1
|
Xia R, Jian X, Rodrigue AL, Bressler J, Boerwinkle E, Cui B, Daviglus ML, DeCarli C, Gallo LC, Glahn DC, Knowles EEM, Moon J, Mosley TH, Satizabal CL, Sofer T, Tarraf W, Testai F, Blangero J, Seshadri S, González HM, Fornage M. Admixture mapping of cognitive function in diverse Hispanic and Latino adults: Results from the Hispanic Community Health Study/Study of Latinos. Alzheimers Dement 2024; 20:6070-6081. [PMID: 38946675 PMCID: PMC11497725 DOI: 10.1002/alz.14082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION We conducted admixture mapping and fine-mapping analyses to identify ancestry-of-origin loci influencing cognitive abilities. METHODS We estimated the association of local ancestry intervals across the genome with five neurocognitive measures in 7140 diverse Hispanic and Latino adults (mean age 55 years). We prioritized genetic variants in associated loci and tested them for replication in four independent cohorts. RESULTS We identified nine local ancestry-associated regions for the five neurocognitive measures. There was strong biological support for the observed associations to cognitive function at all loci and there was statistical evidence of independent replication at 4q12, 9p22.1, and 13q12.13. DISCUSSION Our study identified multiple novel loci harboring genes implicated in cognitive functioning and dementia, and uncovered ancestry-relevant genetic variants. It adds to our understanding of the genetic architecture of cognitive function in Hispanic and Latino adults and demonstrates the power of admixture mapping to discover unique haplotypes influencing cognitive function, complementing genome-wide association studies. HIGHLIGHTS We identified nine ancestry-of-origin chromosomal regions associated with five neurocognitive traits. In each associated region, we identified single nucleotide polymorphisms (SNPs) that explained, at least in part, the admixture signal and were tested for replication in independent samples of Black, non-Hispanic White, and Hispanic/Latino adults with the same or similar neurocognitive tests. Statistical evidence of independent replication of the prioritized SNPs was observed for three of the nine associations, at chr4q12, chr9p22.1, and chr13q12.13. At all loci, there was strong biological support for the observed associations to cognitive function and dementia, prioritizing genes such as KIT, implicated in autophagic clearance of neurotoxic proteins and on mast cell and microglial-mediated inflammation; SLC24A2, implicated in synaptic plasticity associated with learning and memory; and MTMR6, implicated in phosphoinositide lipids metabolism.
Collapse
Affiliation(s)
- Rui Xia
- Institute of Molecular Medicine, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Xueqiu Jian
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Amanda L. Rodrigue
- Department of Psychiatry, Harvard Medical SchoolBoston Children's HospitalBostonMassachusettsUSA
| | - Jan Bressler
- Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Biqi Cui
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Martha L. Daviglus
- Institute for Minority Health ResearchUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Charles DeCarli
- Department of NeurologyUniversity of California DavisSacramentoCaliforniaUSA
| | - Linda C. Gallo
- Department of PsychologySan Diego State UniversitySan DiegoCaliforniaUSA
| | - David C. Glahn
- Department of Psychiatry, Harvard Medical SchoolBoston Children's HospitalBostonMassachusettsUSA
| | - Emma E. M. Knowles
- Department of Psychiatry, Harvard Medical SchoolBoston Children's HospitalBostonMassachusettsUSA
| | - Jee‐Young Moon
- Department of Epidemiology & Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Thomas H. Mosley
- The MIND CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Department of Population Health SciencesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Tamar Sofer
- Department of MedicineHarvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
- CardioVascular InstituteBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Wassim Tarraf
- Institute of Gerontology & Department of Healthcare SciencesWayne State UniversityDetroitMichiganUSA
| | - Fernando Testai
- Department of Neurology and RehabilitationUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - John Blangero
- Department of Human Genetics, South Texas Diabetes and Obesity InstituteUniversity of Texas Rio Grande ValleyBrownsvilleTexasUSA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesThe University of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Hector M. González
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
- Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
2
|
Thorin E, Labbé P, Lambert M, Mury P, Dagher O, Miquel G, Thorin-Trescases N. Angiopoietin-Like Proteins: Cardiovascular Biology and Therapeutic Targeting for the Prevention of Cardiovascular Diseases. Can J Cardiol 2023; 39:1736-1756. [PMID: 37295611 DOI: 10.1016/j.cjca.2023.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Despite the best pharmacologic tools available, cardiovascular diseases (CVDs) remain a major cause of morbidity and mortality in developed countries. After 2 decades of research, new therapeutic targets, such as angiopoietin-like proteins (ANGPTLs), are emerging. ANGPTLs belong to a family of 8 members, from ANGPTL1 to ANGPTL8; they have structural homology with angiopoietins and are secreted in the circulation. ANGPTLs display a multitude of physiological and pathologic functions; they contribute to inflammation, angiogenesis, cell death, senescence, hematopoiesis, and play a role in repair, maintenance, and tissue homeostasis. ANGPTLs-particularly the triad ANGPTL3, 4, and 8-have an established role in lipid metabolism through the regulation of triacylglycerol trafficking according to the nutritional status. Some ANGPTLs also contribute to glucose metabolism. Therefore, dysregulation in ANGPTL expression associated with abnormal circulating levels are linked to a plethora of CVD and metabolic disorders including atherosclerosis, heart diseases, diabetes, but also obesity and cancers. Because ANGPTLs bind to different receptors according to the cell type, antagonists are therapeutically inadequate. Recently, direct inhibitors of ANGPTLs, mainly ANGPTL3, have been developed, and specific monoclonal antibodies and antisense oligonucleotides are currently being tested in clinical trials. The aim of the current review is to provide an up-to-date preclinical and clinical overview on the function of the 8 members of the ANGPTL family in the cardiovascular system, their contribution to CVD, and the therapeutic potential of manipulating some of them.
Collapse
Affiliation(s)
- Eric Thorin
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Pharmacology, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada.
| | - Pauline Labbé
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Mélanie Lambert
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Pharmacology, Université de Montréal, Montréal, Québec, Canada
| | - Pauline Mury
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Pharmacology, Université de Montréal, Montréal, Québec, Canada
| | - Olina Dagher
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Faculty of Medicine, Department of Surgery, Université de Montréal, Montréal, Québec, Canada; Department of Cardiac Sciences, Libin Cardiovascular Institute, Calgary, Alberta, Canada
| | - Géraldine Miquel
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | | |
Collapse
|
3
|
Fang J, Wang Z, Miao CY. Angiogenesis after ischemic stroke. Acta Pharmacol Sin 2023; 44:1305-1321. [PMID: 36829053 PMCID: PMC10310733 DOI: 10.1038/s41401-023-01061-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/01/2023] [Indexed: 02/26/2023] Open
Abstract
Owing to its high disability and mortality rates, stroke has been the second leading cause of death worldwide. Since the pathological mechanisms of stroke are not fully understood, there are few clinical treatment strategies available with an exception of tissue plasminogen activator (tPA), the only FDA-approved drug for the treatment of ischemic stroke. Angiogenesis is an important protective mechanism that promotes neural regeneration and functional recovery during the pathophysiological process of stroke. Thus, inducing angiogenesis in the peri-infarct area could effectively improve hemodynamics, and promote vascular remodeling and recovery of neurovascular function after ischemic stroke. In this review, we summarize the cellular and molecular mechanisms affecting angiogenesis after cerebral ischemia registered in PubMed, and provide pro-angiogenic strategies for exploring the treatment of ischemic stroke, including endothelial progenitor cells, mesenchymal stem cells, growth factors, cytokines, non-coding RNAs, etc.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Zhi Wang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
4
|
Kunze R, Marti HH. Angioneurins - Key regulators of blood-brain barrier integrity during hypoxic and ischemic brain injury. Prog Neurobiol 2019; 178:101611. [PMID: 30970273 DOI: 10.1016/j.pneurobio.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
The loss of blood-brain barrier (BBB) integrity leading to vasogenic edema and brain swelling is a common feature of hypoxic/ischemic brain diseases such as stroke, but is also central to the etiology of other CNS disorders. In the past decades, numerous proteins, belonging to the family of angioneurins, have gained increasing attention as potential therapeutic targets for ischemic stroke, but also other CNS diseases attributed to BBB dysfunction. Angioneurins encompass mediators that affect both neuronal and vascular function. Recently, increasing evidence has been accumulated that certain angioneurins critically determine disease progression and outcome in stroke among others through multifaceted effects on the compromised BBB. Here, we will give a concise overview about the family of angioneurins. We further describe the most important cellular and molecular components that contribute to structural integrity and low permeability of the BBB under steady-state conditions. We then discuss BBB alterations in ischemic stroke, and highlight underlying cellular and molecular mechanisms. For the most prominent angioneurin family members including vascular endothelial growth factors, angiopoietins, platelet-derived growth factors and erythropoietin, we will summarize current scientific literature from experimental studies in animal models, and if available from clinical trials, on the following points: (i) spatiotemporal expression of these factors in the healthy and hypoxic/ischemic CNS, (ii) impact of loss- or gain-of-function during cerebral hypoxia/ischemia for BBB integrity and beyond, and (iii) potential underlying molecular mechanisms. Moreover, we will highlight novel therapeutic strategies based on the activation of endogenous angioneurins that might improve BBB dysfuntion during ischemic stroke.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany.
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany
| |
Collapse
|
5
|
Guebel DV, Torres NV. Sexual Dimorphism and Aging in the Human Hyppocampus: Identification, Validation, and Impact of Differentially Expressed Genes by Factorial Microarray and Network Analysis. Front Aging Neurosci 2016; 8:229. [PMID: 27761111 PMCID: PMC5050216 DOI: 10.3389/fnagi.2016.00229] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/14/2016] [Indexed: 01/09/2023] Open
Abstract
Motivation: In the brain of elderly-healthy individuals, the effects of sexual dimorphism and those due to normal aging appear overlapped. Discrimination of these two dimensions would powerfully contribute to a better understanding of the etiology of some neurodegenerative diseases, such as “sporadic” Alzheimer. Methods: Following a system biology approach, top-down and bottom-up strategies were combined. First, public transcriptome data corresponding to the transition from adulthood to the aging stage in normal, human hippocampus were analyzed through an optimized microarray post-processing (Q-GDEMAR method) together with a proper experimental design (full factorial analysis). Second, the identified genes were placed in context by building compatible networks. The subsequent ontology analyses carried out on these networks clarify the main functionalities involved. Results: Noticeably we could identify large sets of genes according to three groups: those that exclusively depend on the sex, those that exclusively depend on the age, and those that depend on the particular combinations of sex and age (interaction). The genes identified were validated against three independent sources (a proteomic study of aging, a senescence database, and a mitochondrial genetic database). We arrived to several new inferences about the biological functions compromised during aging in two ways: by taking into account the sex-independent effects of aging, and considering the interaction between age and sex where pertinent. In particular, we discuss the impact of our findings on the functions of mitochondria, autophagy, mitophagia, and microRNAs. Conclusions: The evidence obtained herein supports the occurrence of significant neurobiological differences in the hippocampus, not only between adult and elderly individuals, but between old-healthy women and old-healthy men. Hence, to obtain realistic results in further analysis of the transition from the normal aging to incipient Alzheimer, the features derived from the sexual dimorphism in hippocampus should be explicitly considered.
Collapse
Affiliation(s)
- Daniel V Guebel
- Biotechnology Counselling ServicesBuenos Aires, Argentina; Systems Biology and Mathematical Modelling Group, Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Facultad de Ciencias, Universidad de La LagunaSan Cristóbal de La Laguna, España
| | - Néstor V Torres
- Systems Biology and Mathematical Modelling Group, Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Facultad de Ciencias, Universidad de La Laguna San Cristóbal de La Laguna, España
| |
Collapse
|
6
|
Wevers NR, de Vries HE. Morphogens and blood-brain barrier function in health and disease. Tissue Barriers 2015; 4:e1090524. [PMID: 27141417 DOI: 10.1080/21688370.2015.1090524] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/26/2015] [Accepted: 08/30/2015] [Indexed: 12/22/2022] Open
Abstract
The microvasculature of the brain forms a protective blood-brain barrier (BBB) that ensures a homeostatic environment for the central nervous system (CNS), which is essential for optimal brain functioning. The barrier properties of the brain endothelial cells are maintained by cells surrounding the capillaries, such as astrocytes and pericytes. Together with the endothelium and a basement membrane, these supporting cells form the neurovascular unit (NVU). Accumulating evidence indicates that the supporting cells of the NVU release a wide variety of soluble factors that induce and control barrier properties in a concentration-dependent manner. The current review provides a comprehensive overview of how such factors, called morphogens, influence BBB integrity and functioning. Since impaired BBB function is apparent in numerous CNS disorders and is often associated with disease severity, we also discuss the potential therapeutic value of these morphogens, as they may represent promising therapies for a wide variety of CNS disorders.
Collapse
Affiliation(s)
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology; Neuroscience Campus Amsterdam, VU University Medical Center ; Amsterdam, The Netherlands
| |
Collapse
|
7
|
Kim MK, Choi WY, Lee HY. Enhancement of the neuroprotective activity of Hericium erinaceus mycelium co-cultivated with Allium sativum extract. Arch Physiol Biochem 2015; 121:19-25. [PMID: 25354984 DOI: 10.3109/13813455.2014.974618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study investigated the neuroprotective effects of Hericium erinaceus mycelium enriched with garlic extract (HGE) on rat pheochromocytoma nerve cells (PC12). The survival rates of the PC12 nerve cells and the neurite-bearing cells after the addition of HGE were estimated as 3.5 × 10(3) viable cells/ml and 2.3 × 10(3) viable cells/ml, respectively, which were 50% and 30% higher, respectively, compared with the untreated group. For the in vivo ischemia experiments, after treatment with the HGE extract, the hippocampal CA1 region was more strongly stained (>20%) than the control group, and the HGE extract also promoted higher staining levels than HFB, HM and HGEF, and even the garlic extract. This result indicates that HGE must have neuroprotective effects. Furthermore, HGE greatly decreased p21 gene expression to approximately 70% of the control and decreased p21 gene expression to even lower levels compared with HM, HGEF and the garlic extract. This work suggests that a synergistic effect of the H. erinaceus mycelium and the garlic extract (mainly allicin) exist because the amount of allicin in HGE (5.81 µg/ml) was lower than the garlic extract itself (6.89 µg/ml).
Collapse
Affiliation(s)
- Myong Ki Kim
- Department of Natural Medicine Resources, Semyung University , Chungbuk , South Korea
| | | | | |
Collapse
|
8
|
Moon SW, Dinov ID, Zamanyan A, Shi R, Genco A, Hobel S, Thompson PM, Toga AW. Gene interactions and structural brain change in early-onset Alzheimer's disease subjects using the pipeline environment. Psychiatry Investig 2015; 12:125-35. [PMID: 25670955 PMCID: PMC4310910 DOI: 10.4306/pi.2015.12.1.125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE This article investigates subjects aged 55 to 65 from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database to broaden our understanding of early-onset (EO) cognitive impairment using neuroimaging and genetics biomarkers. METHODS Nine of the subjects had EO-AD (Alzheimer's disease) and 27 had EO-MCI (mild cognitive impairment). The 15 most important neuroimaging markers were extracted with the Global Shape Analysis (GSA) Pipeline workflow. The 20 most significant single nucleotide polymorphisms (SNPs) were chosen and were associated with specific neuroimaging biomarkers. RESULTS We identified associations between the neuroimaging phenotypes and genotypes for a total of 36 subjects. Our results for all the subjects taken together showed the most significant associations between rs7718456 and L_hippocampus (volume), and between rs7718456 and R_hippocampus (volume). For the 27 MCI subjects, we found the most significant associations between rs6446443 and R_superior_frontal_gyrus (volume), and between rs17029131 and L_Precuneus (volume). For the nine AD subjects, we found the most significant associations between rs16964473 and L_rectus gyrus (surface area), and between rs12972537 and L_rectus_gyrus (surface area). CONCLUSION We observed significant correlations between the SNPs and the neuroimaging phenotypes in the 36 EO subjects in terms of neuroimaging genetics. However, larger sample sizes are needed to ensure that the effects will be detectable for a reasonable false-positive error rate using the GSA and Plink Pipeline workflows.
Collapse
Affiliation(s)
- Seok Woo Moon
- Department of Psychiatry, Konkuk University School of Medicine, Chungju, Republic of Korea
| | - Ivo D. Dinov
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
- Statistics Online Computational Resource, UMSM, University of Michigan, Ann Arbor, MI, USA
| | - Alen Zamanyan
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
| | - Ran Shi
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
| | - Alex Genco
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
| | - Sam Hobel
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
| | - Paul M. Thompson
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
9
|
Moon SW, Dinov ID, Kim J, Zamanyan A, Hobel S, Thompson PM, Toga AW. Structural Neuroimaging Genetics Interactions in Alzheimer's Disease. J Alzheimers Dis 2015; 48:1051-63. [PMID: 26444770 PMCID: PMC4730943 DOI: 10.3233/jad-150335] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This article investigates late-onset cognitive impairment using neuroimaging and genetics biomarkers for Alzheimer's Disease Neuroimaging Initiative (ADNI) participants. Eight-hundred and eight ADNI subjects were identified and divided into three groups: 200 subjects with Alzheimer's disease (AD), 383 subjects with mild cognitive impairment (MCI), and 225 asymptomatic normal controls (NC). Their structural magnetic resonance imaging (MRI) data were parcellated using BrainParser, and the 80 most important neuroimaging biomarkers were extracted using the global shape analysis Pipeline workflow. Using Plink via the Pipeline environment, we obtained 80 SNPs highly-associated with the imaging biomarkers. In the AD cohort, rs2137962 was significantly associated bilaterally with changes in the hippocampi and the parahippocampal gyri, and rs1498853, rs288503, and rs288496 were associated with the left and right hippocampi, the right parahippocampal gyrus, and the left inferior temporal gyrus. In the MCI cohort, rs17028008 and rs17027976 were significantly associated with the right caudate and right fusiform gyrus, rs2075650 (TOMM40) was associated with the right caudate, and rs1334496 and rs4829605 were significantly associated with the right inferior temporal gyrus. In the NC cohort, Chromosome 15 [rs734854 (STOML1), rs11072463 (PML), rs4886844 (PML), and rs1052242 (PML)] was significantly associated with both hippocampi and both insular cortices, and rs4899412 (RGS6) was significantly associated with the caudate. We observed significant correlations between genetic and neuroimaging phenotypes in the 808 ADNI subjects. These results suggest that differences between AD, MCI, and NC cohorts may be examined by using powerful joint models of morphometric, imaging and genotypic data.
Collapse
Affiliation(s)
- Seok Woo Moon
- Department of Psychiatry, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Ivo D. Dinov
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
- University of Michigan, School of Nursing, Ann Arbor, Michigan, United States of America
| | - Jaebum Kim
- Department of Animal Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Alen Zamanyan
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| | - Sam Hobel
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| | - Paul M. Thompson
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| | - Arthur W. Toga
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
10
|
Abstract
Angiopoietin-like proteins (ANGPTLs) are a family of proteins structurally similar to the angiopoietins. To date, eight ANGPTLs have been discovered, namely ANGPTL1 to ANGPTL8. Emerging evidence implies a key role for ANGPTLs in the regulation of a plethora of physiological and pathophysiological processes. Most of the ANGPTLs exhibit multibiological properties, including established functional roles in lipid and glucose metabolism, inflammation, hematopoiesis, and cancer. This report represents a systematic and updated appraisal of this class of proteins, focusing on the main features of each ANGPTL.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
- College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
- *Correspondence: Gaetano Santulli, College of Physicians and Surgeons, Columbia University Medical Center, St. Nicholas Avenue, RB-5-513, Manhattan, NY 10032, USA e-mail:
| |
Collapse
|
11
|
Meng F, Liu R, Gao M, Wang Y, Yu X, Xuan Z, Sun J, Yang F, Wu C, Du G. Pinocembrin attenuates blood-brain barrier injury induced by global cerebral ischemia-reperfusion in rats. Brain Res 2011; 1391:93-101. [PMID: 21435338 DOI: 10.1016/j.brainres.2011.03.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 03/01/2011] [Accepted: 03/03/2011] [Indexed: 10/18/2022]
Abstract
Blood-brain barrier (BBB) disruption is a major consequence of cerebral ischemia/reperfusion. Several studies have reported the neuroprotection of pinocembrin on cerebral ischemia in vivo and in vitro, but the effects of pinocembrin on BBB and its underlying mechanisms are not clear. In this study, we investigated the effects of pinocembrin on BBB functions in the global cerebral ischemia/reperfusion (GCI/R) model in rats. Neurological scores and brain edema were evaluated. BBB permeability was assessed by detecting the concentrations of Evan's blue (EB) and fluorescein sodium (NaF) in brain tissue. The pathological changes of BBB ultrastructure were observed by transmission electron microscopy. Cerebral blood flow (CBF) was measured by laser Doppler flowmetry. The effects of pinocembrin on primary cultured rat cerebral microvascular endothelial cells (RCMECs) against oxygen-glucose deprivation/reoxygenation (OGD/R) were also investigated. The results showed pinocembrin decreased neurological score and lessened brain edema induced by GCI/R. Pinocembrin also reduced the concentrations of EB and NaF in brain tissue of the GCI/R rats. And pinocembrin alleviated the ultrastructural changes of cerebral microvessels, astrocyte end-feet and neurons, and improved CBF in the GCI/R rats. In addition, pinocembrin increased the viability and mitochondrial membrane potential of cultured RCMECs induced by OGD/R. In conclusion, these data demonstrate that pinocembrin alleviates blood-brain barrier injury induced by GCI/R in rats.
Collapse
Affiliation(s)
- Fanrui Meng
- National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Fujihara C, Yamada S, Ozaki N, Takeshita N, Kawaki H, Takano-Yamamoto T, Murakami S. Role of mechanical stress-induced glutamate signaling-associated molecules in cytodifferentiation of periodontal ligament cells. J Biol Chem 2010; 285:28286-97. [PMID: 20576613 DOI: 10.1074/jbc.m109.097303] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In this study, we analyzed the effects of tensile mechanical stress on the gene expression profile of in vitro-maintained human periodontal ligament (PDL) cells. A DNA chip analysis identified 17 up-regulated genes in human PDL cells under the mechanical stress, including HOMER1 (homer homolog 1) and GRIN3A (glutamate receptor ionotropic N-methyl-d-aspartate 3A), which are related to glutamate signaling. RT-PCR and real-time PCR analyses revealed that human PDL cells constitutively expressed glutamate signaling-associated genes and that mechanical stress increased the expression of these mRNAs, leading to release of glutamate from human PDL cells and intracellular glutamate signal transduction. Interestingly, exogenous glutamate increased the mRNAs of cytodifferentiation and mineralization-related genes as well as the ALP (alkaline phosphatase) activities during the cytodifferentiation of the PDL cells. On the other hand, the glutamate signaling inhibitors riluzole and (+)-MK801 maleate suppressed the alkaline phosphatase activities and mineralized nodule formation during the cytodifferentiation and mineralization. Riluzole inhibited the mechanical stress-induced glutamate signaling-associated gene expressions in human PDL cells. Moreover, in situ hybridization analyses showed up-regulation of glutamate signaling-associated gene expressions at tension sites in the PDL under orthodontic tooth movement in a mouse model. The present data demonstrate that the glutamate signaling induced by mechanical stress positively regulates the cytodifferentiation and mineralization of PDL cells.
Collapse
Affiliation(s)
- Chiharu Fujihara
- Department of Periodontology, Division of Oral Biology and Disease Control, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Hicks K, O'Neil RG, Dubinsky WS, Brown RC. TRPC-mediated actin-myosin contraction is critical for BBB disruption following hypoxic stress. Am J Physiol Cell Physiol 2010; 298:C1583-93. [PMID: 20164382 DOI: 10.1152/ajpcell.00458.2009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Hypoxia-induced disruption of the blood-brain barrier (BBB) is the result of many different mechanisms, including alterations to the cytoskeleton. In this study, we identified actin-binding proteins involved in cytoskeletal dynamics with quantitative proteomics and assessed changes in subcellular localization of two proteins involved in actin polymerization [vasodilator-stimulated phosphoprotein (VASP)] and cytoskeleton-plasma membrane cross-linking (moesin). We found significant redistribution of both VASP and moesin to the cytoskeletal and membrane fractions of BBB endothelial cells after 1-h hypoxic stress. We also investigated activation of actin-myosin contraction through assessment of phosphorylated myosin light chain (pMLC) with confocal microscopy. Hypoxia caused a rapid and transient increase in pMLC. Blocking MLC phosphorylation through inhibition of myosin light chain kinase (MLCK) with ML-7 prevented hypoxia-induced BBB disruption and relocalization of the tight junction protein ZO-1. Finally, we implicate the transient receptor potential (TRP)C family of channels in mediating these events since blockade of TRPC channels and the associated calcium influx with SKF-96365 prevents hypoxia-induced permeability changes and the phosphorylation of MLC needed for actin-myosin contraction. These data suggest that hypoxic stress triggers alterations to cytoskeletal structure that contribute to BBB disruption and that calcium influx through TRPC channels contributes to these events.
Collapse
Affiliation(s)
- Kali Hicks
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
14
|
Kugathasan L, Ray JB, Deng Y, Rezaei E, Dumont DJ, Stewart DJ. The angiopietin-1-Tie2 pathway prevents rather than promotes pulmonary arterial hypertension in transgenic mice. ACTA ACUST UNITED AC 2009; 206:2221-34. [PMID: 19737862 PMCID: PMC2757882 DOI: 10.1084/jem.20090389] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The role of the angiopoietin-1 (Ang1)-Tie2 pathway in the pathogenesis of pulmonary arterial hypertension (PAH) is controversial. Although Ang1 is well known to prevent endothelial activation and injury in systemic vascular beds, this pathway has been suggested to mediate pulmonary vascular remodeling in PAH. Therefore, we used transgenic models to determine the effect of increased or decreased Tie2 activity on the development of PAH. We now report modest spontaneous elevation in right ventricular systolic pressure in Tie2-deficient mice (Tie2(+/-)) compared with wild-type (WT) littermate controls, which was exacerbated upon chronic exposure to the clinically relevant PAH triggers, serotonin (5-HT) or interleukin-6 (IL-6). Moreover, overexpression of Ang1 in transgenic mice had no deleterious effect on pulmonary hemodynamics and, if anything, blunted the response to 5-HT. Exposure to 5-HT or IL-6 also decreased lung Ang1 expression, further reducing Tie2 activity and inducing pulmonary apoptosis in the Tie2(+/-) group only. Similarly, cultured pulmonary artery endothelial cells subjected to Tie2 silencing demonstrated increased susceptibility to apoptosis after 5-HT treatment. Finally, treatment of Tie2-deficient mice with Z-VAD, a pan-caspase inhibitor, prevented the pulmonary hypertensive response to 5-HT. Thus, these findings firmly establish that endothelial survival signaling via the Ang1-Tie2 pathway is protective in PAH.
Collapse
Affiliation(s)
- Lakshmi Kugathasan
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5G 1L5, Canada
| | | | | | | | | | | |
Collapse
|
15
|
Indraswari F, Wong PTH, Yap E, Ng YK, Dheen ST. Upregulation of Dpysl2 and Spna2 gene expression in the rat brain after ischemic stroke. Neurochem Int 2009; 55:235-42. [PMID: 19524114 DOI: 10.1016/j.neuint.2009.03.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Accepted: 03/09/2009] [Indexed: 01/21/2023]
Abstract
Ischemia activates the synthesis of potentially damaging and protective proteins in the central nervous system. Dihydropyrimidinase-like 2 (Dpysl2), a protein involved in neuronal differentiation and axonal guidance, and alpha-spectrin 2 (Spna2), a protein involved in maintaining neuronal membrane integrity, were found altered in various nervous system diseases. Modifications of Dpysl2 and Spna2 proteins have been reported in focal ischemic stroke, but their significance is not yet established. Therefore, this study was aimed to investigate the temporal expression of Dpysl2 and Spna2 genes in normal and stroke rat brain and to characterize stroke brains for cell areas, apoptosis, and microglia cells. The middle cerebral artery of rat brain was occluded and the brain tissue was sectioned for in situ hybridization of Dpysl2 and Spna2 genes, TUNEL, and OX-42 immunofluorescence staining. Dpysl2 and Spna2 mRNA expression was quantified by real-time RT-PCR. Characterization of stroke brain for apoptosis and microglia cells showed apoptotic cells and activated microglia, mainly in the infarct core of ipsilateral cortex and striatum of stroke brain. Significant upregulation of Dpysl2 and Spna2 mRNA expression in the penumbra region after stroke was observed predominantly in injured swollen cells in the cortex and striatum. Upregulation of Dpysl2 and Spna2 expression in hypertrophic cells in the penumbra regions of cortex and striatum of stroke brain indicates an early neuronal defense mechanism involving active neuronal repair, regeneration and development, as these genes are known to be involved in neurite outgrowth and plasticity.
Collapse
Affiliation(s)
- Fransisca Indraswari
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | | | | | | | | |
Collapse
|
16
|
Sandoval KE, Witt KA. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis 2008; 32:200-19. [PMID: 18790057 DOI: 10.1016/j.nbd.2008.08.005] [Citation(s) in RCA: 745] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 07/29/2008] [Accepted: 08/10/2008] [Indexed: 01/01/2023] Open
Abstract
The blood-brain barrier (BBB) is formed by the endothelial cells of cerebral microvessels, providing a dynamic interface between the peripheral circulation and the central nervous system. The tight junctions (TJs) between the endothelial cells serve to restrict blood-borne substances from entering the brain. Under ischemic stroke conditions decreased BBB TJ integrity results in increased paracellular permeability, directly contributing to cerebral vasogenic edema, hemorrhagic transformation, and increased mortality. This loss of TJ integrity occurs in a phasic manner, which is contingent on several interdependent mechanisms (ionic dysregulation, inflammation, oxidative and nitrosative stress, enzymatic activity, and angiogenesis). Understanding the inter-relation of these mechanisms is critical for the development of new therapies. This review focuses on those aspects of ischemic stroke impacting BBB TJ integrity and the principle regulatory pathways, respective to the phases of paracellular permeability.
Collapse
Affiliation(s)
- Karin E Sandoval
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL 62026, USA
| | | |
Collapse
|