1
|
Catto F, Dadgar-Kiani E, Kirschenbaum D, Economides A, Reuss AM, Trevisan C, Caredio D, Mirzet D, Frick L, Weber-Stadlbauer U, Litvinov S, Koumoutsakos P, Hyung Lee J, Aguzzi A. Quantitative 3D histochemistry reveals region-specific amyloid-β reduction by the antidiabetic drug netoglitazone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608042. [PMID: 39185170 PMCID: PMC11343181 DOI: 10.1101/2024.08.15.608042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
A hallmark of Alzheimer's disease (AD) is the extracellular aggregation of toxic amyloid-beta (Aβ) peptides in form of plaques. Here, we identify netoglitazone, an antidiabetic compound previously tested in humans, as an Aβ aggregation antagonist. Netoglitazone improved cognition and reduced microglia activity in a mouse model of AD. Using quantitative whole-brain three-dimensional histology (Q3D), we precisely identified brain regions where netoglitazone reduced the number and size of Aβ plaques. We demonstrate the utility of Q3D in preclinical drug evaluation for AD by providing a high-resolution brain-wide view of drug efficacy. Applying Q3D has the potential to improve pre-clinical drug evaluation by providing information that can help identify mechanisms leading to brain region-specific drug efficacy.
Collapse
Affiliation(s)
- Francesca Catto
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
- IMAI MedTech, Wagistrasse 18, 8952 Schlieren, Zurich, Switzerland
| | - Ehsan Dadgar-Kiani
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 260, 8057 Zürich
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| | - Daniel Kirschenbaum
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Athena Economides
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Anna Maria Reuss
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Chiara Trevisan
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Davide Caredio
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Delic Mirzet
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Lukas Frick
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 260, 8057 Zürich
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| | - Sergey Litvinov
- Computational Science and Engineering Laboratory, ETH Zürich, Clausiusstrasse 33, 8092, Zurich, Switzerland
- Computational Science and Engineering Laboratory, Harvard University, Cambridge, MA 02138, United States
| | - Petros Koumoutsakos
- Computational Science and Engineering Laboratory, Harvard University, Cambridge, MA 02138, United States
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, CA 94305, USA
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| |
Collapse
|
2
|
Espargaró A, Álvarez-Berbel I, Busquets MA, Sabate R. In Vivo Assays for Amyloid-Related Diseases. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:433-458. [PMID: 38598824 DOI: 10.1146/annurev-anchem-061622-023326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Amyloid-related diseases, such as Alzheimer's and Parkinson's disease, are devastating conditions caused by the accumulation of abnormal protein aggregates known as amyloid fibrils. While assays involving animal models are essential for understanding the pathogenesis and developing therapies, a wide array of standard analytical techniques exists to enhance our understanding of these disorders. These techniques provide valuable information on the formation and propagation of amyloid fibrils, as well as the pharmacokinetics and pharmacodynamics of candidate drugs. Despite ethical concerns surrounding animal use, animal models remain vital tools in the search for treatments. Regardless of the specific animal model chosen, the analytical methods used are usually standardized. Therefore, the main objective of this review is to categorize and outline the primary analytical methods used in in vivo assays for amyloid-related diseases, highlighting their critical role in furthering our understanding of these disorders and developing effective therapies.
Collapse
Affiliation(s)
- Alba Espargaró
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Irene Álvarez-Berbel
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
| | - Maria Antònia Busquets
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Raimon Sabate
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
3
|
Rabanal-Ruiz Y, Pedrero-Prieto CM, Sanchez-Rodriguez L, Flores-Cuadrado A, Saiz-Sanchez D, Frontinan-Rubio J, Ubeda-Banon I, Duran Prado M, Martinez-Marcos A, Peinado JR. Differential accumulation of human β-amyloid and tau from enriched extracts in neuronal and endothelial cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167204. [PMID: 38679217 DOI: 10.1016/j.bbadis.2024.167204] [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: 10/30/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
While Aβ and Tau cellular distribution has been largely studied, the comparative internalization and subcellular accumulation of Tau and Aβ isolated from human brain extracts in endothelial and neuronal cells has not yet been unveiled. We have previously demonstrated that controlled enrichment of Aβ from human brain extracts constitutes a valuable tool to monitor cellular internalization in vitro and in vivo. Herein, we establish an alternative method to strongly enrich Aβ and Tau aggregates from human AD brains, which has allowed us to study and compare the cellular internalization, distribution and toxicity of both proteins within brain barrier endothelial (bEnd.3) and neuronal (Neuro2A) cells. Our findings demonstrate the suitability of human enriched brain extracts to monitor the intracellular distribution of human Aβ and Tau, which, once internalized, show dissimilar sorting to different organelles within the cell and differential toxicity, exhibiting higher toxic effects on neuronal cells than on endothelial cells. While tau is strongly concentrated preferentially in mitochondria, Aβ is distributed predominantly within the endolysosomal system in endothelial cells, whereas the endoplasmic reticulum was its preferential location in neurons. Altogether, our findings display a picture of the interactions that human Aβ and Tau might establish in these cells.
Collapse
Affiliation(s)
- Y Rabanal-Ruiz
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - C M Pedrero-Prieto
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - L Sanchez-Rodriguez
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - A Flores-Cuadrado
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - D Saiz-Sanchez
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - J Frontinan-Rubio
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - I Ubeda-Banon
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - M Duran Prado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - A Martinez-Marcos
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Juan R Peinado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| |
Collapse
|
4
|
Liu K, Aierken A, Liu M, Parhat N, Kong W, Yin X, Liu G, Yu D, Hong J, Ni J, Quan Z, Liu X, Ji S, Mao J, Peng W, Chen C, Yan Y, Qing H. The decreased astrocyte-microglia interaction reflects the early characteristics of Alzheimer's disease. iScience 2024; 27:109281. [PMID: 38455972 PMCID: PMC10918213 DOI: 10.1016/j.isci.2024.109281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/29/2024] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease often associated with olfactory dysfunction. Aβ is a typical AD hall marker, but Aβ-induced molecular alterations in olfactory memory remain unclear. In this study, we used a 5xFAD mouse model to investigate Aβ-induced olfactory changes. Results showed that 4-month-old 5xFAD have olfactory memory impairment accompanied by piriform cortex neuron activity decline and no sound or working memory impairment. In addition, synapse and glia functional alteration is consistent across different ages at the proteomic level. Microglia and astrocyte specific proteins showed strong interactions in the conserved co-expression network module. Moreover, this interaction declines only in mild cognitive impairment patients in human postmortem brain proteomic data. This suggests that astrocytes-microglia interaction may play a leading role in the early stage of Aβ-induced olfactory memory impairment, and the decreasing of their synergy may accelerate the neurodegeneration.
Collapse
Affiliation(s)
- Kefu Liu
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410083, Hunan, China
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Ailikemu Aierken
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410083, Hunan, China
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Mengyao Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Nazakat Parhat
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Wei Kong
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Xingyu Yin
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410083, Hunan, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Ding Yu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Jie Hong
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaoyun Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Simei Ji
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China
| | - Jian Mao
- Zhengzhou Tobacco Research Institute of China National Tobacco Company, Zhengzhou 450001, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- National Clinical Research Center for Metabolic Diseases, Changsha, Hunan 410011, China
| | - Chao Chen
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410083, Hunan, China
| | - Yan Yan
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China
| |
Collapse
|
5
|
da Silva EMG, Fischer JSG, Souza IDLS, Andrade ACC, Souza LDCE, Andrade MKD, Carvalho PC, Souza RLR, Vital MABF, Passetti F. Proteomic Analysis of a Rat Streptozotocin Model Shows Dysregulated Biological Pathways Implicated in Alzheimer's Disease. Int J Mol Sci 2024; 25:2772. [PMID: 38474019 DOI: 10.3390/ijms25052772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's Disease (AD) is an age-related neurodegenerative disorder characterized by progressive memory loss and cognitive impairment, affecting 35 million individuals worldwide. Intracerebroventricular (ICV) injection of low to moderate doses of streptozotocin (STZ) in adult male Wistar rats can reproduce classical physiopathological hallmarks of AD. This biological model is known as ICV-STZ. Most studies are focused on the description of behavioral and morphological aspects of the ICV-STZ model. However, knowledge regarding the molecular aspects of the ICV-STZ model is still incipient. Therefore, this work is a first attempt to provide a wide proteome description of the ICV-STZ model based on mass spectrometry (MS). To achieve that, samples from the pre-frontal cortex (PFC) and hippocampus (HPC) of the ICV-STZ model and control (wild-type) were used. Differential protein abundance, pathway, and network analysis were performed based on the protein identification and quantification of the samples. Our analysis revealed dysregulated biological pathways implicated in the early stages of late-onset Alzheimer's disease (LOAD), based on differentially abundant proteins (DAPs). Some of these DAPs had their mRNA expression further investigated through qRT-PCR. Our results shed light on the AD onset and demonstrate the ICV-STZ as a valid model for LOAD proteome description.
Collapse
Affiliation(s)
- Esdras Matheus Gomes da Silva
- Instituto Carlos Chagas, FIOCRUZ, Curitiba 81310-020, PR, Brazil
- Laboratory of Toxinology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-361, RJ, Brazil
| | | | | | | | | | | | - Paulo C Carvalho
- Instituto Carlos Chagas, FIOCRUZ, Curitiba 81310-020, PR, Brazil
| | | | | | - Fabio Passetti
- Instituto Carlos Chagas, FIOCRUZ, Curitiba 81310-020, PR, Brazil
| |
Collapse
|
6
|
Plaschke K, Kopitz J, Gebert J, Wolf ND, Wolf RC. Proteomic Analysis Reveals Potential Exosomal Biomarkers in Patients With Sporadic Alzheimer Disease. Alzheimer Dis Assoc Disord 2023; 37:315-321. [PMID: 38015424 DOI: 10.1097/wad.0000000000000589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/18/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Despite substantial progress made in the past decades, the pathogenesis of sporadic Alzheimer disease (sAD) and related biological markers of the disease are still controversially discussed. Cerebrospinal fluid and functional brain imaging markers have been established to support the clinical diagnosis of sAD. Yet, due to the invasiveness of such diagnostics, less burdensome markers have been increasingly investigated in the past years. Among such markers, extracellular vesicles may yield promise in (early) diagnostics and treatment monitoring in sAD. MATERIALS AND METHODS In this pilot study, we collected the blood plasma of 18 patients with sAD and compared the proteome of extracted extracellular vesicles with the proteome of 11 age-matched healthy controls. The resulting proteomes were characterized by Gene Ontology terms and between-group statistics. RESULTS Ten distinct proteins were found to significantly differ between sAD patients and controls (P<0.05, False Discovery Rate, corrected). These proteins included distinct immunoglobulins, fibronectin, and apolipoproteins. CONCLUSIONS These findings lend further support for exosomal changes in neurodegenerative disorders, and particularly in sAD. Further proteomic research could decisively advance our knowledge of sAD pathophysiology as much as it could foster the development of clinically meaningful biomarkers.
Collapse
Affiliation(s)
| | | | | | - Nadine D Wolf
- Center for Psychosocial Medicine, Department of General Psychiatry, University Hospital Heidelberg, Heidelberg, Germany
| | - Robert Christian Wolf
- Center for Psychosocial Medicine, Department of General Psychiatry, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
7
|
Lacham-Hartman S, Moshe R, Ben-Zichri S, Shmidov Y, Bitton R, Jelinek R, Papo N. APPI-Derived Cyclic Peptide Enhances Aβ42 Aggregation and Reduces Aβ42-Mediated Membrane Destabilization and Cytotoxicity. ACS Chem Neurosci 2023; 14:3385-3397. [PMID: 37579500 DOI: 10.1021/acschemneuro.3c00208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
An amyloid precursor protein inhibitor (APPI) and amyloid beta 42 (Aβ42) are both subdomains of the human transmembrane amyloid precursor protein (APP). In the brains of patients with Alzheimer's disease (AD), Aβ42 oligomerizes into aggregates of various sizes, with intermediate, low-molecular-weight Aβ42 oligomers currently being held to be the species responsible for the most neurotoxic effects associated with the disease. Strategies to ameliorate the toxicity of these intermediate Aβ42 oligomeric species include the use of short, Aβ42-interacting peptides that either inhibit the formation of the Aβ42 oligomeric species or promote their conversion to high-molecular-weight aggregates. We therefore designed such an Aβ42-interacting peptide that is based on the β-hairpin amino acid sequence of the APPI, which exhibits high similarity to the β-sheet-like aggregation site of Aβ42. Upon tight binding of this 20-mer cyclic peptide to Aβ42 (in a 1:1 molar ratio), the formation of Aβ42 aggregates was enhanced, and consequently, Aβ42-mediated cell toxicity was ameliorated. We showed that in the presence of the cyclic peptide, interactions of Aβ42 with both plasma and mitochondrial membranes and with phospholipid vesicles that mimic these membranes were inhibited. Specifically, the cyclic peptide inhibited Aβ42-mediated mitochondrial membrane depolarization and reduced Aβ42-mediated apoptosis and cell death. We suggest that the cyclic peptide modulates Aβ42 aggregation by enhancing the formation of large aggregates─as opposed to low-molecular-weight intermediates─and as such has the potential for further development as an AD therapeutic.
Collapse
Affiliation(s)
- Shiran Lacham-Hartman
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Reut Moshe
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Shani Ben-Zichri
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Yulia Shmidov
- Department of Chemical Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Ronit Bitton
- Department of Chemical Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science &Technology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Raz Jelinek
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science &Technology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| |
Collapse
|
8
|
Gonzalez‐Rodriguez M, Villar‐Conde S, Astillero‐Lopez V, Villanueva‐Anguita P, Ubeda‐Banon I, Flores‐Cuadrado A, Martinez‐Marcos A, Saiz‐Sanchez D. Human amygdala involvement in Alzheimer's disease revealed by stereological and dia-PASEF analysis. Brain Pathol 2023; 33:e13180. [PMID: 37331354 PMCID: PMC10467039 DOI: 10.1111/bpa.13180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/06/2023] [Indexed: 06/20/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of pathological amyloid-β (Aβ) and Tau proteins. According to the prion-like hypothesis, both proteins can seed and disseminate through brain regions through neural connections and glial cells. The amygdaloid complex (AC) is involved early in the disease, and its widespread connections with other brain regions indicate that it is a hub for propagating pathology. To characterize changes in the AC as well as the involvement of neuronal and glial cells in AD, a combined stereological and proteomic analysis was performed in non-Alzheimer's disease and AD human samples. The synaptic alterations identified by proteomic data analysis could be related to the volume reduction observed in AD by the Cavalieri probe without neuronal loss. The pathological markers appeared in a gradient pattern with the medial region (cortical nucleus, Co) being more affected than lateral regions, suggesting the relevance of connections in the distribution of the pathology among different brain regions. Generalized astrogliosis was observed in every AC nucleus, likely related to deposits of pathological proteins. Astrocytes might mediate phagocytic microglial activation, whereas microglia might play a dual role since protective and toxic phenotypes have been described. These results highlight the potential participation of the amygdala in the disease spreading from/to olfactory areas, the temporal lobe and beyond. Proteomic data are available via ProteomeXchange with identifier PXD038322.
Collapse
Affiliation(s)
- Melania Gonzalez‐Rodriguez
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Sandra Villar‐Conde
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Veronica Astillero‐Lopez
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Patricia Villanueva‐Anguita
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Isabel Ubeda‐Banon
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Alicia Flores‐Cuadrado
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Alino Martinez‐Marcos
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| | - Daniel Saiz‐Sanchez
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical SchoolUniversity of Castilla‐La ManchaCiudad RealSpain
| |
Collapse
|
9
|
Kang YJ, Xue Y, Shin JH, Cho H. Human mini-brains for reconstituting central nervous system disorders. LAB ON A CHIP 2023; 23:964-981. [PMID: 36644973 DOI: 10.1039/d2lc00897a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Neurological disorders in the central nervous system (CNS) are progressive and irreversible diseases leading to devastating impacts on patients' life as they cause cognitive impairment, dementia, and even loss of essential body functions. The development of effective medicines curing CNS disorders is, however, one of the most ambitious challenges due to the extremely complex functions and structures of the human brain. In this regard, there are unmet needs to develop simplified but physiopathologically-relevant brain models. Recent advances in the microfluidic techniques allow multicellular culture forming miniaturized 3D human brains by aligning parts of brain regions with specific cells serving suitable functions. In this review, we overview designs and strategies of microfluidics-based human mini-brains for reconstituting CNS disorders, particularly Alzheimer's disease (AD), Parkinson's disease (PD), traumatic brain injury (TBI), vascular dementia (VD), and environmental risk factor-driven dementia (ERFD). Afterward, the applications of the mini-brains in the area of medical science are introduced in terms of the clarification of pathogenic mechanisms and identification of promising biomarkers. We also present expanded model systems ranging from the CNS to CNS-connecting organ axes to study the entry pathways of pathological risk factors into the brain. Lastly, the advantages and potential challenges of current model systems are addressed with future perspectives.
Collapse
Affiliation(s)
- You Jung Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yingqi Xue
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Hee Shin
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
10
|
Frontiñán-Rubio J, Llanos-González E, García-Carpintero S, Peinado JR, Ballesteros-Yáñez I, Rayo MV, de la Fuente J, Pérez-García VM, Perez-Romasanta LA, Malumbres M, Alcaín FJ, Durán-Prado M. CoQ 10 reduces glioblastoma growth and infiltration through proteome remodeling and inhibition of angiogenesis and inflammation. Cell Oncol (Dordr) 2023; 46:65-77. [PMID: 36319818 PMCID: PMC9947058 DOI: 10.1007/s13402-022-00734-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
PURPOSE Most monotherapies available against glioblastoma multiforme (GBM) target individual hallmarks of this aggressive brain tumor with minimal success. In this article, we propose a therapeutic strategy using coenzyme Q10 (CoQ10) as a pleiotropic factor that crosses the blood-brain barrier and accumulates in cell membranes acting as an antioxidant, and in mitochondrial membranes as a regulator of cell bioenergetics and gene expression. METHODS Xenografts of U251 cells in nu/nu mice were used to assay tumor growth, hypoxia, angiogenesis, and inflammation. An orthotopic model was used to explore microglial infiltration, tumor growth, and invasion into the brain parenchyma. Cell proliferation, migration, invasion, proteome remodeling, and secretome were assayed in vitro. Conditioned media were used to assay angiogenesis, monocyte chemoattraction, and differentiation into macrophages in vitro. RESULTS CoQ10 treatment decreased tumor volume in xenografts and orthotopic models, although its effect on tumor cell proliferation was not direct. Tumors from mice treated with CoQ10 were less hypoxic and vascularized, having less infiltration from inflammatory cells. Treatment-induced downregulation of HIF-1α and NF-kB led to a complete remodeling of the tumor cells proteome and secretome, impacting angiogenesis, monocyte infiltration, and their differentiation into macrophages. Besides, tumor cell migration and invasion were drastically restricted by mechanisms involving modulation of the actin cytoskeleton and downregulation of matrix metalloproteases (MMPs). CONCLUSIONS CoQ10 has a pleiotropic effect on GBM growth, targeting several hallmarks simultaneously. Thus, its integration into current treatments of this fatal disease should be considered.
Collapse
Affiliation(s)
- Javier Frontiñán-Rubio
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Emilio Llanos-González
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Sonia García-Carpintero
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Juan Ramón Peinado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Inmaculada Ballesteros-Yáñez
- EMAS Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Margarita Villar Rayo
- SaBio Research Group, Hunting Resources Research Institute (IREC), Ciudad Real, Spain
| | - José de la Fuente
- SaBio Research Group, Hunting Resources Research Institute (IREC), Ciudad Real, Spain
| | - Víctor M Pérez-García
- Laboratory of Mathematical Oncology, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Luis A Perez-Romasanta
- Radiology and Medicinal Physics, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Marcos Malumbres
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Francisco J Alcaín
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain.
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Mario Durán-Prado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain.
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| |
Collapse
|
11
|
Gonzalez-Rodriguez M, Villar-Conde S, Astillero-Lopez V, Villanueva-Anguita P, Ubeda-Banon I, Flores-Cuadrado A, Martinez-Marcos A, Saiz-Sanchez D. Neurodegeneration and Astrogliosis in the Human CA1 Hippocampal Subfield Are Related to hsp90ab1 and bag3 in Alzheimer's Disease. Int J Mol Sci 2021; 23:165. [PMID: 35008592 PMCID: PMC8745315 DOI: 10.3390/ijms23010165] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD), the most prevalent neurodegenerative disorder, is characterized by executive dysfunction and memory impairment mediated by the accumulation of extracellular amyloid-β peptide (Aβ) and intracellular hyperphosphorylated tau protein. The hippocampus (HIPP) is essential for memory formation and is involved in early stages of disease. In fact, hippocampal atrophy is used as an early biomarker of neuronal injury and to evaluate disease progression. It is not yet well-understood whether changes in hippocampal volume are due to neuronal or glial loss. The aim of the study was to assess hippocampal atrophy and/or gliosis using unbiased stereological quantification and to obtain hippocampal proteomic profiles related to neurodegeneration and gliosis. Hippocampal volume measurement, stereological quantification of NeuN-, Iba-1- and GFAP-positive cells, and sequential window acquisition of all theoretical mass spectrometry (SWATH-MS) analysis were performed in AD and non-AD cases. Reduced hippocampal volume was identified using the Cavalieri probe, particularly in the CA1 region, where it correlated with neuronal loss and astrogliosis. A total of 102 downregulated and 47 upregulated proteins were identified in the SWATH-MS analysis after restrictive filtering based on an FC > 1.5 and p value < 0.01. The Hsp90 family of chaperones, particularly BAG3 and HSP90AB1, are closely related to astrocytes, indicating a possible role in degrading Aβ and tau through chaperone-mediated autophagy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alino Martinez-Marcos
- CRIB, Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (M.G.-R.); (S.V.-C.); (V.A.-L.); (P.V.-A.); (I.U.-B.); (A.F.-C.)
| | - Daniel Saiz-Sanchez
- CRIB, Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (M.G.-R.); (S.V.-C.); (V.A.-L.); (P.V.-A.); (I.U.-B.); (A.F.-C.)
| |
Collapse
|
12
|
Matuszyk MM, Garwood CJ, Ferraiuolo L, Simpson JE, Staniforth RA, Wharton SB. Biological and methodological complexities of beta-amyloid peptide: Implications for Alzheimer's disease research. J Neurochem 2021; 160:434-453. [PMID: 34767256 DOI: 10.1111/jnc.15538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/01/2023]
Abstract
Although controversial, the amyloid cascade hypothesis remains central to the Alzheimer's disease (AD) field and posits amyloid-beta (Aβ) as the central factor initiating disease onset. In recent years, there has been an increase in emphasis on studying the role of low molecular weight aggregates, such as oligomers, which are suggested to be more neurotoxic than fibrillary Aβ. Other Aβ isoforms, such as truncated Aβ, have also been implicated in disease. However, developing a clear understanding of AD pathogenesis has been hampered by the complexity of Aβ biochemistry in vitro and in vivo. This review explores factors contributing to the lack of consistency in experimental approaches taken to model Aβ aggregation and toxicity and provides an overview of the different techniques available to analyse Aβ, such as electron and atomic force microscopy, nuclear magnetic resonance spectroscopy, dye-based assays, size exclusion chromatography, mass spectrometry and SDS-PAGE. The review also explores how different types of Aβ can influence Aβ aggregation and toxicity, leading to variation in experimental outcomes, further highlighting the need for standardisation in Aβ preparations and methods used in current research.
Collapse
Affiliation(s)
- Martyna M Matuszyk
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Claire J Garwood
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
13
|
Wang Y, Li Z, Yang G, Cai L, Yang F, Zhang Y, Zeng Y, Ma Q, Zeng F. The Study of Alternative Splicing Events in Human Induced Pluripotent Stem Cells From a Down's Syndrome Patient. Front Cell Dev Biol 2021; 9:661381. [PMID: 34660567 PMCID: PMC8516071 DOI: 10.3389/fcell.2021.661381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/16/2021] [Indexed: 12/03/2022] Open
Abstract
Down's syndrome (DS) is one of the most commonly known disorders with multiple congenital disabilities. Besides severe cognitive impairment and intellectual disability, individuals with DS also exhibit additional phenotypes of variable penetrance and severity, with one or more comorbid conditions, including Alzheimer's disease, congenital heart disease, or leukemia. Various vital genes and regulatory networks had been studied to reveal the pathogenesis of the disease. Nevertheless, very few studies have examined alternative splicing. Alternative splicing (AS) is a regulatory mechanism of gene expression when making one multi-exon protein-coding gene produce more than one unique mature mRNA. We employed the GeneChip Human Transcriptome Array 2.0 (HTA 2.0) for the global gene analysis with hiPSCs from DS and healthy individuals. Examining differentially expressed genes (DEGs) in these groups and focusing on specific transcripts with AS, 466 up-regulated and 722 down-regulated genes with AS events were identified. These genes were significantly enriched in biological processes, such as cell adhesion, cardiac muscle contraction, and immune response, through gene ontology (GO) analysis of DEGs. Candidate genes, such as FN1 were further explored for potentially playing a key role in DS. This study provides important insights into the potential role that AS plays in DS.
Collapse
Affiliation(s)
- Yunjie Wang
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Zexu Li
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Guanheng Yang
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Linlin Cai
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Fan Yang
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Yaqiong Zhang
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Yitao Zeng
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Qingwen Ma
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Fanyi Zeng
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,National Health Commission Key Laboratory of Embryo Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China.,Department of Histoembryology, Genetics & Development, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Analysis of Differentially Expressed Genes in the Dentate Gyrus and Anterior Cingulate Cortex in a Mouse Model of Depression. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5013565. [PMID: 33628784 PMCID: PMC7892236 DOI: 10.1155/2021/5013565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/11/2020] [Accepted: 01/23/2021] [Indexed: 12/18/2022]
Abstract
Major depressive disorder (MDD) is a prevalent, chronic, and relapse-prone psychiatric disease. However, the intermediate molecules resulting from stress and neurological impairment in different brain regions are still unclear. To clarify the pathological changes in the dentate gyrus (DG) and anterior cingulate cortex (ACC) regions of the MDD brain, which are the most closely related to the disease, we investigated the published microarray profile dataset GSE84183 to identify unpredictable chronic mild stress- (UCMS-) induced differentially expressed genes (DEGs) in the DG and ACC regions. Based on the DEG data, functional annotation, protein-protein interaction, and transcription factor (TF) analyses were performed. In this study, 1071 DEGs (679 upregulated and 392 downregulated) and 410 DEGs (222 upregulated and 188 downregulated) were identified in DG and ACC, respectively. The pathways and GO terms enriched by the DEGs in the DG, such as cell adhesion, proteolysis, ion transport, transmembrane transport, chemical synaptic transmission, immune system processes, response to lipopolysaccharide, and nervous system development, may reveal the molecular mechanism of MDD. However, the DEGs in the ACC involved metabolic processes, proteolysis, visual learning, DNA methylation, innate immune responses, cell migration, and circadian rhythm. Sixteen hub genes in the DG (Fn1, Col1a1, Anxa1, Penk, Ptgs2, Cdh1, Timp1, Vim, Rpl30, Rps21, Dntt, Ptk2b, Jun, Avp, Slit1, and Sema5a) were identified. Eight hub genes in the ACC (Prkcg, Grin1, Syngap1, Rrp9, Grwd1, Pik3r1, Hnrnpc, and Prpf40a) were identified. In addition, eleven TFs (Chd2, Zmiz1, Myb, Etv4, Rela, Tcf4, Tcf12, Chd1, Mef2a, Ubtf, and Mxi1) were predicted to regulate more than two of these hub genes. The expression levels of ten randomly selected hub genes that were specifically differentially expressed in the MDD-like animal model were verified in the corresponding regions in the human brain. These hub genes and TFs may be regarded as potential targets for future MDD treatment strategies, thus aiding in the development of new therapeutic approaches to MDD.
Collapse
|
15
|
Liang Y, Ma C, Li F, Nie G, Zhang H. The Role of Contactin 1 in Cancers: What We Know So Far. Front Oncol 2020; 10:574208. [PMID: 33194679 PMCID: PMC7658624 DOI: 10.3389/fonc.2020.574208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022] Open
Abstract
Cancers are among the difficult-to-treat diseases despite advances in diagnosis and treatment. Although newer effective targets remain to be discovered, targeted therapy has emerged as a promising field. In the last decade, contactin 1 (CNTN1) has surfaced as an important cancer-related molecule. CNTN1 is a neuronal membrane glycoprotein, which, if overexpressed, is found in different cancer cell lines, cancer tissues, and transgenic mice. It is positively associated with lymphatic invasion, metastasis, late TNM stage, and a short overall survival time. However, the role of CNTN1 in cancer cell proliferation remains unclear. In addition, CNTN1 is involved in cancer cell invasion, migration, metastasis, and chemoresistance by promoting epithelial-mesenchymal transition and mediating several signal transduction pathways. Several studies suggest CNTN1 as a new therapeutic target for cancers. This review aims to summarize the research developments on CNTN1 in various cancers, to establish its role in epithelial-mesenchymal transition and signal transduction pathways, and to identify promising areas for further investigation.
Collapse
Affiliation(s)
- Yumei Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Cui Ma
- Department of Pediatric Hematology, The First Hospital of Jilin University, Changchun, China
| | - Fengjuan Li
- Oncology Department of Tumor Center, The First Hospital of Jilin University, Changchun, China
| | - Guanhua Nie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Haining Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Integrative Multi-Omics Analysis in Calcific Aortic Valve Disease Reveals a Link to the Formation of Amyloid-Like Deposits. Cells 2020; 9:cells9102164. [PMID: 32987857 PMCID: PMC7600313 DOI: 10.3390/cells9102164] [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: 04/23/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is the most prevalent valvular heart disease in the developed world, yet no pharmacological therapy exists. Here, we hypothesize that the integration of multiple omic data represents an approach towards unveiling novel molecular networks in CAVD. Databases were searched for CAVD omic studies. Differentially expressed molecules from calcified and control samples were retrieved, identifying 32 micro RNAs (miRNA), 596 mRNAs and 80 proteins. Over-representation pathway analysis revealed platelet degranulation and complement/coagulation cascade as dysregulated pathways. Multi-omics integration of overlapping proteome/transcriptome molecules, with the miRNAs, identified a CAVD protein–protein interaction network containing seven seed genes (apolipoprotein A1 (APOA1), hemoglobin subunit β (HBB), transferrin (TF), α-2-macroglobulin (A2M), transforming growth factor β-induced protein (TGFBI), serpin family A member 1 (SERPINA1), lipopolysaccharide binding protein (LBP), inter-α-trypsin inhibitor heavy chain 3 (ITIH3) and immunoglobulin κ constant (IGKC)), four input miRNAs (miR-335-5p, miR-3663-3p, miR-21-5p, miR-93-5p) and two connector genes (amyloid beta precursor protein (APP) and transthyretin (TTR)). In a metabolite–gene–disease network, Alzheimer’s disease exhibited the highest degree of betweenness. To further strengthen the associations based on the multi-omics approach, we validated the presence of APP and TTR in calcified valves from CAVD patients by immunohistochemistry. Our study suggests a novel molecular CAVD network potentially linked to the formation of amyloid-like structures. Further investigations on the associated mechanisms and therapeutic potential of targeting amyloid-like deposits in CAVD may offer significant health benefits.
Collapse
|
17
|
András IE, Sewell BB, Toborek M. HIV-1 and Amyloid Beta Remodel Proteome of Brain Endothelial Extracellular Vesicles. Int J Mol Sci 2020; 21:ijms21082741. [PMID: 32326569 PMCID: PMC7215366 DOI: 10.3390/ijms21082741] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 01/01/2023] Open
Abstract
Amyloid beta (Aβ) depositions are more abundant in HIV-infected brains. The blood-brain barrier, with its backbone created by endothelial cells, is assumed to be a core player in Aβ homeostasis and may contribute to Aβ accumulation in the brain. Exposure to HIV increases shedding of extracellular vesicles (EVs) from human brain endothelial cells and alters EV-Aβ levels. EVs carrying various cargo molecules, including a complex set of proteins, can profoundly affect the biology of surrounding neurovascular unit cells. In the current study, we sought to examine how exposure to HIV, alone or together with Aβ, affects the surface and total proteomic landscape of brain endothelial EVs. By using this unbiased approach, we gained an unprecedented, high-resolution insight into these changes. Our data suggest that HIV and Aβ profoundly remodel the proteome of brain endothelial EVs, altering the pathway networks and functional interactions among proteins. These events may contribute to the EV-mediated amyloid pathology in the HIV-infected brain and may be relevant to HIV-1-associated neurocognitive disorders.
Collapse
|