1
|
Almutairi JA, Kidd EJ. Biological Sex Disparities in Alzheimer's Disease. Curr Top Behav Neurosci 2024. [PMID: 39485650 DOI: 10.1007/7854_2024_545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Alzheimer's disease is a highly complex and multifactorial neurodegenerative disorder, with age being the most significant risk factor. The incidence of Alzheimer's disease doubles every 5 years after the age of 65. Consequently, one of the major challenges in Alzheimer's disease research is understanding how the brain changes with age. Gaining insights into these changes could help identify individuals who are more prone to developing Alzheimer's disease as they age. Over the past 25 years, studies on brain aging have examined thousands of human brains to explore the neuronal basis of age-related cognitive decline. However, most of these studies have focused on adults over 60, often neglecting the critical menopause transition period. During menopause, women experience a substantial decline in ovarian sex hormone production, with a decrease of about 90% in estrogen levels. Estrogen is known for its neuroprotective effects, and its significant loss during menopause affects various biological systems, including the brain. Importantly, despite known differences in dementia risk between sexes, the impact of biological sex and sex hormones on brain aging and the development of Alzheimer's disease remains underexplored.
Collapse
Affiliation(s)
- Jawza A Almutairi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
- Department of Pharmaceutical Science, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Emma J Kidd
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
2
|
Dua I, Pearson AC, Lowman RL, Peshkin L, Yampolsky LY. Post-senescence reproductive rebound in Daphnia associated with reversal of age-related transcriptional changes. GeroScience 2024:10.1007/s11357-024-01401-y. [PMID: 39460850 DOI: 10.1007/s11357-024-01401-y] [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: 08/08/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
A long-lived species of zooplankton microcrustaceans, Daphnia magna, sometimes exhibits late-life rebound of reproduction, briefly reversing reproductive senescence. Such events are often interpreted as terminal investments in anticipation of imminent mortality. We demonstrate that such post-senescence reproductive events (PSREs) neither cause nor anticipate increased mortality. We analyze an RNAseq experiment comparing young, old reproductively senescent, and old PSRE Daphnia females. We first show that overall age-related transcriptional changes are dominated by the increased transcription of guanidine monophosphate synthases and guanylate cyclases, as well as two groups of presumed transposon-encoded proteins, and by a drop in transcription of protein synthesis-related genes. We then focus on gene families and functional groups in which full or partial reversal of age-related transcriptional changes occur. This analysis reveals a reversal, in the PSRE individuals, of age-related up-regulation of apolipoproteins D, lysosomal lipases, and peptidases as well as several proteins related to mitochondrial and muscle functions. While it is not certain which of these changes enable reproductive rejuvenation, and which are by-products of processes that lead to it, we present some evidence that post-senescence reproductive events are associated with the reversal of age-related protein and lipid aggregates removal and apoptosis.
Collapse
Affiliation(s)
- Ishaan Dua
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37614, USA
| | - A Catherine Pearson
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Rachael L Lowman
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Leonid Peshkin
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lev Y Yampolsky
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
3
|
Reitsema VA, Schreuder L, Gerrits E, Eggen BJL, Goris M, Laman JD, de Rooij SE, Wesseling EM, Bouma HR, Henning RH. Calorie restriction increases the sensitivity of progeroid Ercc1 Δ/- mice to acute (neuro)inflammation. GeroScience 2024:10.1007/s11357-024-01347-1. [PMID: 39287878 DOI: 10.1007/s11357-024-01347-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Hospitalized elderly patients frequently suffer from delirium, especially in the context of sepsis-associated encephalopathy. Current treatments of delirium are merely symptomatic. Calorie restriction (CR) is both a promising strategy to protect against sepsis and has beneficial effects on aging-induced neurodegeneration. In this study, we investigated whether six weeks of 30% CR had protective effects on lipopolysaccharide (LPS) induced (neuro)inflammation in wild-type (WT) and progeroid mice deficient in the DNA excision-repair gene Ercc1 (Ercc1Δ/-). While CR did not affect the LPS-induced inflammatory response in WT mice, CR exaggerated the peripheral inflammatory response in Ercc1Δ/- mice, as evidenced by an increase of pro-inflammatory serum cytokines (TNF-α, IL-1β, and IFN-γ) and kidney injury marker Ngal. Neuroinflammatory effects were assessed by RNA-sequencing of isolated microglia. Similarly, CR did not affect microglia gene expression in WT mice, but increased neuroinflammation-associated gene expression in Ercc1Δ/- mice. In conclusion, CR increases the peripheral and brain inflammatory response of Ercc1Δ/- mice to a systemic inflammatory stimulus.
Collapse
Affiliation(s)
- V A Reitsema
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - L Schreuder
- Department of Internal Medicine, University Center for Geriatric Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E Gerrits
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B J L Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M Goris
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J D Laman
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - S E de Rooij
- Department of Internal Medicine, University Center for Geriatric Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E M Wesseling
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - H R Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
- Department of Acute Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - R H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Zhou X, Azimi M, Handin N, Riselli A, Vora B, Chun E, Yee SW, Artursson P, Giacomini KM. Proteomic Profiling Reveals Age-Related Changes in Transporter Proteins in the Human Blood-Brain Barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.604313. [PMID: 39091855 PMCID: PMC11291171 DOI: 10.1101/2024.07.26.604313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The Blood-Brain Barrier (BBB) is a crucial, selective barrier that regulates the entry of molecules including nutrients, environmental toxins, and therapeutic medications into the brain. This function relies heavily on brain endothelial cell proteins, particularly transporters and tight junction proteins. The BBB continues to develop postnatally, adapting its selective barrier function across different developmental phases, and alters with aging and disease. Here we present a global proteomics analysis focused on the ontogeny and aging of proteins in human brain microvessels (BMVs), predominantly composed of brain endothelial cells. Our proteomic profiling quantified 6,223 proteins and revealed possible age-related alteration in BBB permeability due to basement membrane component changes through the early developmental stage and age-dependent changes in transporter expression. Notable changes in expression levels were observed with development and age in nutrient transporters and transporters that play critical roles in drug disposition. This research 1) provides important information on the mechanisms that drive changes in the metabolic content of the brain with age and 2) enables the creation of physiologically based pharmacokinetic models for CNS drug distribution across different life stages.
Collapse
Affiliation(s)
- Xujia Zhou
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Mina Azimi
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Niklas Handin
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Andrew Riselli
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Bianca Vora
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Eden Chun
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| |
Collapse
|
5
|
Pagani A, Duscher D, Kempa S, Ghods M, Prantl L. Preliminary Single-Cell RNA-Sequencing Analysis Uncovers Adipocyte Heterogeneity in Lipedema. Cells 2024; 13:1028. [PMID: 38920656 PMCID: PMC11201579 DOI: 10.3390/cells13121028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
Background: Despite its increasing incidence and prevalence throughout Western countries, lipedema continues to be a very enigmatic disease, often misunderstood or misdiagnosed by the medical community and with an intrinsic pathology that is difficult to trace. The nature of lipedemic tissue is one of hypertrophic adipocytes and poor tissue turnover. So far, there are no identified pathways responsible, and little is known about the cell populations of lipedemic fat. Methods: Adipose tissue samples were collected from affected areas of both lipedema and healthy participants. For single-cell RNA sequencing analysis, the samples were dissociated into single-cell suspensions using enzymatic digestion and then encapsulated into nanoliter-sized droplets containing barcoded beads. Within each droplet, cellular mRNA was converted into complementary DNA. Complementary DNA molecules were then amplified for downstream analysis. Results: The single-cell RNA-sequencing analysis revealed three distinct adipocyte populations at play in lipedema. These populations have unique gene signatures which can be characterized as a lipid generating adipocyte, a disease catalyst adipocyte, and a lipedemic adipocyte. Conclusions: The single-cell RNA sequencing of lipedemic tissue samples highlights a triad of distinct adipocyte subpopulations, each characterized by unique gene signatures and functional roles. The interplay between these adipocyte subtypes offers promising insights into the complex pathophysiology of lipedema.
Collapse
Affiliation(s)
- Andrea Pagani
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany
| | - Dominik Duscher
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany
| | - Sally Kempa
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany
| | - Mojtaba Ghods
- Department of Plastic, Aesthetic and Reconstructive Surgery, Clinic Ernst von Bergmann, Charlottenstraße 71, 14467 Potsdam, Germany
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
6
|
Zhang G, Huang C, Wang R, Guo J, Qin Y, Lv S. Chondroprotective effects of Apolipoprotein D in knee osteoarthritis mice through the PI3K/AKT/mTOR signaling pathway. Int Immunopharmacol 2024; 133:112005. [PMID: 38626543 DOI: 10.1016/j.intimp.2024.112005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND Because the pathophysiology of osteoarthritis (OA) has not been fully elucidated, targeted treatments are lacking. In this study, we assessed the role and underlying mechanism apolipoprotein D (APOD) on the development of OA. METHODS To establish an in vitro OA model, we extracted primary chondrocytes from the cartilage of C57BL/6 mice and stimulated the chondrocytes with IL-1β. After APOD intervention or incubation with an overexpressing plasmid, we detected inflammatory-related markers using RT-qPCR, Western blotting, and ELISA. To detect apoptosis and autophagy-related markers, we used flow cytometry, immunofluorescence, and transmission electron microscopy (TEM). Finally, we measured the level of oxidative stress. We also used RNA-seq to identify the APOD-regulated downstream signaling pathways. We used an in vivo mice OA model of the anterior cruciate ligament transection (ACLT) and administered intra-articular adenovirus overexpressing APOD. To examine cartilage damage severity, we used immunohistochemical analysis (IHC), micro-CT, scanning electron microscopy (SEM), and Safranin O-fast green staining. RESULTS Our results showed that APOD inhibited chondrocyte inflammation, degeneration, and apoptosis induced by IL-1β. Additionally, APOD reversed autophagy inhibition and oxidative stress and also blocked activation of the PI3K/AKT/mTOR signaling pathway induced by IL-1β. Finally, overexpression of the APOD gene through adenovirus was sufficient to mitigate OA progression. CONCLUSIONS Our findings revealed that APOD had a chondroprotective role in OA progression by the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; Department of Orthopedics, Harbin First Hospital, Harbin, Heilongjiang Province, China; Future Medical Laboratory of the Second Affiliated Hospital of Harbin Medical University, China
| | - Chao Huang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ren Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jiangrong Guo
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yong Qin
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - Songcen Lv
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
7
|
Lu JB, Ren PP, Li Q, He F, Xu ZT, Wang SN, Chen JP, Li JM, Zhang CX. The evolution and functional divergence of 10 Apolipoprotein D-like genes in Nilaparvata lugens. INSECT SCIENCE 2024; 31:91-105. [PMID: 37334667 DOI: 10.1111/1744-7917.13216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 04/16/2023] [Accepted: 04/22/2023] [Indexed: 06/20/2023]
Abstract
Apolipoprotein D (ApoD), a member of the lipocalin superfamily of proteins, is involved in lipid transport and stress resistance. Whereas only a single copy of the ApoD gene is found in humans and some other vertebrates, there are typically several ApoD-like genes in insects. To date, there have been relatively few studies that have examined the evolution and functional differentiation of ApoD-like genes in insects, particularly hemi-metabolous insects. In this study, we identified 10 ApoD-like genes (NlApoD1-10) with distinct spatiotemporal expression patterns in Nilaparvata lugens (BPH), which is an important pest of rice. NlApoD1-10 were found to be distributed on 3 chromosomes in a tandem array of NlApoD1/2, NlApoD3-5, and NlApoD7/8, and show sequence and gene structural divergence in the coding regions, indicating that multiple gene duplication events occurred during evolution. Phylogenetic analysis revealed that NlApoD1-10 can be clustered into 5 clades, with NlApoD3-5 and NlApoD7/8 potentially evolving exclusively in the Delphacidae family. Functional screening using an RNA interference approach revealed that only NlApoD2 was essential for BPH development and survival, whereas NlApoD4/5 are highly expressed in testes, and might play roles in reproduction. Moreover, stress response analysis revealed that NlApoD3-5/9, NlApoD3-5, and NlApoD9 were up-regulated after treatment with lipopolysaccharide, H2 O2 , and ultraviolet-C, respectively, indicating their potential roles in stress resistance.
Collapse
Affiliation(s)
- Jia-Bao Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, China
| | - Peng-Peng Ren
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, China
| | - Qiao Li
- Technology Center of Wuhan Customs District, Hubei, China
- Institute of Insect Science, Zhejiang University, Hangzhou, China
| | - Fang He
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, China
| | - Zhong-Tian Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, China
| | - Sai-Nan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, China
| | - Jian-Ping Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, China
| | - Jun-Min Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, China
| | - Chuan-Xi Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang Province, China
- Institute of Insect Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Rehman H, Ang TFA, Tao Q, Espenilla AL, Au R, Farrer LA, Zhang X, Qiu WQ. Comparison of Commonly Measured Plasma and Cerebrospinal Fluid Proteins and Their Significance for the Characterization of Cognitive Impairment Status. J Alzheimers Dis 2024; 97:621-633. [PMID: 38143358 DOI: 10.3233/jad-230837] [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: 12/26/2023]
Abstract
BACKGROUND Although cerebrospinal fluid (CSF) amyloid-β42 peptide (Aβ42) and phosphorylated tau (p-tau) and blood p-tau are valuable for differential diagnosis of Alzheimer's disease (AD) from cognitively normal (CN) there is a lack of validated biomarkers for mild cognitive impairment (MCI). OBJECTIVE This study sought to determine how plasma and CSF protein markers compared in the characterization of MCI and AD status. METHODS This cohort study included Alzheimer's Disease Neuroimaging Initiative (ADNI) participants who had baseline levels of 75 proteins measured commonly in plasma and CSF (257 total, 46 CN, 143 MCI, and 68 AD). Logistic regression, least absolute shrinkage and selection operator (LASSO) and Random Forest (RF) methods were used to identify the protein candidates for the disease classification. RESULTS We observed that six plasma proteins panel (APOE, AMBP, C3, IL16, IGFBP2, APOD) outperformed the seven CSF proteins panel (VEGFA, HGF, PRL, FABP3, FGF4, CD40, RETN) as well as AD markers (CSF p-tau and Aβ42) to distinguish the MCI from AD [area under the curve (AUC) = 0.75 (plasma proteins), AUC = 0.60 (CSF proteins) and AUC = 0.56 (CSF p-tau and Aβ42)]. Also, these six plasma proteins performed better than the CSF proteins and were in line with CSF p-tau and Aβ42 in differentiating CN versus MCI subjects [AUC = 0.89 (plasma proteins), AUC = 0.85 (CSF proteins) and AUC = 0.89 (CSF p-tau and Aβ42)]. These results were adjusted for age, sex, education, and APOEϵ4 genotype. CONCLUSIONS This study suggests that the combination of 6 plasma proteins can serve as an effective marker for differentiating MCI from AD and CN.
Collapse
Affiliation(s)
- Habbiburr Rehman
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ting Fang Alvin Ang
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Arielle Lauren Espenilla
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Rhoda Au
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
9
|
del Valle E, Rubio-Sardón N, Menéndez-Pérez C, Martínez-Pinilla E, Navarro A. Apolipoprotein D as a Potential Biomarker in Neuropsychiatric Disorders. Int J Mol Sci 2023; 24:15631. [PMID: 37958618 PMCID: PMC10650001 DOI: 10.3390/ijms242115631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Neuropsychiatric disorders (NDs) are a diverse group of pathologies, including schizophrenia or bipolar disorders, that directly affect the mental and physical health of those who suffer from them, with an incidence that is increasing worldwide. Most NDs result from a complex interaction of multiple genes and environmental factors such as stress or traumatic events, including the recent Coronavirus Disease (COVID-19) pandemic. In addition to diverse clinical presentations, these diseases are heterogeneous in their pathogenesis, brain regions affected, and clinical symptoms, making diagnosis difficult. Therefore, finding new biomarkers is essential for the detection, prognosis, response prediction, and development of new treatments for NDs. Among the most promising candidates is the apolipoprotein D (Apo D), a component of lipoproteins implicated in lipid metabolism. Evidence suggests an increase in Apo D expression in association with aging and in the presence of neuropathological processes. As a part of the cellular neuroprotective defense machinery against oxidative stress and inflammation, changes in Apo D levels have been demonstrated in neuropsychiatric conditions like schizophrenia (SZ) or bipolar disorders (BPD), not only in some brain areas but in corporal fluids, i.e., blood or serum of patients. What is not clear is whether variation in Apo D quantity could be used as an indicator to detect NDs and their progression. This review aims to provide an updated view of the clinical potential of Apo D as a possible biomarker for NDs.
Collapse
Affiliation(s)
- Eva del Valle
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Nuria Rubio-Sardón
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Carlota Menéndez-Pérez
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Eva Martínez-Pinilla
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Ana Navarro
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| |
Collapse
|
10
|
McMullen E, Hertenstein H, Strassburger K, Deharde L, Brankatschk M, Schirmeier S. Glycolytically impaired Drosophila glial cells fuel neural metabolism via β-oxidation. Nat Commun 2023; 14:2996. [PMID: 37225684 DOI: 10.1038/s41467-023-38813-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/17/2023] [Indexed: 05/26/2023] Open
Abstract
Neuronal function is highly energy demanding and thus requires efficient and constant metabolite delivery by glia. Drosophila glia are highly glycolytic and provide lactate to fuel neuronal metabolism. Flies are able to survive for several weeks in the absence of glial glycolysis. Here, we study how Drosophila glial cells maintain sufficient nutrient supply to neurons under conditions of impaired glycolysis. We show that glycolytically impaired glia rely on mitochondrial fatty acid breakdown and ketone body production to nourish neurons, suggesting that ketone bodies serve as an alternate neuronal fuel to prevent neurodegeneration. We show that in times of long-term starvation, glial degradation of absorbed fatty acids is essential to ensure survival of the fly. Further, we show that Drosophila glial cells act as a metabolic sensor and can induce mobilization of peripheral lipid stores to preserve brain metabolic homeostasis. Our study gives evidence of the importance of glial fatty acid degradation for brain function, and survival, under adverse conditions in Drosophila.
Collapse
Affiliation(s)
- Ellen McMullen
- Department of Molecular Biology and Genetics, University of South Bohemia, České Budějovice, Czech Republic
| | - Helen Hertenstein
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Katrin Strassburger
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Leon Deharde
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Marko Brankatschk
- Biotechnologisches Zentrum, Technische Universität Dresden, Dresden, Germany.
| | - Stefanie Schirmeier
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
11
|
First trimester serum biomarker discovery study for early onset, preterm onset and preeclampsia at term. Placenta 2022; 128:39-48. [DOI: 10.1016/j.placenta.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/05/2022] [Accepted: 08/21/2022] [Indexed: 11/20/2022]
|
12
|
Pan X. The Roles of Fatty Acids and Apolipoproteins in the Kidneys. Metabolites 2022; 12:metabo12050462. [PMID: 35629966 PMCID: PMC9145954 DOI: 10.3390/metabo12050462] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
The kidneys are organs that require energy from the metabolism of fatty acids and glucose; several studies have shown that the kidneys are metabolically active tissues with an estimated energy requirement similar to that of the heart. The kidneys may regulate the normal and pathological function of circulating lipids in the body, and their glomerular filtration barrier prevents large molecules or large lipoprotein particles from being filtered into pre-urine. Given the permeable nature of the kidneys, renal lipid metabolism plays an important role in affecting the rest of the body and the kidneys. Lipid metabolism in the kidneys is important because of the exchange of free fatty acids and apolipoproteins from the peripheral circulation. Apolipoproteins have important roles in the transport and metabolism of lipids within the glomeruli and renal tubules. Indeed, evidence indicates that apolipoproteins have multiple functions in regulating lipid import, transport, synthesis, storage, oxidation and export, and they are important for normal physiological function. Apolipoproteins are also risk factors for several renal diseases; for example, apolipoprotein L polymorphisms induce kidney diseases. Furthermore, renal apolipoprotein gene expression is substantially regulated under various physiological and disease conditions. This review is aimed at describing recent clinical and basic studies on the major roles and functions of apolipoproteins in the kidneys.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA;
- Diabetes and Obesity Research Center, NYU Langone Hospital—Long Island, Mineola, New York, NY 11501, USA
| |
Collapse
|
13
|
Becirovic-Agic M, Chalise U, Jung M, Rodriguez-Paar JR, Konfrst SR, Flynn ER, Salomon JD, Hall ME, Lindsey ML. Faster skin wound healing predicts survival after myocardial infarction. Am J Physiol Heart Circ Physiol 2022; 322:H537-H548. [PMID: 35089808 PMCID: PMC8917917 DOI: 10.1152/ajpheart.00612.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/06/2022] [Accepted: 01/24/2022] [Indexed: 12/20/2022]
Abstract
Both skin wound healing and the cardiac response to myocardial infarction (MI) progress through similar pathways involving inflammation, resolution, tissue repair, and scar formation. Due to the similarities, we hypothesized that the healing response to skin wounding would predict future response to MI. Mice were given a 3-mm skin wound using a disposable biopsy punch and the skin wound was imaged daily until closure. The same set of animals was given MI by permanent coronary artery ligation 28 days later and followed for 7 days. Cardiac physiology was measured by echocardiography at baseline and MI days 3 and 7. Animals that survived until day 7 were grouped as survivors, and animals that died from MI were grouped as nonsurvivors. Survivors had faster skin wound healing than nonsurvivors. Faster skin wound healing predicted MI survival better than commonly used cardiac functional variables (e.g., infarct size, fractional shortening, and end diastolic dimension). N-glycoproteome profiling of MI day 3 plasma revealed α2-macroglobulin and ELL-associated factor 1 as strong predictors of future MI death and progression to heart failure. A second cohort of MI mice validated these findings. To investigate the clinical relevance of α2-macroglobulin, we mapped the plasma glycoproteome in patients with MI 48 h after admission and in healthy controls. In patients, α2-macroglobulin was increased 48 h after MI. Apolipoprotein D, another plasma glycoprotein, detrimentally regulated both skin and cardiac wound healing in male but not female mice by promoting inflammation. Our results reveal that the skin is a mirror to the heart and common pathways link wound healing across organs.NEW & NOTEWORTHY Faster skin wound healers had more efficient cardiac healing after myocardial infarction (MI). Two plasma proteins at D3 MI, EAF1 and A2M, predicted MI death in 66% of cases. ApoD regulated both skin and cardiac wound healing in male mice by promoting inflammation. The skin was a mirror to the heart and common pathways linked wound healing across organs.
Collapse
Affiliation(s)
- Mediha Becirovic-Agic
- University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Upendra Chalise
- University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Jocelyn R Rodriguez-Paar
- University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Shelby R Konfrst
- University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Elizabeth R Flynn
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jeffrey D Salomon
- University of Nebraska Medical Center, Omaha, Nebraska
- Division of Pediatric Critical Care, Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michael E Hall
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Merry L Lindsey
- University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| |
Collapse
|
14
|
Elevated levels of apolipoprotein D predict poor outcome in patients with suspected or established coronary artery disease. Atherosclerosis 2021; 341:27-33. [PMID: 34959206 DOI: 10.1016/j.atherosclerosis.2021.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/23/2021] [Accepted: 12/17/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS Apolipoprotein D (apoD) is a lipocalin exerting neuroprotective effects. However, the relevance of apoD in respect to cardiovascular risk is largely unexplored. Therefore, this study aimed to evaluate the ability of apoD to predict future all-cause mortality, cardiovascular mortality, and cardiovascular events. METHODS Serum apoD levels were measured in a cohort of 531 Caucasian individuals who underwent coronary angiography (356 males, 175 females; mean age 65 ± 10 years). Fatal and non-fatal events were recorded over a median follow-up period of 5.8 years. RESULTS ApoD concentrations at baseline correlated significantly with age, presence of the metabolic syndrome, body mass index, lipoprotein levels, fasting glucose, and estimated glomerular filtration rate. Kaplan-Meier curve analyses by gender-stratified quartiles of apoD revealed that the cumulative incidence rates of mortality and cardiovascular events become higher with increasing apoD levels. The adjusted hazard ratios for participants in the highest quartile of apoD compared to those in the lowest quartile were 4.00 (95% confidence interval [CI] 1.49-10.74) for overall mortality, 5.47 (95% CI 1.20-25.00) for cardiovascular mortality, and 2.52 (95% CI 1.28-5.00) for cardiovascular events. CONCLUSIONS High circulating levels of apoD are an indicator of poor prognosis in patients with suspected or established coronary artery disease.
Collapse
|
15
|
Zalesak-Kravec S, Huang W, Wang P, Yu J, Liu T, Defnet AE, Moise AR, Farese AM, MacVittie TJ, Kane MA. Multi-omic Analysis of Non-human Primate Heart after Partial-body Radiation with Minimal Bone Marrow Sparing. HEALTH PHYSICS 2021; 121:352-371. [PMID: 34546217 PMCID: PMC8554778 DOI: 10.1097/hp.0000000000001478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
ABSTRACT High-dose radiation exposure results in hematopoietic and gastrointestinal acute radiation syndromes followed by delayed effects of acute radiation exposure, which encompasses multiple organs, including heart, kidney, and lung. Here we sought to further characterize the natural history of radiation-induced heart injury via determination of differential protein and metabolite expression in the heart. We quantitatively profiled the proteome and metabolome of left and right ventricle from non-human primates following 12 Gy partial body irradiation with 2.5% bone marrow sparing over a time period of 3 wk. Global proteome profiling identified more than 2,200 unique proteins, with 220 and 286 in the left and right ventricles, respectively, showing significant responses across at least three time points compared to baseline levels. High-throughput targeted metabolomics analyzed a total of 229 metabolites and metabolite combinations, with 18 and 22 in the left and right ventricles, respectively, showing significant responses compared to baseline levels. Bioinformatic analysis performed on metabolomic and proteomic data revealed pathways related to inflammation, energy metabolism, and myocardial remodeling were dysregulated. Additionally, we observed dysregulation of the retinoid homeostasis pathway, including significant post-radiation decreases in retinoic acid, an active metabolite of vitamin A. Significant differences between left and right ventricles in the pathology of radiation-induced injury were identified. This multi-omic study characterizes the natural history and molecular mechanisms of radiation-induced heart injury in NHP exposed to PBI with minimal bone marrow sparing.
Collapse
Affiliation(s)
- Stephanie Zalesak-Kravec
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Weiliang Huang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Pengcheng Wang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Jianshi Yu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Tian Liu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Amy E. Defnet
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Alexander R. Moise
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON, Canada; Departments of Chemistry and Biochemistry, and Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON, Canada
| | - Ann M. Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Maureen A. Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| |
Collapse
|
16
|
Demichev V, Tober-Lau P, Lemke O, Nazarenko T, Thibeault C, Whitwell H, Röhl A, Freiwald A, Szyrwiel L, Ludwig D, Correia-Melo C, Aulakh SK, Helbig ET, Stubbemann P, Lippert LJ, Grüning NM, Blyuss O, Vernardis S, White M, Messner CB, Joannidis M, Sonnweber T, Klein SJ, Pizzini A, Wohlfarter Y, Sahanic S, Hilbe R, Schaefer B, Wagner S, Mittermaier M, Machleidt F, Garcia C, Ruwwe-Glösenkamp C, Lingscheid T, Bosquillon de Jarcy L, Stegemann MS, Pfeiffer M, Jürgens L, Denker S, Zickler D, Enghard P, Zelezniak A, Campbell A, Hayward C, Porteous DJ, Marioni RE, Uhrig A, Müller-Redetzky H, Zoller H, Löffler-Ragg J, Keller MA, Tancevski I, Timms JF, Zaikin A, Hippenstiel S, Ramharter M, Witzenrath M, Suttorp N, Lilley K, Mülleder M, Sander LE, Ralser M, Kurth F. A time-resolved proteomic and prognostic map of COVID-19. Cell Syst 2021; 12:780-794.e7. [PMID: 34139154 PMCID: PMC8201874 DOI: 10.1016/j.cels.2021.05.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/24/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
COVID-19 is highly variable in its clinical presentation, ranging from asymptomatic infection to severe organ damage and death. We characterized the time-dependent progression of the disease in 139 COVID-19 inpatients by measuring 86 accredited diagnostic parameters, such as blood cell counts and enzyme activities, as well as untargeted plasma proteomes at 687 sampling points. We report an initial spike in a systemic inflammatory response, which is gradually alleviated and followed by a protein signature indicative of tissue repair, metabolic reconstitution, and immunomodulation. We identify prognostic marker signatures for devising risk-adapted treatment strategies and use machine learning to classify therapeutic needs. We show that the machine learning models based on the proteome are transferable to an independent cohort. Our study presents a map linking routinely used clinical diagnostic parameters to plasma proteomes and their dynamics in an infectious disease.
Collapse
Affiliation(s)
- Vadim Demichev
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany; The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK; The University of Cambridge, Department of Biochemistry and Cambridge Centre for Proteomics, Cambridge CB21GA, UK
| | - Pinkus Tober-Lau
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Oliver Lemke
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Tatiana Nazarenko
- University College London, Department of Mathematics, London WC1E 6BT, UK; University College London, Department of Women's Cancer, EGA Institute for Women'S Health, London WC1E 6BT, UK
| | - Charlotte Thibeault
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Harry Whitwell
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW72AZ, UK; Lobachevsky University, Department of Applied Mathematics, Nizhny Novgorod 603105, Russia; Imperial College London, Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, London SW7 2AZ, UK
| | - Annika Röhl
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Anja Freiwald
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Lukasz Szyrwiel
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Daniela Ludwig
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Clara Correia-Melo
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Simran Kaur Aulakh
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Elisa T Helbig
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Paula Stubbemann
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Lena J Lippert
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Nana-Maria Grüning
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Oleg Blyuss
- Lobachevsky University, Department of Applied Mathematics, Nizhny Novgorod 603105, Russia; University of Hertfordshire, School of Physics, Astronomy and Mathematics, Hatfield AL10 9AB, UK; Sechenov First Moscow State Medical University, Department of Paediatrics and Paediatric Infectious Diseases, Moscow 119435, Russia
| | - Spyros Vernardis
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Matthew White
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Christoph B Messner
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany; The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK
| | - Michael Joannidis
- Medical University Innsbruck, Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, 6020 Innsbruck, Austria
| | - Thomas Sonnweber
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Sebastian J Klein
- Medical University Innsbruck, Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, 6020 Innsbruck, Austria
| | - Alex Pizzini
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Yvonne Wohlfarter
- Medical University of Innsbruck, Institute of Human Genetics, 6020 Innsbruck, Austria
| | - Sabina Sahanic
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Richard Hilbe
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Benedikt Schaefer
- Medical University of Innsbruck, Christian Doppler Laboratory for Iron and Phosphate Biology, Department of Internal Medicine I, 6020 Innsbruck, Austria
| | - Sonja Wagner
- Medical University of Innsbruck, Christian Doppler Laboratory for Iron and Phosphate Biology, Department of Internal Medicine I, 6020 Innsbruck, Austria
| | - Mirja Mittermaier
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; Berlin Institute of Health, 10178 Berlin, Germany
| | - Felix Machleidt
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Carmen Garcia
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Christoph Ruwwe-Glösenkamp
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Tilman Lingscheid
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Laure Bosquillon de Jarcy
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Miriam S Stegemann
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Moritz Pfeiffer
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Linda Jürgens
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Sophy Denker
- Charité Universitätsmedizin Berlin, Medical Department of Hematology, Oncology & Tumor Immunology, Virchow Campus & Molekulares Krebsforschungszentrum, 13353 Berlin, Germany; Berlin Institute of Health, 10178 Berlin, Germany
| | - Daniel Zickler
- Charité Universitätsmedizin Berlin, Department of Nephrology and Internal Intensive Care Medicine, 10117 Berlin, Germany
| | - Philipp Enghard
- Charité Universitätsmedizin Berlin, Department of Nephrology and Internal Intensive Care Medicine, 10117 Berlin, Germany
| | - Aleksej Zelezniak
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK; Chalmers Tekniska Högskola, Department of Biology and Biological Engineering, SE-412 96 Gothenburg, Sweden
| | - Archie Campbell
- University of Edinburgh, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK; University of Edinburgh, Usher Institute, Edinburgh EH16 4UX, UK
| | - Caroline Hayward
- University of Edinburgh, MRC Human Genetics Unit, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK
| | - David J Porteous
- University of Edinburgh, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK; University of Edinburgh, Usher Institute, Edinburgh EH16 4UX, UK
| | - Riccardo E Marioni
- University of Edinburgh, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK
| | - Alexander Uhrig
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Holger Müller-Redetzky
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany
| | - Heinz Zoller
- Medical University of Innsbruck, Christian Doppler Laboratory for Iron and Phosphate Biology, Department of Internal Medicine I, 6020 Innsbruck, Austria
| | - Judith Löffler-Ragg
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - Markus A Keller
- Medical University of Innsbruck, Institute of Human Genetics, 6020 Innsbruck, Austria
| | - Ivan Tancevski
- Medical University of Innsbruck, Department of Internal Medicine II, 6020 Innsbruck, Austria
| | - John F Timms
- University College London, Department of Women's Cancer, EGA Institute for Women'S Health, London WC1E 6BT, UK
| | - Alexey Zaikin
- University College London, Department of Mathematics, London WC1E 6BT, UK; University College London, Department of Women's Cancer, EGA Institute for Women'S Health, London WC1E 6BT, UK; Lobachevsky University, Laboratory of Systems Medicine of Healthy Ageing, Nizhny Novgorod 603105, Russia
| | - Stefan Hippenstiel
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; German Centre for Lung Research, 35392 Gießen, Germany
| | - Michael Ramharter
- Bernhard Nocht Institute for Tropical Medicine, Department of Tropical Medicine, and University Medical Center Hamburg-Eppendorf, Department of Medicine, 20359 Hamburg, Germany
| | - Martin Witzenrath
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; German Centre for Lung Research, 35392 Gießen, Germany
| | - Norbert Suttorp
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; German Centre for Lung Research, 35392 Gießen, Germany
| | - Kathryn Lilley
- The University of Cambridge, Department of Biochemistry and Cambridge Centre for Proteomics, Cambridge CB21GA, UK
| | - Michael Mülleder
- Charité - Universitätsmedizin Berlin, Core Facility - High-Throughput Mass Spectrometry, 10117 Berlin, Germany
| | - Leif Erik Sander
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; German Centre for Lung Research, 35392 Gießen, Germany
| | - Markus Ralser
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany; The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London NW11AT, UK.
| | - Florian Kurth
- Charité Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, 10117 Berlin, Germany; Bernhard Nocht Institute for Tropical Medicine, Department of Tropical Medicine, and University Medical Center Hamburg-Eppendorf, Department of Medicine, 20359 Hamburg, Germany
| |
Collapse
|
17
|
Molecular underpinnings of the early brain developmental response to differential feeding in the honey bee Apis mellifera. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2021; 1864:194732. [PMID: 34242825 DOI: 10.1016/j.bbagrm.2021.194732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/14/2022]
Abstract
Brain differential morphogenesis in females is one of the major phenotypic manifestations of caste development in honey bees. Brain diphenism appears at the fourth larval phase as a result of the differential feeding regime developing females are submitted during early phases of larval development. Here, we used a forward genetics approach to test the early brain molecular response to differential feeding leading to the brain diphenism observed at later developmental phases. Using RNA sequencing analysis, we identified 53 differentially expressed genes (DEGs) between the brains of queens and workers at the third larval phase. Since miRNAs have been suggested to play a role in caste differentiation after horizontal and vertical transmission, we tested their potential participation in regulating the DEGs. The miRNA-mRNA interaction network, including the DEGs and the royal- and worker-jelly enriched miRNA populations, revealed a subset of miRNAs potentially involved in regulating the expression of DEGs. The interaction of miR-34, miR-210, and miR-317 with Takeout, Neurotrophin-1, Forked, and Masquerade genes was experimentally confirmed using a luciferase reporter system. Taken together, our results reconstruct the regulatory network that governs the development of the early brain diphenism in honey bees.
Collapse
|
18
|
Hernandez-Leon SG, Sarabia Sainz JAI, Ramos-Clamont Montfort G, Huerta-Ocampo JÁ, Ballesteros MN, Guzman-Partida AM, Robles-Burgueño MDR, Vazquez-Moreno L. Nanoproteomic Approach for Isolation and Identification of Potential Biomarkers in Human Urine from Adults with Normal Weight, Overweight and Obesity. Molecules 2021; 26:1803. [PMID: 33806905 PMCID: PMC8004714 DOI: 10.3390/molecules26061803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
In this work, previously synthesized and characterized core-shell silica nanoparticles (FCSNP) functionalized with immobilized molecular bait, Cibacron blue, and a porous polymeric bis-acrylamide shell were incubated with pooled urine samples from adult women or men with normal weight, overweight or obesity for the isolation of potential biomarkers. A total of 30 individuals (15 woman and 15 men) were included. FCSNP allowed the capture of a variety of low molecular weight (LMW) proteins as evidenced by mass spectrometry (MS) and the exclusion of high molecular weight (HMW) proteins (>34 kDa) as demonstrated by SDS-PAGE and 2D SDS-PAGE. A total of 36 proteins were successfully identified by MS and homology database searching against the Homo sapiens subset of the Swiss-Prot database. Identified proteins were grouped into different clusters according to their abundance patterns. Four proteins were found only in women and five only in men, whereas 27 proteins were in urine from both genders with different abundance patterns. Based on these results, this new approach represents an alternative tool for isolation and identification of urinary biomarkers.
Collapse
Affiliation(s)
- Sergio G. Hernandez-Leon
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas, No. 46, col. La Victoria, Hermosillo, Sonora 83304, Mexico; (S.G.H.-L.); (G.R.-C.M.); (M.N.B.); (A.M.G.-P.); (M.d.R.R.-B.)
| | - Jose Andre-i Sarabia Sainz
- Departamento de Investigación en Física, Universidad de Sonora, Blvd. Luis Encinas y Rosales S/N, Col. Centro, Hermosillo, Sonora 83190, Mexico;
| | - Gabriela Ramos-Clamont Montfort
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas, No. 46, col. La Victoria, Hermosillo, Sonora 83304, Mexico; (S.G.H.-L.); (G.R.-C.M.); (M.N.B.); (A.M.G.-P.); (M.d.R.R.-B.)
| | - José Ángel Huerta-Ocampo
- CONACyT-Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas, No. 46, col. La Victoria, Hermosillo, Sonora 83304, Mexico;
| | - Martha Nydia Ballesteros
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas, No. 46, col. La Victoria, Hermosillo, Sonora 83304, Mexico; (S.G.H.-L.); (G.R.-C.M.); (M.N.B.); (A.M.G.-P.); (M.d.R.R.-B.)
| | - Ana M. Guzman-Partida
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas, No. 46, col. La Victoria, Hermosillo, Sonora 83304, Mexico; (S.G.H.-L.); (G.R.-C.M.); (M.N.B.); (A.M.G.-P.); (M.d.R.R.-B.)
| | - María del Refugio Robles-Burgueño
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas, No. 46, col. La Victoria, Hermosillo, Sonora 83304, Mexico; (S.G.H.-L.); (G.R.-C.M.); (M.N.B.); (A.M.G.-P.); (M.d.R.R.-B.)
| | - Luz Vazquez-Moreno
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera Gustavo Enrique Astiazarán Rosas, No. 46, col. La Victoria, Hermosillo, Sonora 83304, Mexico; (S.G.H.-L.); (G.R.-C.M.); (M.N.B.); (A.M.G.-P.); (M.d.R.R.-B.)
| |
Collapse
|
19
|
Martínez-Pinilla E, Rubio-Sardón N, Peláez R, García-Álvarez E, del Valle E, Tolivia J, Larráyoz IM, Navarro A. Neuroprotective Effect of Apolipoprotein D in Cuprizone-Induced Cell Line Models: A Potential Therapeutic Approach for Multiple Sclerosis and Demyelinating Diseases. Int J Mol Sci 2021; 22:1260. [PMID: 33514021 PMCID: PMC7866080 DOI: 10.3390/ijms22031260] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Apolipoprotein D (Apo D) overexpression is a general finding across neurodegenerative conditions so the role of this apolipoprotein in various neuropathologies such as multiple sclerosis (MS) has aroused a great interest in last years. However, its mode of action, as a promising compound for the development of neuroprotective drugs, is unknown. The aim of this work was to address the potential of Apo D to prevent the action of cuprizone (CPZ), a toxin widely used for developing MS models, in oligodendroglial and neuroblastoma cell lines. On one hand, immunocytochemical quantifications and gene expression measures showed that CPZ compromised neural mitochondrial metabolism but did not induce the expression of Apo D, except in extremely high doses in neurons. On the other hand, assays of neuroprotection demonstrated that antipsychotic drug, clozapine, induced an increase in Apo D synthesis only in the presence of CPZ, at the same time that prevented the loss of viability caused by the toxin. The effect of the exogenous addition of human Apo D, once internalized, was also able to directly revert the loss of cell viability caused by treatment with CPZ by a reactive oxygen species (ROS)-independent mechanism of action. Taken together, our results suggest that increasing Apo D levels, in an endo- or exogenous way, moderately prevents the neurotoxic effect of CPZ in a cell model that seems to replicate some features of MS which would open new avenues in the development of interventions to afford MS-related neuroprotection.
Collapse
Affiliation(s)
- Eva Martínez-Pinilla
- Department of Morphology and Cell Biology, University of Oviedo, 33003 Oviedo, Spain; (N.R.-S.); (E.G.-Á.); (E.d.V.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Núria Rubio-Sardón
- Department of Morphology and Cell Biology, University of Oviedo, 33003 Oviedo, Spain; (N.R.-S.); (E.G.-Á.); (E.d.V.); (A.N.)
| | - Rafael Peláez
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain; (R.P.); (I.M.L.)
| | - Enrique García-Álvarez
- Department of Morphology and Cell Biology, University of Oviedo, 33003 Oviedo, Spain; (N.R.-S.); (E.G.-Á.); (E.d.V.); (A.N.)
| | - Eva del Valle
- Department of Morphology and Cell Biology, University of Oviedo, 33003 Oviedo, Spain; (N.R.-S.); (E.G.-Á.); (E.d.V.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Jorge Tolivia
- Department of Morphology and Cell Biology, University of Oviedo, 33003 Oviedo, Spain; (N.R.-S.); (E.G.-Á.); (E.d.V.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Ignacio M. Larráyoz
- Biomarkers and Molecular Signaling Group, Neurodegeneration Area, Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain; (R.P.); (I.M.L.)
| | - Ana Navarro
- Department of Morphology and Cell Biology, University of Oviedo, 33003 Oviedo, Spain; (N.R.-S.); (E.G.-Á.); (E.d.V.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
20
|
Seo YA, Choi EK, Aring L, Paschall M, Iwase S. Transcriptome Analysis of the Cerebellum of Mice Fed a Manganese-Deficient Diet. Front Genet 2020; 11:558725. [PMID: 33408735 PMCID: PMC7780674 DOI: 10.3389/fgene.2020.558725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 11/13/2020] [Indexed: 11/13/2022] Open
Abstract
Manganese (Mn), primarily acquired through diet, is required for brain function and development. Epidemiological studies have found an association between both low and high levels of Mn and impaired neurodevelopment in children. Recent genetic studies have revealed that patients with congenital Mn deficiency display severe psychomotor disability and cerebral and cerebellar atrophy. Although the impact of Mn on gene expression is beginning to be appreciated, Mn-dependent gene expression remains to be explored in vertebrate animals. The goal of this study was to use a mouse model to define the impact of a low-Mn diet on brain metal levels and gene expression. We interrogated gene expression changes in the Mn-deficient mouse brain at the genome-wide scale by RNA-seq analysis of the cerebellum of mice fed low or normal Mn diets. A total of 137 genes were differentially expressed in Mn-deficient cerebellums compared with Mn-adequate cerebellums (Padj < 0.05). Mn-deficient mice displayed downregulation of key pathways involved with "focal adhesion," "neuroactive ligand-receptor interaction," and "cytokine-cytokine receptor interaction" and upregulation of "herpes simplex virus 1 infection," "spliceosome," and "FoxO signaling pathway." Reactome pathway analysis identified upregulation of the splicing-related pathways and transcription-related pathways, as well as downregulation of "metabolism of carbohydrate," and "extracellular matrix organization," and "fatty acid metabolism" reactomes. The recurrent identifications of splicing-related pathways suggest that Mn deficiency leads to upregulation of splicing machineries and downregulation of diverse biological pathways.
Collapse
Affiliation(s)
- Young Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Eun-Kyung Choi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Luisa Aring
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Molly Paschall
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Shigeki Iwase
- Department of Human Genetics, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
21
|
Hao D, Wang H, Niu L. Activation of six lipocalins genes' transcription under PCB18 stress in OsTIL-silenced Oryza sativa L. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 204:111063. [PMID: 32791358 DOI: 10.1016/j.ecoenv.2020.111063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 07/12/2020] [Accepted: 07/21/2020] [Indexed: 06/11/2023]
Abstract
The lipocalins genes have been assigned for involving in the responses of organisms to various stress factors. The function of lipocalins under PCB18 stress was addressed by pathway complementation in the Oryza sativa L. OsTIL-silenced mutant. The growth of wild type (WT) and OsTIL-silenced mutant (MT) callus were suppressed by PCB18, and MT varieties were inhibited more seriously than WT varieties. Meanwhile, only WT varieties showed "Hormesis" effect. Compared with WT (3 day > 90.0%, 6 day ≤45.5%), MT varieties kept high removing efficiency by HPLC analysis. Varied gene transcription after OsTIL silencing was demonstrated between two varieties, especially obvious under PCB stress. Silenced OsTIL induced more protective gene transcriptions by qPCR analysis, OsVDE at 3 day, OsCHL, OsZEP1, OsZEP2 and OsUN at 6 day and OsZEP2 at 9 day. PCB18 stress further irritated these genes transcription in MT varieties. The defense stagy in WT varieties was that the transcriptions of lipocalins were inhibited to reduce PCB18 accumulation and toxicity. OsTIL could effectively limit PCB18 accumulation and toxicity. After TIL lacking, OsCHL, OsZEP1, OsZEP2 and OsUN in mutant were strongly evoked to against PCB stress. Remarkably, OsUN and OsZEP2 gene expressions were responded to PCB18 stress in both two varieties.
Collapse
Affiliation(s)
- Dehou Hao
- University of Science and Technology Beijing School of Chemistry and Biological Engineering, Department of Biological Science and Engineering, 30 Xueyuan Road, Haidian District, Beijing, 100083 China
| | - Haiou Wang
- University of Science and Technology Beijing School of Chemistry and Biological Engineering, Department of Biological Science and Engineering, 30 Xueyuan Road, Haidian District, Beijing, 100083 China.
| | - Lin Niu
- University of Science and Technology Beijing School of Chemistry and Biological Engineering, 30 Xueyuan Road, Haidian District, Beijing, 100083 China
| |
Collapse
|
22
|
Santana MFM, Lira ALA, Pinto RS, Minanni CA, Silva ARM, Sawada MIBAC, Nakandakare ER, Correa-Giannella MLC, Queiroz MS, Ronsein GE, Passarelli M. Enrichment of apolipoprotein A-IV and apolipoprotein D in the HDL proteome is associated with HDL functions in diabetic kidney disease without dialysis. Lipids Health Dis 2020; 19:205. [PMID: 32921312 PMCID: PMC7488728 DOI: 10.1186/s12944-020-01381-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/01/2020] [Indexed: 12/27/2022] Open
Abstract
Background and aims Diabetic kidney disease (DKD) is associated with lipid derangements that worsen kidney function and enhance cardiovascular (CVD) risk. The management of dyslipidemia, hypertension and other traditional risk factors does not completely prevent CVD complications, bringing up the participation of nontraditional risk factors such as advanced glycation end products (AGEs), carbamoylation and changes in the HDL proteome and functionality. The HDL composition, proteome, chemical modification and functionality were analyzed in nondialysis subjects with DKD categorized according to the estimated glomerular filtration rate (eGFR) and urinary albumin excretion rate (AER). Methods Individuals with DKD were divided into eGFR> 60 mL/min/1.73 m2 plus AER stages A1 and A2 (n = 10) and eGFR< 60 plus A3 (n = 25) and matched by age with control subjects (eGFR> 60; n = 8). Results Targeted proteomic analyses quantified 28 proteins associated with HDL in all groups, although only 2 were more highly expressed in the eGFR< 60 + A3 group than in the controls: apolipoprotein D (apoD) and apoA-IV. HDL from the eGFR< 60 + A3 group presented higher levels of total AGEs (20%), pentosidine (6.3%) and carbamoylation (4.2 x) and a reduced ability to remove 14C-cholesterol from macrophages (33%) in comparison to HDL from controls. The antioxidant role of HDL (lag time for LDL oxidation) was similar among groups, but HDL from the eGFR< 60 + A3 group presented a greater ability to inhibit the secretion of IL-6 and TNF-alpha (95%) in LPS-elicited macrophages in comparison to the control group. Conclusion The increase in apoD and apoA-IV could contribute to counteracting the HDL chemical modification by AGEs and carbamoylation, which contributes to HDL loss of function in well-established DKD.
Collapse
Affiliation(s)
- Monique F M Santana
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil
| | - Aécio L A Lira
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil
| | - Raphael S Pinto
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil.,Centro Universitário CESMAC, Maceio, Alagoas, Brazil
| | - Carlos A Minanni
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil.,Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein (HIAE), São Paulo, Brazil
| | - Amanda R M Silva
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Maria I B A C Sawada
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil
| | - Edna R Nakandakare
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil
| | - Maria L C Correa-Giannella
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil.,Laboratório de Carboidratos e Radioimunoensaio (LIM 18), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marcia S Queiroz
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil
| | - Graziella E Ronsein
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Marisa Passarelli
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo 455, room 3305; CEP, São Paulo, 01246-000, Brazil. .,Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil.
| |
Collapse
|
23
|
Narang D, Lento C, J. Wilson D. HDX-MS: An Analytical Tool to Capture Protein Motion in Action. Biomedicines 2020; 8:biomedicines8070224. [PMID: 32709043 PMCID: PMC7399943 DOI: 10.3390/biomedicines8070224] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 01/07/2023] Open
Abstract
Virtually all protein functions in the cell, including pathogenic processes, require coordinated motion of atoms or domains, i.e., conformational dynamics. Understanding protein dynamics is therefore critical both for drug development and to learn about the underlying molecular causes of many diseases. Hydrogen–Deuterium Exchange Mass Spectrometry (HDX-MS) provides valuable information about protein dynamics, which is highly complementary to the static picture provided by conventional high-resolution structural tools (i.e., X-ray crystallography and structural NMR). The amount of protein required to carry out HDX-MS experiments is a fraction of the amount required by alternative biophysical techniques, which are also usually lower resolution. Use of HDX-MS is growing quickly both in industry and academia, and it has been successfully used in numerous drug and vaccine development efforts, with important roles in understanding allosteric effects and mapping binding sites.
Collapse
Affiliation(s)
- Dominic Narang
- Department of Chemistry, York University, Toronto, ON M3J 1P3, Canada; (D.N.); (C.L.)
| | - Cristina Lento
- Department of Chemistry, York University, Toronto, ON M3J 1P3, Canada; (D.N.); (C.L.)
| | - Derek J. Wilson
- Department of Chemistry, York University, Toronto, ON M3J 1P3, Canada; (D.N.); (C.L.)
- Centre for Research of Biomolecular Interactions, York University, Toronto, ON M3J 1P3, Canada
- Centre for Research in Mass Spectrometry, York University, Toronto, ON M3J 1P3, Canada
- Correspondence:
| |
Collapse
|
24
|
De Micheli AJ, Spector JA, Elemento O, Cosgrove BD. A reference single-cell transcriptomic atlas of human skeletal muscle tissue reveals bifurcated muscle stem cell populations. Skelet Muscle 2020; 10:19. [PMID: 32624006 PMCID: PMC7336639 DOI: 10.1186/s13395-020-00236-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
Single-cell RNA-sequencing (scRNA-seq) facilitates the unbiased reconstruction of multicellular tissue systems in health and disease. Here, we present a curated scRNA-seq dataset of human muscle samples from 10 adult donors with diverse anatomical locations. We integrated ~ 22,000 single-cell transcriptomes using Scanorama to account for technical and biological variation and resolved 16 distinct populations of muscle-resident cells using unsupervised clustering of the data compendium. These cell populations included muscle stem/progenitor cells (MuSCs), which bifurcated into discrete "quiescent" and "early-activated" MuSC subpopulations. Differential expression analysis identified transcriptional profiles altered in the activated MuSCs including genes associated with aging, obesity, diabetes, and impaired muscle regeneration, as well as long non-coding RNAs previously undescribed in human myogenic cells. Further, we modeled ligand-receptor cell-communication interactions and observed enrichment of the TWEAK-FN14 pathway in activated MuSCs, a characteristic signature of muscle wasting diseases. In contrast, the quiescent MuSCs have enhanced expression of the EGFR receptor, a recognized human MuSC marker. This work provides a new benchmark reference resource to examine human muscle tissue heterogeneity and identify potential targets in MuSC diversity and dysregulation in disease contexts.
Collapse
Affiliation(s)
- Andrea J De Micheli
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jason A Spector
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Division of Plastic Surgery, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
25
|
Upadhya R, Zingg W, Shetty S, Shetty AK. Astrocyte-derived extracellular vesicles: Neuroreparative properties and role in the pathogenesis of neurodegenerative disorders. J Control Release 2020; 323:225-239. [PMID: 32289328 DOI: 10.1016/j.jconrel.2020.04.017] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) released by neural cells play an essential role in brain homeostasis and the crosstalk between neural cells and the periphery. EVs are diverse, nano-sized vesicles, which transport proteins, nucleic acids, and lipids between cells over short and long expanses and hence are proficient for modulating the target cells. EVs released from neural cells are implicated in synaptic plasticity, neuron-glia interface, neuroprotection, neuroregeneration, and the dissemination of neuropathological molecules. This review confers the various properties of EVs secreted by astrocytes and their potential role in health and disease with a focus on evolving concepts. Naïve astrocytes shed EVs containing a host of neuroprotective compounds, which include fibroblast growth factor-2, vascular endothelial growth factor, and apolipoprotein-D. Stimulated astrocytes secrete EVs with neuroprotective molecules including heat shock proteins, synapsin 1, unique microRNAs, and glutamate transporters. Well-characterized astrocyte-derived EVs (ADEVs) generated in specific culture conditions and ADEVs that are engineered to carry the desired miRNAs or proteins are likely useful for treating brain injury and neurogenerative diseases. On the other hand, in conditions such as Alzheimer's disease (AD), stroke, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), and other neuroinflammatory conditions, EVs released by activated astrocytes appear to mediate or exacerbate the pathological processes. The examples include ADEVs spreading the dysregulated complement system in AD, mediating motoneuron toxicity in ALS, and stimulating peripheral leukocyte migration into the brain in inflammatory conditions. Strategies restraining the release of EVs by activated astrocytes or modulating the composition of ADEVs are likely beneficial for treating neurodegenerative diseases. Also, periodic analyses of ADEVs in the blood is useful for detecting astrocyte-specific biomarkers in different neurological conditions and for monitoring disease progression and remission with distinct therapeutic approaches.
Collapse
Affiliation(s)
- Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Winston Zingg
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Siddhant Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.
| |
Collapse
|
26
|
Cirulli F, Musillo C, Berry A. Maternal Obesity as a Risk Factor for Brain Development and Mental Health in the Offspring. Neuroscience 2020; 447:122-135. [PMID: 32032668 DOI: 10.1016/j.neuroscience.2020.01.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/30/2022]
Abstract
Maternal obesity plays a key role in the health trajectory of the offspring. Although research on this topic has largely focused on the potential of this condition to increase the risk for child obesity, it is becoming more and more evident that it can also significantly impact cognitive function and mental health. The mechanisms underlying these effects are starting to be elucidated and point to the placenta as a critical organ that may mediate changes in the response to stress, immune function and oxidative stress. Long-term effects of maternal obesity may rely upon epigenetic changes in selected genes that are involved in metabolic and trophic regulations of the brain. More recent evidence also indicates the gut microbiota as a potential mediator of these effects. Overall, understanding cause-effect relationships can allow the development of preventive measures that could rely upon dietary changes in the mother and the offspring. Addressing diets appears more feasible than developing new pharmacological targets and has the potential to affect the multiple interconnected physiological pathways engaged by these complex regulations, allowing prevention of both metabolic and mental disorders.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
27
|
Liu H, Anders F, Funke S, Mercieca K, Grus F, Prokosch V. Proteome alterations in aqueous humour of primary open angle glaucoma patients. Int J Ophthalmol 2020; 13:176-179. [PMID: 31956586 DOI: 10.18240/ijo.2020.01.24] [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: 07/23/2019] [Accepted: 11/20/2019] [Indexed: 01/10/2023] Open
Abstract
AIM To unravel the primary open angle glaucoma (POAG) related proteomic changes in aqueous humour (AH). METHODS Totally 35 patients listed for cataract surgery (controls: n=12, age: 67.4±13.6y) or trabeculectomy for POAG (n=23, age: 72.5±8.3y) were included. AH samples of those patients were obtained during cataract surgery or trabeculectomy. AH samples were subsequently pooled into the experimental groups under equal contribution in terms of protein amount of each individual patient. Protein samples were analyzed by a linear trap quadrupol Orbitrap Mass Spectrometry device with an upstream liquid chromatography system. The obtained raw data were analyzed using the Maxquant proteome software and compared. Proteins with a fold-change ratio higher than a cut-off of 2 were considered as noticeably altered. RESULTS A total number of 175 proteins could be identified out of the AH from POAG and cataract by means of quantitative mass spectrometric analysis. Apolipoprotein D (fold change, 3.16 times), complement C3 (2.96), pigment epithelium-derived factor (2.86), dickkopf-related protein 3 (2.18) and wingless-related integration (Wnt) inhibitory factor 1 (2.35) were significantly upregulated within the AH of glaucoma compared to cataract serving as controls. CONCLUSION AH provides a tool to analyze changes in glaucoma and shows striking changes in Wnt signaling inhibitory molecules and other proteins.
Collapse
Affiliation(s)
- Hanhan Liu
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany.,Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | - Fabian Anders
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | - Sebastian Funke
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | - Karl Mercieca
- Royal Eye Hospital, School of Medicine, University of Manchester, Manchester M202UL, United Kingdom
| | - Franz Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | - Verena Prokosch
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| |
Collapse
|
28
|
Abstract
The apolipoproteins are well known for their roles in both health and disease, as components of plasma lipoprotein particles, such as high-density lipoprotein (HDL), low-density lipoprotein (LDL), very-low-density lipoprotein (VLDL), chylomicrons, and metabolic, vascular- and inflammation-related disorders, such as cardiovascular disease, atherosclerosis, metabolic syndrome, and diabetes. Increasingly, their roles in neurovascular and neurodegenerative disorders are also being elucidated. They play major roles in lipid and cholesterol transport between blood and organs and are, therefore, critical to maintenance and homeostasis of the lipidome, with apolipoprotein-lipid interactions, including cholesterol, fatty acids, triglycerides, phospholipids, and isoprostanes. Further, they have important pleiotropic roles related to aging and longevity, which are largely managed through their many structural variants, including multiple isoforms, and a diversity of post-translational modifications. Consequently, tools for the characterization and accurate quantification of apolipoproteins, including their diverse array of variant forms, are required to understand their salutary and disease related roles. In this chapter we outline three distinct quantitative approaches suitable for targeting apolipoproteins: (1) multiplex immunoassays, (2) mass spectrometric immunoassay, and (3) multiple reaction monitoring, mass spectrometric quantification. We also discuss management of pre-analytical and experimental design variables.
Collapse
|
29
|
Medeiros ADM, Silva RH. Sex Differences in Alzheimer’s Disease: Where Do We Stand? J Alzheimers Dis 2019; 67:35-60. [DOI: 10.3233/jad-180213] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- André de Macêdo Medeiros
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Center of Health and Biological Sciences, Universidade Federal Rural do Semiárido, Mossoró, Brazil
| | - Regina Helena Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
30
|
Desmarais F, Bergeron KF, Rassart E, Mounier C. Apolipoprotein D overexpression alters hepatic prostaglandin and omega fatty acid metabolism during the development of a non-inflammatory hepatic steatosis. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:522-531. [PMID: 30630053 DOI: 10.1016/j.bbalip.2019.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/17/2018] [Accepted: 01/06/2019] [Indexed: 12/31/2022]
Abstract
Apolipoprotein D (ApoD) is a secreted lipocalin associated with neuroprotection and lipid metabolism. Overexpression of ApoD in mouse neural tissue induces the development of a non-inflammatory hepatic steatosis in 12-month-old transgenic animals. Previous data indicates that accumulation of arachidonic acid, ApoD's preferential ligand, and overactivation of PPARγ are likely the driving forces in the development of the pathology. However, the lack of inflammation under those conditions is surprising. Hence, we further investigated the apparent repression of inflammation during hepatic steatosis development in aging transgenic animals. The earliest modulation of lipid metabolism and inflammation occurred at 6 months with a transient overexpression of L-PGDS and concomitant overproduction of 15d-PGJ2, a PPARγ agonist. Hepatic lipid accumulation was detectable as soon as 9 months. Inflammatory polarization balance varied in time, with a robust anti-inflammatory profile at 6 months coinciding with 15d-PGJ2 overproduction. Omega-3 and omega-6 fatty acids were preferentially stored in the liver of 12-month-old transgenic mice and resulted in a higher omega-3/omega-6 ratio compared to wild type mice of the same age. Thus, inflammation seems to be controlled by several mechanisms in the liver of transgenic mice: first by an increase in 15d-PGJ2 production and later by a beneficial omega-3/omega-6 ratio. PPARγ seems to play important roles in these processes. The accumulation of several omega fatty acids species in the transgenic mouse liver suggests that ApoD might bind to a broader range of fatty acids than previously thought.
Collapse
Affiliation(s)
- Frederik Desmarais
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Canada
| | - Karl-F Bergeron
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Canada
| | - Eric Rassart
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Canada
| | - Catherine Mounier
- Molecular Metabolism of Lipids Laboratory, BioMed Research Center, Biological Sciences Department, University of Quebec in Montreal (UQAM), Canada.
| |
Collapse
|
31
|
Semmouri I, Asselman J, Van Nieuwerburgh F, Deforce D, Janssen CR, De Schamphelaere KAC. The transcriptome of the marine calanoid copepod Temora longicornis under heat stress and recovery. MARINE ENVIRONMENTAL RESEARCH 2019; 143:10-23. [PMID: 30415781 DOI: 10.1016/j.marenvres.2018.10.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/10/2018] [Accepted: 10/30/2018] [Indexed: 06/09/2023]
Abstract
Understanding the impacts of global change in zooplankton communities is crucial, as alterations in the zooplankton communities can affect entire marine ecosystems. Despite the economic and ecological importance of the calanoid copepod Temora longicornis in the Belgian part of the North Sea, molecular data is still very limited for this species. Using HiSeq Illumina sequencing, we sequenced the whole transcriptome of T. longicornis, after being exposed to realistic temperatures of 14 and 17 °C. After both an acute (1 day) and a more sustained (5 days) thermal exposure to 17 °C, we investigated gene expression differences with animals exposed to 14 °C, which may be critical for the thermal acclimation and resilience of this copepod species. We also studied the possibility of a short term stress recovery of a heat shock. A total of 179,569 transcripts were yielded, of which 44,985 putative ORF transcripts were identified. These transcripts were subsequently annotated into roughly 22,000 genes based on known sequences using Gene Ontology (GO) and KEGG databases. Temora only showed a mild response to both the temperature and the duration of the exposure. We found that the expression of 27 transcripts varied significantly with an increase in temperature of 3 °C, of which eight transcripts were differentially expressed after acute exposure only. Gene set enrichment analysis revealed that, overall, T. longicornis was more impacted by a sustained thermal exposure, rather than an immediate (acute) exposure, with two times as many enriched GO terms in the sustained treatment. We also identified several general stress responses independent of exposure time, such as modified protein synthesis, energy mobilisation, cuticle and chaperone proteins. Finally, we highlighted candidate genes of a possible recovery from heat exposure, identifying similar terms as those enriched in the heat treatments, i.e. related to for example energy metabolism, cuticle genes and extracellular matrix. The data presented in this study provides the first transcriptome available for T. longicornis which can be used for future genomic studies.
Collapse
Affiliation(s)
- Ilias Semmouri
- Ghent University, Laboratory of Environmental Toxicology and Aquatic Ecology, Faculty of Bioscience Engineering, 9000, Ghent, Belgium.
| | - Jana Asselman
- Ghent University, Laboratory of Environmental Toxicology and Aquatic Ecology, Faculty of Bioscience Engineering, 9000, Ghent, Belgium
| | - Filip Van Nieuwerburgh
- Ghent University, Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, 9000, Ghent, Belgium
| | - Dieter Deforce
- Ghent University, Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, 9000, Ghent, Belgium
| | - Colin R Janssen
- Ghent University, Laboratory of Environmental Toxicology and Aquatic Ecology, Faculty of Bioscience Engineering, 9000, Ghent, Belgium
| | - Karel A C De Schamphelaere
- Ghent University, Laboratory of Environmental Toxicology and Aquatic Ecology, Faculty of Bioscience Engineering, 9000, Ghent, Belgium
| |
Collapse
|
32
|
Navarro A, Rioseras B, Del Valle E, Martínez-Pinilla E, Astudillo A, Tolivia J. Expression Pattern of Myelin-Related Apolipoprotein D in Human Multiple Sclerosis Lesions. Front Aging Neurosci 2018; 10:254. [PMID: 30186153 PMCID: PMC6110904 DOI: 10.3389/fnagi.2018.00254] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 08/02/2018] [Indexed: 11/13/2022] Open
Abstract
Apolipoprotein D (Apo D) is a key molecule in the lipid transport during homeostasis and repair processes in normal and pathological conditions of the nervous system with a putative neuroprotective effect. In the last decades, huge experimental efforts have been made to know the exact mechanism of action of Apo D, even though, it remains an open question. In this regard, studies in mammals and flies have suggested that Apo D seems to act through a variety of cellular mechanisms related with its ability to selectively bind different lipid ligands. For instance, this apolipoprotein is required to myelin compaction, it participates in axon regeneration/remyelination, and it can control the magnitude and timing of the inflammatory response after injury, promoting myelin clearance, and regulating the number of immune cells recruited to the damaged area. These, among others, are some of the reasons to study Apo D in multiple sclerosis (MS) pathology, where it could be particularly important since the autoimmune reaction against oligodendrocytes (OLGs) and myelin is generally assumed as the most plausible cause of this pathology. The aim of this work was to investigate the Apo D expression pattern in MS lesions, including active and inactive demyelinating plaques, and also remyelinating ones. Human brain tissues with inflammatory demyelination consistent with MS were used to quantify Apo D immunosignal in different lesions. Our results show a clear decrease of Apo D expression in all sclerosis plaques, being lower in the inactive than in active areas but recovers in the remyelination ones. Apo D is mainly produced by the matured OLGs of white matter and is located in cell processes surrounding the myelin sheath. All these data seem to indicate an important role of Apo D in myelination/remyelination processes as a molecule with a neuroprotective potential, and may serve as a good starting point for its study in MS.
Collapse
Affiliation(s)
- Ana Navarro
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.,Departamento de Morfología y Biología Celular, Facultad de Biología y Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Beatriz Rioseras
- Departamento de Morfología y Biología Celular, Facultad de Biología y Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Eva Del Valle
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.,Departamento de Morfología y Biología Celular, Facultad de Biología y Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Eva Martínez-Pinilla
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.,Departamento de Morfología y Biología Celular, Facultad de Biología y Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Aurora Astudillo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.,Servicio de Anatomía Patológica, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Jorge Tolivia
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.,Departamento de Morfología y Biología Celular, Facultad de Biología y Medicina, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
33
|
Kosarussavadi S, Pennington ZT, Covell J, Blaisdell AP, Schlinger BA. Across sex and age: Learning and memory and patterns of avian hippocampal gene expression. Behav Neurosci 2018; 131:483-491. [PMID: 29189019 DOI: 10.1037/bne0000222] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Age-related decrements in cognitive ability have been proposed to stem from deteriorating function of the hippocampus. Many birds are long lived, especially for their relatively small body mass and elevated metabolism, making them a unique model of resilience to aging. Nevertheless, little is known about avian age-related changes in cognition and hippocampal physiology. We studied spatial cognition and hippocampal expression of the age-related gene, Apolipoprotein D (ApoD), and the immediate early gene Egr-1 in zebra finches at various developmental time points. In a first experiment, middle-aged adult males outperformed middle-aged females in learning correct food locations in a four-arm maze, but all birds remembered the task equally well after a 5- or 10-day delay. In a second experiment comparing young and old birds, aged birds showed minimal evidence for deterioration in spatial cognition or motivation relative to young birds, except that aged females showed less rapid gains in accuracy during spatial learning than young females. These findings indicate that sex differences in hippocampus-dependent spatial learning and decline with age are phylogenetically conserved. With respect to hippocampal gene expression, adult females expressed Egr-1 at significantly greater levels than males after memory retrieval, perhaps reflecting a neurobiological compensation. Contrary to mammals, ApoD expression was elevated in young zebra finches compared with aged birds. This may explain the near absence of decrements in spatial memory due to age, possibly indicating an alternative mechanism of managing oxidative stress in aged birds. (PsycINFO Database Record
Collapse
Affiliation(s)
- Saritha Kosarussavadi
- Department of Integrative Biology and Physiology, University of California, Los Angeles
| | | | - Jeremy Covell
- Department of Integrative Biology and Physiology, University of California, Los Angeles
| | | | - Barney A Schlinger
- Department of Integrative Biology and Physiology, University of California, Los Angeles
| |
Collapse
|
34
|
Zucca FA, Vanna R, Cupaioli FA, Bellei C, De Palma A, Di Silvestre D, Mauri P, Grassi S, Prinetti A, Casella L, Sulzer D, Zecca L. Neuromelanin organelles are specialized autolysosomes that accumulate undegraded proteins and lipids in aging human brain and are likely involved in Parkinson's disease. NPJ Parkinsons Dis 2018; 4:17. [PMID: 29900402 PMCID: PMC5988730 DOI: 10.1038/s41531-018-0050-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/10/2018] [Accepted: 04/17/2018] [Indexed: 01/08/2023] Open
Abstract
During aging, neuronal organelles filled with neuromelanin (a dark-brown pigment) and lipid bodies accumulate in the brain, particularly in the substantia nigra, a region targeted in Parkinson's disease. We have investigated protein and lipid systems involved in the formation of these organelles and in the synthesis of the neuromelanin of human substantia nigra. Membrane and matrix proteins characteristic of lysosomes were found in neuromelanin-containing organelles at a lower number than in typical lysosomes, indicating a reduced enzymatic activity and likely impaired capacity for lysosomal and autophagosomal fusion. The presence of proteins involved in lipid transport may explain the accumulation of lipid bodies in the organelle and the lipid component in neuromelanin structure. The major lipids observed in lipid bodies of the organelle are dolichols with lower amounts of other lipids. Proteins of aggregation and degradation pathways were present, suggesting a role for accumulation by this organelle when the ubiquitin-proteasome system is inadequate. The presence of proteins associated with aging and storage diseases may reflect impaired autophagic degradation or impaired function of lysosomal enzymes. The identification of typical autophagy proteins and double membranes demonstrates the organelle's autophagic nature and indicates that it has engulfed neuromelanin precursors from the cytosol. Based on these data, it appears that the neuromelanin-containing organelle has a very slow turnover during the life of a neuron and represents an intracellular compartment of final destination for numerous molecules not degraded by other systems.
Collapse
Affiliation(s)
- Fabio A. Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Renzo Vanna
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
- IRCCS Don Carlo Gnocchi ONLUS Foundation, Milan, Italy
| | - Francesca A. Cupaioli
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Chiara Bellei
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Antonella De Palma
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Dario Di Silvestre
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Pierluigi Mauri
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Luigi Casella
- Department of Chemistry, University of Pavia, Pavia, Italy
| | - David Sulzer
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY USA
- Department of Neurology, Columbia University Medical Center, New York, NY USA
- Department of Pharmacology, Columbia University Medical Center, New York, NY USA
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY USA
| |
Collapse
|
35
|
Deibel SH, Zelinski EL, Keeley RJ, Kovalchuk O, McDonald RJ. Epigenetic alterations in the suprachiasmatic nucleus and hippocampus contribute to age-related cognitive decline. Oncotarget 2016; 6:23181-203. [PMID: 26252151 PMCID: PMC4695111 DOI: 10.18632/oncotarget.4036] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 12/31/1969] [Indexed: 12/16/2022] Open
Abstract
Circadian rhythm dysfunction and cognitive decline, specifically memory loss, frequently accompany natural aging. Circadian rhythms and memory are intertwined, as circadian rhythms influence memory formation and recall in young and old rodents. Although, the precise relationship between circadian rhythms and memory is still largely unknown, it is hypothesized that circadian rhythm disruption, which occurs during aging, contributes to age-associated cognitive decline, specifically memory loss. While there are a variety of mechanisms that could mediate this effect, changes in the epigenome that occur during aging has been proposed as a potential candidate. Interestingly, epigenetic mechanisms, such as DNA methylation and sirtuin1 (SIRT1) are necessary for both circadian rhythms and memory. During aging, similar alterations of epigenetic mechanisms occur in the suprachiasmatic nucleus (SCN) and hippocampus, which are necessary for circadian rhythm generation and memory, respectively. Recently, circadian rhythms have been linked to epigenetic function in the hippocampus, as some of these epigenetic mechanisms oscillate in the hippocampus and are disrupted by clock gene deletion. The current paper will review how circadian rhythms and memory change with age, and will suggest how epigenetic changes in these processes might contribute to age-related cognitive decline.
Collapse
Affiliation(s)
- Scott H Deibel
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Erin L Zelinski
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Robin J Keeley
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, Canada
| | - Robert J McDonald
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
36
|
Cheow ESH, Cheng WC, Lee CN, de Kleijn D, Sorokin V, Sze SK. Plasma-derived Extracellular Vesicles Contain Predictive Biomarkers and Potential Therapeutic Targets for Myocardial Ischemic (MI) Injury. Mol Cell Proteomics 2016; 15:2628-40. [PMID: 27234505 DOI: 10.1074/mcp.m115.055731] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Indexed: 12/21/2022] Open
Abstract
Myocardial infarction (MI) triggers a potent inflammatory response via the release of circulatory mediators, including extracellular vesicles (EVs) by damaged cardiac cells, necessary for myocardial healing. Timely repression of inflammatory response are critical to prevent and minimize cardiac tissue injuries, nonetheless, progression in this aspect remains challenging. The ability of EVs to trigger a functional response upon delivery of carried bioactive cargos, have made them clinically attractive diagnostic biomarkers and vectors for therapeutic interventions. Using label-free quantitative proteomics approach, we compared the protein cargo of plasma EVs between patients with MI and from patients with stable angina (NMI). We report, for the first time, the proteomics profiling on 252 EV proteins that were modulated with >1.2-fold after MI. We identified six up-regulated biomarkers with potential for clinical applications; these reflected post-infarct pathways of complement activation (Complement C1q subcomponent subunit A (C1QA), 3.23-fold change, p = 0.012; Complement C5 (C5), 1.27-fold change, p = 0.087), lipoprotein metabolism (Apoliporotein D (APOD), 1.86-fold change, p = 0.033; Apolipoprotein C-III (APOCC3), 2.63-fold change, p = 0.029) and platelet activation (Platelet glycoprotein Ib alpha chain (GP1BA), 9.18-fold change, p < 0.0001; Platelet basic protein (PPBP), 4.72-fold change, p = 0.027). The data have been deposited to the ProteomeXchange with identifier PXD002950. This novel biomarker panel was validated in 43 patients using antibody-based assays (C1QA (p = 0.005); C5 (p = 0.0047), APOD (p = 0.0267); APOC3 (p = 0.0064); GP1BA (p = 0.0031); PPBP (p = 0.0465)). We further present that EV-derived fibrinogen components were paradoxically down-regulated in MI, suggesting that a compensatory mechanism may suppress post-infarct coagulation pathways, indicating potential for therapeutic targeting of this mechanism in MI. Taken together, these data demonstrated that plasma EVs contain novel diagnostic biomarkers and therapeutic targets that can be further developed for clinical use to benefit patients with coronary artery diseases (CADs).
Collapse
Affiliation(s)
- Esther Sok Hwee Cheow
- From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Woo Chin Cheng
- §Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, and Cardiovascular Research Institute, Singapore 119228
| | - Chuen Neng Lee
- §Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, and Cardiovascular Research Institute, Singapore 119228; ¶National University Heart Centre, Department of Cardiac, Thoracic & Vascular Surgery, Singapore 119228; ‖Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Dominique de Kleijn
- §Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, and Cardiovascular Research Institute, Singapore 119228; **Experimental Cardiology Laboratory, Cardiology, University Medical Center Utrecht, the Netherlands & Interuniversity Cardiovascular Institute of the Netherlands, Utrecht, the Netherlands
| | - Vitaly Sorokin
- §Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, and Cardiovascular Research Institute, Singapore 119228; ¶National University Heart Centre, Department of Cardiac, Thoracic & Vascular Surgery, Singapore 119228
| | - Siu Kwan Sze
- From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551;
| |
Collapse
|
37
|
Del Valle E, Navarro A, Martínez-Pinilla E, Torices S, Tolivia J. Apo J and Apo D: Complementary or Antagonistic Roles in Alzheimer's Disease? J Alzheimers Dis 2016; 53:639-50. [PMID: 27197790 DOI: 10.3233/jad-160032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Apolipoprotein D (Apo D) and Apolipoprotein J (Apo J) are among the only nine apolipoproteins synthesized in the nervous system. Apart from development, these apolipoproteins are implicated in the normal aging process as well as in different neuropathologies as Alzheimer's disease (AD), where a neuroprotective role has been postulated. Different authors have proposed that Apo D and Apo J could be biomarkers for AD but as far as we know, there are no studies about the relationship between them as well as their expression pattern along the progression of the disease. In this paper, using double immunohistochemistry techniques, we have demonstrated that Apo D is mainly located in glial cells while Apo J expression preferentially occurs in neurons; both proteins are also present in AD diffuse and mature senile plaques but without signal overlap. In addition, we have observed that Apo J and Apo D immunostaining shows a positive correlation with the progression of the disease and the Braak's stages. These results suggest complementary and cell-dependent neuroprotective roles for each apolipoprotein during AD progress.
Collapse
|
38
|
Brosel S, Laub C, Averdam A, Bender A, Elstner M. Molecular aging of the mammalian vestibular system. Ageing Res Rev 2016; 26:72-80. [PMID: 26739358 DOI: 10.1016/j.arr.2015.12.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 12/16/2015] [Accepted: 12/21/2015] [Indexed: 12/18/2022]
Abstract
Dizziness and imbalance frequently affect the elderly and contribute to falls and frailty. In many geriatric patients, clinical testing uncovers a dysfunction of the vestibular system, but no specific etiology can be identified. Neuropathological studies have demonstrated age-related degeneration of peripheral and central vestibular neurons, but the molecular mechanisms are poorly understood. In contrast, recent studies into age-related hearing loss strongly implicate mitochondrial dysfunction, oxidative stress and apoptotic cell death of cochlear hair cells. While some data suggest that analogous biological pathomechanisms may underlie vestibular dysfunction, actual proof is missing. In this review, we summarize the available data on the molecular causes of vestibular dysfunction.
Collapse
Affiliation(s)
- Sonja Brosel
- German Center for Vertigo and Balance Disorders, Department of Neurology, Klinikum Grosshadern, Ludwig-Maximilians-University, Marchioninistr. 15, 81377 Munich, Germany.
| | - Christoph Laub
- Department of Neurology with Friedrich-Baur-Institute, Klinikum Grosshadern, Ludwig-Maximilians-University, Marchioninistr. 15, 81377 Munich, Germany
| | - Anne Averdam
- Department of Neurology with Friedrich-Baur-Institute, Klinikum Grosshadern, Ludwig-Maximilians-University, Marchioninistr. 15, 81377 Munich, Germany
| | - Andreas Bender
- Department of Neurology, Therapiezentrum Burgau, Kapuzinerstr.34, 89331 Burgau, Germany
| | - Matthias Elstner
- Department of Neurology with Friedrich-Baur-Institute, Klinikum Grosshadern, Ludwig-Maximilians-University, Marchioninistr. 15, 81377 Munich, Germany; Department of Neurology and Clinical Neurophysiology, Academic Hospital Munich-Bogenhausen, Technical University of Munich, Englschalkingerstr. 77, 81925 Munich, Germany
| |
Collapse
|
39
|
del Caño-Espinel M, Acebes JR, Sanchez D, Ganfornina MD. Lazarillo-related Lipocalins confer long-term protection against type I Spinocerebellar Ataxia degeneration contributing to optimize selective autophagy. Mol Neurodegener 2015; 10:11. [PMID: 25888134 PMCID: PMC4374295 DOI: 10.1186/s13024-015-0009-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 03/02/2015] [Indexed: 12/22/2022] Open
Abstract
Background A diverse set of neurodegenerative disorders are caused by abnormal extensions of polyglutamine (poly-Q) stretches in various, functionally unrelated proteins. A common feature of these diseases is altered proteostasis. Autophagy induction is part of the endogenous response to poly-Q protein expression. However, if autophagy is not resolved properly, clearance of toxic proteins or aggregates cannot occur effectively. Likewise, excessive autophagy induction can cause autophagic stress and neurodegeneration. The Lipocalins ApoD, Glial Lazarillo (GLaz) and Neural Lazarillo (NLaz) are neuroprotectors upon oxidative stress or aging. In this work we test whether these Lipocalins also protect against poly-Q-triggered deterioration of protein quality control systems. Results Using a Drosophila retinal degeneration model of Type-1 Spinocerebellar Ataxia (SCA1) combined with genetic manipulation of NLaz and GLaz expression, we demonstrate that both Lipocalins protect against SCA1 neurodegeneration. They are part of the endogenous transcriptional response to SCA1, and their effect is non-additive, suggesting participation in a similar mechanism. GLaz beneficial effects persist throughout aging, and appears when expressed by degenerating neurons or by retinal support and glial cells. GLaz gain-of-function reduces cell death and the extent of ubiquitinated proteins accumulation, and decreases the expression of Atg8a/LC3, p62 mRNA and protein levels, and GstS1 induction. Over-expression of GLaz is able to reduce p62 and ubiquitinated proteins levels when rapamycin-dependent and SCA1-dependent inductions of autophagy are combined. In the absence of neurodegeneration, GLaz loss-of-function increases Atg8a/LC3 mRNA and p62 protein levels without altering p62 mRNA levels. Knocking-down autophagy, by interfering with Atg8a or p62 expression or by expressing dominant-negative Atg1/ULK1 or Atg4a transgenes, rescues SCA1-dependent neurodegeneration in a similar extent to the protective effect of GLaz. Further GLaz-dependent improvement is concealed. Conclusions This work shows for the first time that a Lipocalin rescues neurons from pathogenic SCA1 degeneration by optimizing clearance of aggregation-prone proteins. GLaz modulates key autophagy genes and lipid-peroxide clearance responsive genes. Down-regulation of selective autophagy causes similar and non-additive rescuing effects. These data suggest that SCA1 neurodegeneration concurs with autophagic stress, and places Lazarillo-related Lipocalins as valuable players in the endogenous protection against the two major contributors to aging and neurodegeneration: ROS-dependent damage and proteostasis deterioration. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0009-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manuela del Caño-Espinel
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| | - Judith R Acebes
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| | - Diego Sanchez
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| | - Maria D Ganfornina
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| |
Collapse
|
40
|
Braesch-Andersen S, Beckman L, Paulie S, Kumagai-Braesch M. ApoD mediates binding of HDL to LDL and to growing T24 carcinoma. PLoS One 2014; 9:e115180. [PMID: 25513803 PMCID: PMC4267786 DOI: 10.1371/journal.pone.0115180] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 11/19/2014] [Indexed: 11/28/2022] Open
Abstract
Apolipoprotein (Apo) D is an important protein produced in many parts of the body. It is necessary for the development and repair of the brain and protection from oxidative stress. The purpose of this study was to investigate the extent to which apoD interacts with lipoproteins in human plasma. By using detergent-free ELISA, we show that immobilized monoclonal antibodies against apoD very efficiently bind to low density lipoprotein (LDL) from plasma; this binding is as equally efficient as binding to an anti-apoB monoclonal antibody. Adding detergent to the plasma inhibited the binding, suggesting that the binding is dependent on the presence of intact lipoprotein particles. Reversing the system by using immobilized anti-apoB revealed that the affinity of apoD for LDL is rather low, suggesting that multiple bindings are needed for a durable connection. Biosensor experiments using purified lipoproteins also showed that purified apoD and high density lipoprotein 3 (HDL3), a lipoprotein fraction rich in apoD, were both able to bind LDL very efficiently, indicating that the HDL3-LDL interaction may be a physiological consequence of the affinity of apoD for LDL. Furthermore, we found that apoD increases the binding of HDL to actively growing T24 bladder carcinoma cells but not to quiescent, contact-inhibited, confluent T24 cells. This result is especially intriguing given that the T24 supernatant only contained detectable levels of apoD after growth inhibition, raising the possibility that alternating the expression of apoD and a putative apoD-receptor could give direction to the flow of lipids. In the current paper, we conclude that apoD mediates binding of HDL to LDL and to growing T24 carcinomas, thereby highlighting the importance of apoD in lipid metabolism.
Collapse
Affiliation(s)
| | - Lena Beckman
- Mabtech, Nacka Strand, Sweden
- Metabolism Unit, Department of Endocrinology, Metabolism and Diabetes, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | |
Collapse
|
41
|
Moskalev AA, Aliper AM, Smit-McBride Z, Buzdin A, Zhavoronkov A. Genetics and epigenetics of aging and longevity. Cell Cycle 2014; 13:1063-77. [PMID: 24603410 PMCID: PMC4013158 DOI: 10.4161/cc.28433] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Evolutionary theories of aging predict the existence of certain genes that provide selective advantage early in life with adverse effect on lifespan later in life (antagonistic pleiotropy theory) or longevity insurance genes (disposable soma theory). Indeed, the study of human and animal genetics is gradually identifying new genes that increase lifespan when overexpressed or mutated: gerontogenes. Furthermore, genetic and epigenetic mechanisms are being identified that have a positive effect on longevity. The gerontogenes are classified as lifespan regulators, mediators, effectors, housekeeping genes, genes involved in mitochondrial function, and genes regulating cellular senescence and apoptosis. In this review we demonstrate that the majority of the genes as well as genetic and epigenetic mechanisms that are involved in regulation of longevity are highly interconnected and related to stress response.
Collapse
Affiliation(s)
- Alexey A Moskalev
- Moscow Institute of Physics and Technology; Moscow, Russian Federation; Institute of Biology; Komi Science Center of Russian Academy of Sciences; Syktyvkar, Russian Federation; Syktyvkar State University; Syktyvkar, Russian Federation
| | - Alexander M Aliper
- Moscow Institute of Physics and Technology; Moscow, Russian Federation; Institute of Biology; Komi Science Center of Russian Academy of Sciences; Syktyvkar, Russian Federation
| | - Zeljka Smit-McBride
- Department of Ophthalmology and Vision Science; School of Medicine; University of California at Davis; Davis, CA USA
| | - Anton Buzdin
- Moscow Institute of Physics and Technology; Moscow, Russian Federation; Federal Clinical Research Center of Pediatric Hematology, Oncology, and Immunology; Moscow, Russian Federation; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry; Moscow, Russian Federation; First Oncology Research and Advisory Center; Moscow, Russian Federation
| | - Alex Zhavoronkov
- Moscow Institute of Physics and Technology; Moscow, Russian Federation; Federal Clinical Research Center of Pediatric Hematology, Oncology, and Immunology; Moscow, Russian Federation; The Biogerontology Research Foundation; London, UK
| |
Collapse
|
42
|
Lifelong expression of apolipoprotein D in the human brainstem: correlation with reduced age-related neurodegeneration. PLoS One 2013; 8:e77852. [PMID: 24167586 PMCID: PMC3805570 DOI: 10.1371/journal.pone.0077852] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/04/2013] [Indexed: 11/19/2022] Open
Abstract
The lipocalin apolipoprotein D (Apo D) is upregulated in peripheral nerves following injury and in regions of the central nervous system, such as the cerebral cortex, hippocampus, and cerebellum, during aging and progression of certain neurological diseases. In contrast, few studies have examined Apo D expression in the brainstem, a region necessary for survival and generally less prone to age-related degeneration. We measured Apo D expression in whole human brainstem lysates by slot-blot and at higher spatial resolution by quantitative immunohistochemistry in eleven brainstem nuclei (the 4 nuclei of the vestibular nuclear complex, inferior olive, hypoglossal nucleus, oculomotor nucleus, facial motor nucleus, nucleus of the solitary tract, dorsal motor nucleus of the vagus nerve, and Roller`s nucleus). In contrast to cortex, hippocampus, and cerebellum, apolipoprotein D was highly expressed in brainstem tissue from subjects (N = 26, 32−96 years of age) with no history of neurological disease, and expression showed little variation with age. Expression was significantly stronger in somatomotor nuclei (hypoglossal, oculomotor, facial) than visceromotor or sensory nuclei. Both neurons and glia expressed Apo D, particularly neurons with larger somata and glia in the periphery of these brainstem centers. Immunostaining was strongest in the neuronal perinuclear region and absent in the nucleus. We propose that strong brainstem expression of Apo D throughout adult life contributes to resistance against neurodegenerative disease and age-related degeneration, possibly by preventing oxidative stress and ensuing lipid peroxidation.
Collapse
|
43
|
Identification of apolipoprotein D as a cardioprotective gene using a mouse model of lethal atherosclerotic coronary artery disease. Proc Natl Acad Sci U S A 2013; 110:17023-8. [PMID: 24082102 DOI: 10.1073/pnas.1315986110] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mice with homozygous null mutations in the HDL receptor (scavenger receptor class B, type I, or SR-BI) and apolipoprotein E (apoE) genes [SR-BI/apoE double KO (SR-BI(-/-)/apoE(-/-) or dKO) mice] spontaneously develop occlusive, atherosclerotic coronary artery disease (CAD) and die prematurely (50% mortality at 42 d of age). Using microarray mRNA expression profiling, we identified genes whose expression in the hearts of dKO mice changed substantially during disease progression [at 21 d of age (no CAD), 31 d of age (small myocardial infarctions), and 43 d of age (extensive myocardial infarctions) vs. CAD-free SR-BI(+/-)/apoE(-/-) controls]. Expression of most genes that increased >sixfold in dKO hearts at 43 d also increased after coronary artery ligation. We examined the influence and potential mechanism of action of apolipoprotein D (apoD) whose expression in dKO hearts increased 80-fold by 43 d. Analysis of ischemia/reperfusion-induced myocardial infarction in both apoD KO mice and wild-type mice with abnormally high plasma levels of apoD (adenovirus-mediated hepatic overexpression) established that apoD reduces myocardial infarction. There was a correlation of apoD's ability to protect primary cultured rat cardiomyocytes from hypoxia/reoxygenation injury with its potent ability to inhibit oxidation in a standard antioxidation assay in vitro. We conclude that dKO mice represent a useful mouse model of CAD and apoD may be part of an intrinsic cardioprotective system, possibly as a consequence of its antioxidation activity.
Collapse
|
44
|
Wang X. Discovery of molecular associations among aging, stem cells, and cancer based on gene expression profiling. CHINESE JOURNAL OF CANCER 2013; 32:155-61. [PMID: 23298462 PMCID: PMC3618551 DOI: 10.5732/cjc.012.10114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The emergence of a huge volume of "omics" data enables a computational approach to the investigation of the biology of cancer. The cancer informatics approach is a useful supplement to the traditional experimental approach. I reviewed several reports that used a bioinformatics approach to analyze the associations among aging, stem cells, and cancer by microarray gene expression profiling. The high expression of aging- or human embryonic stem cell-related molecules in cancer suggests that certain important mechanisms are commonly underlying aging, stem cells, and cancer. These mechanisms are involved in cell cycle regulation, metabolic process, DNA damage response, apoptosis, p53 signaling pathway, immune/inflammatory response, and other processes, suggesting that cancer is a developmental and evolutional disease that is strongly related to aging. Moreover, these mechanisms demonstrate that the initiation, proliferation, and metastasis of cancer are associated with the deregulation of stem cells. These findings provide insights into the biology of cancer. Certainly, the findings that are obtained by the informatics approach should be justified by experimental validation. This review also noted that next-generation sequencing data provide enriched sources for cancer informatics study.
Collapse
Affiliation(s)
- Xiaosheng Wang
- National Cancer Institute, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
45
|
Jaiswal M, Sandoval H, Zhang K, Bayat V, Bellen HJ. Probing mechanisms that underlie human neurodegenerative diseases in Drosophila. Annu Rev Genet 2012; 46:371-96. [PMID: 22974305 DOI: 10.1146/annurev-genet-110711-155456] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The fruit fly, Drosophila melanogaster, is an excellent organism for the study of the genetic and molecular basis of metazoan development. Drosophila provides numerous tools and reagents to unravel the molecular and cellular functions of genes that cause human disease, and the past decade has witnessed a significant expansion of the study of neurodegenerative disease mechanisms in flies. Here we review the interplay between oxidative stress and neuronal toxicity. We cover some of the studies that show how proteasome degradation of protein aggregates, autophagy, mitophagy, and lysosomal function affect the quality control mechanisms required for neuronal survival. We discuss how forward genetic screens in flies have led to the isolation of a few loci that cause neurodegeneration, paving the way for large-scale systematic screens to identify such loci in flies as well as promoting gene discovery in humans.
Collapse
Affiliation(s)
- M Jaiswal
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
46
|
Wang X. Microarray analysis of ageing-related signatures and their expression in tumors based on a computational biology approach. GENOMICS PROTEOMICS & BIOINFORMATICS 2012; 10:136-41. [PMID: 22917186 PMCID: PMC3586943 DOI: 10.1016/j.gpb.2012.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 01/03/2012] [Accepted: 01/30/2012] [Indexed: 12/24/2022]
Abstract
Ageing and cancer have been associated with genetic and genomic changes. The identification of common signatures between ageing and cancer can reveal shared molecular mechanisms underlying them. In this study, we collected ageing-related gene signatures from ten published studies involved in six different human tissues and an online resource. We found that most of these gene signatures were tissue-specific and a few were related to multiple tissues. We performed a genome-wide examination of the expression of these signatures in various human tumor types, and found that a large proportion of these signatures were universally differentially expressed among normal vs. tumor phenotypes. Functional analyses of the highly-overlapping genes between ageing and cancer using DAVID tools have identified important functional categories and pathways linking ageing with cancer. The convergent and divergent mechanisms between ageing and cancer are discussed. This study provides insights into the biology of ageing and cancer, suggesting the possibility of potential interventions aimed at postponing ageing and preventing cancer.
Collapse
Affiliation(s)
- Xiaosheng Wang
- Biometric Research Branch, National Cancer Institute, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
47
|
Pajaniappan M, Glober NK, Kennard S, Liu H, Zhao N, Lilly B. Endothelial cells downregulate apolipoprotein D expression in mural cells through paracrine secretion and Notch signaling. Am J Physiol Heart Circ Physiol 2011; 301:H784-93. [PMID: 21705670 DOI: 10.1152/ajpheart.00116.2011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endothelial and mural cell interactions are vitally important for proper formation and function of blood vessels. These two cell types communicate to regulate multiple aspects of vessel function. In studying genes regulated by this interaction, we identified apolipoprotein D (APOD) as one gene that is downregulated in mural cells by coculture with endothelial cells. APOD is a secreted glycoprotein that has been implicated in governing stress response, lipid metabolism, and aging. Moreover, APOD is known to regulate smooth muscle cells and is found in abundance within atherosclerotic lesions. Our data show that the regulation of APOD in mural cells is bimodal. Paracrine secretion by endothelial cells causes partial downregulation of APOD expression. Additionally, cell contact-dependent Notch signaling plays a role. NOTCH3 on mural cells promotes the downregulation of APOD, possibly through interaction with the JAGGED-1 ligand on endothelial cells. Our results show that NOTCH3 contributes to the downregulation of APOD and by itself is sufficient to attenuate APOD transcript expression. In examining the consequence of decreased APOD expression in mural cells, we show that APOD negatively regulates cell adhesion. APOD attenuates adhesion by reducing focal contacts; however, it has no effect on stress fiber formation. These data reveal a novel mechanism in which endothelial cells control neighboring mural cells through the downregulation of APOD, which, in turn, influences mural cell function by modulating adhesion.
Collapse
|
48
|
Wakasaya Y, Kawarabayashi T, Watanabe M, Yamamoto-Watanabe Y, Takamura A, Kurata T, Murakami T, Abe K, Yamada K, Wakabayashi K, Sasaki A, Westaway D, Hyslop PSG, Matsubara E, Shoji M. Factors responsible for neurofibrillary tangles and neuronal cell losses in tauopathy. J Neurosci Res 2011; 89:576-84. [DOI: 10.1002/jnr.22572] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 11/08/2010] [Accepted: 11/09/2010] [Indexed: 11/09/2022]
|