Yang
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Feng Liu
- YongDing Clinical Institute of Soochow University, Hygeia Suzhou YongDing Hospital, Suzhou, China
| | - Xue Zhang
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Xin Guo
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Hui Niu
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Yi Wang
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Yi-Xuan Liu
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Wen-Long Ye
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Xiu-Ming Li
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Xue-Mei ZhuanSun
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Pu Sun
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Xiao-Shun He
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Guang Hu
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hua Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Guo-Qiang Wu
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| | - Hua Wu
- Suzhou Medical College of Soochow University & Clinical Medicine Research Institute of Soochow University and Suzhou BenQ Medical Center, Soochow University, Suzhou, China
| |
Collapse
|
4
|
Al-Kuraishy HM, Fahad EH, Al-Windy S, El-Sherbeni SA, Negm WA, Batiha GES. The effects of cholesterol and statins on Parkinson's neuropathology: a narrative review. Inflammopharmacology 2024; 32:917-925. [PMID: 38499742 DOI: 10.1007/s10787-023-01400-z] [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: 02/13/2023] [Accepted: 11/14/2023] [Indexed: 03/20/2024]
Abstract
Parkinson disease (PD) is chronic and progressive neurodegenerative disease of the brain characterized by motor symptoms including tremors, rigidity, postural instability, and bradykinesia. PD neuropathology is due to the progressive degeneration of dopaminergic neurons in the substantia nigra and accumulation of Lewy bodies in the survival neurons. The brain contains a largest amount of cholesterol which is mainly synthesized from astrocytes and glial cells. Cholesterol is intricate in the pathogenesis of PD and may be beneficial or deleterious. Therefore, there are controversial points concerning the role of cholesterol in PD neuropathology. In addition, cholesterol-lowering agents' statins can affect brain cholesterol. Different studies highlighted that statins, via inhibition of brain HMG-CoA, can affect neuronal integrity through suppression of neuronal cholesterol, which regulates synaptic plasticity and neurotransmitter release. Furthermore, statins affect the development and progression of different neurodegenerative diseases in bidirectional ways that could be beneficial or detrimental. Therefore, the objective of the present review was to clarify the double-sward effects of cholesterol and statins on PD neuropathology.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, 14132, Iraq
| | - Esraa H Fahad
- Department of Pharmacology and Toxicology, College of Pharmacy, Mustansiriyah University, Baghdad, 14132, Iraq
| | - Salah Al-Windy
- Department of Biology, College of Science, Baghdad University, Baghdad, Iraq
| | - Suzy A El-Sherbeni
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
5
|
Huebbe P, Bilke S, Rueter J, Schloesser A, Campbel G, Glüer CC, Lucius R, Röcken C, Tholey A, Rimbach G. Human APOE4 Protects High-Fat and High-Sucrose Diet Fed Targeted Replacement Mice against Fatty Liver Disease Compared to APOE3. Aging Dis 2024; 15:259-281. [PMID: 37450924 PMCID: PMC10796091 DOI: 10.14336/ad.2023.0530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/30/2023] [Indexed: 07/18/2023] Open
Abstract
Recent genome- and exome-wide association studies suggest that the human APOE ε4 allele protects against non-alcoholic fatty liver disease (NAFLD), while ε3 promotes hepatic steatosis and steatohepatitis. The present study aimed at examining the APOE genotype-dependent development of fatty liver disease and its underlying mechanisms in a targeted replacement mouse model. Male mice expressing the human APOE3 or APOE4 protein isoforms on a C57BL/6J background and unmodified C57BL/6J mice were chronically fed a high-fat and high-sucrose diet to induce obesity. After 7 months, body weight gain was more pronounced in human APOE than endogenous APOE expressing mice with elevated plasma biomarkers suggesting aggravated metabolic dysfunction. APOE3 mice exhibited the highest liver weights and, compared to APOE4, massive hepatic steatosis. An untargeted quantitative proteome analysis of the liver identified a high number of proteins differentially abundant in APOE3 versus APOE4 mice. The majority of the higher abundant proteins in APOE3 mice could be grouped to inflammation and damage-associated response, and lipid storage, amongst others. Results of the targeted qRT-PCR and Western blot analyses contribute to the overall finding that APOE3 as opposed to APOE4 promotes hepatic steatosis, inflammatory- and damage-associated response signaling and fibrosis in the liver of obese mice. Our experimental data substantiate the observation of an increased NAFLD-risk associated with the human APOEε3 allele, while APOEε4 appears protective. The underlying mechanisms of the protection possibly involve a higher capacity of nonectopic lipid deposition in subcutaneous adipose tissue and lower hepatic pathogen recognition in the APOE4 mice.
Collapse
Affiliation(s)
- Patricia Huebbe
- Institute of Human Nutrition and Food Science, Kiel University, D-24118 Kiel, Germany.
| | - Stephanie Bilke
- Institute of Experimental Medicine, Proteomics & Bioanalytics, Kiel University, D-24105 Kiel, Germany.
| | - Johanna Rueter
- Institute of Human Nutrition and Food Science, Kiel University, D-24118 Kiel, Germany.
| | - Anke Schloesser
- Institute of Human Nutrition and Food Science, Kiel University, D-24118 Kiel, Germany.
| | - Graeme Campbel
- Section Biomedical Imaging, Department of Radiology and Neuroradiology, Kiel University, D-24118 Kiel, Germany.
| | - Claus-C. Glüer
- Section Biomedical Imaging, Department of Radiology and Neuroradiology, Kiel University, D-24118 Kiel, Germany.
| | - Ralph Lucius
- Anatomical Institute, Kiel University, D-24118 Kiel, Germany.
| | - Christoph Röcken
- Department of Pathology, Kiel University and University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany.
| | - Andreas Tholey
- Institute of Experimental Medicine, Proteomics & Bioanalytics, Kiel University, D-24105 Kiel, Germany.
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, Kiel University, D-24118 Kiel, Germany.
| |
Collapse
|
6
|
Lozupone M, Panza F. Impact of apolipoprotein E isoforms on sporadic Alzheimer's disease: beyond the role of amyloid beta. Neural Regen Res 2024; 19:80-83. [PMID: 37488848 PMCID: PMC10479857 DOI: 10.4103/1673-5374.375316] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/20/2022] [Accepted: 04/16/2023] [Indexed: 07/26/2023] Open
Abstract
The impact of apolipoprotein E (ApoE) isoforms on sporadic Alzheimer's disease has long been studied; however, the influences of apolipoprotein E gene (APOE) on healthy and pathological human brains are not fully understood. ApoE exists as three common isoforms (ApoE2, ApoE3, and ApoE4), which differ in two amino acid residues. Traditionally, ApoE binds cholesterol and phospholipids and ApoE isoforms display different affinities for their receptors, lipids transport and distribution in the brain and periphery. The role of ApoE in the human depends on ApoE isoforms, brain regions, aging, and neural injury. APOE ε4 is the strongest genetic risk factor for sporadic Alzheimer's disease, considering its role in influencing amyloid-beta metabolism. The exact mechanisms by which APOE gene variants may increase or decrease Alzheimer's disease risk are not fully understood, but APOE was also known to affect directly and indirectly tau-mediated neurodegeneration, lipids metabolism, neurovascular unit, and microglial function. Consistent with the biological function of ApoE, ApoE4 isoform significantly altered signaling pathways associated with cholesterol homeostasis, transport, and myelination. Also, the rare protective APOE variants confirm that ApoE plays an important role in Alzheimer's disease pathogenesis. The objectives of the present mini-review were to describe classical and new roles of various ApoE isoforms in Alzheimer's disease pathophysiology beyond the deposition of amyloid-beta and to establish a functional link between APOE, brain function, and memory, from a molecular to a clinical level. APOE genotype also exerted a heterogeneous effect on clinical Alzheimer's disease phenotype and its outcomes. Not only in learning and memory but also in neuropsychiatric symptoms that occur in a premorbid condition. Clarifying the relationships between Alzheimer's disease-related pathology with neuropsychiatric symptoms, particularly suicidal ideation in Alzheimer's disease patients, may be useful for elucidating also the underlying pathophysiological process and its prognosis. Also, the effects of anti-amyloid-beta drugs, recently approved for the treatment of Alzheimer's disease, could be influenced by the APOE genotype.
Collapse
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Panza
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, Castellana Grotte, Bari, Italy
| |
Collapse
|
7
|
Dai X, Hu Y, Jiang L, Lei L, Fu C, Wu S, Zhang X, Zhu L, Zhang F, Chen J, Zeng Q. Decreased oxidative stress response and oxidant detoxification of skin during aging. Mech Ageing Dev 2023; 216:111878. [PMID: 37827221 DOI: 10.1016/j.mad.2023.111878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/04/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Oxidative stress plays an important role in the skin aging process; however, the mechanisms are not fully elucidated. Especially the changes in various types of skin cells with aging and the key oxidative stress-related genes that play a regulatory role are not clear. In this study, single-cell RNA sequencing data and microarray transcriptome data were used to explore the changes in oxidative stress response and oxidant detoxification capacity of skin cells during aging and oxidative stress-related genes potentially involved in regulating skin aging were searched. The oxidative stress response and oxidant detoxification ability were weakened in the elderly compared with those of the young. Among the different types of skin cells, keratinocytes, melanocytes, vascular endothelial cells, fibroblasts, and lymphatic endothelial cells exhibited a stronger oxidative stress response and oxidant detoxification ability, while immune cells exhibited a weaker oxidative stress response and detoxification capacity. During aging, the oxidative stress response and oxidant detoxification capacity of keratinocytes, fibroblasts, macrophages, and vascular endothelial cells were significantly weakened. Annexin A1 (ANXA1) and Apolipoprotein E (APOE) may be key oxidative stress-related genes affecting skin aging.
Collapse
Affiliation(s)
- Xixia Dai
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yibo Hu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ling Jiang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Li Lei
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Chuhan Fu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Songjiang Wu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xiaolin Zhang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Lu Zhu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Fan Zhang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jing Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
8
|
Gholami A. Alzheimer's disease: The role of proteins in formation, mechanisms, and new therapeutic approaches. Neurosci Lett 2023; 817:137532. [PMID: 37866702 DOI: 10.1016/j.neulet.2023.137532] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/03/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder that affects the central nervous system (CNS), leading to memory and cognitive decline. In AD, the brain experiences three main structural changes: a significant decrease in the quantity of neurons, the development of neurofibrillary tangles (NFT) composed of hyperphosphorylated tau protein, and the formation of amyloid beta (Aβ) or senile plaques, which are protein deposits found outside cells and surrounded by dystrophic neurites. Genetic studies have identified four genes associated with autosomal dominant or familial early-onset AD (FAD): amyloid precursor protein (APP), presenilin 1 (PS1), presenilin 2 (PS2), and apolipoprotein E (ApoE). The formation of plaques primarily involves the accumulation of Aβ, which can be influenced by mutations in APP, PS1, PS2, or ApoE genes. Mutations in the APP and presenilin (PS) proteins can cause an increased amyloid β peptides production, especially the further form of amyloidogenic known as Aβ42. Apart from genetic factors, environmental factors such as cytokines and neurotoxins may also have a significant impact on the development and progression of AD by influencing the formation of amyloid plaques and intracellular tangles. Exploring the causes and implications of protein aggregation in the brain could lead to innovative therapeutic approaches. Some promising therapy strategies that have reached the clinical stage include using acetylcholinesterase inhibitors, estrogen, nonsteroidal anti-inflammatory drugs (NSAIDs), antioxidants, and antiapoptotic agents. The most hopeful therapeutic strategies involve inhibiting activity of secretase and preventing the β-amyloid oligomers and fibrils formation, which are associated with the β-amyloid fibrils accumulation in AD. Additionally, immunotherapy development holds promise as a progressive therapeutic approach for treatment of AD. Recently, the two primary categories of brain stimulation techniques that have been studied for the treatment of AD are invasive brain stimulation (IBS) and non-invasive brain stimulation (NIBS). In this article, the amyloid proteins that play a significant role in the AD formation, the mechanism of disease formation as well as new drugs utilized to treat of AD will be reviewed.
Collapse
Affiliation(s)
- Amirreza Gholami
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
9
|
Balu D, Valencia-Olvera AC, Nguyen A, Patnam M, York J, Peri F, Neumann F, LaDu MJ, Tai LM. A small-molecule TLR4 antagonist reduced neuroinflammation in female E4FAD mice. Alzheimers Res Ther 2023; 15:181. [PMID: 37858252 PMCID: PMC10585767 DOI: 10.1186/s13195-023-01330-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND APOE genotype is the greatest genetic risk factor for sporadic Alzheimer's disease (AD). APOE4 increases AD risk up to 12-fold compared to APOE3, an effect that is greater in females. Evidence suggests that one-way APOE could modulate AD risk and progression through neuroinflammation. Indeed, APOE4 is associated with higher glial activation and cytokine levels in AD patients and mice. Therefore, identifying pathways that contribute to APOE4-associated neuroinflammation is an important approach for understanding and treating AD. Human and in vivo evidence suggests that TLR4, one of the key receptors involved in the innate immune system, could be involved in APOE-modulated neuroinflammation. Consistent with that idea, we previously demonstrated that the TLR4 antagonist IAXO-101 can reduce LPS- and Aβ-induced cytokine secretion in APOE4 glial cultures. Therefore, the goal of this study was to advance these findings and determine whether IAXO-101 can modulate neuroinflammation, Aβ pathology, and behavior in mice that express APOE4. METHODS We used mice that express five familial AD mutations and human APOE3 (E3FAD) or APOE4 (E4FAD). Female and male E4FAD mice and female E3FAD mice were treated with vehicle or IAXO-101 in two treatment paradigms: prevention from 4 to 6 months of age or reversal from 6 to 7 months of age. Learning and memory were assessed by modified Morris water maze. Aβ deposition, fibrillar amyloid deposition, astrogliosis, and microgliosis were assessed by immunohistochemistry. Soluble levels of Aβ and apoE, insoluble levels of apoE and Aβ, and IL-1β were measured by ELISA. RESULTS IAXO-101 treatment resulted in lower Iba-1 coverage, lower number of reactive microglia, and improved memory in female E4FAD mice in both prevention and reversal paradigms. IAXO-101-treated male E4FAD mice also had lower Iba-1 coverage and reactivity in the RVS paradigm, but there was no effect on behavior. There was also no effect of IAXO-101 treatment on neuroinflammation and behavior in female E3FAD mice. CONCLUSION Our data supports that TLR4 is a potential mechanistic therapeutic target for modulating neuroinflammation and cognition in APOE4 females.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ana C Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Austin Nguyen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mehul Patnam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
10
|
Liu ZJ, Yang LY, Lu TC, Huang C, Liang YQ, Xu XW, Xu YF, Liu MM, Lin XH, Chen JY. Precise Differentiation of Wobble-Type Allele via Ratiometric Design of a Ligase Chain Reaction-Based Electrochemical Biosensor for CYP2C19*2 Genotyping of Clinical Samples. Anal Chem 2023; 95:14592-14599. [PMID: 37683102 DOI: 10.1021/acs.analchem.3c01907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Due to the comparable stability between the perfect-base pair and the wobble-base pair, a precise differentiation of the wobble-type allele has remained a challenge, often leading to false results. Herein, we proposed a ligase chain reaction (LCR)-based ratiometric electrochemical DNA sensor, namely, R-eLCR, for a precise typing of the wobble-type allele, in which the traditionally recognized "negative" signal of wobble-base pair-mediated amplification was fully utilized as a "positive" one and a ratiometric readout mode was employed to ameliorated the underlying potential external influence and improved its detection accuracy in the typing of the wobble-type allele. The results showed that the ratio between current of methylene blue (IMB) and current of ferrocene (IFc) was partitioned in three regions and three types of wobble-type allele were thus precisely differentiated (AA homozygote: IMB/IFc > 2; GG homozygote: IMB/IFc < 1; GA heterozygote: 1 < IMB/IFc < 2); the proposed R-eLCR successfully discriminated the three types of CYP2C19*2 allele in nine cases of human whole blood samples, which was consistent with those of the sequencing method. These results evidence that the proposed R-eLCR can serve as an accurate and robust alternative for the identification of wobble-type allele, which lays a solid foundation and holds great potential for precision medicine.
Collapse
Affiliation(s)
- Zhou-Jie Liu
- Department of Pharmacy, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Liang-Yong Yang
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Tai-Cheng Lu
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Chen Huang
- Department of Pharmacy, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yu-Qi Liang
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Xiong-Wei Xu
- Department of Pharmacy, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yan-Fang Xu
- The Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Meng-Meng Liu
- Department of Pharmacy, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xin-Hua Lin
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jin-Yuan Chen
- The Central Laboratory, Fujian Key Laboratory of Precision Medicine for Cancer, Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
11
|
Abondio P, Bruno F, Luiselli D. Apolipoprotein E (APOE) Haplotypes in Healthy Subjects from Worldwide Macroareas: A Population Genetics Perspective for Cardiovascular Disease, Neurodegeneration, and Dementia. Curr Issues Mol Biol 2023; 45:2817-2831. [PMID: 37185708 PMCID: PMC10137191 DOI: 10.3390/cimb45040184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Human APOE is a 299-amino acid long protein expressed and secreted in several tissues and body districts, where it exerts different functions mainly related to lipid metabolism, with specific activities around cholesterol transport and absorption/elimination. It has three main isoforms, determined by the pair of mutations rs7412-C/T and rs429358-C/T, which gives rise to the functionally different APOE variants ε2, ε3, and ε4. These have a distinct impact on lipid metabolism and are differentially implicated in Alzheimer’s disease and neurodegeneration, cardiovascular disease, and dyslipidemia. A plethora of other single nucleotide variants along the sequence of the APOE gene have been studied in cohorts of affected individuals, where they also modulate the influence of the three main isoforms to determine the risk of developing the disease. However, no contextual analysis of gene-long haplotypes has been carried out so far, and never extensively in cohorts of healthy individuals from different worldwide populations. Leveraging a rich population genomics dataset, this study elucidates the distribution of APOE variants and haplotypes that are shared across populations and to specific macroareas, revealing a variety of risk-allele associations that distinguish specific ancestral backgrounds and can be leveraged for specific ancestry-informed screenings in medicine and public health.
Collapse
|
12
|
Rueter J, Rimbach G, Treitz C, Schloesser A, Lüersen K, Tholey A, Huebbe P. The mitochondrial BCKD complex interacts with hepatic apolipoprotein E in cultured cells in vitro and mouse livers in vivo. Cell Mol Life Sci 2023; 80:59. [PMID: 36749362 PMCID: PMC9905200 DOI: 10.1007/s00018-023-04706-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/22/2022] [Accepted: 01/23/2023] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Apolipoprotein E (APOE) is known for its role in lipid metabolism and its association with age-related disease pathology. The aim of the present work was to identify previously unknown functions of APOE based on the detection of novel APOE protein-protein interaction candidates. APPROACH AND RESULTS APOE targeted replacement mice and transfected cultured hepatocytes expressing the human isoforms APOE3 and APOE4 were used. For 7 months, APOE3 and APOE4 mice were fed a high-fat and high-sugar diet to induce obesity, while a subgroup was subjected to 30% dietary restriction. Proteomic analysis of coimmunoprecipitation products from APOE mouse liver extracts revealed 28 APOE-interacting candidate proteins, including branched-chain alpha-keto acid dehydrogenase (BCKD) complex subunit alpha (BCKDHA) and voltage-dependent anion-selective channel 1 (VDAC1). The binding of APOE and BCKDHA was verified in situ by proximity ligation assay in cultured cells. The activity of the BCKD enzyme complex was significantly higher in obese APOE4 mice than in APOE3 mice, while the plasma levels of branched-chain amino acids and mTOR signalling proteins were not different. However, the protein-protein interaction with VDAC1 was strongly induced in APOE3 and APOE4 mice upon dietary restriction, suggesting a prominent role of APOE in mitochondrial function. CONCLUSIONS The protein-protein interactions of APOE with BCKDHA and VDAC1 appear to be of physiological relevance and are modulated upon dietary restriction. Because these are mitochondrial proteins, it may be suggested that APOE is involved in mitochondria-related processes and adaptation to hepatic energy demands.
Collapse
Affiliation(s)
- Johanna Rueter
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118, Kiel, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118, Kiel, Germany.
| | - Christian Treitz
- Institute of Experimental Medicine, University of Kiel, Niemannsweg 11, 24105, Kiel, Germany
| | - Anke Schloesser
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118, Kiel, Germany
| | - Kai Lüersen
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118, Kiel, Germany
| | - Andreas Tholey
- Institute of Experimental Medicine, University of Kiel, Niemannsweg 11, 24105, Kiel, Germany
| | - Patricia Huebbe
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118, Kiel, Germany
| |
Collapse
|
13
|
Gabrielli AP, Weidling I, Ranjan A, Wang X, Novikova L, Chowdhury SR, Menta B, Berkowicz A, Wilkins HM, Peterson KR, Swerdlow RH. Mitochondria Profoundly Influence Apolipoprotein E Biology. J Alzheimers Dis 2023; 92:591-604. [PMID: 36776072 DOI: 10.3233/jad-221177] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND Mitochondria can trigger Alzheimer's disease (AD)-associated molecular phenomena, but how mitochondria impact apolipoprotein E (APOE; apoE) is not well known. OBJECTIVE Consider whether and how mitochondrial biology influences APOE and apoE biology. METHODS We measured APOE expression in human SH-SY5Y neuronal cells with different forms of mitochondrial dysfunction including total, chronic mitochondrial DNA (mtDNA) depletion (ρ0 cells); acute, partial mtDNA depletion; and toxin-induced mitochondrial dysfunction. We further assessed intracellular and secreted apoE protein levels in the ρ0 cells and interrogated the impact of transcription factors and stress signaling pathways known to influence APOE expression. RESULTS SH-SY5Y ρ0 cells exhibited a 65-fold increase in APOE mRNA, an 8-fold increase in secreted apoE protein, and increased intracellular apoE protein. Other models of primary mitochondrial dysfunction including partial mtDNA-depletion, toxin-induced respiratory chain inhibition, and chemical-induced manipulations of the mitochondrial membrane potential similarly increased SH-SY5Y cell APOE mRNA. We explored potential mediators and found in the ρ0 cells knock-down of the C/EBPα and NFE2L2 (Nrf2) transcription factors reduced APOE mRNA. The activity of two mitogen-activated protein kinases, JNK and ERK, also strongly influenced ρ0 cell APOE mRNA levels. CONCLUSION Primary mitochondrial dysfunction either directly or indirectly activates APOE expression in a neuronal cell model by altering transcription factors and stress signaling pathways. These studies demonstrate mitochondrial biology can influence the biology of the APOE gene and apoE protein, which are implicated in AD.
Collapse
Affiliation(s)
- Alexander P Gabrielli
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ian Weidling
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Amol Ranjan
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Xiaowan Wang
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Lesya Novikova
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Subir Roy Chowdhury
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Blaise Menta
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Alexandra Berkowicz
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kenneth R Peterson
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
14
|
Donen G, Milad N, Bernatchez P. Humanization of the mdx Mouse Phenotype for Duchenne Muscular Dystrophy Modeling: A Metabolic Perspective. J Neuromuscul Dis 2023; 10:1003-1012. [PMID: 37574742 PMCID: PMC10657711 DOI: 10.3233/jnd-230126] [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] [Accepted: 07/25/2023] [Indexed: 08/15/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a severe form of muscular dystrophy (MD) that is characterized by early muscle wasting and lethal cardiorespiratory failure. While the mdx mouse is the most common model of DMD, it fails to replicate the severe loss of muscle mass and other complications observed in patients, in part due to the multiple rescue pathways found in mice. This led to several attempts at improving DMD animal models by interfering with these rescue pathways through double transgenic approaches, resulting in more severe phenotypes with mixed relevance to the human pathology. As a growing body of literature depicts DMD as a multi-system metabolic disease, improvements in mdx-based modeling of DMD may be achieved by modulating whole-body metabolism instead of muscle homeostasis. This review provides an overview of the established dual-transgenic approaches that exacerbate the mild mdx phenotype by primarily interfering with muscle homeostasis and highlights how advances in DMD modeling coincide with inducing whole-body metabolic changes. We focus on the DBA2/J strain-based D2.mdx mouse with heightened transforming growth factor (TGF)-β signaling and the dyslipidemic mdx/apolipoprotein E (mdx/ApoE) knock-out (KO) mouse, and summarize how these novel models emulate the metabolic changes observed in DMD.
Collapse
Affiliation(s)
| | | | - Pascal Bernatchez
- Correspondence to: Dr. Pascal Bernatchez, Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, 2176 Health Sciences mall, room 217, Vancouver BC, V6T 1Z3, Canada. Tel.: +1 604 806 8346 /Ext.66060; E-mail:
| |
Collapse
|
15
|
Kodama J, Wilkinson KJ, Otsuru S. Nutrient metabolism of the nucleus pulposus: A literature review. NORTH AMERICAN SPINE SOCIETY JOURNAL 2022; 13:100191. [PMID: 36590450 PMCID: PMC9801222 DOI: 10.1016/j.xnsj.2022.100191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022]
Abstract
Cells take in, consume, and synthesize nutrients for numerous physiological functions. This includes not only energy production but also macromolecule biosynthesis, which will further influence cellular signaling, redox homeostasis, and cell fate commitment. Therefore, alteration in cellular nutrient metabolism is associated with pathological conditions. Intervertebral discs, particularly the nucleus pulposus (NP), are avascular and exhibit unique metabolic preferences. Clinical and preclinical studies have indicated a correlation between intervertebral degeneration (IDD) and systemic metabolic diseases such as diabetes, obesity, and dyslipidemia. However, a lack of understanding of the nutrient metabolism of NP cells is masking the underlying mechanism. Indeed, although previous studies indicated that glucose metabolism is essential for NP cells, the downstream metabolic pathways remain unknown, and the potential role of other nutrients, like amino acids and lipids, is understudied. In this literature review, we summarize the current understanding of nutrient metabolism in NP cells and discuss other potential metabolic pathways by referring to a human NP transcriptomic dataset deposited to the Gene Expression Omnibus, which can provide us hints for future studies of nutrient metabolism in NP cells and novel therapies for IDD.
Collapse
Affiliation(s)
- Joe Kodama
- Corresponding authors at: 670 W Baltimore St. HSFIII 7173, Baltimore, MD 21201, USA.
| | | | - Satoru Otsuru
- Corresponding authors at: 670 W Baltimore St. HSFIII 7173, Baltimore, MD 21201, USA.
| |
Collapse
|