1
|
Pérez-González AP, de Anda-Jáuregui G, Hernández-Lemus E. Differential Transcriptional Programs Reveal Modular Network Rearrangements Associated with Late-Onset Alzheimer's Disease. Int J Mol Sci 2025; 26:2361. [PMID: 40076979 PMCID: PMC11900169 DOI: 10.3390/ijms26052361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is a complex, genetically heterogeneous disorder. The diverse phenotypes associated with AD result from interactions between genetic and environmental factors, influencing multiple biological pathways throughout disease progression. Network-based approaches offer a way to assess phenotype-specific states. In this study, we calculated key network metrics to characterize the network transcriptional structure and organization in LOAD, focusing on genes and pathways implicated in AD pathology within the dorsolateral prefrontal cortex (DLPFC). Our findings revealed disease-specific coexpression markers associated with diverse metabolic functions. Additionally, significant differences were observed at both the mesoscopic and local levels between AD and control networks, along with a restructuring of gene coexpression and biological functions into distinct transcriptional modules. These results show the molecular reorganization of the transcriptional program occurring in LOAD, highlighting specific adaptations that may contribute to or result from cellular responses to pathological stressors. Our findings may support the development of a unified model for the causal mechanisms of AD, suggesting that its diverse manifestations arise from multiple pathways working together to produce the disease's complex clinical patho-phenotype.
Collapse
Affiliation(s)
- Alejandra Paulina Pérez-González
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Programa de Doctorado en Ciencias Biomédicas, Unidad de Posgrado Edificio B Primer Piso, Ciudad Universitaria, Mexico City 04510, Mexico
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Guillermo de Anda-Jáuregui
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Investigadores por M’exico, Conahcyt, Mexico City 03940, Mexico
| | - Enrique Hernández-Lemus
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
2
|
Tripathi A, Bartosh A, Mata J, Jacks C, Madeshiya AK, Hussein U, Hong LE, Zhao Z, Pillai A. Microglial type I interferon signaling mediates chronic stress-induced synapse loss and social behavior deficits. Mol Psychiatry 2025; 30:423-434. [PMID: 39095477 DOI: 10.1038/s41380-024-02675-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
Inflammation and synapse loss have been associated with deficits in social behavior and are involved in pathophysiology of many neuropsychiatric disorders. Synapse loss, characterized by reduction in dendritic spines can significantly disrupt synaptic connectivity and neural circuitry underlying social behavior. Chronic stress is known to induce loss of spines and dendrites in the prefrontal cortex (PFC), a brain region implicated in social behavior. However, the underlying mechanisms are not well understood. In the present study, we investigated the role of type I Interferon (IFN-I) signaling in chronic unpredictable stress (CUS)-induced synapse loss and behavior deficits in mice. We found increased expression of type I IFN receptor (IFNAR) in microglia following CUS. Conditional knockout of microglial IFNAR in adult mice rescued CUS-induced social behavior deficits and synapse loss. Bulk RNA sequencing data show that microglial IFNAR deletion attenuated CUS-mediated changes in the expression of genes such as Keratin 20 (Krt20), Claudin-5 (Cldn5) and Nuclear Receptor Subfamily 4 Group A Member 1 (Nr4a1) in the PFC. Cldn5 and Nr4a1 are known for their roles in synaptic plasticity. Krt20 is an intermediate filament protein responsible for the structural integrity of epithelial cells. The reduction in Krt20 following CUS presents a novel insight into the potential contribution of cytokeratin in stress-induced alterations in neuroplasticity. Overall, these results suggest that microglial IFNAR plays a critical role in regulating synaptic plasticity and social behavior deficits associated with chronic stress conditions.
Collapse
Affiliation(s)
- Ashutosh Tripathi
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alona Bartosh
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jocelyn Mata
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chale Jacks
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Amit Kumar Madeshiya
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Usama Hussein
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Lymphoma & Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Elliot Hong
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhongming Zhao
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anilkumar Pillai
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
3
|
Li Y, Zeng M, Qin Y, Feng F, Wei H. The role of KRT18 in lung adenocarcinoma development: integrative bioinformatics and experimental validation. Discov Oncol 2024; 15:841. [PMID: 39729139 DOI: 10.1007/s12672-024-01728-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Lung adenocarcinoma (LUAD) represents one of the most common subtypes of lung cancer with high rates of incidence and mortality, which contributes to substantial health and economic demand across the globe. Treatment today mainly consists of surgery, radiotherapy, and chemotherapy, but their efficacy in advanced stages is often suboptimal and emphasizes the clear need for new biomarkers and therapeutic targets. Using comprehensive bioinformatics analyses consisting of the Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), Human Protein Atlas (HPA) and Clinical Proteomic Tumor Analysis Consortium (CPTAC), immune infiltration analysis and functional enrichment analysis, and single-cell analysis, we examined the potential of keratin 18 (KRT18) as a candidate biomarker in advanced LUAD. KRT18 was significantly elevated in LUAD tissue relative to normal adjacent tissue (p < 0.05), and its expression was correlated with poor clinical-pathological features and inferior prognostic outcome. Furthermore, KRT18 expression was associated with several populations of immune cells, suggesting KRT18 may contribute to the local tumor microenvironment and potentially pathways of immune evasion. Survival analysis indicated that elevated KRT18 expression correlated with poor overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI), reinforcing its legitimacy as a prognostic tool (AUC = 0.846). Importantly, gene enrichment analysis found KRT18-associated genes enriched for pathways associated with lymphocyte differentiation and immune response pathways, which provides mechanistic insight into biological effects attributed to KRT18. Notably, NU.1025 has demonstrated the capability of reversing KRT18-modulated oncogenic features, and targeted therapeutic strategies can be developed moving forward. In conclusion, our data demonstrate that KRT18 has utility as a potential biomarker but may also serve as a therapeutic target in LUAD and merit further investigation into underlying mechanistic functions and potential therapeutic roles in the clinic.
Collapse
Affiliation(s)
- Yongjie Li
- School of Pharmacy, Shaoyang University, Shaoyang, 422000, Hunan, China
- Southwest Hunan Research Center of Engineering for Development and Utilization of Traditional Chinese Medicine, Shaoyang, 422000, Hunan, China
| | - Min Zeng
- Department of Respiratory and Critical Care Medicine, The Affiliated Shaoyang Hospital, Hengyang Medical School, University of South China, Shaoyang, 422000, Hunan, China.
| | - Yinan Qin
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, Zhejiang, China
| | - Fen Feng
- School of Pharmacy, Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Hailiang Wei
- School of Pharmacy, Shaoyang University, Shaoyang, 422000, Hunan, China
| |
Collapse
|
4
|
Li W, Pergande MR, Crutchfield CA, Searle BC, Backlund PS, Picache JA, Burkert K, Yanjanin-Farhat NM, Blank PS, Toth CL, Wassif CA, Porter FD, Cologna SM. A differential proteomics study of cerebrospinal fluid from individuals with Niemann-Pick disease, Type C1. Proteomics 2023; 23:e2200378. [PMID: 36638187 PMCID: PMC10918788 DOI: 10.1002/pmic.202200378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/18/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Niemann-Pick, type C1 (NPC1) is a fatal, neurodegenerative disease, which belongs to the family of lysosomal diseases. In NPC1, endo/lysosomal accumulation of unesterified cholesterol and sphingolipids arise from improper intracellular trafficking resulting in multi-organ dysfunction. With the proximity between the brain and cerebrospinal fluid (CSF), performing differential proteomics provides a means to shed light to changes occurring in the brain. In this study, CSF samples obtained from NPC1 individuals and unaffected controls were used for protein biomarker identification. A subset of these individuals with NPC1 are being treated with miglustat, a glycosphingolipid synthesis inhibitor. Of the 300 identified proteins, 71 proteins were altered in individuals with NPC1 compared to controls including cathepsin D, and members of the complement family. Included are a report of 10 potential markers for monitoring therapeutic treatment. We observed that pro-neuropeptide Y (NPY) was significantly increased in NPC1 individuals relative to healthy controls; however, individuals treated with miglustat displayed levels comparable to healthy controls. In further investigation, NPY levels in a NPC1 mouse model corroborated our findings. We posit that NPY could be a potential therapeutic target for NPC1 due to its multiple roles in the central nervous system such as attenuating neuroinflammation and reducing excitotoxicity.
Collapse
Affiliation(s)
- Wenping Li
- Department of Chemistry, University of Illinois Chicago
| | | | - Christopher A. Crutchfield
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Brian C. Searle
- Department of Biomedical Informatics, The Ohio State University Medical Center
| | - Peter S. Backlund
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Jaqueline A. Picache
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Kathryn Burkert
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Nicole M. Yanjanin-Farhat
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Paul S. Blank
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Cynthia L. Toth
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Christopher A. Wassif
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | - Forbes D. Porter
- Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health
| | | |
Collapse
|
5
|
François M, Karpe AV, Liu JW, Beale DJ, Hor M, Hecker J, Faunt J, Maddison J, Johns S, Doecke JD, Rose S, Leifert WR. Multi-Omics, an Integrated Approach to Identify Novel Blood Biomarkers of Alzheimer's Disease. Metabolites 2022; 12:949. [PMID: 36295851 PMCID: PMC9610280 DOI: 10.3390/metabo12100949] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
The metabolomic and proteomic basis of mild cognitive impairment (MCI) and Alzheimer's disease (AD) is poorly understood, and the relationships between systemic abnormalities in metabolism and AD/MCI pathogenesis is unclear. This study compared the metabolomic and proteomic signature of plasma from cognitively normal (CN) and dementia patients diagnosed with MCI or AD, to identify specific cellular pathways and new biomarkers altered with the progression of the disease. We analysed 80 plasma samples from individuals with MCI or AD, as well as age- and gender-matched CN individuals, by utilising mass spectrometry methods and data analyses that included combined pathway analysis and model predictions. Several proteins clearly identified AD from the MCI and CN groups and included plasma actins, mannan-binding lectin serine protease 1, serum amyloid A2, fibronectin and extracellular matrix protein 1 and Keratin 9. The integrated pathway analysis showed various metabolic pathways were affected in AD, such as the arginine, alanine, aspartate, glutamate and pyruvate metabolism pathways. Therefore, our multi-omics approach identified novel plasma biomarkers for the MCI and AD groups, identified changes in metabolic processes, and may form the basis of a biomarker panel for stratifying dementia participants in future clinical trials.
Collapse
Affiliation(s)
- Maxime François
- CSIRO Health & Biosecurity, Human Health Program, Molecular Diagnostic Solutions Group, Adelaide, SA 5000, Australia
| | - Avinash V. Karpe
- CSIRO Land & Water, Metabolomics Unit, Ecosciences Precinct, Dutton Park, QLD 4001, Australia
| | - Jian-Wei Liu
- CSIRO Land & Water, Agricultural and Environmental Sciences Precinct, Acton, Canberra, ACT 2601, Australia
| | - David J. Beale
- CSIRO Land & Water, Metabolomics Unit, Ecosciences Precinct, Dutton Park, QLD 4001, Australia
| | - Maryam Hor
- CSIRO Health & Biosecurity, Human Health Program, Molecular Diagnostic Solutions Group, Adelaide, SA 5000, Australia
| | - Jane Hecker
- Department of Internal Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Jeff Faunt
- Department of General Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - John Maddison
- Aged Care Rehabilitation & Palliative Care, SA Health, Modbury Hospital, Modbury, SA 5092, Australia
| | - Sally Johns
- Aged Care Rehabilitation & Palliative Care, SA Health, Modbury Hospital, Modbury, SA 5092, Australia
| | - James D. Doecke
- Australian e-Health Research Centre, CSIRO, Level 7, Surgical Treatment and Rehabilitation Service—STARS, Herston, QLD 4029, Australia
| | - Stephen Rose
- Australian e-Health Research Centre, CSIRO, Level 7, Surgical Treatment and Rehabilitation Service—STARS, Herston, QLD 4029, Australia
| | - Wayne R. Leifert
- CSIRO Health & Biosecurity, Human Health Program, Molecular Diagnostic Solutions Group, Adelaide, SA 5000, Australia
| |
Collapse
|
6
|
Xiong W, Cai J, Li R, Wen C, Tan H. Rare Variant Analysis and Molecular Dynamics Simulation in Alzheimer’s Disease Identifies Exonic Variants in FLG. Genes (Basel) 2022; 13. [PMID: 35627223 PMCID: PMC9141140 DOI: 10.3390/genes13050838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Although an increasing number of common variants contributing to Alzheimer’s disease (AD) are uncovered by genome-wide association studies, they can only explain less than half of the heritability of AD. Rare variant association studies (RVAS) has become an increasingly important area to explain the risk or trait variability of AD. Method: To investigate the potential rare variants that cause AD, we screened 70,209 rare variants from two cohorts of a 175 AD cohort and a 214 cognitively normal cohort from the Alzheimer’s Disease Neuroimaging Initiative database. MIRARE, a novel RVAS method, was performed on 232 non-synonymous variants selected by ANNOVAR annotation. Molecular docking and molecular dynamics (MD) simulation were adopted to verify the interaction between the chosen functional variants and BACE1. Results: MIRAGE analysis revealed significant associations between AD and six potential pathogenic genes, including PREX2, FLG, DHX16, NID2, ZnF585B and ZnF875. Only interactions between FLG (including wild type and rs3120654(SER742TYR)) and BACE1 were verified by molecular docking and MD simulation. The interaction of FLG(SER742TYR) with BACE1 was greater than that of wildtype FLG with BACE1. Conclusions: According to the literature search, bio-informatics analysis, and molecular docking and MD simulation, we find non-synonymous rare variants in six genes, especially FLG(rs3120654), that may play key roles in AD.
Collapse
Affiliation(s)
- Weixue Xiong
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515000, China
| | - Jiahui Cai
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515000, China
| | - Ruijia Li
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei 230000, China;
| | - Canhong Wen
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei 230000, China
| | - Haizhu Tan
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515000, China
| | | |
Collapse
|
7
|
Wang YJ, Downey MA, Choi S, Shoup TM, Elmaleh DR. Cromolyn platform suppresses fibrosis and inflammation, promotes microglial phagocytosis and neurite outgrowth. Sci Rep 2021; 11:22161. [PMID: 34772945 PMCID: PMC8589953 DOI: 10.1038/s41598-021-00465-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases are characterized by chronic neuroinflammation and may perpetuate ongoing fibrotic reactions within the central nervous system. Unfortunately, there is no therapeutic available that treats neurodegenerative inflammation and its sequelae. Here we utilize cromolyn, a mast cell inhibitor with anti-inflammatory capabilities, and its fluorinated analogue F-cromolyn to study fibrosis-related protein regulation and secretion downstream of neuroinflammation and their ability to promote microglial phagocytosis and neurite outgrowth. In this report, RNA-seq analysis shows that administration of the pro-inflammatory cytokine TNF-α to HMC3 human microglia results in a robust upregulation of fibrosis-associated genes. Subsequent treatment with cromolyn and F-cromolyn resulted in reduced secretion of collagen XVIII, fibronectin, and tenascin-c. Additionally, we show that cromolyn and F-cromolyn reduce pro-inflammatory proteins PLP1, PELP1, HSP90, IL-2, GRO-α, Eotaxin, and VEGF-Α, while promoting secretion of anti-inflammatory IL-4 in HMC3 microglia. Furthermore, cromolyn and F-cromolyn augment neurite outgrowth in PC12 neuronal cells in concert with nerve growth factor. Treatment also differentially altered secretion of neurogenesis-related proteins TTL, PROX1, Rab35, and CSDE1 in HMC3 microglia. Finally, iPSC-derived human microglia more readily phagocytose Aβ42 with cromolyn and F-cromolyn relative to controls. We propose the cromolyn platform targets multiple proteins upstream of PI3K/Akt/mTOR, NF-κB, and GSK-3β signaling pathways to affect cytokine, chemokine, and fibrosis-related protein expression.
Collapse
Affiliation(s)
| | | | - Sungwoon Choi
- Department of New Drug Discovery, Chungnam National University, Daejeon, South Korea
| | - Timothy M Shoup
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129-2060, USA
| | - David R Elmaleh
- AZTherapies, Inc., Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129-2060, USA.
| |
Collapse
|
8
|
Gęgotek A, Atalay S, Skrzydlewska E. UV induced changes in proteome of rats plasma are reversed by dermally applied cannabidiol. Sci Rep 2021; 11:20666. [PMID: 34667212 PMCID: PMC8526570 DOI: 10.1038/s41598-021-00134-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/07/2021] [Indexed: 11/20/2022] Open
Abstract
UV radiation is known to induce a multiple changes in the metabolism of skin-building cells, what can affect the functioning not only neighboring cells, but also, following signal transduction releasing into the blood vessels, the entire body. Therefore, the aim of this study was to analyze the proteomic disturbances occurred in plasma of chronically UVA/UVB irradiated rats and define the effect on these changes of skin topically applied cannabidiol (CBD). Obtained results showed significant changes in the expression of numerous anti-inflammatory and signaling proteins including: NFκB inhibitor, 14-3-3 protein, protein kinase C, keratin, and protein S100 after UV irradiation and CBD treatment. Moreover, the effects of UVA and UVB were manifested by increased level of lipid peroxidation products-protein adducts formation. CBD partially prevented all of these changes, but in a various degree depending on the UV radiation type. Moreover, topical treatment with CBD resulted in the penetration of CBD into the blood and, as a consequence, in direct modifications to the plasma protein structure by creating CBD adducts with molecules, such as proline-rich protein 30, transcription factor 19, or N-acetylglucosamine-6-sulfatase, what significantly changed the activity of these proteins. In conclusion, it may be suggested that CBD applied topically may be an effective compound against systemic UV-induced oxidative stress, but its effectiveness requires careful analysis of CBD's effects on other tissues of the living organism.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Sinemyiz Atalay
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| |
Collapse
|
9
|
Herzog DP, Perumal N, Manicam C, Treccani G, Nadig J, Rossmanith M, Engelmann J, Jene T, Hasch A, van der Kooij MA, Lieb K, Gassen NC, Grus FH, Müller MB. Longitudinal CSF proteome profiling in mice to uncover the acute and sustained mechanisms of action of rapid acting antidepressant (2R,6R)-hydroxynorketamine (HNK). Neurobiol Stress 2021; 15:100404. [PMID: 34632008 PMCID: PMC8488754 DOI: 10.1016/j.ynstr.2021.100404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/16/2021] [Accepted: 09/26/2021] [Indexed: 02/04/2023] Open
Abstract
Delayed onset of antidepressant action is a shortcoming in depression treatment. Ketamine and its metabolite (2R,6R)-hydroxynorketamine (HNK) have emerged as promising rapid-acting antidepressants. However, their mechanism of action remains unknown. In this study, we first described the anxious and depression-prone inbred mouse strain, DBA/2J, as an animal model to assess the antidepressant-like effects of ketamine and HNK in vivo. To decode the molecular mechanisms mediating HNK's rapid antidepressant effects, a longitudinal cerebrospinal fluid (CSF) proteome profiling of its acute and sustained effects was conducted using an unbiased, hypothesis-free mass spectrometry-based proteomics approach. A total of 387 proteins were identified, with a major implication of significantly differentially expressed proteins in the glucocorticoid receptor (GR) signaling pathway, providing evidence for a link between HNK and regulation of the stress hormone system. Mechanistically, we identified HNK to repress GR-mediated transcription and reduce hormonal sensitivity of GR in vitro. In addition, mammalian target of rapamycin (mTOR) and brain-derived neurotrophic factor (BDNF) were predicted to be important upstream regulators of HNK treatment. Our results contribute to precise understanding of the temporal dynamics and molecular targets underlying HNK's rapid antidepressant-like effects, which can be used as a benchmark for improved treatment strategies for depression in future.
Collapse
Affiliation(s)
- David P Herzog
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Natarajan Perumal
- Experimental and Translational Ophthalmology, Department of Ophthalmology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Caroline Manicam
- Experimental and Translational Ophthalmology, Department of Ophthalmology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Giulia Treccani
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany.,Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg University Medical Center, Mainz, Germany.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Jens Nadig
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Milena Rossmanith
- Experimental and Translational Ophthalmology, Department of Ophthalmology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Jan Engelmann
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Tanja Jene
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Annika Hasch
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Michael A van der Kooij
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany.,Leibniz Institute for Resilience Research, Mainz, Germany
| | - Klaus Lieb
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany.,Leibniz Institute for Resilience Research, Mainz, Germany
| | - Nils C Gassen
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Medical Center Bonn, Bonn, Germany
| | - Franz H Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Marianne B Müller
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany.,Leibniz Institute for Resilience Research, Mainz, Germany
| |
Collapse
|
10
|
Khan MJ, Desaire H, Lopez OL, Kamboh MI, Robinson RA. Why Inclusion Matters for Alzheimer's Disease Biomarker Discovery in Plasma. J Alzheimers Dis 2021; 79:1327-1344. [PMID: 33427747 PMCID: PMC9126484 DOI: 10.3233/jad-201318] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND African American/Black adults have a disproportionate incidence of Alzheimer's disease (AD) and are underrepresented in biomarker discovery efforts. OBJECTIVE This study aimed to identify potential diagnostic biomarkers for AD using a combination of proteomics and machine learning approaches in a cohort that included African American/Black adults. METHODS We conducted a discovery-based plasma proteomics study on plasma samples (N = 113) obtained from clinically diagnosed AD and cognitively normal adults that were self-reported African American/Black or non-Hispanic White. Sets of differentially-expressed proteins were then classified using a support vector machine (SVM) to identify biomarker candidates. RESULTS In total, 740 proteins were identified of which, 25 differentially-expressed proteins in AD came from comparisons within a single racial and ethnic background group. Six proteins were differentially-expressed in AD regardless of racial and ethnic background. Supervised classification by SVM yielded an area under the curve (AUC) of 0.91 and accuracy of 86%for differentiating AD in samples from non-Hispanic White adults when trained with differentially-expressed proteins unique to that group. However, the same model yielded an AUC of 0.49 and accuracy of 47%for differentiating AD in samples from African American/Black adults. Other covariates such as age, APOE4 status, sex, and years of education were found to improve the model mostly in the samples from non-Hispanic White adults for classifying AD. CONCLUSION These results demonstrate the importance of study designs in AD biomarker discovery, which must include diverse racial and ethnic groups such as African American/Black adults to develop effective biomarkers.
Collapse
Affiliation(s)
- Mostafa J. Khan
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Heather Desaire
- Department of Chemistry, University of Kansas, Lawrence, KS, USA
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - M. Ilyas Kamboh
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Renã A.S. Robinson
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
11
|
Zhang D, Dong X, Liu X, Ye L, Li S, Zhu R, Ye Y, Jiang Y. Proteomic Analysis of Brain Regions Reveals Brain Regional Differences and the Involvement of Multiple Keratins in Chronic Alcohol Neurotoxicity. Alcohol Alcohol 2020; 55:147-156. [PMID: 32047899 DOI: 10.1093/alcalc/agaa007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/22/2019] [Accepted: 01/13/2020] [Indexed: 12/29/2022] Open
Abstract
AIMS Alcohol abuse has attracted public attention and chronic alcohol exposure can result in irreversible structural changes in the brain. The molecular mechanisms underlying alcohol neurotoxicity are complex, mandating comprehensive mining of spatial protein expression profile. METHODS In this study, mice models of chronic alcohol intoxication were established after 95% alcohol vapor administration for 30 consecutive days. On Day 30, striatum (the dorsal and ventral striatum) and hippocampus, the two major brain regions responsible for learning and memorizing while being sensitive to alcohol toxicity, were collected. After that, isobaric tags for relative and absolute quantitation -based quantitative proteomic analysis were carried out for further exploration of the novel mechanisms underlying alcohol neurotoxicity. RESULTS Proteomic results showed that in the striatum, 29 proteins were significantly up-regulated and 17 proteins were significantly down-regulated. In the hippocampus, 72 proteins were significantly up-regulated, while 2 proteins were significantly down-regulated. Analysis of the overlay proteins revealed that a total of 102 proteins were consistently altered (P < 0.05) in both hippocampus and striatum regions, including multiple keratins such as Krt6a, Krt17 and Krt5. Ingenuity pathway analysis revealed that previously reported diseases/biofunctions such as dermatological diseases and developmental disorders were enriched in those proteins. Interestingly, the glucocorticoid receptor (GR) signaling was among the top enriched pathways in both brain regions, while multiple keratins from the GR signaling such as Krt1 and Krt17 exhibited significantly opposite expression patterns in the two brain nuclei. Moreover, there are several other involved pathways significantly differed between the hippocampus and striatum. CONCLUSIONS Our data revealed brain regional differences upon alcohol consumption and indicated the critical involvement of keratins from GR signaling in alcohol neurotoxicity. The differences in proteomic results between the striatum and hippocampus suggested a necessity of taking into consideration brain regional differences and intertwined signaling pathways rather than merely focusing on single nuclei or molecule during the study of drug-induced neurotoxicity in the future.
Collapse
Affiliation(s)
- Dingang Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoru Dong
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaochen Liu
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lin Ye
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shuhao Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Rongzhe Zhu
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yonghong Ye
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yan Jiang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Carbonate Apatite and Hydroxyapatite Formulated with Minimal Ingredients to Deliver SiRNA into Breast Cancer Cells In Vitro and In Vivo. J Funct Biomater 2020; 11:jfb11030063. [PMID: 32927738 PMCID: PMC7565062 DOI: 10.3390/jfb11030063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/24/2020] [Accepted: 09/07/2020] [Indexed: 01/22/2023] Open
Abstract
Introduction: Cancer is one of the top-ranked noncommunicable diseases causing deaths to nine million people and affecting almost double worldwide in 2018. Tremendous advancement in surgery, chemotherapy, radiation and targeted immunotherapy have improved the rate of cure and disease-free survival. As genetic mutations vary in different cancers, potential of customized treatment to silence the problem gene/s at the translational level is being explored too. Yet delivering therapeutics at the required dosage only to the affected cells without affecting the healthy ones, is a big hurdle to be overcome. Scientists worldwide have been working to invent a smart drug delivery system for targeted delivery of therapeutics to tumor tissues only. As part of such an effort, few organic nanocarriers went to clinical trials, while inorganic nanoparticles (NPs) are still in development stage despite their many customizable properties. Carbonate apatite (CA), a pH sensitive nanocarrier has emerged as an efficient delivery system for drugs, plasmids and siRNAs in preclinical models of breast and colon cancers. Like hydroxyapatite (HA) which serves as a classical tool for delivery of genetic materials such as siRNA and plasmid, CA is an apatite-based synthetic carrier. We developed simplified methods of formulating CA-in-DMEM and a DMEM-mimicking buffer and HA in a HEPES-buffered solution and characterized them in terms of size, stability, protein corona (PC) composition, cytotoxicity, siRNA delivery efficiency in breast cancer cells and siRNA biodistribution profile in a mouse model of breast cancer. Methods: Particle growth was analyzed via spectrophotometry and light microscopy, size was measured via dynamic light scattering and scanning electron microscopy and confirmation of functional groups in apatite structures was made by FT-IR. siRNA-binding was analyzed via spectrophotometry. Stability of the formulation solutions/buffers was tested over various time points and at different temperatures to determine their compatibility in the context of practical usage. Cellular uptake was studied via fluorescence microscopy. MTT assay was performed to measure the cytotoxicity of the NPs. Liquid chromatography—mass spectrometry was carried out to analyze the PC formed around all three different NPs in serum-containing media. To explore biodistribution of all the formulations, fluorescence-labeled siRNA-loaded NPs were administered intravenously prior to analysis of fluorescence intensity in the collected organs and tumors of the treated mice. Results: The size of NPs in 10% serum-containing media was dramatically different where CA-in-DMB and HA were much larger than CA-in-DMEM. Effect of media was notable on the PC composition of all three NPs. All three NPs bound albumin and some common protease inhibitors involved in bone metabolism due to their compositional similarity to our bone materials. Moreover, CA also bound heme-binding proteins and opsonins. Unlike CA, HA bound different kinds of keratins. Difference in PC constitution was likely to influence accumulation of NPs in various organs including those of reticuloendothelial system, such as liver and spleen and the tumor. We found 10 times more tumor accumulation of CA-in-DMB than CA-in-DMEM, which could be due to more stable siRNA-binding and distinct PC composition of the former. Conclusion: As a nanocarrier CA is more efficient than HA for siRNA delivery to the tumor. CA prepared in a buffer containing only the mere constituents was potentially more efficient than classical CA prepared in DMEM, owing to the exclusion of interference attributed by the inorganic ions and organic molecules present in DMEM.
Collapse
|
13
|
Screening for the Proteins That Can Interact with Grouper Nervous Necrosis Virus Capsid Protein. Viruses 2020; 12:v12090985. [PMID: 32899810 PMCID: PMC7552068 DOI: 10.3390/v12090985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/29/2022] Open
Abstract
Nervous necrosis virus (NNV) can infect many species of fish and has an 80-100% mortality rate. NNV capsid protein (NNVCP) is the only structural protein of NNV, but there are few studies on the protein-protein interaction between NNVCP and the host cell. To investigate NNV morphogenesis, native NNV capsid protein (NNVCP) was used to screen for protein-protein interactions in this study. The results identified that 49 grouper optic nerve proteins can interact with NNVCP and may function as putative receptor or co-receptor, cytoskeleton, glucose metabolism and ATP generation, immunity, mitochondrial ion regulation, and ribosomal proteins. Creatine kinase B-type (CKB) is one of those 49 optic nerve proteins. CKB, a kind of enzyme of ATP generation, was confirmed to interact with NNVCP by far-Western blot and showed to colocalize with NNVCP in GF-1 cells. Compared to the control, the expression of CKB was significantly induced in the brain and eyes infected with NNV. Moreover, the amount of replication of NNV is relatively high in cells expressing CKB. In addition to providing the database of proteins that can interact with NNVCP for subsequent analysis, the results of this research also verified that CKB plays an important role in the morphogenesis of NNV.
Collapse
|
14
|
Zolochevska O, Bjorklund N, Woltjer R, Wiktorowicz JE, Taglialatela G. Postsynaptic Proteome of Non-Demented Individuals with Alzheimer's Disease Neuropathology. J Alzheimers Dis 2019; 65:659-682. [PMID: 30103319 PMCID: PMC6130411 DOI: 10.3233/jad-180179] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Some individuals, here referred to as Non-Demented with Alzheimer’s Neuropathology (NDAN), retain their cognitive function despite the presence of amyloid plaques and tau tangles typical of symptomatic Alzheimer’s disease (AD). In NDAN, unlike AD, toxic amyloid-β oligomers do not localize to the postsynaptic densities (PSDs). Synaptic resistance to amyloid-β in NDAN may thus enable these individuals to remain cognitively intact despite the AD-like pathology. The mechanism(s) responsible for this resistance remains unresolved and understanding such protective biological processes could reveal novel targets for the development of effective treatments for AD. The present study uses a proteomic approach to compare the hippocampal postsynaptic densities of NDAN, AD, and healthy age-matched persons to identify protein signatures characteristic for these groups. Subcellular fractionation followed by 2D gel electrophoresis and mass spectrometry were used to analyze the PSDs. We describe fifteen proteins which comprise the unique proteomic signature of NDAN PSDs, thus setting them apart from control subjects and AD patients.
Collapse
Affiliation(s)
- Olga Zolochevska
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nicole Bjorklund
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Randall Woltjer
- Department of Pathology, Oregon Health and Science University, Portland, OR, USA
| | - John E Wiktorowicz
- Department of Biochemistry and Molecular Biology, Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Giulio Taglialatela
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
15
|
Saffarian A, Tarokh A, Reza Haghshenas M, Taghipour M, Chenari N, Ghaderi A, Razmkhah M. Proteomics Study of Mesenchymal Stem Cell-Like Cells Isolated from Cerebrospinal Fluid of Patients with Meningioma. CURR PROTEOMICS 2019. [DOI: 10.2174/1570164616666190204161453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:Cerebrospinal fluid (CSF) contains pro-growth factors that can affect proliferation, migration and differentiation of Mesenchymal Stem Cells (MSCs).Objective:This study aimed to isolate MSC like cells from CSF of patients with meningioma and psudotumorcerebri (PTC) and identify differentially expressed proteins in these cells.Methods:Five patients with newly diagnosed intracranial meningioma and five patients with PTC were recruited in this comparative proteomics study. MSCs were isolated from CSF and validated by mesenchyml and non-mesenchyml fluorochrome antibodies, and flow cytometer analysis. Two- Dimensional Gel Electrophoresis (2-DE) coupled with Mass Spectrometry (MS) was performed to identify differentially expressed proteins.Results:Microscopic views of the isolated cells as well as flow cytometer analysis were found to be compatible with MSC-like cells. Eight distinct protein spots were differentially and reproducibly expressed among the stained gels of two studied groups. The identified proteins were Phosphoglycerate Mutase 1 (PGAM1), LIM and SH3 domain protein (LASP1), peroxiredoxin-6 (PRDX-6), type I cytoskeletal 9 (KRT9), Superoxide Dismutase (SOD), endoplasmin, Stathmin 1 (STMN1), and glutathione S-transferase (GST).Conclusion:This study provides new insights into the plausible role of CSF derived MSCs in cancer progression, and reveals a promising therapeutic opportunity for targeting of MSC proteins in patients with meningioma.
Collapse
Affiliation(s)
- Arash Saffarian
- Department of Neurosurgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tarokh
- Department of Neurosurgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Haghshenas
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mousa Taghipour
- Department of Neurosurgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nooshafarin Chenari
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
Transcriptional Analysis Shows a Robust Host Response to Toxoplasma gondii during Early and Late Chronic Infection in Both Male and Female Mice. Infect Immun 2019; 87:IAI.00024-19. [PMID: 30858341 DOI: 10.1128/iai.00024-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/01/2019] [Indexed: 01/01/2023] Open
Abstract
The long-term host effects caused by the protozoan parasite Toxoplasma gondii are poorly understood. High-throughput RNA sequencing analysis previously determined that the host response in the brain was greater and more complex at 28 days than at 10 days postinfection. Here, we analyzed the host transcriptional profile of age- and sex-matched mice during very early (21 days), early (28 days), mid (3 months), and late (6 months) chronic infection. We found that a majority of the host genes which increase in abundance at day 21 postinfection are still increased at 6 months postinfection for both male and female mice. While most of the differentially expressed genes were similar between sexes, females had far fewer genes that were significantly less abundant, which may have led to the slightly increased cyst burden in males. Transcripts for C-X-C motif chemokine ligand 13 and a C-C motif chemokine receptor 2 (CCR2) were significantly higher in females than in males during infection. As T. gondii chronic infection and profilin (PRF) confer resistance to Listeria monocytogenes infection in a CCR2-dependent manner, the differences in CCR2 expression led us to retest the protection of PRF in both sexes. Male mice were nearly as effective as female mice at reducing the bacterial burden either with a chronic infection or when treated with PRF. These data show that most of the host genes differentially expressed in response to T. gondii infection are similar between males and females. While differences in transcript abundance exist between the sexes, the infection phenotypes tested here did not show significant differences.
Collapse
|
17
|
Deng L, Pushpitha K, Joseph C, Gupta V, Rajput R, Chitranshi N, Dheer Y, Amirkhani A, Kamath K, Pascovici D, Wu JX, Salekdeh GH, Haynes PA, Graham SL, Gupta VK, Mirzaei M. Amyloid β Induces Early Changes in the Ribosomal Machinery, Cytoskeletal Organization and Oxidative Phosphorylation in Retinal Photoreceptor Cells. Front Mol Neurosci 2019; 12:24. [PMID: 30853886 PMCID: PMC6395395 DOI: 10.3389/fnmol.2019.00024] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 01/21/2019] [Indexed: 01/20/2023] Open
Abstract
Amyloid β (Aβ) accumulation and its aggregation is characteristic molecular feature of the development of Alzheimer's disease (AD). More recently, Aβ has been suggested to be associated with retinal pathology associated with AD, glaucoma and drusen deposits in age related macular degeneration (AMD). In this study, we investigated the proteins and biochemical networks that are affected by Aβ in the 661 W photoreceptor cells in culture. Time and dose dependent effects of Aβ on the photoreceptor cells were determined utilizing tandem mass tag (TMT) labeling-based quantitative mass-spectrometric approach. Bioinformatic analysis of the data revealed concentration and time dependent effects of the Aβ peptide stimulation on various key biochemical pathways that might be involved in mediating the toxicity effects of the peptide. We identified increased Tau phosphorylation, GSK3β dysregulation and reduced cell viability in cells treated with Aβ in a dose and time dependent manner. This study has delineated for the first-time molecular networks in photoreceptor cells that are impacted early upon Aβ treatment and contrasted the findings with a longer-term treatment effect. Proteins associated with ribosomal machinery homeostasis, mitochondrial function and cytoskeletal organization were affected in the initial stages of Aβ exposure, which may provide key insights into AD effects on the photoreceptors and specific molecular changes induced by Aβ peptide.
Collapse
Affiliation(s)
- Liting Deng
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Kanishka Pushpitha
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Chitra Joseph
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Waurn Ponds, VIC, Australia
| | - Rashi Rajput
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Yogita Dheer
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ardeshir Amirkhani
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| | - Karthik Kamath
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| | - Dana Pascovici
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| | - Jemma X. Wu
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Cell Science Research Center, Department of Molecular Systems Biology, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Paul A. Haynes
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Stuart L. Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Vivek K. Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
18
|
Andolino C, Hess C, Prince T, Williams H, Chernin M. Drug-induced keratin 9 interaction with Hsp70 in bladder cancer cells. Cell Stress Chaperones 2018; 23:1137-1142. [PMID: 29802537 PMCID: PMC6111075 DOI: 10.1007/s12192-018-0913-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 04/05/2018] [Accepted: 05/08/2018] [Indexed: 12/18/2022] Open
Abstract
A pull-down experiment (co-immunoprecipitation) was performed on a T24 human bladder cancer cell lysate treated with the Hsp inhibitor VER155008 using an Hsp70 antibody attached to Dynabeads. Keratin 9, a cytoskeleton intermediate filament protein, was identified by LC MS/MS analysis. This novel finding was confirmed by Western blotting, RT-PCR, and immunocytochemistry. Other members of the keratin family of proteins have been shown to be involved in cancer progression, most recently identified to be associated with cell invasion and metastasis. The specific role of keratin 9 expression in these cells is yet to be determined.
Collapse
Affiliation(s)
- C Andolino
- Biology Department, Bucknell University, 1 Dent Drive, Lewisburg, PA, 17837, USA
| | - C Hess
- Biology Department, Bucknell University, 1 Dent Drive, Lewisburg, PA, 17837, USA
| | - T Prince
- Department of Urology, Geisinger Clinic, Danville, PA, USA
| | - H Williams
- Department of Urology, Geisinger Clinic, Danville, PA, USA
| | - M Chernin
- Biology Department, Bucknell University, 1 Dent Drive, Lewisburg, PA, 17837, USA.
| |
Collapse
|
19
|
Kumar A, Singh S, Verma A, Mishra VN. Proteomics based identification of differential plasma proteins and changes in white matter integrity as markers in early detection of mild cognitive impaired subjects at high risk of Alzheimer’s disease. Neurosci Lett 2018; 676:71-77. [DOI: 10.1016/j.neulet.2018.04.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/22/2018] [Accepted: 04/06/2018] [Indexed: 01/07/2023]
|
20
|
He L, He R, Liang R, Li Y, Li X, Li C, Zhang S. Protein expression profiling in the hippocampus after focal cerebral ischemia injury in rats. J Integr Neurosci 2018. [DOI: 10.3233/jin-170047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Lichan He
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, No. 396 Tongfu Zhong Road, Guangzhou 510220, China. E-mail:
| | - Rui He
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, No. 396 Tongfu Zhong Road, Guangzhou 510220, China. E-mail:
| | - Ruihua Liang
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, No. 396 Tongfu Zhong Road, Guangzhou 510220, China. E-mail:
| | - Yi Li
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, No. 396 Tongfu Zhong Road, Guangzhou 510220, China. E-mail:
| | - Xiaoqiang Li
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, No. 396 Tongfu Zhong Road, Guangzhou 510220, China. E-mail:
| | - Chuqiao Li
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, No. 396 Tongfu Zhong Road, Guangzhou 510220, China. E-mail:
| | - Suping Zhang
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, No. 396 Tongfu Zhong Road, Guangzhou 510220, China. E-mail:
| |
Collapse
|
21
|
Kolialexi A, Tsangaris GT, Sifakis S, Gourgiotis D, Katsafadou A, Lykoudi A, Marmarinos A, Mavreli D, Pergialiotis V, Fexi D, Mavrou A, Papaioanou GK, Papantoniou N. Plasma biomarkers for the identification of women at risk for early-onset preeclampsia. Expert Rev Proteomics 2017; 14:269-276. [PMID: 28222616 DOI: 10.1080/14789450.2017.1291345] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND To identify potential biomarkers in the 1st trimester of pregnancy for the identification of women destined to develop early onset preeclampsia (EOPE). METHODS Blood samples were obtained from pregnant women at 11-13 weeks of gestation. Women were followed up until delivery. Five samples from EOPE complicated pregnancies and 5 from unaffected ones were analysed using 2-DE and MALDI-TOF-TOF MS/MS. The altered expression of selected proteins was verified by ELISA in an extended sample cohort. RESULTS Twelve proteins were differentially expressed in the plasma of women who subsequently developed EOPE as compared to controls. Alpha-1-antitrypsin (A1AT), CD5 antigen-like molecule (CD5L) Keratin, type I cytoskeletal 9 (K1C9), Myeloid cell nuclear differentiation antigen (MNDA), Transferrin (TRFE) and Vitamin D-binding protein (VTDB) were up-regulated with fold changes 3.14, 2.18, 1.53, 1.53, 4.26 3.38 respectively, whereas Alpha-2-HS-glycoprotein (FETUA), Beta-2-glycoprotein 1 (APOH), Complement factor B (CFAB), Haptoglobin (HPT), Vitronectin (VTNC) and Zinc-alpha-2-glycoprotein (ZA2G) were down-regulated with fold changes -0.38, -0.76, -0.24, -0.47, -0.23, and -0.50 respectively. The down-regulation of APOH, VTNC and HPT was verified using ELISA. CONCLUSIONS The differentially expressed proteins represent potential biomarkers for the early screening for EOPE. Follow-up experiments however are necessary for evaluation.
Collapse
Affiliation(s)
- Aggeliki Kolialexi
- a 3rd Department of Obstetrics Gynecology , Athens University school of Medicine , Athens , Greece.,b Department of Medical Genetics , Athens University school of Medicine , Athens , Greece
| | - George Th Tsangaris
- c Proteomics Research Unit , Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | - Stavros Sifakis
- d Department of Obstetrics & Gynecology , University of Crete , Heraklion , Greece
| | - Dimitris Gourgiotis
- e 2nd Department of Paediatrics , Athens University school of Medicine , Athens , Greece
| | - Aggeliki Katsafadou
- c Proteomics Research Unit , Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | - Alexandra Lykoudi
- a 3rd Department of Obstetrics Gynecology , Athens University school of Medicine , Athens , Greece.,b Department of Medical Genetics , Athens University school of Medicine , Athens , Greece
| | - Antonios Marmarinos
- e 2nd Department of Paediatrics , Athens University school of Medicine , Athens , Greece
| | - Danai Mavreli
- a 3rd Department of Obstetrics Gynecology , Athens University school of Medicine , Athens , Greece
| | - Vassilis Pergialiotis
- a 3rd Department of Obstetrics Gynecology , Athens University school of Medicine , Athens , Greece
| | - Dimitra Fexi
- a 3rd Department of Obstetrics Gynecology , Athens University school of Medicine , Athens , Greece
| | - Ariadni Mavrou
- b Department of Medical Genetics , Athens University school of Medicine , Athens , Greece
| | - George K Papaioanou
- a 3rd Department of Obstetrics Gynecology , Athens University school of Medicine , Athens , Greece
| | - Nikolas Papantoniou
- a 3rd Department of Obstetrics Gynecology , Athens University school of Medicine , Athens , Greece
| |
Collapse
|