1
|
Pham TNM, Behl C. Cellular models of stress resistance may pave ways to fight neurodegenerative diseases. Neural Regen Res 2025; 20:2579-2580. [PMID: 39503421 PMCID: PMC11801301 DOI: 10.4103/nrr.nrr-d-24-00476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/01/2024] [Accepted: 08/15/2024] [Indexed: 02/08/2025] Open
Affiliation(s)
- Thu Nguyen Minh Pham
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Christian Behl
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
2
|
Yang X, Yao K, Zhang M, Zhang W, Zu H. New insight into the role of altered brain cholesterol metabolism in the pathogenesis of AD: A unifying cholesterol hypothesis and new therapeutic approach for AD. Brain Res Bull 2025; 224:111321. [PMID: 40164234 DOI: 10.1016/j.brainresbull.2025.111321] [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: 01/30/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
The dysregulation of cholesterol metabolism homeostasis has been universally suggested in the aeotiology of Alzheimer's disease (AD). Initially, studies indicate that alteration of serum cholesterol level might contribute to AD. However, because blood-brain barrier impedes entry of plasma cholesterol, brain cells are not directly influenced by plasma cholesterol. Furthermore, mounting evidences suggest a link between alteration of brain cholesterol metabolism and AD. Interestingly, Amyloid-β proteins (Aβ) can markedly inhibit cellular cholesterol biosynthesis and lower cellular cholesterol content in cultured cells. And Aβ overproduction/overload induces a significant decrease of brain cellular cholesterol content in familial AD (FAD) animals. Importantly, mutations or polymorphisms of genes related to brain cholesterol transportation, such as ApoE4, ATP binding cassette (ABC) transporters, low-density lipoprotein receptor (LDLR) family and Niemann-Pick C disease 1 or 2 (NPC1/2), obviously lead to decreased brain cholesterol transport, resulting in brain cellular cholesterol loss, which could be tightly associated with AD pathological impairments. Additionally, accumulating data show that there are reduction of brain cholesterol biosynthesis and/or disorder of brain cholesterol trafficking in a variety of sporadic AD (SAD) animals and patients. Collectively, compelling evidences indicate that FAD and SAD could share one common and overlapping neurochemical mechanism: brain neuronal/cellular cholesterol deficiency. Therefore, accumulated evidences strongly support a novel hypothesis that deficiency of brain cholesterol contributes to the onset and progression of AD. This review highlights the pivotal role of brain cholesterol deficiency in the pathogenesis of AD. The hypothesis offers valuable insights for the future development of AD treatment.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China; Department of Neurology, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200237, China
| | - Kai Yao
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Mengqi Zhang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Wenbin Zhang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China.
| |
Collapse
|
3
|
Wu Y, Song W, Su M, He J, Hu R, Zhao Y. The Role of Cholesterol Metabolism and Its Regulation in Tumor Development. Cancer Med 2025; 14:e70783. [PMID: 40145543 PMCID: PMC11948085 DOI: 10.1002/cam4.70783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Within the tumor microenvironment, tumor cells undergo metabolic reprogramming of cholesterol due to intrinsic cellular alterations and changes in the extracellular milieu. Furthermore, cholesterol reprogramming within this microenvironment influences the immune landscape of tumors, facilitating immune evasion and consequently promoting tumorigenesis. These biological changes involve modifications in numerous enzymes associated with cholesterol uptake and synthesis, including NPC1L1, SREBP, HMGCR, SQLE, and PCSK9. REVIEW This review systematically summarizes the role of cholesterol metabolism and its associated enzymes in cancer progression, examines the mechanisms through which dysregulation of cholesterol metabolism affects immune cells within the tumor microenvironment, and discusses recent advancements in cancer therapies that target cholesterol metabolism. CONCLUSION Targeting cholesterol metabolism-related enzymes can inhibit tumor growth, reshape immune landscapes, and rejuvenate antitumor immunity, offering potential therapeutic avenues in cancer treatment.
Collapse
Affiliation(s)
- Yongmei Wu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Wenqian Song
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Min Su
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Jing He
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Rong Hu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Youbo Zhao
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| |
Collapse
|
4
|
Cai P, Zhang W, Jiang S, Xiong Y, Qiao H, Jin S, Fu H. A study on the functional role of the DHCR24 gene in gonadal differentiation and development of Macrobrachium nipponense. Sci Rep 2024; 14:29443. [PMID: 39604403 PMCID: PMC11603370 DOI: 10.1038/s41598-024-80651-4] [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: 07/10/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
Sex differentiation in crustaceans is a complex process influenced by various factors, including the androgenic gland and sex-related genes. This study characterized the role of the Mn-DHCR24 gene in the oriental river prawn (Macrobrachium nipponense). We used bioinformatics to analyze sequence features and phylogenetic relationships of a single Mn-DHCR24 gene. The expression patterns of Mn-DHCR24 across different tissues and developmental stages were determined by real-time PCR, and its localization in testis was determined by in situ hybridization. RNA interference was used to knock down Mn-DHCR24 expression, followed by examining changes in sex ratio and gonadal development at the PL10 stage. Additionally, an enzyme-linked immunosorbent assay measured 17α-methyltestosterone levels, and tissue sections were used to characterize gonadal development. The results indicated that Mn-DHCR24 was high expression in testis, which was critical for sperm maturation and gonadal differentiation. RNAi experiments showed the role of Mn-DHCR24 during reproductive regulation rather than as a master gene for sex differentiation. This study further showed that Mn-DHCR24 regulated sex and hormone-related genes, influencing steroid biosynthesis pathways. Together, these findings provided valuable insights into the genetic and hormonal mechanisms of gonadal differentiation in M. nipponense, and supported the development of monosex culture technology.
Collapse
Affiliation(s)
- Pengfei Cai
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China
| | - Wenyi Zhang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Sufei Jiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Yiwei Xiong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Hui Qiao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Shubo Jin
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China.
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
| | - Hongtuo Fu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China.
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
| |
Collapse
|
5
|
Mistry H, Richardson CD, Higginbottom A, Ashford B, Ahamed SU, Moore Z, Matthews FE, Brayne C, Simpson JE, Wharton SB. Relationships of brain cholesterol and cholesterol biosynthetic enzymes to Alzheimer's pathology and dementia in the CFAS population-derived neuropathology cohort. Neurosci Res 2024; 204:22-33. [PMID: 38278219 PMCID: PMC11192635 DOI: 10.1016/j.neures.2024.01.003] [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: 06/01/2023] [Revised: 01/05/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
Altered cholesterol metabolism is implicated in brain ageing and Alzheimer's disease. We examined whether key genes regulating cholesterol metabolism and levels of brain cholesterol are altered in dementia and Alzheimer's disease neuropathological change (ADNC). Temporal cortex (n = 99) was obtained from the Cognitive Function and Ageing Study. Expression of the cholesterol biosynthesis rate-limiting enzyme HMG-CoA reductase (HMGCR) and its regulator, SREBP2, were detected using immunohistochemistry. Expression of HMGCR, SREBP2, CYP46A1 and ABCA1 were quantified by qPCR in samples enriched for astrocyte and neuronal RNA following laser-capture microdissection. Total cortical cholesterol was measured using the Amplex Red assay. HMGCR and SREBP2 proteins were predominantly expressed in pyramidal neurones, and in glia. Neuronal HMGCR did not vary with ADNC, oxidative stress, neuroinflammation or dementia status. Expression of HMGCR neuronal mRNA decreased with ADNC (p = 0.022) and increased with neuronal DNA damage (p = 0.049), whilst SREBP2 increased with ADNC (p = 0.005). High or moderate tertiles for cholesterol levels were associated with increased dementia risk (OR 1.44, 1.58). APOE ε4 allele was not associated with cortical cholesterol levels. ADNC is associated with gene expression changes that may impair cholesterol biosynthesis in neurones but not astrocytes, whilst levels of cortical cholesterol show a weak relationship to dementia status.
Collapse
Affiliation(s)
- Hemant Mistry
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | | | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | - Bridget Ashford
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | - Saif U Ahamed
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | - Zoe Moore
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | | | - Carol Brayne
- Cambridge Public Health, University of Cambridge, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK.
| |
Collapse
|
6
|
Zhao J, Wei M, Guo M, Wang M, Niu H, Xu T, Zhou Y. GSK3: A potential target and pending issues for treatment of Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14818. [PMID: 38946682 PMCID: PMC11215492 DOI: 10.1111/cns.14818] [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: 04/22/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Glycogen synthase kinase-3 (GSK3), consisting of GSK3α and GSK3β subtypes, is a complex protein kinase that regulates numerous substrates. Research has observed increased GSK3 expression in the brains of Alzheimer's disease (AD) patients and models. AD is a neurodegenerative disorder with diverse pathogenesis and notable cognitive impairments, characterized by Aβ aggregation and excessive tau phosphorylation. This article provides an overview of GSK3's structure and regulation, extensively analyzing its relationship with AD factors. GSK3 overactivation disrupts neural growth, development, and function. It directly promotes tau phosphorylation, regulates amyloid precursor protein (APP) cleavage, leading to Aβ formation, and directly or indirectly triggers neuroinflammation and oxidative damage. We also summarize preclinical research highlighting the inhibition of GSK3 activity as a primary therapeutic approach for AD. Finally, pending issues like the lack of highly specific and affinity-driven GSK3 inhibitors, are raised and expected to be addressed in future research. In conclusion, GSK3 represents a target in AD treatment, filled with hope, challenges, opportunities, and obstacles.
Collapse
Affiliation(s)
- Jiahui Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Mengying Wei
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Future Health Laboratory, Innovation Center of Yangtze River DeltaZhejiang UniversityJiaxingChina
| | - Minsong Guo
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouChina
| | - Mengyao Wang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Hongxia Niu
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Laboratory of Blood‐stasis‐toxin Syndrome of Zhejiang ProvinceHangzhouChina
| | - Tengfei Xu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouChina
| | - Yuan Zhou
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Laboratory of Blood‐stasis‐toxin Syndrome of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
7
|
Staurenghi E, Testa G, Leoni V, Cecci R, Floro L, Giannelli S, Barone E, Perluigi M, Leonarduzzi G, Sottero B, Gamba P. Altered Brain Cholesterol Machinery in a Down Syndrome Mouse Model: A Possible Common Feature with Alzheimer's Disease. Antioxidants (Basel) 2024; 13:435. [PMID: 38671883 PMCID: PMC11047305 DOI: 10.3390/antiox13040435] [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: 03/06/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Down syndrome (DS) is a complex chromosomal disorder considered as a genetically determined form of Alzheimer's disease (AD). Maintenance of brain cholesterol homeostasis is essential for brain functioning and development, and its dysregulation is associated with AD neuroinflammation and oxidative damage. Brain cholesterol imbalances also likely occur in DS, concurring with the precocious AD-like neurodegeneration. In this pilot study, we analyzed, in the brain of the Ts2Cje (Ts2) mouse model of DS, the expression of genes encoding key enzymes involved in cholesterol metabolism and of the levels of cholesterol and its main precursors and products of its metabolism (i.e., oxysterols). The results showed, in Ts2 mice compared to euploid mice, the downregulation of the transcription of the genes encoding the enzymes 3-hydroxy-3-methylglutaryl-CoA reductase and 24-dehydrocholesterol reductase, the latter originally recognized as an indicator of AD, and the consequent reduction in total cholesterol levels. Moreover, the expression of genes encoding enzymes responsible for brain cholesterol oxidation and the amounts of the resulting oxysterols were modified in Ts2 mouse brains, and the levels of cholesterol autoxidation products were increased, suggesting an exacerbation of cerebral oxidative stress. We also observed an enhanced inflammatory response in Ts2 mice, underlined by the upregulation of the transcription of the genes encoding for α-interferon and interleukin-6, two cytokines whose synthesis is increased in the brains of AD patients. Overall, these results suggest that DS and AD brains share cholesterol cycle derangements and altered oxysterol levels, which may contribute to the oxidative and inflammatory events involved in both diseases.
Collapse
Affiliation(s)
- Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Valerio Leoni
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, 20832 Desio, Italy;
| | - Rebecca Cecci
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Lucrezia Floro
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University, 00185 Roma, Italy; (E.B.); (M.P.)
| | - Marzia Perluigi
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University, 00185 Roma, Italy; (E.B.); (M.P.)
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Barbara Sottero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| |
Collapse
|
8
|
Peeples ES, Mirnics K, Korade Z. Chemical Inhibition of Sterol Biosynthesis. Biomolecules 2024; 14:410. [PMID: 38672427 PMCID: PMC11048061 DOI: 10.3390/biom14040410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Cholesterol is an essential molecule of life, and its synthesis can be inhibited by both genetic and nongenetic mechanisms. Hundreds of chemicals that we are exposed to in our daily lives can alter sterol biosynthesis. These also encompass various classes of FDA-approved medications, including (but not limited to) commonly used antipsychotic, antidepressant, antifungal, and cardiovascular medications. These medications can interfere with various enzymes of the post-lanosterol biosynthetic pathway, giving rise to complex biochemical changes throughout the body. The consequences of these short- and long-term homeostatic disruptions are mostly unknown. We performed a comprehensive review of the literature and built a catalogue of chemical agents capable of inhibiting post-lanosterol biosynthesis. This process identified significant gaps in existing knowledge, which fall into two main areas: mechanisms by which sterol biosynthesis is altered and consequences that arise from the inhibitions of the different steps in the sterol biosynthesis pathway. The outcome of our review also reinforced that sterol inhibition is an often-overlooked mechanism that can result in adverse consequences and that there is a need to develop new safety guidelines for the use of (novel and already approved) medications with sterol biosynthesis inhibiting side effects, especially during pregnancy.
Collapse
Affiliation(s)
- Eric S. Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Child Health Research Institute, Omaha, NE 68198, USA;
- Division of Neonatology, Children’s Nebraska, Omaha, NE 68114, USA
| | - Karoly Mirnics
- Child Health Research Institute, Omaha, NE 68198, USA;
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zeljka Korade
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Child Health Research Institute, Omaha, NE 68198, USA;
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
9
|
Cocciadiferro D, Mazza T, Vecchio D, Biagini T, Petrizzelli F, Agolini E, Villani A, Minervino D, Martinelli D, Rizzo C, Boenzi S, Panfili FM, Buonuomo PS, Macchiaiolo M, Bartuli A, Novelli A. Exploiting in silico structural analysis to introduce emerging genotype-phenotype correlations in DHCR24-related sterol biosynthesis disorder: a case study. Front Genet 2024; 14:1307934. [PMID: 38239854 PMCID: PMC10795535 DOI: 10.3389/fgene.2023.1307934] [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: 10/05/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Desmosterolosis is a rare sterol biosynthesis disorder characterized by multiple congenital anomalies, failure to thrive, severe developmental delay, progressive epileptic encephalopathy, and elevated levels of desmosterol caused by biallelic mutations of DHCR24 encoding 3-β-hydroxysterol Δ-24-reductase. DHCR24 is regarded as the key enzyme of cholesterol synthesis in the metabolism of brain cholesterol as it catalyzes the reduction of the Δ-24 double bond of sterol intermediates during cholesterol biosynthesis. To date, 15 DHCR24 variants, detected in 2 related and 14 unrelated patients, have been associated with the desmosterolosis disorder. Here, we describe a proband harboring the never-described DHCR24 homozygous missense variant NM_014762.4:c.506T>C, NP_055577.1:p.M169T, whose functional validation was confirmed through biochemical assay. By using molecular dynamics simulation techniques, we investigated the impact of this variant on the protein stability and interaction network with the flavin adenine dinucleotide cofactor, thereby providing a preliminary assessment of its mechanistic role in comparison to all known pathogenic variants, the wild-type protein, and a known benign DHCR24 variant. This report expands the clinical and molecular spectra of the DHCR24-related disorder, reports on a novel DHCR24 deleterious variant associated with desmosterolosis, and gives new insights into genotype-phenotype correlations.
Collapse
Affiliation(s)
- Dario Cocciadiferro
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Davide Vecchio
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Tommaso Biagini
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesco Petrizzelli
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Emanuele Agolini
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Andrea Villani
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Daniele Minervino
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Diego Martinelli
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Cristiano Rizzo
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Sara Boenzi
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Filippo Maria Panfili
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paola Sabrina Buonuomo
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Andrea Bartuli
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
10
|
Fu X, Wang Z. DHCR24 in Tumor Diagnosis and Treatment: A Comprehensive Review. Technol Cancer Res Treat 2024; 23:15330338241259780. [PMID: 38847653 PMCID: PMC11162140 DOI: 10.1177/15330338241259780] [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] [Indexed: 06/10/2024] Open
Abstract
As an important nutrient in the human body, cholesterol can not only provide structural components for the body's cells, but also can be transformed into a variety of active substances to regulate cell signaling pathways. As an important cholesterol synthase, DHCR24 participates in important regulatory processes in the body. The application of DHCR24 in tumor clinical diagnosis and treatment also attracts much attention. This article reviews the structure and regulatory characteristics of DHCR24, and the research of DHCR24 on tumor progression. We summarize the possible mechanisms of DHCR24 promoting tumor progression through reactive oxygen species (ROS), p53, Ras and PI3K-AKT pathways. Through our review, we hope to provide more research ideas and reference value for the application of DHCR24 in tumor prevention and treatment.
Collapse
Affiliation(s)
- Xin Fu
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhaosong Wang
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Laboratory Animal Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
11
|
Liu Q, Chen S, Tian R, Xue B, Li H, Guo M, Liu S, Yan M, You R, Wang L, Yang D, Wan M, Zhu H. 3β-hydroxysteroid-Δ24 reductase dampens anti-viral innate immune responses by targeting K27 ubiquitination of MAVS and STING. J Virol 2023; 97:e0151323. [PMID: 38032198 PMCID: PMC10734464 DOI: 10.1128/jvi.01513-23] [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: 09/27/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
IMPORTANCE The precise regulation of the innate immune response is essential for the maintenance of homeostasis. MAVS and STING play key roles in immune signaling pathways activated by RNA and DNA viruses, respectively. Here, we showed that DHCR24 impaired the antiviral response by targeting MAVS and STING. Notably, DHCR24 interacts with MAVS and STING and inhibits TRIM21-triggered K27-linked ubiquitination of MAVS and AMFR-triggered K27-linked ubiquitination of STING, restraining the activation of MAVS and STING, respectively. Together, this study elucidates how one cholesterol key enzyme orchestrates two antiviral signal transduction pathways.
Collapse
Affiliation(s)
- Qian Liu
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Shengwen Chen
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Renyun Tian
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Binbin Xue
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Huiyi Li
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Mengmeng Guo
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Shun Liu
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Ming Yan
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Ruina You
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Luoling Wang
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Di Yang
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Mengyu Wan
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Haizhen Zhu
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
- />Department of Pathogen Biology and Immunology, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Institute of Pathogen Biology and Immunology, School of Basic Medicine and Life Science, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Hainan, China
- Department of Clinical Laboratory of the Second Affiliated Hospital of Hainan Medical University, Hainan Medical University, Hainan, China
| |
Collapse
|
12
|
Pham TNM, Perumal N, Manicam C, Basoglu M, Eimer S, Fuhrmann DC, Pietrzik CU, Clement AM, Körschgen H, Schepers J, Behl C. Adaptive responses of neuronal cells to chronic endoplasmic reticulum (ER) stress. Redox Biol 2023; 67:102943. [PMID: 37883843 PMCID: PMC10618786 DOI: 10.1016/j.redox.2023.102943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Accumulation of misfolded proteins or perturbation of calcium homeostasis leads to endoplasmic reticulum (ER) stress and is linked to the pathogenesis of neurodegenerative diseases. Hence, understanding the ability of neuronal cells to cope with chronic ER stress is of fundamental interest. Interestingly, several brain areas uphold functions that enable them to resist challenges associated with neurodegeneration. Here, we established novel clonal mouse hippocampal (HT22) cell lines that are resistant to prolonged (chronic) ER stress induced by thapsigargin (TgR) or tunicamycin (TmR) as in vitro models to study the adaption to ER stress. Morphologically, we observed a significant increase in vesicular und autophagosomal structures in both resistant lines and 'giant lysosomes', especially striking in TgR cells. While autophagic activity increased under ER stress, lysosomal function appeared slightly impaired; in both cell lines, we observed enhanced ER-phagy. However, proteomic analyses revealed that various protein clusters and signaling pathways were differentially regulated in TgR versus TmR cells in response to chronic ER stress. Additionally, bioenergetic analyses in both resistant cell lines showed a shift toward aerobic glycolysis ('Warburg effect') and a defective complex I of the oxidative phosphorylation (OXPHOS) machinery. Furthermore, ER stress-resistant cells differentially activated the unfolded protein response (UPR) comprising IRE1α and ATF6 pathways. These findings display the wide portfolio of adaptive responses of neuronal cells to chronic ER stress. ER stress-resistant neuronal cells could be the basis to uncover molecular modulators of adaptation, resistance, and neuroprotection as potential pharmacological targets for preventing neurodegeneration.
Collapse
Affiliation(s)
- Thu Nguyen Minh Pham
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Natarajan Perumal
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Marion Basoglu
- Department of Structural Cell Biology, Institute for Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
| | - Stefan Eimer
- Department of Structural Cell Biology, Institute for Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Albrecht M Clement
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hagen Körschgen
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jana Schepers
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Christian Behl
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
13
|
Chaparro CIP, Simões BT, Borges JP, Castanho MARB, Soares PIP, Neves V. A Promising Approach: Magnetic Nanosystems for Alzheimer's Disease Theranostics. Pharmaceutics 2023; 15:2316. [PMID: 37765284 PMCID: PMC10536416 DOI: 10.3390/pharmaceutics15092316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Among central nervous system (CNS) disorders, Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and a major cause of dementia worldwide. The yet unclear etiology of AD and the high impenetrability of the blood-brain barrier (BBB) limit most therapeutic compounds from reaching the brain. Although many efforts have been made to effectively deliver drugs to the CNS, both invasive and noninvasive strategies employed often come with associated side effects. Nanotechnology-based approaches such as nanoparticles (NPs), which can act as multifunctional platforms in a single system, emerged as a potential solution for current AD theranostics. Among these, magnetic nanoparticles (MNPs) are an appealing strategy since they can act as contrast agents for magnetic resonance imaging (MRI) and as drug delivery systems. The nanocarrier functionalization with specific moieties, such as peptides, proteins, and antibodies, influences the particles' interaction with brain endothelial cell constituents, facilitating transport across the BBB and possibly increasing brain penetration. In this review, we introduce MNP-based systems, combining surface modifications with the particles' physical properties for molecular imaging, as a novel neuro-targeted strategy for AD theranostics. The main goal is to highlight the potential of multifunctional MNPs and their advances as a dual nanotechnological diagnosis and treatment platform for neurodegenerative disorders.
Collapse
Affiliation(s)
- Catarina I. P. Chaparro
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (C.I.P.C.); (B.T.S.); (M.A.R.B.C.)
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University of Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - Beatriz T. Simões
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (C.I.P.C.); (B.T.S.); (M.A.R.B.C.)
| | - João P. Borges
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University of Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - Miguel A. R. B. Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (C.I.P.C.); (B.T.S.); (M.A.R.B.C.)
| | - Paula I. P. Soares
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University of Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal;
| | - Vera Neves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (C.I.P.C.); (B.T.S.); (M.A.R.B.C.)
| |
Collapse
|
14
|
Lu Z, Wang H, Zhang X, Huang X, Jiang S, Li Y, Liu T, Lu X, Gao B. High fat diet induces brain injury and neuronal apoptosis via down-regulating 3-β hydroxycholesterol 24 reductase (DHCR24). Cell Tissue Res 2023; 393:471-487. [PMID: 37458798 DOI: 10.1007/s00441-023-03804-3] [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: 01/16/2023] [Accepted: 06/29/2023] [Indexed: 09/08/2023]
Abstract
Hyperlipidemia (HLP) is one of the risk factors for memory impairment and cognitive impairment. However, its pathological molecular mechanism remained unclear. 3β-hydroxysterol Δ24- reductase (DHCR24) is a key enzyme in cholesterol synthesis and has been reported to decrease in the affected areas in the brain of neurodegenerative disorders. In this study, hyperlipidemic mouse model was established to study the effect of high blood lipid on brain. The data obtained from HPLC analysis demonstrated that the cholesterol level in the brain of mice with hyperlipidemia was significantly elevated compared to the control group. While the pathological damages were observed in both cerebral cortex and hippocampus in the brain of hyperlipidemic mice. Furthermore, the protein level of DHCR24 was downregulated accompanied by elevated ubiquitination level in the hyperlipidemic mice brain. The mouse neuroblastoma cells N2a were exposed to the excess cholesterol loading, the cells underwent apoptosis and the mRNA and protein of DHCR24 in cholesterol-loaded N2a cells were significantly reduced. In addition, the expression level of endoplasmic reticulum stress marker protein (Bip and Chop) was markedly increased in response to the cholesterol loading. More importantly, overexpression of DHCR24 in N2a reversed neuronal apoptosis induced by the cholesterol loading. Conclusively, these findings suggested that hyperlipidemia could cause brain tissue injuries via down-regulating DHCR24, and overexpression of DHCR24 may alleviate hyperlipidemia-induced neuronal cells damage by reversing the endoplasmic reticulum stress-mediated apoptosis.
Collapse
Affiliation(s)
- Ziyin Lu
- The School of Life Science, Liaoning University, Chongshanzhong-Lu No.66, Huanggu-Qu, Shenyang, 110036, China
| | - Haozhen Wang
- The School of Life Science, Liaoning University, Chongshanzhong-Lu No.66, Huanggu-Qu, Shenyang, 110036, China
| | - Xiujin Zhang
- The School of Life Science, Liaoning University, Chongshanzhong-Lu No.66, Huanggu-Qu, Shenyang, 110036, China
| | - Xiuting Huang
- The School of Life Science, Liaoning University, Chongshanzhong-Lu No.66, Huanggu-Qu, Shenyang, 110036, China
| | - Shan Jiang
- The School of Life Science, Liaoning University, Chongshanzhong-Lu No.66, Huanggu-Qu, Shenyang, 110036, China
| | - Yang Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shenyang Medical College, Huang-He-Bei-Dajie, No.146, Shenyang, 110034, China
| | - Ting Liu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shenyang Medical College, Huang-He-Bei-Dajie, No.146, Shenyang, 110034, China
| | - Xiuli Lu
- The School of Life Science, Liaoning University, Chongshanzhong-Lu No.66, Huanggu-Qu, Shenyang, 110036, China.
| | - Bing Gao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shenyang Medical College, Huang-He-Bei-Dajie, No.146, Shenyang, 110034, China.
| |
Collapse
|
15
|
Zhang X, Wolinska J, Blair D, Hu W, Yin M. Responses to predation pressure involve similar sets of genes in two divergent species of Daphnia. J Anim Ecol 2023; 92:1743-1758. [PMID: 37337454 DOI: 10.1111/1365-2656.13969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 06/02/2023] [Indexed: 06/21/2023]
Abstract
Species that are not closely related can express similar inducible traits, but molecular mechanisms underlying the observed responses are often unknown, nor is it known if these mechanisms are shared between such species. Here, we compared transcriptional profiles of two Daphnia species (D. mitsukuri and D. sinensis) from different subgenera, at both juvenile and adult developmental stages. Both species were exposed to the same predation threat (fish kairomones), and both showed similar induced morphological changes (reduced body length). At the early developmental stage, response to predation risk resulted in similar changes in expression levels of 23 orthologues in both species. These orthologues, involved in 107 GO categories, changed in the same direction in both species (over- or underexpressed), in comparison to non-exposed controls. Several of these orthologues were associated with DNA replication, structural constituents of cuticle or innate immune response. In both species, the differentially expressed (DE) genes on average had higher ω (dN /dS ) values than non-DE genes, suggesting that these genes had experienced greater positive selection or lower purifying selection than non-DE genes. Overall, our results suggest that similar suites of genes, responding in similar ways to predation pressure, have been retained in Daphnia for many millions of years.
Collapse
Affiliation(s)
- Xiuping Zhang
- MOE Key Laboratory for Biodiversity Science and Ecological Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Justyna Wolinska
- Department of Evolutionary and Integrative Ecology, Leibniz Institute of Freshwater Ecology and Inland Fisheries, Berlin, Germany
- Department of Biology, Chemistry, Pharmacy, Institute of Biology, Freie Universität Berlin, Berlin, Germany
| | - David Blair
- College of Marine and Environmental Sciences, James Cook University, Townsville, Queensland, Australia
| | - Wei Hu
- MOE Key Laboratory for Biodiversity Science and Ecological Engineering, School of Life Science, Fudan University, Shanghai, China
- Department of Microbiology and Bioengineering, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Mingbo Yin
- MOE Key Laboratory for Biodiversity Science and Ecological Engineering, School of Life Science, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Zhang WB, Huang Y, Guo XR, Zhang MQ, Yuan XS, Zu HB. DHCR24 reverses Alzheimer's disease-related pathology and cognitive impairment via increasing hippocampal cholesterol levels in 5xFAD mice. Acta Neuropathol Commun 2023; 11:102. [PMID: 37344916 DOI: 10.1186/s40478-023-01593-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023] Open
Abstract
Accumulating evidences reveal that cellular cholesterol deficiency could trigger the onset of Alzheimer's disease (AD). As a key regulator, 24-dehydrocholesterol reductase (DHCR24) controls cellular cholesterol homeostasis, which was found to be downregulated in AD vulnerable regions and involved in AD-related pathological activities. However, DHCR24 as a potential therapeutic target for AD remains to be identified. In present study, we demonstrated the role of DHCR24 in AD by employing delivery of adeno-associated virus carrying DHCR24 gene into the hippocampus of 5xFAD mice. Here, we found that 5xFAD mice had lower levels of cholesterol and DHCR24 expression, and the cholesterol loss was alleviated by DHCR24 overexpression. Surprisingly, the cognitive impairment of 5xFAD mice was significantly reversed after DHCR24-based gene therapy. Moreover, we revealed that DHCR24 knock-in successfully prevented or reversed AD-related pathology in 5xFAD mice, including amyloid-β deposition, synaptic injuries, autophagy, reactive astrocytosis, microglial phagocytosis and apoptosis. In conclusion, our results firstly demonstrated that the potential value of DHCR24-mediated regulation of cellular cholesterol level as a promising treatment for AD.
Collapse
Affiliation(s)
- Wen-Bin Zhang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China
| | - Yue Huang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China
| | - Xiao-Rou Guo
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China
| | - Meng-Qi Zhang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China
| | - Xiang-Shan Yuan
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China.
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Heng-Bing Zu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China.
| |
Collapse
|
17
|
DHCR24, a Key Enzyme of Cholesterol Synthesis, Serves as a Marker Gene of the Mouse Adrenal Gland Inner Cortex. Int J Mol Sci 2023; 24:ijms24020933. [PMID: 36674444 PMCID: PMC9867314 DOI: 10.3390/ijms24020933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Steroid hormones are synthesized through enzymatic reactions using cholesterol as the substrate. In steroidogenic cells, the required cholesterol for steroidogenesis can be obtained from blood circulation or synthesized de novo from acetate. One of the key enzymes that control cholesterol synthesis is 24-dehydrocholesterol reductase (encoded by DHCR24). In humans and rats, DHCR24 is highly expressed in the adrenal gland, especially in the zona fasciculata. We recently reported that DHCR24 was expressed in the mouse adrenal gland's inner cortex and also found that thyroid hormone treatment significantly upregulated the expression of Dhcr24 in the mouse adrenal gland. In the present study, we showed the cellular expression of DHCR24 in mouse adrenal glands in early postnatal stages. We found that the expression pattern of DHCR24 was similar to the X-zone marker gene 20αHSD in most developmental stages. This finding indicates that most steroidogenic adrenocortical cells in the mouse adrenal gland do not synthesize cholesterol locally. Unlike the 20αHSD-positive X-zone regresses during pregnancy, some DHCR24-positive cells remain present in parous females. Conditional knockout mice showed that the removal of Dhcr24 in steroidogenic cells did not affect the overall development of the adrenal gland or the secretion of corticosterone under acute stress. Whether DHCR24 plays a role in conditions where a continuous high amount of corticosterone production is needed requires further investigation.
Collapse
|
18
|
Circular RNA hsa_circ_0012673 Promotes Breast Cancer Progression via miR-576-3p/SOX4 Axis. Mol Biotechnol 2023; 65:61-71. [PMID: 35794450 DOI: 10.1007/s12033-022-00524-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 06/14/2022] [Indexed: 01/11/2023]
Abstract
Circular RNAs (circRNAs) have been reported to exert critical roles in human cancers. In this study, we investigated the role and molecular mechanism of hsa_circ_0012673 in breast cancer. Herein, we found that the expression of hsa_circ_0012673 was upregulated in breast cancer tissues and cell lines. Knockdown of hsa_circ_0012673 using RNA interference technique suppressed the proliferation, migration, and invasion of breast cancer cells. Mechanistically, hsa_circ_0012673 sponged miR-576-3p to stabilize SRY-box transcription factor 4 (SOX4), and thereby facilitating breast cancer cell proliferation, migration and invasion. Collectively, our study identified the oncogenic properties of hsa_circ_0012673/miR-576-3p/SOX4 axis in breast cancer, providing potential and exploitable diagnostic and therapeutic molecules for breast cancer.
Collapse
|
19
|
Augmentation of 3β-hydroxysteroid-Δ24 Reductase (DHCR24) Expression Induced by Bovine Viral Diarrhea Virus Infection Facilitates Viral Replication via Promoting Cholesterol Synthesis. J Virol 2022; 96:e0149222. [PMID: 36468862 PMCID: PMC9769396 DOI: 10.1128/jvi.01492-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV) is the etiologic agent of bovine viral diarrhea-mucosal disease, one of the most important viral diseases of cattle, leading to numerous losses to the cattle rearing industry worldwide. The pathogenicity of BVDV is extremely complex, and many underlying mechanisms involved in BVDV-host interactions are poorly understood, especially how BVDV utilizes host metabolism pathway for efficient viral replication and spread. In our previous study, using an integrative analysis of transcriptomics and proteomics, we found that DHCR24 (3β-hydroxysteroid-Δ24 reductase), a key enzyme in regulating cholesterol synthesis, was significantly upregulated at both gene and protein levels in the BVDV-infected bovine cells, indicating that cholesterol is important for BVDV replication. In the present study, the effects of DHCR24-mediated cholesterol synthesis on BVDV replication was explored. Our results showed that overexpression of the DHCR24 effectively promoted cholesterol synthesis, as well as BVDV replication, while acute cholesterol depletion in the bovine cells by treating cells with methyl-β-cyclodextrin (MβCD) obviously inhibited BVDV replication. In addition, knockdown of DHCR24 (gene silencing with siRNA targeting DHCR24, siDHCR24) or chemical inhibition (treating bovine cells with U18666A, an inhibitor of DHCR24 activity and cholesterol synthesis) significantly suppressed BVDV replication, whereas supplementation with exogenous cholesterol to the siDHCR24-transfected or U18666A-treated bovine cells remarkably restored viral replication. We further confirmed that BVDV nonstructural protein NS5A contributed to the augmentation of DHCR24 expression. Conclusively, augmentation of the DHCR24 induced by BVDV infection plays an important role in BVDV replication via promoting cholesterol production. IMPORTANCE Bovine viral diarrhea virus (BVDV), an important pathogen of cattle, is the causative agent of bovine viral diarrhea-mucosal disease, which causes extensive economic losses in both cow- and beef-rearing industry worldwide. The molecular interactions between BVDV and its host are extremely complex. In our previous study, we found that an essential host factor 3β-hydroxysteroid-δ24 reductase (DHCR24), a key enzyme involved in cholesterol synthesis, was significantly upregulated at both gene and protein levels in BVDV-infected bovine cells. Here, we experimentally explored the function of the DHCR24-mediated cholesterol synthesis in regulating BVDV replication. We elucidated that the augmentation of the DHCR24 induced by BVDV infection played a significant role in viral replication via promoting cholesterol synthesis. Our data provide evidence that BVDV utilizes a host metabolism pathway to facilitate its replication and spread.
Collapse
|
20
|
Li H, Shen J, Ma S, Zhao F, Zhao W, Chen F, Fu Y, Li B, Cheng J, Deng Y. TGF-β1 suppresses de novo cholesterol biosynthesis in granulosa-lutein cells by down-regulating DHCR24 expression via the GSK-3β/EZH2/H3K27me3 signaling pathway. Int J Biol Macromol 2022; 224:1118-1128. [DOI: 10.1016/j.ijbiomac.2022.10.196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022]
|
21
|
Mai M, Guo X, Huang Y, Zhang W, Xu Y, Zhang Y, Bai X, Wu J, Zu H. DHCR24 Knockdown Induces Tau Hyperphosphorylation at Thr181, Ser199, Ser262, and Ser396 Sites via Activation of the Lipid Raft-Dependent Ras/MEK/ERK Signaling Pathway in C8D1A Astrocytes. Mol Neurobiol 2022; 59:5856-5873. [PMID: 35804281 PMCID: PMC9395500 DOI: 10.1007/s12035-022-02945-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 06/28/2022] [Indexed: 02/01/2023]
Abstract
The synthetase 3β-hydroxysterol-Δ24 reductase (DHCR24) is a key regulator involved in cholesterol synthesis and homeostasis. A growing body of evidence indicates that DHCR24 is downregulated in the brain of various models of Alzheimer's disease (AD), such as astrocytes isolated from AD mice. For the past decades, astrocytic tau pathology has been found in AD patients, while the origin of phosphorylated tau in astrocytes remains unknown. A previous study suggests that downregulation of DHCR24 is associated with neuronal tau hyperphosphorylation. Herein, the present study is to explore whether DHCR24 deficiency can also affect tau phosphorylation in astrocytes. Here, we showed that DHCR24 knockdown could induce tau hyperphosphorylation at Thr181, Ser199, Thr231, Ser262, and Ser396 sites in C8D1A astrocytes. Meanwhile, we found that DHCR24-silencing cells had reduced the level of free cholesterol in the plasma membrane and intracellular organelles, as well as cholesterol esters. Furthermore, reduced cellular cholesterol level caused a decreased level of the caveolae-associated protein, cavin1, which disrupted lipid rafts/caveolae and activated rafts/caveolae-dependent Ras/MEK/ERK signaling pathway. In contrast, overexpression of DHCR24 prevented the overactivation of Ras/MEK/ERK signaling by increasing cellular cholesterol content, therefore decreasing tau hyperphosphorylation in C8D1A astrocytes. Herein, we firstly found that DHCR24 knockdown can lead to tau hyperphosphorylation in the astrocyte itself by activating lipid raft-dependent Ras/MEK/ERK signaling, which might contribute to the pathogenesis of AD and other degenerative tauopathies.
Collapse
Affiliation(s)
- Meiting Mai
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Xiaorou Guo
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Yue Huang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Wenbin Zhang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Yixuan Xu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Ying Zhang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Xiaojing Bai
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Junfeng Wu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| |
Collapse
|
22
|
Pflugfelder A, Yong XLH, Jagirdar K, Eigentler TK, Soyer HP, Sturm RA, Flatz L, Duffy DL. Genome-Wide Association Study Suggests the Variant rs7551288*A within the DHCR24 Gene Is Associated with Poor Overall Survival in Melanoma Patients. Cancers (Basel) 2022; 14:cancers14102410. [PMID: 35626014 PMCID: PMC9139953 DOI: 10.3390/cancers14102410] [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: 02/28/2022] [Revised: 04/27/2022] [Accepted: 05/10/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary The aim of this work was to investigate prognostic genetic factors in melanoma patients. Phenotypic and disease data as well as biomaterial were collected after informed consent from patients followed up in a Skin Cancer Center of a University clinic. Genome-wide analysis (GWAS) was performed with survival data of 556 melanoma patients and genetic data including more than 300,000 common polymorphisms. The SNP rs7551288 reached suggestive genome-wide significance (p = 2 × 10−6). This intronic variant of the DHCR24 gene is involved in the cholesterol synthesis pathway. Further analyses and a literature review suggest an important role of this locus for the clinical course of disease in melanoma patients. Abstract Melanoma incidence rates are high among individuals with fair skin and multiple naevi. Established prognostic factors are tumour specific, and less is known about prognostic host factors. A total of 556 stage I to stage IV melanoma patients from Germany with phenotypic and disease-specific data were analysed; 64 of these patients died of melanoma after a median follow-up time of 8 years. Germline DNA was assessed by the HumanCoreExome BeadChip and data of 356,384 common polymorphisms distributed over all 23 chromosomes were used for a genome-wide analysis. A suggestive genome-wide significant association of the intronic allele rs7551288*A with diminished melanoma-specific survival was detected (p = 2 × 10−6). The frequency of rs7551288*A was 0.43 and was not associated with melanoma risk, hair and eye colour, tanning and total naevus count. Cox regression multivariate analyses revealed a 5.31-fold increased risk of melanoma-specific death for patients with the rs7551288 A/A genotype, independent of tumour thickness, ulceration and stage of disease at diagnoses. The variant rs7551288 belongs to the DHCR24 gene, which encodes Seladin-1, an enzyme involved in the biosynthesis of cholesterol. Further investigations are needed to confirm this genetic variant as a novel prognostic biomarker and to explore whether specific treatment strategies for melanoma patients might be derived from it.
Collapse
Affiliation(s)
- Annette Pflugfelder
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Center of Dermatooncology, Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany;
- Correspondence:
| | - Xuan Ling Hilary Yong
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, QLD 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kasturee Jagirdar
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Biochemistry and Molecular Biology Department, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Thomas K. Eigentler
- Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10177 Berlin, Germany;
| | - H. Peter Soyer
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Department of Dermatology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | - Richard A. Sturm
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
| | - Lukas Flatz
- Center of Dermatooncology, Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany;
| | - David L. Duffy
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Genetic Epidemiology, QIMR Berghofer Institute of Medical Research, Herston, QLD 4006, Australia
| |
Collapse
|
23
|
Knapp B, Roedig J, Roedig H, Krzysko J, Horn N, Güler BE, Kusuluri DK, Yildirim A, Boldt K, Ueffing M, Liebscher I, Wolfrum U. Affinity Proteomics Identifies Interaction Partners and Defines Novel Insights into the Function of the Adhesion GPCR VLGR1/ADGRV1. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103108. [PMID: 35630584 PMCID: PMC9146371 DOI: 10.3390/molecules27103108] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/20/2022]
Abstract
The very large G-protein-coupled receptor 1 (VLGR1/ADGRV1) is the largest member of the adhesion G-protein-coupled receptor (ADGR) family. Mutations in VLGR1/ADGRV1 cause human Usher syndrome (USH), a form of hereditary deaf-blindness, and have been additionally linked to epilepsy. In the absence of tangible knowledge of the molecular function and signaling of VLGR1, the pathomechanisms underlying the development of these diseases are still unknown. Our study aimed to identify novel, previously unknown protein networks associated with VLGR1 in order to describe new functional cellular modules of this receptor. Using affinity proteomics, we have identified numerous new potential binding partners and ligands of VLGR1. Tandem affinity purification hits were functionally grouped based on their Gene Ontology terms and associated with functional cellular modules indicative of functions of VLGR1 in transcriptional regulation, splicing, cell cycle regulation, ciliogenesis, cell adhesion, neuronal development, and retinal maintenance. In addition, we validated the identified protein interactions and pathways in vitro and in situ. Our data provided new insights into possible functions of VLGR1, related to the development of USH and epilepsy, and also suggest a possible role in the development of other neuronal diseases such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Barbara Knapp
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Jens Roedig
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Heiko Roedig
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Jacek Krzysko
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Nicola Horn
- Core Facility for Medical Bioanalytics, Institute for Ophthalmic Research, University of Tuebingen, 72076 Tuebingen, Germany; (N.H.); (K.B.); (M.U.)
| | - Baran E. Güler
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Deva Krupakar Kusuluri
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Adem Yildirim
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Karsten Boldt
- Core Facility for Medical Bioanalytics, Institute for Ophthalmic Research, University of Tuebingen, 72076 Tuebingen, Germany; (N.H.); (K.B.); (M.U.)
| | - Marius Ueffing
- Core Facility for Medical Bioanalytics, Institute for Ophthalmic Research, University of Tuebingen, 72076 Tuebingen, Germany; (N.H.); (K.B.); (M.U.)
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany;
| | - Uwe Wolfrum
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
- Correspondence:
| |
Collapse
|
24
|
D’Angiolini S, Chiricosta L, Mazzon E. Sphingolipid Metabolism as a New Predictive Target Correlated with Aging and AD: A Transcriptomic Analysis. Medicina (B Aires) 2022; 58:medicina58040493. [PMID: 35454332 PMCID: PMC9025366 DOI: 10.3390/medicina58040493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background and objectives: Alzheimer’s disease (AD) is the most common form of dementia characterized by memory loss and executive dysfunction. To date, no markers can effectively predict the onset of AD and an early diagnosis is increasingly necessary. Age represents an important risk factor for the disease but it is not known whether it is the trigger event. Materials and Methods: We downloaded transcriptomic data related to post-mortem brain of thirty samples gathered as young without AD (Young), old without AD (Old), and old suffering from AD (OAD) groups. Results: Our results showed that steroid biosynthesis was enriched and associated with aging, while sphingolipid metabolism was related to both aging and AD. Specifically, sphingolipid metabolism is involved in the deregulation of CERS2, UGT8, and PLPP2. These genes are downregulated in Young and Old groups as compared with upregulated between Old and OAD groups. Moreover, the analysis of the interaction networks revealed that GABAergic synapse and Hippo signaling pathways were altered in AD condition along with mitochondrial metabolism and RNA processing. Conclusions: Observing the particular trend of genes related to sphingolipid metabolism that are downregulated during normal aging and start to be upregulated with the onset of AD, we suppose that sphingolipids could be early markers for the disease.
Collapse
|
25
|
Bai X, Mai M, Yao K, Zhang M, Huang Y, Zhang W, Guo X, Xu Y, Zhang Y, Qurban A, Duan L, Bu J, Zhang J, Wu J, Zhao Y, Yuan X, Zu H. The role of DHCR24 in the pathogenesis of AD: re-cognition of the relationship between cholesterol and AD pathogenesis. Acta Neuropathol Commun 2022; 10:35. [PMID: 35296367 PMCID: PMC8925223 DOI: 10.1186/s40478-022-01338-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/27/2022] [Indexed: 02/01/2023] Open
Abstract
Previous studies show that 3β-hydroxysterol-Δ24 reductase (DHCR24) has a remarked decline in the brain of AD patients. In brain cholesterol synthetic metabolism, DHCR24 is known as the heavily key synthetase in cholesterol synthesis. Moreover, mutations of DHCR24 gene result in inhibition of the enzymatic activity of DHCR24, causing brain cholesterol deficiency and desmosterol accumulation. Furthermore, in vitro studies also demonstrated that DHCR24 knockdown lead to the inhibition of cholesterol synthesis, and the decrease of plasma membrane cholesterol and intracellular cholesterol level. Obviously, DHCR24 could play a crucial role in maintaining cholesterol homeostasis via the control of cholesterol synthesis. Over the past two decades, accumulating data suggests that DHCR24 activity is downregulated by major risk factors for AD, suggesting a potential link between DHCR24 downregulation and AD pathogenesis. Thus, the brain cholesterol loss seems to be induced by the major risk factors for AD, suggesting a possible causative link between brain cholesterol loss and AD. According to previous data and our study, we further found that the reduced cholesterol level in plasma membrane and intracellular compartments by the deficiency of DHCR24 activity obviously was involved in β-amyloid generation, tau hyperphosphorylation, apoptosis. Importantly, increasing evidences reveal that the brain cholesterol loss and lipid raft disorganization are obviously linked to neuropathological impairments which are associated with AD pathogenesis. Therefore, based on previous data and research on DHCR24, we suppose that the brain cholesterol deficiency/loss might be involved in the pathogenesis of AD.
Collapse
|
26
|
Yao M, Li F, Xu L, Ma L, Zhang S. 24-Dehydrocholesterol Reductase alleviates oxidative damage-induced apoptosis in alveolar epithelial cells via regulating Phosphatidylinositol-3-Kinase/Protein Kinase B activation. Bioengineered 2021; 13:155-163. [PMID: 34949154 PMCID: PMC8805900 DOI: 10.1080/21655979.2021.2011634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Apoptosis of alveolar epithelial cells during acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a critical pathological event that seriously endangers the health of patients. Suppressing apoptosis of alveolar epithelial cells was shown to alleviate functional damage of lung, and modulator of the reactive oxygen species (ROS)-induced apoptosis becomes a promising approach to the ALI therapy. Previous little studies showed that DHCR24 possessed anti-oxidative and anti-apoptotic property in ALI. Thus, H2O2 was utilized to mimic oxidative damage in vitro in alveolar epithelial cell line A549 in the present study. Our results exhibited that H2O2 treatment of A549 cells reduced the level of SOD and increased the level of ROS. Moreover, H2O2 inhibited Bcl-2 expression in A549 cells, but increased Bax and the activity of Caspase-3. In addition, the apoptosis rate in the H2O2 treatment group also increased significantly. And the expression of 24-dehydrocholesterol reductase (DHCR24) was markedly reduced in the H2O2 treatment group. Overexpression of DHCR24 can remarkably inhibit H2O2-induced oxidative stress and apoptosis. We found that overexpression of DHCR24 increased the phosphorylation level of PI3K and AKT, however, the PI3K inhibitor LY294002 (LY) eliminated the protective effect of DHCR24 in ALI. DHCR24 was down-regulated in H2O2-induced ALI, and overexpression of DHCR24 could inhibit H2O2-induced oxidative stress and apoptosis of A549 cells by activating the Phosphatidylinositol-3-Kinase/Protein Kinase B (PI3K/AKT) signaling pathway. The above exhibited a protective effect of DHCR24 on alveolar epithelial cells exposed to oxidative stress-mediated apoptosis and provided a novel therapeutic method for ALI.
Collapse
Affiliation(s)
- Ming Yao
- Department of Critical Care Medicine, Wuhan Puren Hospital, Wuhan, China
| | - Feng Li
- Department of Critical Care Medicine, Wuhan Puren Hospital, Wuhan, China
| | - Liang Xu
- Department of Critical Care Medicine, Wuhan Puren Hospital, Wuhan, China
| | - Li Ma
- Department of Cardiovascular Medical, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Shutong Zhang
- Department of Medical Image, The Central Hospital of Wuhan, Wuhan, China
| |
Collapse
|
27
|
Wang N, Gong Z, Wang J, Xu W, Yang Q, Chen S. Characterization of Chinese tongue sole (Cynoglossus semilaevis) 24-dehydrocholesterol reductase: Expression profile, epigenetic modification, and its knock-down effect. Gen Comp Endocrinol 2021; 312:113870. [PMID: 34324841 DOI: 10.1016/j.ygcen.2021.113870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 11/27/2022]
Abstract
The sexual size dimorphism of the Chinese tongue sole (Cynoglossus semilaevis) has greatly obstructed its sustainable development; however, the underlying mechanism remains unclear. Based on C. semilaevis transcriptomic information, 24-dehydrocholesterol reductase (dhcr24) was identified in steroid biosynthesis, showing female-liver-biased expression. Dhcr24 has been reported to participate in various processes, such as cholesterol synthesis, oxidative stress response, neuroprotection, and cell survival. The present study assessed its role in the sexual size dimorphism in fish. First, detailed expression pattern analysis showed that dhcr24 mRNAs were extensively expressed in tissues and the highest levels were found in the liver and gonads of females. Analysis of the dhcr24 promoter region demonstrated different DNA methylation statuses in female, male, and pseudomale gonads with higher epigenetic modification in males. The confirmation of transcription activity of the dhcr24 promoter and putative transcription factors (e.g., ER, AR, SREBP, and POU1F1a) provides the foundation for studying its regulatory mechanism. Finally, dhcr24-siRNA mediated knock-down assay using C. semilaevis liver cells showed that steroid biosynthesis related genes (e.g., ebp, dhcr7, and sc5d), core component of PI3K/Akt pathway (e.g., pi3k), and igf1r exhibited different expression patterns. Further investigation on the interplay between steroid hormones, dhcr24, PI3K/Akt, and IGF-1 systems will be valuable to better understand the mechanism underlying the sexual size dimorphism in C. semilaevis.
Collapse
Affiliation(s)
- Na Wang
- Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Shandong Key Laboratory of Marine Fisheries Biotechnology and Genetic Breeding, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China.
| | - Zhihong Gong
- Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Jialin Wang
- Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Wenteng Xu
- Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Shandong Key Laboratory of Marine Fisheries Biotechnology and Genetic Breeding, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Qian Yang
- Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Songlin Chen
- Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Shandong Key Laboratory of Marine Fisheries Biotechnology and Genetic Breeding, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China.
| |
Collapse
|
28
|
Zhang H, Zhu W, Niu T, Wang Z, An K, Cao W, Shi J, Wang S. Inverted U-shaped correlation between serum low-density lipoprotein cholesterol levels and cognitive functions of patients with type 2 diabetes mellitus. Lipids Health Dis 2021; 20:103. [PMID: 34511118 PMCID: PMC8436464 DOI: 10.1186/s12944-021-01534-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL-C) metabolic disorder is common in individuals with diabetes. The role of LDL-C in mild cognitive impairment (MCI) remains to be explored. We aim to investigate the associations between LDL-C at different levels and details of cognition decline in patients with type 2 diabetes mellitus (T2DM). METHODS Patients with T2DM (n = 497) were recruited. Clinical parameters and neuropsychological tests were compared between patients with MCI and controls. Goodness of fit was assessed to determine the linear or U-shaped relationship between LDL-C and cognitive function. The cut-off point of LDL-C was calculated. Correlation and regression were carried out to explore the relationship between cognitive dysfunction and LDL-C levels above and below the cut-off point. RESULTS Although no significant difference in LDL-C levels was detected in 235 patients with MCI, compared with 262 patients without MCI, inverted-U-shaped association was determined between LDL-C and Montreal Cognitive Assessment (MoCA). The cut-off point of LDL-C is 2.686 mmol/l. LDL-C (>2.686 mmol/l) is positively related to Trail Making Test B (TMTB) indicating executive function. LDL-C (<2.686 mmol/l) is positively associated with Clock Drawing Test (CDT) reflecting visual space function in patients with T2DM. CONCLUSION Inverted U-shaped correlation was found between serum LDL-C and cognitive function in patients with T2DM. Despite that the mechanisms of different LDL-C levels involved in special cognitive dysfunctions remain incompletely clarified, excessive LDL-C damages executive function, while the deficient LDL-C impairs visual space function. TRIAL REGISTRATION ChiCTR-OCC-15006060 .
Collapse
Affiliation(s)
- Haoqiang Zhang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Wenwen Zhu
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Tong Niu
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Zheng Wang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Ke An
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Wuyou Cao
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Jijing Shi
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Shaohua Wang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China. .,School of Medicine, Southeast University, Nanjing, 210009, PR China.
| |
Collapse
|
29
|
Identification and Characterization of 24-Dehydrocholesterol Reductase (DHCR24) in the Two-Spotted Cricket, Gryllus bimaculatus. INSECTS 2021; 12:insects12090782. [PMID: 34564222 PMCID: PMC8471071 DOI: 10.3390/insects12090782] [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: 07/29/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022]
Abstract
Simple Summary DHCR24 (24-dehydrocholesterol reductase) is a key enzyme for producing cholesterol from desmosterol and that is also involved in the conversion of plant sterols to cholesterol in most plant-feeding insects. This study extensively examined the possibility of DHCR24 involved in the sterol conversion in omnivorous insects, which feed on multiple food origins. Homologs of DHCR24 (GbDHCR24-1 and -2) were identified and characterized by using the two-spotted cricket, Gryllus bimaculatus, as an experimental model. The quantitative expression analyses and RNA interference experiments revealed that GbDHCR24-1 rather than GbDHCR24-2 facilitates the desmosterol-to-cholesterol conversion in crickets. Our data suggested that the omnivorous species produced cholesterol from desmosterol in the same manner as the plant-feeding species do. Abstract Arthropods, including insects, convert sterols into cholesterol due to the inability to synthesise cholesterol de novo. 24-dehydrocholesterol reductase (DHCR24) plays an important role in the conversion. Not only involving the cholesterol biosynthesis in vertebrates, DHCR24 is required for the conversion of desmosterol into cholesterol in phytophagous insects. The current study extensively examined DHCR24 in omnivorous insects, which feed on both plants and animals, using Gryllus bimaculatus as the experimental model. We identified cDNAs encoding two homologues of DHCR24 from G. bimaculatus, which were designated as GbDHCR24-1 and GbDHCR24-2. Both homologues contained the flavin adenine dinucleotide binding domain, which is a feature of DHCR24. Quantitative polymerase chain reaction revealed that among tissues of adult crickets, fat body and anterior midgut expressed high levels of GbDHCR24s. Both fat body and anterior midgut demonstrated DHCR24 activities in which one of the functions is the conversion of desmosterol into cholesterol in vitro. Knockdown of GbDHCR24-1 significantly reduced the conversion activity in the anterior midgut while knockdown of the GbDHCR24-2 did not. Additionally, the accumulation of desmosterol was detected in a feeding experiment with a specific DHCR24 inhibitor, azacosterol. We finally concluded that GbDHCR24-1 is the major enzyme that facilitates the desmosterol-to-cholesterol-conversion in crickets.
Collapse
|
30
|
Uddin MS, Yu WS, Lim LW. Exploring ER stress response in cellular aging and neuroinflammation in Alzheimer's disease. Ageing Res Rev 2021; 70:101417. [PMID: 34339860 DOI: 10.1016/j.arr.2021.101417] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
One evident hallmark of Alzheimer's disease (AD) is the irregular accumulation of proteins due to changes in proteostasis involving endoplasmic reticulum (ER) stress. To alleviate ER stress and reinstate proteostasis, cells undergo an integrated signaling cascade called the unfolded protein response (UPR) that reduces the number of misfolded proteins and inhibits abnormal protein accumulation. Aging is associated with changes in the expression of ER chaperones and folding enzymes, leading to the impairment of proteostasis, and accumulation of misfolded proteins. The disrupted initiation of UPR prevents the elimination of unfolded proteins, leading to ER stress. In AD, the accumulation of misfolded proteins caused by sustained cellular stress leads to neurodegeneration and neuronal death. Current research has revealed that ER stress can trigger an inflammatory response through diverse transducers of UPR. Although the involvement of a neuroinflammatory component in AD has been documented for decades, whether it is a contributing factor or part of the neurodegenerative events is so far unknown. Besides, a feedback loop occurs between neuroinflammation and ER stress, which is strongly associated with neurodegenerative processes in AD. In this review, we focus on the current research on ER stress and UPR in cellular aging and neuroinflammatory processes, leading to memory impairment and synapse dysfunction in AD.
Collapse
|
31
|
Valproic acid suppresses cuprizone-induced hippocampal demyelination and anxiety-like behavior by promoting cholesterol biosynthesis. Neurobiol Dis 2021; 158:105489. [PMID: 34461265 DOI: 10.1016/j.nbd.2021.105489] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 11/21/2022] Open
Abstract
Myelin consists of several layers of tightly compacted membranes that form an insulating sheath around axons. These membranes are highly enriched in cholesterol, which is essential for the myelination process. Proper myelination is crucial for various neurophysiological functions while demyelination may cause CNS disease, such as multiple sclerosis (MS). Recent studies demonstrated that demyelination occurs not only in the white matter but also in the grey matter, such as the hippocampus, which may cause cognitive deficits and mental disorders. Valproic acid (VPA) is an anticonvulsant agent prescribed for the treatment of epilepsy and seizure. Recently, VPA was reported to alter cholesterol metabolism in neural cells, suggesting that it may play an important role in myelin biogenesis. Here in this study, we found significant demyelination in the hippocampus of the mouse cuprizone model, which is accompanied by reduced cholesterol biosynthesis and increased anxiety-like behavior. VPA treatment, however, suppressed cuprizone-induced hippocampal demyelination and anxiety-like behavior by promoting cholesterol biosynthesis. These data identify an important role of VPA in the hippocampal demyelination process and the hippocampal demyelination-related behavior deficit via regulation of cholesterol biosynthesis, which provides new insights into the mechanisms of VPA as a protective agent against CNS demyelination.
Collapse
|
32
|
Sliz E, Shin J, Syme C, Patel Y, Parker N, Richer L, Gaudet D, Bennett S, Paus T, Pausova Z. A variant near DHCR24 associates with microstructural properties of white matter and peripheral lipid metabolism in adolescents. Mol Psychiatry 2021; 26:3795-3805. [PMID: 31900429 PMCID: PMC7332371 DOI: 10.1038/s41380-019-0640-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/01/2019] [Accepted: 12/12/2019] [Indexed: 12/17/2022]
Abstract
Visceral adiposity has been associated with altered microstructural properties of white matter in adolescents. Previous evidence suggests that circulating phospholipid PC(16:0/2:0) may mediate this association. To investigate the underlying biology, we performed a genome-wide association study (GWAS) of the shared variance of visceral fat, PC(16:0/2:0), and white matter microstructure in 872 adolescents from the Saguenay Youth Study. We further studied the metabolomic profile of the GWAS-lead variant in 931 adolescents. Visceral fat and white matter microstructure were assessed with magnetic resonance imaging. Circulating metabolites were quantified with serum lipidomics and metabolomics. We identified a genome-wide significant association near DHCR24 (Seladin-1) encoding a cholesterol-synthesizing enzyme (rs588709, p = 3.6 × 10-8); rs588709 was also associated nominally with each of the three traits (white matter microstructure: p = 2.1 × 10-6, PC(16:0/2:0): p = 0.005, visceral fat: p = 0.010). We found that the metabolic profile associated with rs588709 resembled that of a TM6SF2 variant impacting very low-density lipoprotein (VLDL) secretion and was only partially similar to that of a HMGCR variant. This suggests that the effect of rs588709 on VLDL lipids may arise due to altered phospholipid rather than cholesterol metabolism. The rs588709 was also nominally associated with circulating concentrations of omega-3 fatty acids in interaction with visceral fat and PC(16:0/2:0), and these fatty acid measures showed robust associations with white matter microstructure. Overall, the present study provides evidence that the DHCR24 locus may link peripheral metabolism to brain microstructure, an association with implications for cognitive impairment.
Collapse
Affiliation(s)
- Eeva Sliz
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Center for Life-Course Health Research and Computational Medicine, Faculty of Medicine, University of Oulu, and Biocenter Oulu, Oulu, Finland
| | - Jean Shin
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Catriona Syme
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Yash Patel
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Nadine Parker
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Louis Richer
- Department of Health Sciences, Université du Québec à Chicoutimi, Chicoutimi, QC, Canada
| | - Daniel Gaudet
- Clinical Lipidology and rare lipid disorders Unit, Community Genetic Medicine Center, Department of Medicine, Université de Montréal, ECOGENE-21, Chicoutimi, QC, Canada
| | - Steffany Bennett
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Tomas Paus
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
33
|
Yavuz U, Alaylıoğlu M, Şengül B, Karras SN, Gezen-Ak D, Dursun E. Protein disulfide isomerase A3 might be involved in the regulation of 24-dehydrocholesterol reductase via vitamin D equilibrium in primary cortical neurons. In Vitro Cell Dev Biol Anim 2021; 57:704-714. [PMID: 34338991 DOI: 10.1007/s11626-021-00602-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/11/2021] [Indexed: 11/28/2022]
Abstract
Vitamin D is a secosteroid hormone mediating its functions via vitamin D receptor (VDR) and an endoplasmic reticulum chaperone, protein disulfide isomerase A3 (PDIA3). From a physiological perspective, there is also a well-established association of cholesterol and vitamin D synthesis, since both share a common metabolic substrate, 7 dehydrocholesterol (7-DHC). Yet, the potential basic pathways, of the biological interplay of DHCR24 and vitamin D equilibrium, on neuronal level, are yet to be determined. In this study, we aimed to investigate the relation between vitamin D pathways and DHCR24 in primary cortical neuron cultures. The neocortex of Sprague-Dawley rat embryos (E16) was used for the preparation of primary cortical neuron cultures. DHCR24 mRNA and protein expression levels were determined by qRT-PCR, Western blotting, and immunofluorescent labeling in 1,25-dihydroxyvitamin D3-treated or VDR/PDIA3-silenced primary cortical neurons. The mRNA expression of DHCR24 was significantly decreased in the cortical neurons treated with 10-8M 1,25-dihydroxyvitamin D3 (p<0.001). In parallel with the mRNA results, DHCR24 protein expression in cortical neurons treated with 10-8M 1,25-dihydroxyvitamin D3 was also significantly lower than untreated neurons (p<0.05). These data were also confirmed with immunofluorescent labeling and fluorescence intensity measurements of DHCR24 (p<0.001). Finally, DHCR24 mRNA expression level was significantly increased in PDIA3 siRNA-treated neurons (p<0.05). Similar to the mRNA results, the DHCR24 protein expression of PDIA3 siRNA-treated neurons was also statistically higher than the other groups (p<0.05). Results of this mechanistic experimental basic study demonstrate that DHCR24 mRNA expression and protein concentrations attenuated in response to vitamin D treatment. Furthermore, we observed that PDIA3 might be involved in this modulatory effect. Our findings indicate a complex interaction of DHCR24 and vitamin D equilibrium, through the involvement of PDIA3 and vitamin D in the modulation of cholesterol metabolism in neuronal cells, requiring future studies on the field.
Collapse
Affiliation(s)
- Ulaş Yavuz
- Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Merve Alaylıoğlu
- Brain and Neurodegenerative Disorders Research Laboratory, Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Büşra Şengül
- Brain and Neurodegenerative Disorders Research Laboratory, Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | | | - Duygu Gezen-Ak
- Brain and Neurodegenerative Disorders Research Laboratory, Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Erdinç Dursun
- Brain and Neurodegenerative Disorders Research Laboratory, Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
- Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| |
Collapse
|
34
|
Abnormal brain cholesterol homeostasis in Alzheimer's disease-a targeted metabolomic and transcriptomic study. NPJ Aging Mech Dis 2021; 7:11. [PMID: 34075056 PMCID: PMC8169871 DOI: 10.1038/s41514-021-00064-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 03/18/2021] [Indexed: 02/02/2023] Open
Abstract
The role of brain cholesterol metabolism in Alzheimer's disease (AD) remains unclear. Peripheral and brain cholesterol levels are largely independent due to the impermeability of the blood brain barrier (BBB), highlighting the importance of studying the role of brain cholesterol homeostasis in AD. We first tested whether metabolite markers of brain cholesterol biosynthesis and catabolism were altered in AD and associated with AD pathology using linear mixed-effects models in two brain autopsy samples from the Baltimore Longitudinal Study of Aging (BLSA) and the Religious Orders Study (ROS). We next tested whether genetic regulators of brain cholesterol biosynthesis and catabolism were altered in AD using the ANOVA test in publicly available brain tissue transcriptomic datasets. Finally, using regional brain transcriptomic data, we performed genome-scale metabolic network modeling to assess alterations in cholesterol biosynthesis and catabolism reactions in AD. We show that AD is associated with pervasive abnormalities in cholesterol biosynthesis and catabolism. Using transcriptomic data from Parkinson's disease (PD) brain tissue samples, we found that gene expression alterations identified in AD were not observed in PD, suggesting that these changes may be specific to AD. Our results suggest that reduced de novo cholesterol biosynthesis may occur in response to impaired enzymatic cholesterol catabolism and efflux to maintain brain cholesterol levels in AD. This is accompanied by the accumulation of nonenzymatically generated cytotoxic oxysterols. Our results set the stage for experimental studies to address whether abnormalities in cholesterol metabolism are plausible therapeutic targets in AD.
Collapse
|
35
|
Stamatakos S, Beretta GL, Vergani E, Dugo M, Corno C, Corna E, Tinelli S, Frigerio S, Ciusani E, Rodolfo M, Perego P, Gatti L. Deregulated FASN Expression in BRAF Inhibitor-Resistant Melanoma Cells Unveils New Targets for Drug Combinations. Cancers (Basel) 2021; 13:cancers13092284. [PMID: 34068792 PMCID: PMC8126202 DOI: 10.3390/cancers13092284] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Metabolic changes promoting cell survival are involved in metastatic melanoma progression and in the development of drug resistance. In BRAF-inhibitor resistant melanoma cells, we explored the role of FASN, an enzyme involved in lipogenesis overexpressed in metastatic melanoma. Resistant melanoma cells displaying enhanced migratory and pro-invasive abilities increased sensitivity to the BRAF inhibitor PLX4032 upon the molecular targeting of FASN and upon treatment with the FASN inhibitor orlistat. This behavior was associated with a marked apoptosis and caspase 3/7 activation observed for the drug combination. The expression of FASN was found to be inversely associated with drug resistance in BRAF-mutant cell lines, both in a set of six resistant/sensitive matched lines and in the Cancer Cell Line Encyclopedia. A favorable drug interaction in resistant cells was also observed with U18666 A inhibiting DHCR24, which increased upon FASN targeting. The simultaneous combination of the two inhibitors showed a synergistic interaction with PLX4032 in resistant cells. In conclusion, FASN plays a role in BRAF-mutated melanoma progression, thereby creating novel therapeutic opportunities for the treatment of melanoma.
Collapse
Affiliation(s)
- Serena Stamatakos
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Giovanni Luca Beretta
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Elisabetta Vergani
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (E.V.); (S.F.)
| | - Matteo Dugo
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Cristina Corno
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Elisabetta Corna
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Stella Tinelli
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Simona Frigerio
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (E.V.); (S.F.)
| | - Emilio Ciusani
- Department of Diagnostic and Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Monica Rodolfo
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (E.V.); (S.F.)
- Correspondence:
| | - Paola Perego
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Laura Gatti
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| |
Collapse
|
36
|
Bai X, Wu J, Zhang M, Xu Y, Duan L, Yao K, Zhang J, Bo J, Zhao Y, Xu G, Zu H. DHCR24 Knock-Down Induced Tau Hyperphosphorylation at Thr181, Ser199, Thr231, Ser262, Ser396 Epitopes and Inhibition of Autophagy by Overactivation of GSK3β/mTOR Signaling. Front Aging Neurosci 2021; 13:513605. [PMID: 33967735 PMCID: PMC8098657 DOI: 10.3389/fnagi.2021.513605] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/26/2021] [Indexed: 02/01/2023] Open
Abstract
Accumulating evidences supported that knock-down of DHCR24 is linked to the pathological risk factors of AD, suggesting a potential role of DHCR24 in AD pathogenesis. However, the molecular mechanism link between DHCR24 and tauopathy remains unknown. Here, in order to elucidate the relationship between DHCR24 and tauopathy, we will focus on the effect of DHCR24 on the tau hyperphosphorylation at some toxic sites. In present study, we found that DHCR24 knock-down significantly lead to the hyperphosphorylation of tau sites at Thr181, Ser199, Thr231, Ser262, Ser396. Moreover, DHCR24 knock-down also increase the accumulation of p62 protein, simultaneously decreased the ratio of LC3-II/LC3-I and the number of autophagosome compared to the control groups, suggesting the inhibition of autophagy activity. In contrast, DHCR24 knock-in obviously abolished the effect of DHCR24 knock-down on tau hyperphosphrylation and autophagy. In addition, to elucidate the association between DHCR24 and tauopathy, we further showed that the level of plasma membrane cholesterol, lipid raft-anchored protein caveolin-1, and concomitantly total I class PI3-K (p110α), phospho-Akt (Thr308 and Ser473) were significantly decreased, resulting in the disruption of lipid raft/caveola and inhibition of PI3-K/Akt signaling in silencing DHCR24 SH-SY5Y cells compared to control groups. At the same time, DHCR24 knock-down simultaneously decreased the level of phosphorylated GSK3β at Ser9 (inactive form) and increased the level of phosphorylated mTOR at Ser2448 (active form), leading to overactivation of GSK3β and mTOR signaling. On the contrary, DHCR24 knock-in largely increased the level of membrane cholesterol and caveolin-1, suggesting the enhancement of lipid raft/caveola. And synchronously DHCR24 knock-in also abolished the effect of DHCR24 knock-down on the inhibition of PI3-K/Akt signaling as well as the overactivation of GSK3β and mTOR signaling. Collectively, our data strongly supported DHCR24 knock-down lead to tau hyperphosphorylation and the inhibition of autophagy by a lipid raft-dependent PI3-K/Akt-mediated GSK3β and mTOR signaling. Taking together, our results firstly demonstrated that the decrease of plasma membrane cholesterol mediated by DHCR24 deficiency might contribute to the tauopathy in AD and other tauopathies.
Collapse
Affiliation(s)
- Xiaojing Bai
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Junfeng Wu
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Mengqi Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yixuan Xu
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lijie Duan
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Kai Yao
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jianfeng Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jimei Bo
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yongfei Zhao
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Guoxiong Xu
- The Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Qi Z, Zhang Y, Yao K, Zhang M, Xu Y, Zhang J, Bai X, Zu H. DHCR24 Knockdown Lead to Hyperphosphorylation of Tau at Thr181, Thr231, Ser262, Ser396, and Ser422 Sites by Membrane Lipid-Raft Dependent PP2A Signaling in SH-SY5Y Cells. Neurochem Res 2021; 46:1627-1640. [PMID: 33710538 DOI: 10.1007/s11064-021-03273-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 11/27/2022]
Abstract
Accumulating data suggest that the downregulation of DHCR24 is linked to the pathological risk factors of AD, denoting a potential role of DHCR24 in AD pathogenesis. However, it remains unclear whether the downregulation of DHCR24 affects the abnormal heper-phosphorylation of tau protein, which is involved in tauopathy. In present papers, immunofluorescence and Filipin III fluorescence results showed that DHCR24 knockdown significantly lowered the level of plasma membrane cholesterol and expression level of membrane lipid-raft structural protein caveolin-1; and overexpression of DHCR24 could increase the plasma membrane cholesterol levels and facilitating caveolae structure through increase the expression of caveolin-1. PP2A is the key phosphatase involving in tau phosphorylation, which is localized in cholesterol-dependent caveola/raft lipid domains. Here, the PP2A activity was detected by western blot assay. Interestingly, the level of p-PP2Ac at Y307 (inactive) and p-GSK3β at Y216 (active) in the downstream of the PP2A signal pathway were both significantly increased in silencing DHCR24 SH-SY5Y cells, which denoted an inhibition of the PP2A and activation of GSK3β signaling. Conversely, overexpression of DHCR24 blunted the inhibition effect of PP2A and activation of GSK3β. Besides, in the SH-SY5Y cell lines we demonstrated that DHCR24 knockdown obviously induced hyperphosphorylation of tau at Thr181, Thr231, Ser262, Ser396, and Ser422 Sites. In contrast, DHCR24 overexpression protects neuronal SH-SY5Y cells against the hyperphosphorylation of tau at Thr181, Thr231, Ser262, Ser396, and Ser422 Sites. Furthermore, PP2A activator D-erythro-Sphingosine (DES) also obviously inhibited the hyperphosphorylation of tau induced by DHCR24 knockdown. Collectively, our findings firstly confirmed that DHCR24 knockdown obviously induced abnormal hyperphosphorylation of tau by a novel lipid raft-dependent PP2A signaling. We propose that DHCR24 downregulation led to altered cholesterol synthesis as a potential mechanism in the progression of tau hyperphosphorylation involving in AD and other tauopathies.
Collapse
Affiliation(s)
- Zihan Qi
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Ying Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Kai Yao
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Mengqi Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yixuan Xu
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Jianfeng Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Xiaojing Bai
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
38
|
Walker RM, Vaher K, Bermingham ML, Morris SW, Bretherick AD, Zeng Y, Rawlik K, Amador C, Campbell A, Haley CS, Hayward C, Porteous DJ, McIntosh AM, Marioni RE, Evans KL. Identification of epigenome-wide DNA methylation differences between carriers of APOE ε4 and APOE ε2 alleles. Genome Med 2021; 13:1. [PMID: 33397400 PMCID: PMC7784364 DOI: 10.1186/s13073-020-00808-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 11/12/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late onset Alzheimer's disease, whilst the ε2 allele confers protection. Previous studies report differential DNA methylation of APOE between ε4 and ε2 carriers, but associations with epigenome-wide methylation have not previously been characterised. METHODS Using the EPIC array, we investigated epigenome-wide differences in whole blood DNA methylation patterns between Alzheimer's disease-free APOE ε4 (n = 2469) and ε2 (n = 1118) carriers from the two largest single-cohort DNA methylation samples profiled to date. Using a discovery, replication and meta-analysis study design, methylation differences were identified using epigenome-wide association analysis and differentially methylated region (DMR) approaches. Results were explored using pathway and methylation quantitative trait loci (meQTL) analyses. RESULTS We obtained replicated evidence for DNA methylation differences in a ~ 169 kb region, which encompasses part of APOE and several upstream genes. Meta-analytic approaches identified DNA methylation differences outside of APOE: differentially methylated positions were identified in DHCR24, LDLR and ABCG1 (2.59 × 10-100 ≤ P ≤ 2.44 × 10-8) and DMRs were identified in SREBF2 and LDLR (1.63 × 10-4 ≤ P ≤ 3.01 × 10-2). Pathway and meQTL analyses implicated lipid-related processes and high-density lipoprotein cholesterol was identified as a partial mediator of the methylation differences in ABCG1 and DHCR24. CONCLUSIONS APOE ε4 vs. ε2 carrier status is associated with epigenome-wide methylation differences in the blood. The loci identified are located in trans as well as cis to APOE and implicate genes involved in lipid homeostasis.
Collapse
Affiliation(s)
- Rosie M. Walker
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
- Present Address: Centre for Clinical Brain Sciences, Chancellor’s Building, 49 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4SB UK
| | - Kadi Vaher
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
- Present Address: MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ UK
| | - Mairead L. Bermingham
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Stewart W. Morris
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Andrew D. Bretherick
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Yanni Zeng
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
- Present address: Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080 China
| | - Konrad Rawlik
- Division of Genetics and Genomics, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, UK
| | - Carmen Amador
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Archie Campbell
- Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Chris S. Haley
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - David J. Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
- Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Andrew M. McIntosh
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF UK
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Kathryn L. Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| |
Collapse
|
39
|
Marbouti L, Zahmatkesh M, Riahi E, Shafiee Sabet M. GnRH protective effects against amyloid β-induced cognitive decline: A potential role of the 17β-estradiol. Mol Cell Endocrinol 2020; 518:110985. [PMID: 32805333 DOI: 10.1016/j.mce.2020.110985] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/04/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The 17β-estradiol (E2) enhances hippocampal dendritic spine synapses, facilitates learning processes, and exerts neuroprotection. Brain estrogen decline has been reported in Alzheimer's disease. The role of GnRH in modulating steroid biosynthesis convinced us to examine whether hippocampal GnRH administration could enhance the local E2 levels and overcome the development of cognition decline in amyloid β (Aβ) neurotoxicity. To explore if GnRH acts through regulating E2 synthesis, letrozole, an aromatase inhibitor, has been applied in combination with GnRH. METHODS Female rats received an intracerebroventricular injection of Aβ. The GnRH and, or letrozole were injected into the CA1 for 14 consecutive days. Working memory, novel object recognition memory, and anxiety-like behavior were evaluated. Serum and hippocampal E2 levels were measured. Hippocampal mRNA expression of GnRH (GnRH-R) and E2 (ERα and ERβ) receptors was assessed. GnRH effect on the excitability of pyramidal cells was studied by in vivo single-unit recording. RESULTS GnRH increased hippocampal E2 levels, evoked an increase in the spontaneous firing of pyramidal neurons, and caused mRNA overexpression of hippocampal GnRH receptors. GnRH prevented the adverse effects of Aβ on working memory, NOR index, and anxiogenic behavior. Letrozole did not reverse GnRH modulatory effects on hippocampal E2 levels and neuroprotection. CONCLUSION GnRH prevented the Aβ-induced memory deficit, which may be mediated through hippocampal E2 levels enhancement. The electrophysiological analysis revealed the enhanced neuronal excitability in the CA1 region. All these data suggest that GnRH might be a promising candidate that reduces anxiety and improves memory indices in the context of Aβ neurotoxicity.
Collapse
Affiliation(s)
- Ladan Marbouti
- Neuroscience and Addiction Studies Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Zahmatkesh
- Neuroscience and Addiction Studies Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Cognitive and Behavioral Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Esmail Riahi
- Physiology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Shafiee Sabet
- Family Medicine Department, Ziaeian Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Önmez A, Alpay M, Torun S, Şahin IE, Öneç K, Değirmenci Y. Serum seladin-1 levels in diabetes mellitus and Alzheimer's disease patients. Acta Neurol Belg 2020; 120:1399-1404. [PMID: 32506354 DOI: 10.1007/s13760-020-01393-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Diabetes mellitus (DM) has been shown to increase the risk of Alzheimer's disease (AD). Downregulation of selective Alzheimer's disease indicator-1 (seladin-1) occurs in the cerebral regions affected by AD. However, inconsistent results have been reported for the relationship between seladin-1 levels and AD. The effect of DM on serum seladin-1 levels in AD is unknown. The present study is aimed to investigate serum seladin-1 levels in diabetic and non-diabetic patients with AD. METHODS Forty-six patients with AD and 25 healthy volunteers over 65 years of age were included in this study. The patients were divided into three groups-those with AD only, those with DM and AD, and control groups. Demographic characteristics and serum seladin-1 levels were compared among the groups. RESULTS There was no statistically significant difference in seladin-1 levels in the AD only group compared to the control group (p = 0.376). However, seladin-1 levels were significantly lower in the DM and AD group compared to the AD only and control groups (p = 0.002, p = 0.001; respectively). Negative correlations were present between seladin-1 and fasting glucose, postprandial glucose, HbA1c, and insulin (p < 0.05; all). CONCLUSION Decreased serum seladin-1 values in the presence of DM and inverse correlations with diabetic parameters in patients with AD, together with a non-significant difference from the control group, suggests that seladin-1 may be altered only in the presence of DM in patients with AD. Lower serum seladin-1 levels may also play a role in the pathogenesis of AD in patients with DM.
Collapse
Affiliation(s)
- Attila Önmez
- Department of Internal Medicine, Duzce University Medical Faculty, Duzce, Turkey.
| | - Merve Alpay
- Department of Biochemistry, Duzce University Medical Faculty, Duzce, Turkey
| | - Serkan Torun
- Department of Internal Medicine, Duzce University Medical Faculty, Duzce, Turkey
| | - I Ethem Şahin
- Department of Biochemistry, Duzce University Medical Faculty, Duzce, Turkey
| | - Kürşad Öneç
- Department of Internal Medicine, Duzce University Medical Faculty, Duzce, Turkey
| | - Yıldız Değirmenci
- Department of Neurology, Duzce University Medical Faculty, Duzce, Turkey
| |
Collapse
|
41
|
Yang A, Alrosan AZ, Sharpe LJ, Brown AJ, Callaghan R, Gelissen IC. Regulation of ABCG4 transporter expression by sterols and LXR ligands. Biochim Biophys Acta Gen Subj 2020; 1865:129769. [PMID: 33141061 DOI: 10.1016/j.bbagen.2020.129769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/01/2020] [Accepted: 10/19/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Oxysterols, which are derivatives of cholesterol produced by enzymic or non-enzymic pathways, are potent regulators of cellular lipid homeostasis. Sterol homeostasis in the brain is an important area of interest with regards to neurodegenerative conditions like Alzheimer's disease (AD). Brain cells including neurons and astrocytes express sterol transporters belonging to the ABC transporter family of proteins, including ABCA1, ABCG1 and ABCG4, and these transporters are considered of interest as therapeutic targets. Although regulation of ABCA1 and ABCG1 is well established, regulation of ABCG4 is still controversial, in particular whether the transporter is an Liver X receptor (LXR) target. ABCG4 is thought to transport cholesterol, oxysterols and cholesterol synthesis intermediates, and was recently found on the blood brain barrier (BBB), implicated in amyloid-beta export. In this study, we investigate the regulation of ABCG4 by oxysterols, cholesterol-synthesis intermediates and cholesterol itself. METHODS ABC transporter expression was measured in neuroblastoma and gliablastoma cell lines and cells overexpressing ABCG4 in response to synthetic LXR ligands, oxysterols and cholesterol-synthesis intermediates. RESULTS In contrast to previous reports, ABCG4 expression was induced by a synthetic LXR ligand in U87-MG astrocytes but not in neuroblastoma and BBB endothelial cell lines. In addition, ABCG4 protein was stabilized by cholesterol as was previously shown for ABCG1. ABCG4 protein was furthermore stabilized by cholesterol-synthesis intermediates, desmosterol, lathosterol and lanosterol. CONCLUSIONS These results identify new aspects of the post-translational control of ABCG4 that warrant further exploration into the role of this transporter in the maintenance of sterol homeostasis in the brain.
Collapse
Affiliation(s)
- Alryel Yang
- Sydney Pharmacy School, Faculty of Medicine and Health, Pharmacy Bank Building A15, Science Road, The University of Sydney, Sydney, NSW 2006, Australia
| | - Amjad Z Alrosan
- Sydney Pharmacy School, Faculty of Medicine and Health, Pharmacy Bank Building A15, Science Road, The University of Sydney, Sydney, NSW 2006, Australia
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, Chancellery Walk, The University of New South Wales, Kensington, NSW 2033, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, Chancellery Walk, The University of New South Wales, Kensington, NSW 2033, Australia
| | - Richard Callaghan
- Research School of Biology and Medical School, Linnaeus Way, Australian National University, ACT 2600, Australia
| | - Ingrid C Gelissen
- Sydney Pharmacy School, Faculty of Medicine and Health, Pharmacy Bank Building A15, Science Road, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
42
|
ÖNMEZ A, EŞBAH O, ŞAHİN İE. Investigation of serum Seladin-1 / DHCR24 levels in breast cancer patients. KONURALP TIP DERGISI 2020. [DOI: 10.18521/ktd.785524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
43
|
Zu HB, Liu XY, Yao K. DHCR24 overexpression modulates microglia polarization and inflammatory response via Akt/GSK3β signaling in Aβ 25-35 treated BV-2 cells. Life Sci 2020; 260:118470. [PMID: 32950573 DOI: 10.1016/j.lfs.2020.118470] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 11/30/2022]
Abstract
Microglial phenotypic polarization, divided into pro-inflammatory "M1" phenotype and anti-inflammatory "M2" phenotype, played a crucial role in the pathogenesis of Alzheimer's disease (AD). Facilitating microglial polarization from M1 to M2 phenotype was shown to alleviate AD-associate pathologic damage, and modulator of the microglial phenotype has become a promising therapeutic approach for the treatment of AD. Previous little evidence showed that DHCR24 (3-β-hydroxysteroid-Δ-24-reductase), also known as seladin-1 (selective Alzheimer's disease indicator-1), exerted potential anti-inflammatory property, however, the link between DHCR24 and microglial polarization has never been reported. Thus, the role of DHCR24 in microglial polarization in amyloid-beta 25-35 (Aβ25-35) treated BV-2 cells was evaluated in this study. Our results demonstrated that Aβ25-35 aggravated inflammatory response and facilitated the transition of microglia phenotype from M2 to M1 in BV-2 cells, by upregulating M1 marker (i-NOS, IL-1β and TNF-α) and downregulating M2 marker (arginase-1, IL-4 and TGF-β). DHCR24 overexpression by lentivirus transfection could significantly reverse these effects, meanwhile, activated Akt/GSK3β signaling pathway via increasing the protein expression of P-Akt and P-GSK3β. Furthermore, when co-treated with Akt inhibitor MK2206, the effect of DHCR24 was obviously reversed. The study exhibited the neuroprotective function of DHCR24 in AD-related inflammatory injury and provided a novel therapeutic target for AD in the future.
Collapse
Affiliation(s)
- Heng-Bing Zu
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Xin-Ying Liu
- Department of Endoscopy, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Kai Yao
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China.
| |
Collapse
|
44
|
Nikpour M, Sharafi A, Hamidi M, Andalib S. Effect of Colloidal Aqueous Solution of Fullerene (C60) in the Presence of a P-Glycoprotein Inhibitor (Verapamil) on Spatial Memory and Hippocampal Expression of Sirtuin6, SELADIN1, and AQP1 Genes in a Rat Model of Alzheimer's Disease. ACS Chem Neurosci 2020; 11:2549-2565. [PMID: 32631043 DOI: 10.1021/acschemneuro.0c00213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common types of neurodegenerative diseases which is accompanied by irreversible neuronal damage, learning difficulties, memory impairments, and cognitive disorders. The cholinergic system is destroyed during AD pathogenesis, leading to the major symptoms of the disease. Although in severe stages AD is life threatening, to date no absolute treatment has been found for this illness and some palliative options are available. The aim of this study was to investigate the effect of fullerene (C60) aqueous suspension (FAS) on improving spatial memory in amnesic male Wistar rats (weighing 200 ± 20 g) and to further compare the results with that of donepezil (DNPZL) as a standard drug. FAS was prepared via a solvent exchange method. The particle size was in the 119.14 ± 3.38 nm range with polydispersity index of 0.15 ± 0.02 and zeta potential of -12.22 ± 5.98 mV. A simple and high sensitive reversed phase high performance liquid chromatography (HPLC) method was developed to identify the C60 concentration in FAS (21 μg/mL). Efficiencies of drugs were examined in both pretreatment and post-treatment groups of animals to better understand how they participate in affecting AD symptoms. Seeing that previous studies have presented antithetical declarations about whether C60 is a P-glycoprotein (P-gp) substrate, we studied FAS effects in both conditions of the presence and absence of a P-gp inhibitor (verapamil HCl, 25 mg/kg). In order to clarify the molecular mechanisms of action of two drugs, their effects on the expression of three principal genes involved in AD, including Sirtuin6, SELADIN1, and AQP1, and as well as their total antioxidant capacities (TACs) were studied. In order to induce memory impairment, scopolamine HBr (SCOP) was administered for 10 days (2 mg/kg/i.p.). FAS and DNPZL administration regimens were 21 μg/mL, BID (i.p.) and 10 mg/kg (p.o.) for 10 days, respectively. Our results introduce FAS as a promising nanoformulation for improving AD symptoms, especially memory impairment, and further assert that more studies are needed to elucidate C60 and P-gp interaction type.
Collapse
Affiliation(s)
- Mehrnoosh Nikpour
- Department of Pharmaceutics, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Sharafi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehrdad Hamidi
- Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Sina Andalib
- Department of Pharmacology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
45
|
Baranowska M, Suliborska K, Todorovic V, Kusznierewicz B, Chrzanowski W, Sobajic S, Bartoszek A. Interactions between bioactive components determine antioxidant, cytotoxic and nutrigenomic activity of cocoa powder extract. Free Radic Biol Med 2020; 154:48-61. [PMID: 32360591 DOI: 10.1016/j.freeradbiomed.2020.04.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/25/2022]
Abstract
Numerous studies have shown, rather disappointingly, that isolated bioactive phytochemicals are not as biologically effective as natural plant products. Such a discrepancy may be explained by the concept of food synergy, which was verified in this research for cocoa extract versus its major components with regard to cancer chemoprevention. The evaluation embraced the relationship between redox properties evaluated in cell-free systems with the aid of free radicals scavenging method and differential pulse voltammetry, and redox associated anticarcinogenic activities (cellular antioxidant activity, cytotoxicity, nutrigenomic activity) in human colon adenocarcinoma cell line exposed to either cocoa powder extract or artificial mixtures of cocoa bioactives at matching concentrations. In contrast to expectations, our results showed that the stepwise enrichment with antioxidants caused no gradual increase in the antioxidant activity of the model mixtures; also, these model mixtures did not reach the reducing potential of cocoa in the cell-free systems or cellular model employed. Further, the biological activities examined in colon adenocarcinoma cells did not alter in a stepwise manner that could reflect the gradual changes in composition of bioactive ingredients. In conclusion, the experiments presented here showed that the growing complexity of a mixture of phytochemicals seems to create a new redox bioactive substance rather than enrich the mixture with new activities, characteristic of the compound added. It follows that no simple, predictable relationship can be expected between the chemopreventive potential and the composition of real food items containing a complicated set of non-toxic redox active ingredients. Our observations suggest that the interactions between different bioactive compounds and food matrix components are cooperating factors determining the final bioactivity of foods.
Collapse
Affiliation(s)
- Monika Baranowska
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland.
| | - Klaudia Suliborska
- Department of Physical Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Vanja Todorovic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Barbara Kusznierewicz
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Wojciech Chrzanowski
- Department of Physical Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Sladjana Sobajic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Agnieszka Bartoszek
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| |
Collapse
|
46
|
Kulas JA, Weigel TK, Ferris HA. Insulin resistance and impaired lipid metabolism as a potential link between diabetes and Alzheimer's disease. Drug Dev Res 2020; 81:194-205. [PMID: 32022298 DOI: 10.1002/ddr.21643] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 12/20/2019] [Accepted: 01/23/2020] [Indexed: 12/13/2022]
Abstract
Diabetes disrupts organs throughout the body including the brain. Evidence suggests diabetes is a risk factor for Alzheimer's disease (AD) and neurodegeneration. In this review, we focus on understanding how diabetes contributes to the progression of neurodegeneration by influencing several aspects of the disease process. We emphasize the potential roles of brain insulin resistance, as well as cholesterol and lipid disruption, as factors which worsen AD.
Collapse
Affiliation(s)
- Joshua A Kulas
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, Virginia
| | - Thaddeus K Weigel
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Heather A Ferris
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, Virginia.,Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
47
|
Mahakizadeh S, Mokhtari T, Navaee F, Poorhassan M, Tajik A, Hassanzadeh G. Effects of chronic hypoxia on the expression of seladin-1/Tuj1 and the number of dark neurons of hippocampus. J Chem Neuroanat 2020; 104:101744. [PMID: 31926979 DOI: 10.1016/j.jchemneu.2020.101744] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/23/2019] [Accepted: 01/08/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND There are evidences showing the relation between chronic hypoxia and Alzheimer's disease (AD) as a metabolic neurodegenerative disease. This study was designed to evaluate the effects of chronic hypoxia on factors which characterized in AD to introduce a new model of AD-dementia. METHODS AND MATERIALS Twenty-four male rats were randomly divided in three groups: Control group (Co), Sham group (Sh), Hypoxia induction group (Hx, exposed to hypoxic chamber [oxygen 8% and nitrogen 92%] for 30 days, 4 h/day). Spatial learning and memory were analyzed using the Morris water maze task. At day 30 after hypoxia period, animals were sacrificed and serum was gathered for pro-inflammatory cytokines (interleukin-1β and tumor necrosis factor) measurements and brains were used for molecular and histopathological investigations. RESULTS According to behavioral studies, a significant impairment was seen in Hx group (P < 0.05). TNF-α and IL-1β showed a significant enhanced in Hx group comparing with Co group and Sh group (P < 0.05). As well, the gene expression of seladin-1, Tuj1 and the number of seladin-1+, Tuj1+neurons significantly decreased and also the mean number of dark neurons significantly increased in CA1 and CA3 regions of hippocampus. CONCLUSIONS In this study, a new model of AD was developed which showed the underlying mechanisms of AD and its relations with chronic hypoxia. Hypoxia for 30 days decreased seladin-1, Tuj1 expression, increased the number of dark neurons, and also induced memory impairment. These results indicated that chronic hypoxia mediated the dementia underlying AD and AD-related pathogenesis in rat.
Collapse
Affiliation(s)
- Simin Mahakizadeh
- Department of Anatomy, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Tahmineh Mokhtari
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Fatemeh Navaee
- Department of Anatomy, School of Medicine, Shahidbeheshti University of Medical Sciences, Tehran, Iran
| | - Mahnaz Poorhassan
- Department of Anatomy, School of Medicine, Shahidbeheshti University of Medical Sciences, Tehran, Iran
| | - Armin Tajik
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Li Y, Wang X, Yang B, Wang H, Ma Z, Lu Z, Lu X, Gao B. 3 β-Hydroxysteroid- Δ24 Reductase (DHCR24) Protects Pancreatic β Cells from Endoplasmic Reticulum Stress-Induced Apoptosis by Scavenging Excessive Intracellular Reactive Oxygen Species. J Diabetes Res 2020; 2020:3426902. [PMID: 32724824 PMCID: PMC7382746 DOI: 10.1155/2020/3426902] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/17/2020] [Indexed: 12/20/2022] Open
Abstract
There is accumulating evidence showing that apoptosis induced by endoplasmic reticulum (ER) stress plays a key role in pancreatic β cell dysfunction and insulin resistance. 3β-Hydroxysteroid-Δ24 Reductase (DHCR24) is a multifunctional enzyme located in the endoplasmic reticulum (ER), which has been previously shown to protect neuronal cells from ER stress-induced apoptosis. However, the role of DHCR24 in type 2 diabetes is only incompletely understood so far. In the present study, we induced ER stress by tunicamycin (TM) treatment and showed that infection of MIN6 cells with Ad-DHCR24-myc rendered these cells resistant to caspase-3-mediated apoptosis induced by TM, while cells transfected with siRNAs targeting DHCR24 were more sensitive to TM. Western blot analysis showed that TM treatment induced upregulation of Bip protein levels in both cells infected with Ad-LacZ (the control group) and Ad-DHCR24-myc, indicating substantial ER stress. Cells infected with Ad-LacZ exhibited a rapid and strong activation of ATF6 and p38, peaking at 3 h after TM exposure. Conversely, cells infected with Ad-DHCR24-myc showed a higher and more sustained activation of ATF6 and Bip than control cells. DHCR24 overexpression also inhibited the generation of intracellular reactive oxygen species (ROS) induced by ER stress and protected cells from apoptosis caused by treatment with both cholesterol and hydrogen peroxide. In summary, these data demonstrate, for the first time, that DHCR24 protects pancreatic β cells from apoptosis induced by ER stress.
Collapse
Affiliation(s)
- Yang Li
- Department of Biochemistry and Molecular Biology, Life Science School, Liaoning University, Shenyang 110036, China
| | - Xude Wang
- Department of Biochemistry and Molecular Biology, Life Science School, Liaoning University, Shenyang 110036, China
| | - Baoyu Yang
- Department of Biochemistry and Molecular Biology, Life Science School, Liaoning University, Shenyang 110036, China
| | - Haozhen Wang
- Department of Biochemistry and Molecular Biology, Life Science School, Liaoning University, Shenyang 110036, China
| | - Zhenzhong Ma
- Department of Biochemistry and Molecular Biology, Life Science School, Liaoning University, Shenyang 110036, China
| | - Ziyin Lu
- Department of Biochemistry and Molecular Biology, Life Science School, Liaoning University, Shenyang 110036, China
| | - Xiuli Lu
- Department of Biochemistry and Molecular Biology, Life Science School, Liaoning University, Shenyang 110036, China
| | - Bing Gao
- Department of Cell Biology and Genetics, Shenyang Medical College, Shenyang 110034, China
| |
Collapse
|
49
|
Kelicen-Ugur P, Cincioğlu-Palabıyık M, Çelik H, Karahan H. Interactions of Aromatase and Seladin-1: A Neurosteroidogenic and Gender Perspective. Transl Neurosci 2019; 10:264-279. [PMID: 31737354 PMCID: PMC6843488 DOI: 10.1515/tnsci-2019-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022] Open
Abstract
Aromatase and seladin-1 are enzymes that have major roles in estrogen synthesis and are important in both brain physiology and pathology. Aromatase is the key enzyme that catalyzes estrogen biosynthesis from androgen precursors and regulates the brain’s neurosteroidogenic activity. Seladin-1 is the enzyme that catalyzes the last step in the biosynthesis of cholesterol, the precursor of all hormones, from desmosterol. Studies indicated that seladin-1 is a downstream mediator of the neuroprotective activity of estrogen. Recently, we also showed that there is an interaction between aromatase and seladin-1 in the brain. Therefore, the expression of local brain aromatase and seladin-1 is important, as they produce neuroactive steroids in the brain for the protection of neuronal damage. Increasing steroid biosynthesis specifically in the central nervous system (CNS) without affecting peripheral hormone levels may be possible by manipulating brain-specific promoters of steroidogenic enzymes. This review emphasizes that local estrogen, rather than plasma estrogen, may be responsible for estrogens’ protective effects in the brain. Therefore, the roles of aromatase and seladin-1 and their interactions in neurodegenerative events such as Alzheimer’s disease (AD), ischemia/reperfusion injury (stroke), and epilepsy are also discussed in this review.
Collapse
Affiliation(s)
- Pelin Kelicen-Ugur
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Sıhhiye Ankara Turkey
| | - Mehtap Cincioğlu-Palabıyık
- Turkish Medicines and Medical Devices Agency (TITCK), Department of Regulatory Affairs, Division of Pharmacological Assessment, Ankara, Turkey
| | - Hande Çelik
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Sıhhiye Ankara Turkey
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
50
|
Tang KS. The cellular and molecular processes associated with scopolamine-induced memory deficit: A model of Alzheimer's biomarkers. Life Sci 2019; 233:116695. [DOI: 10.1016/j.lfs.2019.116695] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
|