1
|
Choi YK. Detrimental Roles of Hypoxia-Inducible Factor-1α in Severe Hypoxic Brain Diseases. Int J Mol Sci 2024; 25:4465. [PMID: 38674050 PMCID: PMC11050730 DOI: 10.3390/ijms25084465] [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: 03/14/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Hypoxia stabilizes hypoxia-inducible factors (HIFs), facilitating adaptation to hypoxic conditions. Appropriate hypoxia is pivotal for neurovascular regeneration and immune cell mobilization. However, in central nervous system (CNS) injury, prolonged and severe hypoxia harms the brain by triggering neurovascular inflammation, oxidative stress, glial activation, vascular damage, mitochondrial dysfunction, and cell death. Diminished hypoxia in the brain improves cognitive function in individuals with CNS injuries. This review discusses the current evidence regarding the contribution of severe hypoxia to CNS injuries, with an emphasis on HIF-1α-mediated pathways. During severe hypoxia in the CNS, HIF-1α facilitates inflammasome formation, mitochondrial dysfunction, and cell death. This review presents the molecular mechanisms by which HIF-1α is involved in the pathogenesis of CNS injuries, such as stroke, traumatic brain injury, and Alzheimer's disease. Deciphering the molecular mechanisms of HIF-1α will contribute to the development of therapeutic strategies for severe hypoxic brain diseases.
Collapse
Affiliation(s)
- Yoon Kyung Choi
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
2
|
Giuliani G, Sborgia G, Niro A, Castellana F, Lampignano L, Puzo P, Pascale A, Pastore V, Buonamassa R, Galati R, Bordinone M, Cassano F, Clemente A, Landini L, Scotti G, Gaudiomonte M, Guglielmi A, Semeraro R, Santoro M, Alessio G, Sardone R, Boscia F. Correlation between retinal vessel rarefaction and psychometric measures in an older Southern Italian population. Front Aging Neurosci 2022; 14:999796. [PMID: 36212041 PMCID: PMC9541429 DOI: 10.3389/fnagi.2022.999796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/06/2022] [Indexed: 12/22/2022] Open
Abstract
Objective To explore the linear association between inner retinal layers thickness and macular capillary density compared to variations of global cognition evaluated by psychometric measures in a cohort of Mediterranean subjects aged 65+ years. Materials and methods We performed a cross-sectional analysis of 574 participants aged 65 years+ drawn from a population-based Southern Italian study. All subjects underwent neurological evaluations, including global cognitive screening, the Mini-Mental State Examination (MMSE) and frontal assessment battery (FAB), together with an ophthalmic examination including optical coherence tomography (OCT) and OCT-Angiography. We assessed the average thickness of the ganglion cell complex (GCC) and the retinal nerve fiber layer (RNFL), the foveal avascular zone area, and vascular density (VD) of superficial (SVD) and deep (DVD) capillary plexi at the foveal and parafoveal area. Linear regression was applied to assess associations of ocular measurements with MMSE and FAB scores. Results In the linear regression model, foveal DVD (beta = 0.01, 95% CI:0.004–0.052), whole DVD (beta = 0.04, 95% CI:0.02–0.08), and whole SVD (beta = 0.04, 95% CI:0.02–0.07) showed a positive association with MMSE. In addition, foveal SVD (beta = 0.01, 95% CI:0.003–0.05) and whole SVD (beta = 0.03, 95% CI:0.004–0.08) were positively associated with the FAB score. We found no further significant association between the MMSE score or the FAB score and the average thickness of the GCC and RNFL, and FAZ area. Conclusion A direct linear association between the VD of the macular capillary plexi with global and frontal cognitive functions was observed in elderly subjects.
Collapse
Affiliation(s)
- Gianluigi Giuliani
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Giancarlo Sborgia
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Alfredo Niro
- Eye Clinic, Hospital “SS Annunziata,” ASL Taranto, Taranto, Italy
- *Correspondence: Alfredo Niro,
| | - Fabio Castellana
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study,” National Institute of Gastroenterology “Saverio de Bellis,” Research Hospital, Bari, Italy
| | - Luisa Lampignano
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study,” National Institute of Gastroenterology “Saverio de Bellis,” Research Hospital, Bari, Italy
| | - Pasquale Puzo
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Angelo Pascale
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Valentina Pastore
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Rosa Buonamassa
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Roberta Galati
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Marco Bordinone
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Flavio Cassano
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Arcangelo Clemente
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Luca Landini
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Giacomo Scotti
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Marida Gaudiomonte
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Antonella Guglielmi
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Roberto Semeraro
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Michele Santoro
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Giovanni Alessio
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Rodolfo Sardone
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study,” National Institute of Gastroenterology “Saverio de Bellis,” Research Hospital, Bari, Italy
| | - Francesco Boscia
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| |
Collapse
|
3
|
Snyder B, Wu HK, Tillman B, Floyd TF. Aged Mouse Hippocampus Exhibits Signs of Chronic Hypoxia and an Impaired HIF-Controlled Response to Acute Hypoxic Exposures. Cells 2022; 11:cells11030423. [PMID: 35159233 PMCID: PMC8833982 DOI: 10.3390/cells11030423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
Altered hypoxia-inducible factor-alpha (HIF-α) activity may have significant consequences in the hippocampus, which mediates declarative memory, has limited vascularization, and is vulnerable to hypoxic insults. Previous studies have reported that neurovascular coupling is reduced in aged brains and that diseases which cause hypoxia increase with age, which may render the hippocampus susceptible to acute hypoxia. Most studies have investigated the actions of HIF-α in aging cortical structures, but few have focused on the role of HIF-α within aged hippocampus. This study tests the hypothesis that aging is associated with impaired hippocampal HIF-α activity. Dorsal hippocampal sections from mice aged 3, 9, 18, and 24 months were probed for the presence of HIF-α isoforms or their associated gene products using immunohistochemistry and fluorescent in situ hybridization (fISH). A subset of each age was exposed to acute hypoxia (8% oxygen) for 3 h to investigate changes in the responsiveness of HIF-α to hypoxia. Basal mean intensity of fluorescently labeled HIF-1α protein increases with age in the hippocampus, whereas HIF-2α intensity only increases in the 24-month group. Acute hypoxic elevation of HIF-1α is lost with aging and is reversed in the 24-month group. fISH reveals that glycolytic genes induced by HIF-1α (lactose dehydrogenase-a, phosphoglycerate kinase 1, and pyruvate dehydrogenase kinase 1) are lower in aged hippocampus than in 3-month hippocampus, and mRNA for monocarboxylate transporter 1, a lactose transporter, increases. These results indicate that lactate, used in neurotransmission, may be limited in aged hippocampus, concurrent with impaired HIF-α response to hypoxic events. Therefore, impaired HIF-α may contribute to age-associated cognitive decline during hypoxic events.
Collapse
Affiliation(s)
- Brina Snyder
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.S.); (H.-K.W.); (B.T.)
| | - Hua-Kang Wu
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.S.); (H.-K.W.); (B.T.)
| | - Brianna Tillman
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.S.); (H.-K.W.); (B.T.)
| | - Thomas F. Floyd
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.S.); (H.-K.W.); (B.T.)
- Department of Cardiothoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence:
| |
Collapse
|
4
|
Burtscher J, Mallet RT, Burtscher M, Millet GP. Hypoxia and brain aging: Neurodegeneration or neuroprotection? Ageing Res Rev 2021; 68:101343. [PMID: 33862277 DOI: 10.1016/j.arr.2021.101343] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022]
Abstract
The absolute reliance of the mammalian brain on oxygen to generate ATP renders it acutely vulnerable to hypoxia, whether at high altitude or in clinical settings of anemia or pulmonary disease. Hypoxia is pivotal to the pathogeneses of myriad neurological disorders, including Alzheimer's, Parkinson's and other age-related neurodegenerative diseases. Conversely, reduced environmental oxygen, e.g. sojourns or residing at high altitudes, may impart favorable effects on aging and mortality. Moreover, controlled hypoxia exposure may represent a treatment strategy for age-related neurological disorders. This review discusses evidence of hypoxia's beneficial vs. detrimental impacts on the aging brain and the molecular mechanisms that mediate these divergent effects. It draws upon an extensive literature search on the effects of hypoxia/altitude on brain aging, and detailed analysis of all identified studies directly comparing brain responses to hypoxia in young vs. aged humans or rodents. Special attention is directed toward the risks vs. benefits of hypoxia exposure to the elderly, and potential therapeutic applications of hypoxia for neurodegenerative diseases. Finally, important questions for future research are discussed.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland; Institute of Sport Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland.
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland
| |
Collapse
|
5
|
Wilson DF, Matschinsky FM. Cerebrovascular Blood Flow Design and Regulation; Vulnerability in Aging Brain. Front Physiol 2020; 11:584891. [PMID: 33178048 PMCID: PMC7596697 DOI: 10.3389/fphys.2020.584891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/24/2020] [Indexed: 12/24/2022] Open
Abstract
Nutrient delivery to the brain presents a unique challenge because the tissue functions as a computer system with in the order of 200,000 neurons/mm3. Penetrating arterioles bud from surface arteries of the brain and penetrate downward through the cortex. Capillary networks spread from penetrating arterioles through the surrounding tissue. Each penetrating arteriole forms a vascular unit, with little sharing of flow among vascular units (collateral flow). Unlike cells in other tissues, neurons have to be operationally isolated, interacting with other neurons through specific electrical connections. Neuronal activation typically involves only a few of the cells within a vascular unit, but the local increase in nutrient consumption is substantial. The metabolic response to activation is transmitted to the feeding arteriole through the endothelium of neighboring capillaries and alters calcium permeability of smooth muscle in the wall resulting in modulation of flow through the entire vascular unit. Many age and trauma related brain pathologies can be traced to vascular malfunction. This includes: 1. Physical damage such as in traumatic injury with imposed shear stress as soft tissue moves relative to the skull. Lack of collateral flow among vascular units results in death of the cells in that vascular unit and loss of brain tissue. 2. Age dependent changes lead to progressive increase in vascular resistance and decrease in tissue levels of oxygen and glucose. Chronic hypoxia/hypoglycemia compromises tissue energy metabolism and related regulatory processes. This alters stem cell proliferation and differentiation, undermines vascular integrity, and suppresses critical repair mechanisms such as oligodendrocyte generation and maturation. Reduced structural integrity results in local regions of acute hypoxia and microbleeds, while failure of oligodendrocytes to fully mature leads to poor axonal myelination and defective neuronal function. Understanding and treating age related pathologies, particularly in brain, requires better knowledge of why and how vasculature changes with age. That knowledge will, hopefully, make possible drugs/methods for protecting vascular function, substantially alleviating the negative health and cognitive deficits associated with growing old.
Collapse
Affiliation(s)
- David F Wilson
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Franz M Matschinsky
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
6
|
Bjedov I, Rallis C. The Target of Rapamycin Signalling Pathway in Ageing and Lifespan Regulation. Genes (Basel) 2020; 11:E1043. [PMID: 32899412 PMCID: PMC7565554 DOI: 10.3390/genes11091043] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
Ageing is a complex trait controlled by genes and the environment. The highly conserved mechanistic target of rapamycin signalling pathway (mTOR) is a major regulator of lifespan in all eukaryotes and is thought to be mediating some of the effects of dietary restriction. mTOR is a rheostat of energy sensing diverse inputs such as amino acids, oxygen, hormones, and stress and regulates lifespan by tuning cellular functions such as gene expression, ribosome biogenesis, proteostasis, and mitochondrial metabolism. Deregulation of the mTOR signalling pathway is implicated in multiple age-related diseases such as cancer, neurodegeneration, and auto-immunity. In this review, we briefly summarise some of the workings of mTOR in lifespan and ageing through the processes of transcription, translation, autophagy, and metabolism. A good understanding of the pathway's outputs and connectivity is paramount towards our ability for genetic and pharmacological interventions for healthy ageing and amelioration of age-related disease.
Collapse
Affiliation(s)
- Ivana Bjedov
- UCL Cancer Institute, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Charalampos Rallis
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| |
Collapse
|
7
|
Alique M, Sánchez-López E, Bodega G, Giannarelli C, Carracedo J, Ramírez R. Hypoxia-Inducible Factor-1α: The Master Regulator of Endothelial Cell Senescence in Vascular Aging. Cells 2020; 9:cells9010195. [PMID: 31941032 PMCID: PMC7016968 DOI: 10.3390/cells9010195] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/07/2020] [Accepted: 01/11/2020] [Indexed: 12/11/2022] Open
Abstract
Aging is one of the hottest topics in biomedical research. Advances in research and medicine have helped to preserve human health, leading to an extension of life expectancy. However, the extension of life is an irreversible process that is accompanied by the development of aging-related conditions such as weakness, slower metabolism, and stiffness of vessels. It also debated that aging can be considered an actual disease with aging-derived comorbidities, including cancer or cardiovascular disease. Currently, cardiovascular disorders, including atherosclerosis, are considered as premature aging and represent the first causes of death in developed countries, accounting for 31% of annual deaths globally. Emerging evidence has identified hypoxia-inducible factor-1α as a critical transcription factor with an essential role in aging-related pathology, in particular, regulating cellular senescence associated with cardiovascular aging. In this review, we will focus on the regulation of senescence mediated by hypoxia-inducible factor-1α in age-related pathologies, with particular emphasis on the crosstalk between endothelial and vascular cells in age-associated atherosclerotic lesions. More specifically, we will focus on the characteristics and mechanisms by which cells within the vascular wall, including endothelial and vascular cells, achieve a senescent phenotype.
Collapse
Affiliation(s)
- Matilde Alique
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud (IRYCIS), Universidad de Alcalá, Alcalá de Henares, 28805 Madrid, Spain;
- Correspondence: (M.A.); (J.C.); Tel.: +34-91-885-6436 (M.A.); +34-91-394-5005 (J.C.)
| | - Elsa Sánchez-López
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA 92037, USA;
| | - Guillermo Bodega
- Departamento de Biomedicina y Biotecnología, Facultad de Biología, Química y Ciencias Ambientales, Universidad de Alcalá, Alcalá de Henares, 28805 Madrid, Spain;
| | - Chiara Giannarelli
- Cardiovascular Research Center, Institute for Genomics and Multiscale Biology, New York, NY 10029, USA;
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28041 Madrid, Spain
- Correspondence: (M.A.); (J.C.); Tel.: +34-91-885-6436 (M.A.); +34-91-394-5005 (J.C.)
| | - Rafael Ramírez
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud (IRYCIS), Universidad de Alcalá, Alcalá de Henares, 28805 Madrid, Spain;
| |
Collapse
|
8
|
Desmettre T. [Epigenetics in age-related macular degeneration (AMD) - French translation of the article]. J Fr Ophtalmol 2018; 41:981-990. [PMID: 30454959 DOI: 10.1016/j.jfo.2018.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/02/2018] [Accepted: 06/07/2018] [Indexed: 02/02/2023]
Abstract
Age-related macular degeneration (AMD) is a complex multifactorial condition involving multiple genetic, environmental and constitutional factors. Inflammation, oxidative stress and lipid metabolism seem to be the most important factors in the pathogenesis of the disease. The importance of genetic factors has mainly been revealed with the influence of histocompatibility complement factor H (CFH) variations and the ARSM2 susceptibility gene. Another component, epigenetics, could help to explain some of the relationships between environmental and genetic factors. Epigenetics is defined as the study of modulations of gene activity that can be transmitted over cell divisions without involving mutation of the DNA sequence. The molecules that are involved in these mechanisms are referred to as the epigenome. The mechanisms involve DNA methylation, histone modification, chromatin remodeling, and gene inhibition by non-coding RNA. Epigenetics could explain how the environment may induce relatively stable changes in traits or even diseases, possibly inheritable over several generations. Epigenetic traits established during development, and/or acquired under the influence of nutritional factors or other environmental factors, could influence the interactions between genes and the environment. Several authors have recently shown the influence of epigenetic factors in the pathogenesis of ocular diseases such as cataract, dry eye, glaucoma, diabetic retinopathy and more recently AMD. A better understanding of the involvement of genetic variants at risk, their relationship with epigenetics and environmental factors would certainly help to better assess the risk of developing AMD or better understand recent changes in the incidence of the disease.
Collapse
Affiliation(s)
- T Desmettre
- Centre de rétine médicale, 187, rue de Menin, 59520 Marquette-Lez-Lille, France; London International Medical Centre, 18-22 Queen Anne Street, London, W1G 8HU, Royaume-Uni.
| |
Collapse
|
9
|
Leiton CV, Chen E, Cutrone A, Conn K, Mellanson K, Malik DM, Klingener M, Lamm R, Cutrone M, Petrie J, Sheikh J, DiBua A, Cohen B, Floyd TF. Astrocyte HIF-2α supports learning in a passive avoidance paradigm under hypoxic stress. HYPOXIA (AUCKLAND, N.Z.) 2018; 6:35-56. [PMID: 30519596 PMCID: PMC6234990 DOI: 10.2147/hp.s173589] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The brain is extensively vascularized, useŝ20% of the body's oxygen, and is highly sensitive to changes in oxygen. While synaptic plasticity and memory are impaired in healthy individuals by exposure to mild hypoxia, aged individuals appear to be even more sensitive. Aging is associated with progressive failure in pulmonary and cardiovascular systems, exposing the aged to both chronic and superimposed acute hypoxia. The HIF proteins, the "master regulators" of the cellular response to hypoxia, are robustly expressed in neurons and astrocytes. Astrocytes support neurons and synaptic plasticity via complex metabolic and trophic mechanisms. The activity of HIF proteins in the brain is diminished with aging, and the increased exposure to chronic and acute hypoxia with aging combined with diminished HIF activity may impair synaptic plasticity. PURPOSE Herein, we test the hypothesis that astrocyte HIF supports synaptic plasticity and learning upon hypoxia. MATERIALS AND METHODS An Astrocyte-specific HIF loss-of-function model was employed, where knock-out of HIF-1α or HIF-2α in GFAP expressing cells was accomplished by cre-mediated recombination. Animals were tested for behavioral (open field and rotarod), learning (passive avoidance paradigm), and electrophysiological (long term potentiation) responses to mild hypoxic challenge. RESULTS In an astrocyte-specific HIF loss-of-function model followed by mild hypoxia, we identified that the depletion of HIF-2α resulted in an impaired passive avoidance learning performance. This was accompanied by an attenuated response to induction in long-term potentiation (LTP), suggesting that the hippocampal circuitry was perturbed upon hypoxic exposure following HIF-2α loss in astrocytes, and not due to hippocampal cell death. We investigated HIF-regulated trophic and metabolic target genes and found that they were not regulated by HIF-2α, suggesting that these specific targets may not be involved in mediating the phenotypes observed. CONCLUSION Together, these results point to a role for HIF-2α in the astrocyte's regulatory role in synaptic plasticity and learning under hypoxia and suggest that even mild, acute hypoxic challenges can impair cognitive performance in the aged population who harbor impaired HIF function.
Collapse
Affiliation(s)
- Cindy V Leiton
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Elyssa Chen
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Alissa Cutrone
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kristy Conn
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Kennelia Mellanson
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
| | - Dania M Malik
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Klingener
- Department of Genetics, Stony Brook University, Stony Brook, NY, USA
| | - Ryan Lamm
- Department of General Surgery, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Michael Cutrone
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - John Petrie
- Department of Biochemistry & Molecular Biology, Bloomberg School of Public health, Johns Hopkins University, Baltimore, MD, USA
| | - Joher Sheikh
- Department of Physiology and Biophysics, Georgetown University, Washington, DC, USA
| | - Adriana DiBua
- Department of Chemistry, Hofstra University, Hempstead, NY, USA
| | - Betsy Cohen
- Computer Science Department, Swarthmore College, Swarthmore, PA, USA
| | - Thomas F Floyd
- Department of Anesthesiology and Pain Management, University of Texas Southwestern, Dallas, TX, USA,
- Department of Cardiothoracic Surgery, University of Texas Southwestern, Dallas, TX, USA,
- Department of Radiology, University of Texas Southwestern, Dallas, TX, USA,
| |
Collapse
|
10
|
Desmettre TJ. Epigenetics in Age-related Macular Degeneration (AMD). J Fr Ophtalmol 2018; 41:e407-e415. [PMID: 30458925 DOI: 10.1016/j.jfo.2018.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 01/03/2023]
Abstract
Age-related Macular Degeneration (AMD) is a complex multifactorial condition involving multiple genetic, environmental and constitutional factors. Inflammation, oxidative stress and lipid metabolism seem to be the most important factors in the pathogenesis of the disease. The importance of genetic factors has mainly been revealed with the influence of histocompatibility complement factor H (CFH) variations and the ARSM2 susceptibility gene. Another component, epigenetics, could help to explain some of the relationships between environmental and genetic factors. Epigenetics is defined as the study of modulations of gene activity that can be transmitted over cell divisions without involving mutation of the DNA sequence. The molecules that are involved in these mechanisms are referred to as the epigenome. The mechanisms involve DNA methylation, histone modification, chromatin remodeling, and gene inhibition by non-coding RNA. Epigenetics could explain how the environment may induce relatively stable changes in traits or even diseases, possibly inheritable over several generations. Epigenetic traits established during development, and/or acquired under the influence of nutritional factors or other environmental factors, could influence the interactions between genes and the environment. Several authors have recently shown the influence of epigenetic factors in the pathogenesis of ocular diseases such as cataract, dry eye, glaucoma, diabetic retinopathy and more recently AMD. A better understanding of the involvement of genetic variants at risk, their relationship with epigenetics and environmental factors would certainly help to better assess the risk of developing AMD or better understand recent changes in the incidence of the disease.
Collapse
Affiliation(s)
- T J Desmettre
- Centre de rétine médicale, 187, rue de Menin, 59520 Marquette-Lez-Lille, France; London International Medical Centre, 18-22 Queen Anne Street, London, W1G 8HU, United Kingdom.
| |
Collapse
|
11
|
Moeini M, Lu X, Avti PK, Damseh R, Bélanger S, Picard F, Boas D, Kakkar A, Lesage F. Compromised microvascular oxygen delivery increases brain tissue vulnerability with age. Sci Rep 2018; 8:8219. [PMID: 29844478 PMCID: PMC5974237 DOI: 10.1038/s41598-018-26543-w] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/16/2018] [Indexed: 11/22/2022] Open
Abstract
Despite the possible role of impaired cerebral tissue oxygenation in age-related cognition decline, much is still unknown about the changes in brain tissue pO2 with age. Using a detailed investigation of the age-related changes in cerebral tissue oxygenation in the barrel cortex of healthy, awake aged mice, we demonstrate decreased arteriolar and tissue pO2 with age. These changes are exacerbated after middle-age. We further uncovered evidence of the presence of hypoxic micro-pockets in the cortex of awake old mice. Our data suggests that from young to middle-age, a well-regulated capillary oxygen supply maintains the oxygen availability in cerebral tissue, despite decreased tissue pO2 next to arterioles. After middle-age, due to decreased hematocrit, reduced capillary density and higher capillary transit time heterogeneity, the capillary network fails to compensate for larger decreases in arterial pO2. The substantial decrease in brain tissue pO2, and the presence of hypoxic micro-pockets after middle-age are of significant importance, as these factors may be related to cognitive decline in elderly people.
Collapse
Affiliation(s)
- Mohammad Moeini
- Biomedical Engineering Institute, École Polytechnique de Montréal, Montréal, QC, Canada.,Research Center of Montreal Heart Institute, Montréal, QC, Canada.,Department of Chemistry, McGill University, Montréal, QC, Canada
| | - Xuecong Lu
- Biomedical Engineering Institute, École Polytechnique de Montréal, Montréal, QC, Canada.,Research Center of Montreal Heart Institute, Montréal, QC, Canada
| | - Pramod K Avti
- Biomedical Engineering Institute, École Polytechnique de Montréal, Montréal, QC, Canada.,Research Center of Montreal Heart Institute, Montréal, QC, Canada.,Department of Biophysics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rafat Damseh
- Biomedical Engineering Institute, École Polytechnique de Montréal, Montréal, QC, Canada
| | - Samuel Bélanger
- Biomedical Engineering Institute, École Polytechnique de Montréal, Montréal, QC, Canada.,Research Center of Montreal Heart Institute, Montréal, QC, Canada
| | - Frédéric Picard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec (IUCPQ), Québec, QC, Canada
| | - David Boas
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Biomedical Engineering Department, College of Engineering, Boston University, Boston, MA, USA
| | - Ashok Kakkar
- Department of Chemistry, McGill University, Montréal, QC, Canada
| | - Frédéric Lesage
- Biomedical Engineering Institute, École Polytechnique de Montréal, Montréal, QC, Canada. .,Research Center of Montreal Heart Institute, Montréal, QC, Canada.
| |
Collapse
|
12
|
Zhou YH, Han QF, Wang LH, Liu T, Meng XY, Wu L, Li T, Jiao YR, Yao HC, Zhang DY. High mobility group box 1 protein attenuates myocardial ischemia reperfusion injury via inhibition of the p38 mitogen-activated protein kinase signaling pathway. Exp Ther Med 2017; 14:1582-1588. [PMID: 28810624 PMCID: PMC5525636 DOI: 10.3892/etm.2017.4653] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 04/28/2017] [Indexed: 01/07/2023] Open
Abstract
The present study aimed to determine the effects of high mobility group box 1 protein (HMGB1) on myocardial ischemia reperfusion (I/R) injury in rats following acute myocardial ischemia and investigate the underlying molecular mechanisms of these effects. Male Wistar rats were randomly divided into the following groups (n=10/group): Sham operation; I/R; HMGB50 (50 ng/kg HMGB1 before I/R); HMGB100 (100 ng/kg HMGB1 before I/R); and HMGB200 (200 ng/kg HMGB1 before I/R). Serum cardiac troponin I (cTnI), interleukin (IL)-6 and tumor necrosis factor (TNF)-α levels were subsequently measured. Myocardial levels of malondialdehyde (MDA) and superoxide dismutase (SOD) were also determined. Myocardial infarction size (IS) was determined by 2,3,5-triphenyltetrazolium chloride staining. Myocardial expression of hypoxia inducible factor (HIF)-1α and phosphorylated p38 mitogen-activated protein kinase (P-p38 MAPK) protein was measured using western blotting. The results demonstrated that HMGB1 significantly decreased serum levels of cTnI, IL-6 and TNF-α and myocardial IS in I/R rats compared with the sham group (all P<0.05). HMGB1 also significantly decreased and increased myocardial levels of MDA and SOD, respectively (both P<0.05). HMGB1 significantly increased myocardial expression of HIF-1α and decreased expression of P-p38 MAPK following I/R (both P<0.05). These effects of HMGB1 occurred in a dose-dependent manner. The results of the current study indicate that the cardioprotective effects of intravenous HMGB1 are associated with increased myocardial expression of HIF-1α via inhibition of P-p38 MAPK expression, leading to inhibition of the P-p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Yan-Hong Zhou
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Qian-Feng Han
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Lan-Hua Wang
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Tao Liu
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Xiao-Yan Meng
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Lei Wu
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Tai Li
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Yue-Ru Jiao
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Heng-Chen Yao
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - De-Yong Zhang
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
13
|
Robust gene expression changes in the ganglia following subclinical reactivation in rhesus macaques infected with simian varicella virus. J Neurovirol 2017; 23:520-538. [PMID: 28321697 DOI: 10.1007/s13365-017-0522-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/03/2017] [Accepted: 02/17/2017] [Indexed: 12/20/2022]
Abstract
Varicella zoster virus (VZV) causes varicella during acute infection and establishes latency in the sensory ganglia. Reactivation of VZV results in herpes zoster, a debilitating and painful disease. It is believed that VZV reactivates due to a decline in cell-mediated immunity; however, the roles that CD4 versus CD8 T cells play in the prevention of herpes zoster remain poorly understood. To address this question, we used a well-characterized model of VZV infection where rhesus macaques are intrabronchially infected with the homologous simian varicella virus (SVV). Latently infected rhesus macaques were thymectomized and depleted of either CD4 or CD8 T cells to induce selective senescence of each T cell subset. After T cell depletion, the animals were transferred to a new housing room to induce stress. SVV reactivation (viremia in the absence of rash) was detected in three out of six CD8-depleted and two out of six CD4-depleted animals suggesting that both CD4 and CD8 T cells play a critical role in preventing SVV reactivation. Viral loads in multiple ganglia were higher in reactivated animals compared to non-reactivated animals. In addition, reactivation results in sustained transcriptional changes in the ganglia that enriched to gene ontology and diseases terms associated with neuronal function and inflammation indicative of potential damage as a result of viral reactivation. These studies support the critical role of cellular immunity in preventing varicella virus reactivation and indicate that reactivation results in long-lasting remodeling of the ganglia transcriptome.
Collapse
|
14
|
Haller M, Khalid S, Kremser L, Fresser F, Furlan T, Hermann M, Guenther J, Drasche A, Leitges M, Giorgio M, Baier G, Lindner H, Troppmair J. Novel Insights into the PKCβ-dependent Regulation of the Oxidoreductase p66Shc. J Biol Chem 2016; 291:23557-23568. [PMID: 27624939 PMCID: PMC5095410 DOI: 10.1074/jbc.m116.752766] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Indexed: 12/12/2022] Open
Abstract
Dysfunctional mitochondria contribute to the development of many diseases and pathological conditions through the excessive production of reactive oxygen species (ROS), and, where studied, ablation of p66Shc (p66) was beneficial. p66 translocates to the mitochondria and oxidizes cytochrome c to yield H2O2, which in turn initiates cell death. PKCβ-mediated phosphorylation of serine 36 in p66 has been implicated as a key regulatory step preceding mitochondrial translocation, ROS production, and cell death, and PKCβ thus may provide a target for therapeutic intervention. We performed a reassessment of PKCβ regulation of the oxidoreductase activity of p66. Although our experiments did not substantiate Ser36 phosphorylation by PKCβ, they instead provided evidence for Ser139 and Ser213 as PKCβ phosphorylation sites regulating the pro-oxidant and pro-apoptotic function of p66. Mutation of another predicted PKCβ phosphorylation site also located in the phosphotyrosine binding domain, threonine 206, had no phenotype. Intriguingly, p66 with Thr206 and Ser213 mutated to glutamic acid showed a gain-of-function phenotype with significantly increased ROS production and cell death induction. Taken together, these data argue for a complex mechanism of PKCβ-dependent regulation of p66 activation involving Ser139 and a motif surrounding Ser213.
Collapse
Affiliation(s)
- Martina Haller
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | - Sana Khalid
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | - Leopold Kremser
- Division of Clinical Biochemistry, Protein Micro-Analysis Facility
| | - Friedrich Fresser
- Department for Pharmacology and Genetics, Division of Translational Cell Genetics, and
| | - Tobias Furlan
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | - Martin Hermann
- Department for Anesthesiology and Intensive Care, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Julia Guenther
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | - Astrid Drasche
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | | | - Marco Giorgio
- the European Institute of Oncology, 20139 Milano, Italy
| | - Gottfried Baier
- Department for Pharmacology and Genetics, Division of Translational Cell Genetics, and
| | - Herbert Lindner
- Division of Clinical Biochemistry, Protein Micro-Analysis Facility
| | - Jakob Troppmair
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery,
| |
Collapse
|
15
|
Sakita M, Murakami S, Fujino H. Age-related morphological regression of myelinated fibers and capillary architecture of distal peripheral nerves in rats. BMC Neurosci 2016; 17:39. [PMID: 27342571 PMCID: PMC4919893 DOI: 10.1186/s12868-016-0277-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/16/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Regression of myelinated peripheral nerve fibers in the lower extremities contributes to sarcopenia and balance dysfunction in normal aging. This subclinical regression of myelinated fibers (MFs) is heavily influenced by alterations in microvasculature, though the mechanism underlying these age-related degenerative phenomena remains unclear. The aim of the present study was to examine age-related regressions in myelinated distal peripheral nerve fibers as well as capillary architecture in rats using both morphological and histochemical methods. RESULTS MFs were categorized into tertiles of 'large', 'medium', and 'small' sizes based on the distribution of MF diameters. A two-way ANOVA was used to assess effects of fiber size (large/medium/small) and group (young/elderly) on myelin thickness, axon diameter, myelin perimeter, axon perimeter, and G-ratio (axon diameter/fiber diameter). Significant main effects were observed for both MF size and group with respect to all dimensions except for G-ratio. Values for fiber diameter (P < 0.01), myelin thickness (P < 0.01), axon diameter (P < 0.01), myelin perimeter (P < 0.01), and axon perimeter (P < 0.01) were significantly lower than those in the young group. Additionally, mean capillary diameter and number of microvascular branch points were significantly lower in the elderly group than in the young group. CONCLUSIONS The present study demonstrated that spontaneous age-related regression predominantly occurs for all fiber sizes in the distal peripheral nerves and the capillary architecture. The results of the present study further suggest that both the distal MFs and capillaries in the peripheral nerve may simultaneously regress with aging.
Collapse
Affiliation(s)
- Masahiro Sakita
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, 34 Oyakeyamada, Yamashina Ward, Kyoto City, Kyoto, 607-8175, Japan.
| | - Shinichiro Murakami
- Department of Physical Therapy, Faculty of Health Care Sciences, Himeji-Dokkyo University, 7-2-1 Kamiono, Himeji City, Hyogo, 670-0896, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma Ward, Kobe City, Hyogo, 654-0412, Japan
| |
Collapse
|
16
|
Yao HC, Zhou M, Zhou YH, Wang LH, Zhang DY, Han QF, Liu T, Wu L, Tian KL, Zhang M. Intravenous high mobility group box 1 upregulates the expression of HIF-1α in the myocardium via a protein kinase B-dependent pathway in rats following acute myocardial ischemia. Mol Med Rep 2015; 13:1211-9. [PMID: 26648172 PMCID: PMC4732844 DOI: 10.3892/mmr.2015.4648] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 11/10/2015] [Indexed: 12/23/2022] Open
Abstract
The effects of intravenous high mobility group box 1 (HMGB1) on myocardial ischemia/reperfusion (I/R) injury remains to be elucidated. The purpose of the present study was to investigate the effects of intravenous HMGB1 on the expression of hypoxia inducible factor-1α (HIF-1α) in the myocardium of rats following acute myocardial ischemia, and to examine the effects of intravenous HMGB1 on myocardial I/R injury. Male Wistar rats were divided into the following groups: Sham operation group (n=10), a group exposed to ischemia for 30 min and reperfusion for 4 h (I/R group) as a control (n=10), an HMGB group, in which 100 ng/kg HMGB was administered intravenously 30 min prior to ischemia (n=10), an LY group, in whic LY294002, an inhibitor of phosphoinositide 3-kinase (PI3K), was administered intravenously (0.3 mg/kg) 40 min prior to ischemia (n=10), and the HMGB1+LY group, in which HMGB1 (100 ng/kg) and LY294002 (0.3 mg/kg) were administered intravenously 30 min and 40 min prior to ischemia, respectively (n=10). The serum levels of cardiac troponin I (cTnI) and tumor necrosis factor-α (TNF-α), and myocardial infarct size were measured. The expression levels of phosphorylated Akt and HIF-1α were investigated using western blot analyses. The results showed that pre-treatment with HMGB1 significantly decreased serum levels of cTnI, and TNF-α, and reduced myocardial infarct size following 4 h reperfusion (all P<0.05). HMGB1 also increased the expression levels of HIF-1α and p-Akt induced by I/R (P<0.05). LY294002 was found to eliminate the effects of intravenous HMGB1 on myocardial I/R injury (P<0.05). These results suggest that intravenous pre-treatment with HMGB1 may exert its cardioprotective effects via the upregulation of the myocardial expression of HIF-1α, which may be regulated by the PI3K/Akt signaling pathway, in rats following acute myocardial I/R.
Collapse
Affiliation(s)
- Heng-Chen Yao
- Department of Cardiology, Qilu Hospital of Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Min Zhou
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Yan-Hong Zhou
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Lan-Hua Wang
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - De-Yong Zhang
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Qian-Feng Han
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Tao Liu
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Lei Wu
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Ke-Li Tian
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Mei Zhang
- Department of Cardiology, Qilu Hospital of Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
17
|
Gemenetzi M, Lotery AJ. The role of epigenetics in age-related macular degeneration. Eye (Lond) 2014; 28:1407-17. [PMID: 25233816 DOI: 10.1038/eye.2014.225] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 07/31/2014] [Indexed: 12/27/2022] Open
Abstract
It is becoming increasingly evident that epigenetic mechanisms influence gene expression and can explain how interactions between genetics and the environment result in particular phenotypes during development. The extent to which this epigenetic effect contributes to phenotype heritability in age-related macular degeneration (AMD) is currently ill defined. However, emerging evidence suggests that epigenetic changes are relevant to AMD and as such provide an exciting new avenue of research for AMD. This review addresses information on the impact of posttranslational modification of the genome on the pathogenesis of AMD, such as DNA methylation changes affecting antioxidant gene expression, hypoxia-regulated alterations in chromatin structure, and histone acetylation status in relation to angiogenesis and inflammation. It also contains information on the role of non-coding RNA-mediated gene regulation in AMD at a posttranscriptional (before translation) level. Our aim was to review the epigenetic mechanisms that cause heritable changes in gene activity without changing the DNA sequence. We also describe some long-term alterations in the transcriptional potential of a cell, which are not necessarily heritable but remains to be defined in the future. Increasing understanding of the significance of common and rare genetic variants and their relationship to epigenetics and environmental influences may help in establishing methods to assess the risk of AMD. This in turn may allow new therapeutic interventions for the leading cause of central vision impairment in patients over the age of 50 years in developed countries. Search strategy We searched the MEDLINE/PubMed database following MeSH suggestions for articles including the terms: 'ocular epigenetic mechanisms', 'human disease epigenetics', and 'age-related macular degeneration genetics'. The headline used to locate related articles in PubMed was 'epigenetics in ocular disease', and to restrict search, we used the headlines 'DNA methylation in age related macular degeneration', 'altered gene expression in AMD pathogenesis'. A manual search was also based on references from these articles as well as review articles.
Collapse
Affiliation(s)
- M Gemenetzi
- Southampton Eye Unit, Southampton University Hospital, Southampton, UK
| | - A J Lotery
- 1] Southampton Eye Unit, Southampton University Hospital, Southampton, UK [2] Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| |
Collapse
|
18
|
Dalmases M, Torres M, Márquez-Kisinousky L, Almendros I, Planas AM, Embid C, Martínez-Garcia MÁ, Navajas D, Farré R, Montserrat JM. Brain tissue hypoxia and oxidative stress induced by obstructive apneas is different in young and aged rats. Sleep 2014; 37:1249-56. [PMID: 25061253 DOI: 10.5665/sleep.3848] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES To test the hypotheses that brain oxygen partial pressure (PtO2) in response to obstructive apneas changes with age and that it might lead to different levels of cerebral tissue oxidative stress. DESIGN Prospective controlled animal study. SETTING University laboratory. PARTICIPANTS Sixty-four male Wistar rats: 32 young (3 mo old) and 32 aged (18 mo). INTERVENTIONS Protocol 1: Twenty-four animals were subjected to obstructive apneas (50 apneas/h, lasting 15 sec each) or to sham procedure for 50 min. Protocol 2: Forty rats were subjected to obstructive apneas or sham procedure for 4 h. MEASUREMENTS AND RESULTS Protocol 1: Real-time PtO2 measurements were performed using a fast-response oxygen microelectrode. During successive apneas cerebral cortex PtO2 presented a different pattern in the two age groups; there was a fast increase in young rats, whereas it remained without significant changes between the beginning and the end of the protocol in the aged group. Protocol 2: Brain oxidative stress assessed by lipid peroxidation increased after apneas in young rats (1.34 ± 0.17 nmol/mg of protein) compared to old ones (0.63 ± 0.03 nmol/mg), where a higher expression of antioxidant enzymes was observed. CONCLUSIONS The results suggest that brain oxidative stress in aged rats is lower than in young rats in response to recurrent apneas, mimicking obstructive sleep apnea. This could be due to the different PtO2 response observed between age groups and the increased antioxidant expression in aged rats. CITATION Dalmases M, Torres M, Márquez-Kisinousky L, Almendros I, Planas AM, Embid C, Martínez-Garcia MA, Navajas D, Farré R, Montserrat JM. Brain tissue hypoxia and oxidative stress induced by obstructive apneas is different in young and aged rats.
Collapse
Affiliation(s)
- Mireia Dalmases
- Laboratori de la Son, Pneumologia, Hospital Clinic-Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain ; CIBER Enfermedades Respiratorias, Bunyola, Spain
| | - Marta Torres
- Laboratori de la Son, Pneumologia, Hospital Clinic-Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain ; CIBER Enfermedades Respiratorias, Bunyola, Spain
| | | | - Isaac Almendros
- Laboratori de la Son, Pneumologia, Hospital Clinic-Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain ; CIBER Enfermedades Respiratorias, Bunyola, Spain
| | - Anna M Planas
- Departament d'Isquemia cerebral y neurodegeneració, IIBB-CSIC-IDIBAPS, Barcelona, Spain
| | - Cristina Embid
- Laboratori de la Son, Pneumologia, Hospital Clinic-Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain ; CIBER Enfermedades Respiratorias, Bunyola, Spain
| | | | - Daniel Navajas
- CIBER Enfermedades Respiratorias, Bunyola, Spain ; Unitat de Biofisica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain ; Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Ramon Farré
- CIBER Enfermedades Respiratorias, Bunyola, Spain ; Unitat de Biofisica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain
| | - Josep Maria Montserrat
- Laboratori de la Son, Pneumologia, Hospital Clinic-Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain ; CIBER Enfermedades Respiratorias, Bunyola, Spain
| |
Collapse
|
19
|
Developmental and pathological angiogenesis in the central nervous system. Cell Mol Life Sci 2014; 71:3489-506. [PMID: 24760128 DOI: 10.1007/s00018-014-1625-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 01/24/2023]
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing vessels, in the central nervous system (CNS) is seen both as a normal physiological response as well as a pathological step in disease progression. Formation of the blood-brain barrier (BBB) is an essential step in physiological CNS angiogenesis. The BBB is regulated by a neurovascular unit (NVU) consisting of endothelial and perivascular cells as well as vascular astrocytes. The NVU plays a critical role in preventing entry of neurotoxic substances and regulation of blood flow in the CNS. In recent years, research on numerous acquired and hereditary disorders of the CNS has increasingly emphasized the role of angiogenesis in disease pathophysiology. Here, we discuss molecular mechanisms of CNS angiogenesis during embryogenesis as well as various pathological states including brain tumor formation, ischemic stroke, arteriovenous malformations, and neurodegenerative diseases.
Collapse
|
20
|
Sakita M, Murakami S, Fujino H. The Morphological Changes in the Capillary Architecture of the Tibial Nerve Associated with Spontaneous Aging and Aerobic Exercise Intervention during Aging in Rats. J Phys Ther Sci 2014; 26:263-7. [PMID: 24648645 PMCID: PMC3944302 DOI: 10.1589/jpts.26.263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 09/05/2013] [Indexed: 01/26/2023] Open
Abstract
[Purpose] Peripheral nerve degradation associated with aging is linked to failure of
interactions in capillary metabolism. The aim of this study was to morphologically
investigate the age-related changes in the capillary architecture of the tibial nerve in
spontaneous aging and with aerobic exercise intervention in rats. [Subjects] Male
Sprague-Dawley rats (n=15) were used in the present study. [Methods] The rats were divided
into control (Cont, n=5), elderly (Elder, n=5), and elderly with aerobic exercise
(Elder+Ex, n=5) groups. Aerobic training of low intensity was performed for 10 weeks using
a treadmill starting at 96 weeks of age by the Elder+Ex group. The capillary diameter,
cross-sectional area and number of microvascular ramifications in the tibial nerve were
compared among the Cont (20-week-old), Elder (106-week-old) and Elder+Ex groups using
three-dimensional images gained from confocal laser scanning microscopy. [Results] The
capillary diameter, cross-sectional area and number of microvascular ramifications in the
Elder group were significantly smaller than those observed in the Cont and Elder+Ex
groups. [Conclusion] These findings suggest that the capillaries in the peripheral nerve
degrade with spontaneous aging and that aerobic exercise of low intensity promotes
angiogenesis, and protects the capillary from oxidative stress.
Collapse
Affiliation(s)
- Masahiro Sakita
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Japan
| | - Shinichiro Murakami
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Japan
| | - Hidemi Fujino
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Japan
| |
Collapse
|
21
|
Takechi R, Pallebage-Gamarallage MM, Lam V, Giles C, Mamo JC. Nutraceutical agents with anti-inflammatory properties prevent dietary saturated-fat induced disturbances in blood-brain barrier function in wild-type mice. J Neuroinflammation 2013; 10:73. [PMID: 23782872 PMCID: PMC3693897 DOI: 10.1186/1742-2094-10-73] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 06/08/2013] [Indexed: 12/11/2022] Open
Abstract
Background Emerging evidence suggests that disturbances in the blood–brain barrier (BBB) may be pivotal to the pathogenesis and pathology of vascular-based neurodegenerative disorders. Studies suggest that heightened systemic and central inflammations are associated with BBB dysfunction. This study investigated the effect of the anti-inflammatory nutraceuticals garlic extract-aged (GEA), alpha lipoic acid (ALA), niacin, and nicotinamide (NA) in a murine dietary-induced model of BBB dysfunction. Methods C57BL/6 mice were fed a diet enriched in saturated fatty acids (SFA, 40% fat of total energy) for nine months to induce systemic inflammation and BBB disturbances. Nutraceutical treatment groups included the provision of either GEA, ALA, niacin or NA in the positive control SFA-group and in low-fat fed controls. Brain parenchymal extravasation of plasma derived immunoglobulin G (IgG) and large macromolecules (apolipoprotein (apo) B lipoproteins) measured by quantitative immunofluorescent microscopy, were used as markers of disturbed BBB integrity. Parenchymal glial fibrillar acidic protein (GFAP) and cyclooxygenase-2 (COX-2) were considered in the context of surrogate markers of neurovascular inflammation and oxidative stress. Total anti-oxidant status and glutathione reductase activity were determined in plasma. Results Brain parenchymal abundance of IgG and apoB lipoproteins was markedly exaggerated in mice maintained on the SFA diet concomitant with significantly increased GFAP and COX-2, and reduced systemic anti-oxidative status. The nutraceutical GEA, ALA, niacin, and NA completely prevented the SFA-induced disturbances of BBB and normalized the measures of neurovascular inflammation and oxidative stress. Conclusions The anti-inflammatory nutraceutical agents GEA, ALA, niacin, or NA are potent inhibitors of dietary fat-induced disturbances of BBB induced by systemic inflammations.
Collapse
Affiliation(s)
- Ryusuke Takechi
- School of Public Health, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Faculty of Health Sciences, Curtin University, Kent st, Bentley, WA, 6102, Australia
| | | | | | | | | |
Collapse
|
22
|
Wang Z, Si LY. Hypoxia-inducible factor-1α and vascular endothelial growth factor in the cardioprotective effects of intermittent hypoxia in rats. Ups J Med Sci 2013; 118:65-74. [PMID: 23441597 PMCID: PMC3633332 DOI: 10.3109/03009734.2013.766914] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 12/11/2012] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE This study investigated the effects of short-term intermittent hypoxia (IH) preconditioning on cardiac structure and function in rats and the influence of ischemia reperfusion (I/R) injury. Special attention was then paid to the involvement of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF). METHODS Wistar rats were given IH treatment for 1, 7, 14, or 28 days. Some of them were thereafter subject to myocardial infarction surgery. Right ventricle systolic pressure (RVSP), myocardial capillary density (CD), and mRNA/protein expression of HIF-1α, VEGF, and Bcl-2 in rat myocardial tissue were determined. Apoptotic cell number was determined by TUNEL staining, and concentrations of malondialdehyde (MDA) and superoxide dismutase (SOD) were measured. RESULTS IH treatment for 1, 7, 14, and 28 days reduced the myocardial infarction size, whereas IH for 28 days increased the RVSP, ratio of right to left ventricle weight (RV/LV+S), and CD. IH up-regulated the mRNA and protein levels of HIF-1α, VEGF, and Bcl-2 both under normal and I/R conditions. The induced expression of HIF-1α and VEGF by IH reached a peak after 7 days of treatment. Moreover, IH for 28 days induced cardiomyocyte apoptosis, whereas prior treatment with IH for 1, 7, 14, and 28 days all markedly attenuated the apoptosis effected by the subsequent I/R injury. IH also decreased the concentrations of MDA but increased those of SOD in myocardial tissue of both in normal rats and following I/R. CONCLUSIONS The present study demonstrates that short-term IH protects the heart from I/R injury through inhibiting apoptosis and oxidative stress. The up-regulation of HIF-1α and VEGF by short-term IH may participate in the cardioprotective effect of IH.
Collapse
Affiliation(s)
- Zhang Wang
- Department of Geriatrics, The First Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Liang-Yi Si
- Department of Geriatrics, The First Affiliated Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
23
|
Takechi R, Pallebage-Gamarallage MM, Lam V, Giles C, Mamo JC. Aging-related changes in blood-brain barrier integrity and the effect of dietary fat. NEURODEGENER DIS 2012; 12:125-35. [PMID: 23128303 DOI: 10.1159/000343211] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 09/06/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Disturbances in blood-brain barrier (BBB) integrity contribute to the onset and progression of neurodegenerative diseases including Alzheimer's disease (AD) and vascular dementia (VaD). Aging is positively associated with AD and VaD risk, but this may reflect comorbidities or the effects of other chronic modulators of vascular function such as diet. OBJECTIVE To explore putative synergistic effects of aging with diet, in this study genetically unmanipulated mice were maintained on diets enriched in saturated fatty acids (SFA) or cholesterol and compared to mice provided with low-fat (LF) feed formula. METHODS The functional integrity of the BBB was assessed following 3, 6 and 12 months of dietary intervention commenced at 6 weeks of age, by determining the brain parenchymal extravasation of immunoglobulin G (IgG). RESULTS Mice maintained on the SFA- or cholesterol-enriched diet showed significant parenchymal IgG abundance following 3 months of feeding, concomitant with diminished expression of the tight junction protein occludin. LF control mice had essentially no evidence of BBB disturbances. Six months of SFA feeding exacerbated the difference in IgG abundance compared to the LF mice. At 12 months of feeding, the control LF mice also had significant parenchymal IgG that was comparable to mice fed the SFA- or cholesterol-enriched diet for 3 months. However, there may have been an adaptation to the fat-enriched diets because SFA and cholesterol did not exacerbate IgG parenchymal accumulation beyond 6 months of feeding. CONCLUSION Collectively, the study suggests that diets enriched in SFA or cholesterol accelerate the onset of BBB dysfunction that otherwise occurs with aging.
Collapse
Affiliation(s)
- R Takechi
- Curtin Health Innovation Research Institute, Faculty of Health Science, School of Public Health, Curtin University, Perth, W.A., Australia
| | | | | | | | | |
Collapse
|
24
|
Labbé A, Lafleur VN, Patten DA, Robitaille GA, Garand C, Lamalice L, Lebel M, Richard DE. The Werner syndrome gene product (WRN): a repressor of hypoxia-inducible factor-1 activity. Exp Cell Res 2012; 318:1620-32. [PMID: 22659133 DOI: 10.1016/j.yexcr.2012.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 03/19/2012] [Accepted: 04/10/2012] [Indexed: 11/30/2022]
Abstract
Werner syndrome (WS) is a rare autosomal disease characterized by the premature onset of several age-associated pathologies. The protein defective in WS patients (WRN) is a helicase/exonuclease involved in DNA repair, replication, transcription and telomere maintenance. Hypoxia-inducible factor-1 (HIF-1) is a decisive element for the transcriptional regulation of genes essential for adaptation to low oxygen conditions. HIF-1 is also implicated in the molecular mechanisms of ageing. Here, we show that the cellular depletion of WRN protein (by siRNA targeting) leads to increased HIF-1 complex stabilization and activation. HIF-1 activation in the absence of WRN involves the generation of mitochondrial reactive oxygen species (mtROS) since SkQ1, a mitochondrial-targeted antioxidant, and stigmatellin, an inhibitor of mitochondrial complex III, blocked increased HIF-1 levels. Ascorbate, an essential co-factor involved in HIF-1 stability, was decreased in WRN-depleted cells. Interestingly, expression levels of GLUT1, a known dehydroascorbic acid transporter, were also decreased in WRN-depleted cells. Ascorbate supplementation of WRN-depleted cells led to a dose-dependent inhibition of HIF-1 activation. These results indicate that WRN protein regulates HIF-1 activation by affecting mitochondrial ROS production and intracellular ascorbate levels. This work provides a novel mechanistic link between HIF-1 activity and different age-associated pathologies.
Collapse
Affiliation(s)
- Adam Labbé
- Centre de Recherche en Cancérologie de l'Université Laval, Centre de recherche du CHUQ, L'Hôtel-Dieu de Québec, Québec, QC, Canada G1R 2J6
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Cardiorespiratory parameters during submaximal exercise under acute exposure to normobaric and hypobaric hypoxia. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.apunts.2011.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
26
|
Cataldi A, Zara S, Rapino M, Zingariello M, di Giacomo V, Antonucci A. p53 and telomerase control rat myocardial tissue response to hypoxia and ageing. Eur J Histochem 2012; 53:e25. [PMID: 22073357 PMCID: PMC3167339 DOI: 10.4081/ejh.2009.e25] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2009] [Indexed: 01/30/2023] Open
Abstract
Cellular senescence implies loss of proliferative and tissue regenerative capability. Also hypoxia, producing Reactive Oxygen Species (ROS), can damage cellular components through the oxidation of DNA, proteins and lipids, thus influencing the shortening of telomeres. Since ribonucleoprotein Telomerase (TERT), catalyzing the replication of the ends of eukaryotic chromosomes, promotes cardiac muscle cell proliferation, hypertrophy and survival, here we investigated its role in the events regulating apoptosis occurrence and life span in hearts deriving from young and old rats exposed to hypoxia. TUNEL (terminal-deoxinucleotidyl -transferase- mediated dUTP nick end-labeling) analysis reveals an increased apoptotic cell number in both samples after hypoxia exposure, mainly in the young with respect to the old. TERT expression lowers either in the hypoxic young, either in the old in both experimental conditions, with respect to the normoxic young. These events are paralleled by p53 and HIF-1 α expression dramatic increase and by p53/ HIF-1 α co-immunoprecipitation in the hypoxic young, evidencing the young subject as the most stressed by such challenge. These effects could be explained by induction of damage to genomic DNA by ROS that accelerates cell senescence through p53 activation. Moreover, by preventing TERT enzyme down-regulation, cell cycle exit and apoptosis occurrence could be delayed and new possibilities for intervention against cell ageing and hypoxia could be opened.
Collapse
Affiliation(s)
- A Cataldi
- Dipartimento di Biomorfologia, Istituto di Genetica Molecolare del CNR, Chieti, Italy.
| | | | | | | | | | | |
Collapse
|
27
|
Treatment with edaravone attenuates ischemic brain injury and inhibits neurogenesis in the subventricular zone of adult rats after focal cerebral ischemia and reperfusion injury. Neuroscience 2012; 201:297-306. [DOI: 10.1016/j.neuroscience.2011.11.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 10/18/2011] [Accepted: 11/02/2011] [Indexed: 01/18/2023]
|
28
|
Caprara C, Grimm C. From oxygen to erythropoietin: relevance of hypoxia for retinal development, health and disease. Prog Retin Eye Res 2011; 31:89-119. [PMID: 22108059 DOI: 10.1016/j.preteyeres.2011.11.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/01/2011] [Accepted: 11/07/2011] [Indexed: 12/20/2022]
Abstract
Photoreceptors and other cells of the retina consume large quantities of energy to efficiently convert light information into a neuronal signal understandable by the brain. The necessary energy is mainly provided by the oxygen-dependent generation of ATP in the numerous mitochondria of retinal cells. To secure the availability of sufficient oxygen for this process, the retina requires constant blood flow through the vasculature of the retina and the choroid. Inefficient supply of oxygen and nutrients, as it may occur in conditions of disturbed hemodynamics or vascular defects, results in tissue ischemia or hypoxia. This has profound consequences on retinal function and cell survival, requiring an adaptational response by cells to cope with the reduced oxygen tension. Central to this response are hypoxia inducible factors, transcription factors that accumulate under hypoxic conditions and drive the expression of a large variety of target genes involved in angiogenesis, cell survival and metabolism. Prominent among these factors are vascular endothelial growth factor and erythropoietin, which may contribute to normal angiogenesis during development, but may also cause neovascularization and vascular leakage under pathologically reduced oxygen levels. Since ischemia and hypoxia may have a role in various retinal diseases such as diabetic retinopathy and retinopathy of prematurity, studying the cellular and molecular response to reduced tissue oxygenation is of high relevance. In addition, the concept of preconditioning with ischemia or hypoxia demonstrates the capacity of the retina to activate endogenous survival mechanisms, which may protect cells against a following noxious insult. Part of these mechanisms is the local production of protective factors such as erythropoietin. Due to its plethora of effects in the retina including neuro- and vaso-protective activities, erythropoietin has gained strong interest as potential therapeutic factor for retinal degenerative diseases.
Collapse
Affiliation(s)
- Christian Caprara
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
29
|
Brown WR, Thore CR. Review: cerebral microvascular pathology in ageing and neurodegeneration. Neuropathol Appl Neurobiol 2011; 37:56-74. [PMID: 20946471 DOI: 10.1111/j.1365-2990.2010.01139.x] [Citation(s) in RCA: 526] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This review of age-related brain microvascular pathologies focuses on topics studied by this laboratory, including anatomy of the blood supply, tortuous vessels, venous collagenosis, capillary remnants, vascular density and microembolic brain injury. Our studies feature thick sections, large blocks embedded in celloidin, and vascular staining by alkaline phosphatase. This permits study of the vascular network in three dimensions, and the differentiation of afferent from efferent vessels. Current evidence suggests that there is decreased vascular density in ageing, Alzheimer's disease and leukoaraiosis, and cerebrovascular dysfunction precedes and accompanies cognitive dysfunction and neurodegeneration. A decline in cerebrovascular angiogenesis may inhibit recovery from hypoxia-induced capillary loss. Cerebral blood flow is inhibited by tortuous arterioles and deposition of excessive collagen in veins and venules. Misery perfusion due to capillary loss appears to occur before cell loss in leukoaraiosis, and cerebral blood flow is also reduced in the normal-appearing white matter. Hypoperfusion occurs early in Alzheimer's disease, inducing white matter lesions and correlating with dementia. In vascular dementia, cholinergic reductions are correlated with cognitive impairment, and cholinesterase inhibitors have some benefit. Most lipid microemboli from cardiac surgery pass through the brain in a few days, but some remain for weeks. They can cause what appears to be a type of vascular dementia years after surgery. Donepezil has shown some benefit. Emboli, such as clots, cholesterol crystals and microspheres can be extruded through the walls of cerebral vessels, but there is no evidence yet that lipid emboli undergo such extravasation.
Collapse
Affiliation(s)
- W R Brown
- Department of Radiology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA.
| | | |
Collapse
|
30
|
Chan JYH, Tsai CY, Wu CHY, Li FCH, Dai KY, Sun EYH, Chan SHH, Chang AYW. Sumoylation of hypoxia-inducible factor-1α ameliorates failure of brain stem cardiovascular regulation in experimental brain death. PLoS One 2011; 6:e17375. [PMID: 21390240 PMCID: PMC3048406 DOI: 10.1371/journal.pone.0017375] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 02/01/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND One aspect of brain death is cardiovascular deregulation because asystole invariably occurs shortly after its diagnosis. A suitable neural substrate for mechanistic delineation of this aspect of brain death resides in the rostral ventrolateral medulla (RVLM). RVLM is the origin of a life-and-death signal that our laboratory detected from blood pressure of comatose patients that disappears before brain death ensues. At the same time, transcriptional upregulation of heme oxygenase-1 in RVLM by hypoxia-inducible factor-1α (HIF-1α) plays a pro-life role in experimental brain death, and HIF-1α is subject to sumoylation activated by transient cerebral ischemia. It follows that sumoylation of HIF-1α in RVLM in response to hypoxia may play a modulatory role on brain stem cardiovascular regulation during experimental brain death. METHODOLOGY/PRINCIPAL FINDINGS A clinically relevant animal model that employed mevinphos as the experimental insult in Sprague-Dawley rat was used. Biochemical changes in RVLM during distinct phenotypes in systemic arterial pressure spectrum that reflect maintained or defunct brain stem cardiovascular regulation were studied. Western blot analysis, EMSA, ELISA, confocal microscopy and immunoprecipitation demonstrated that drastic tissue hypoxia, elevated levels of proteins conjugated by small ubiquitin-related modifier-1 (SUMO-1), Ubc9 (the only known conjugating enzyme for the sumoylation pathway) or HIF-1α, augmented sumoylation of HIF-1α, nucleus-bound translocation and enhanced transcriptional activity of HIF-1α in RVLM neurons took place preferentially during the pro-life phase of experimental brain death. Furthermore, loss-of-function manipulations by immunoneutralization of SUMO-1, Ubc9 or HIF-1α in RVLM blunted the upregulated nitric oxide synthase I/protein kinase G signaling cascade, which sustains the brain stem cardiovascular regulatory machinery during the pro-life phase. CONCLUSIONS/SIGNIFICANCE We conclude that sumoylation of HIF-1α in RVLM ameliorates brain stem cardiovascular regulatory failure during experimental brain death via upregulation of nitric oxide synthase I/protein kinase G signaling. This information should offer new therapeutic initiatives against this fatal eventuality.
Collapse
Affiliation(s)
- Julie Y. H. Chan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, Republic of China
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan, Republic of China
| | - Ching-Yi Tsai
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan, Republic of China
| | - Carol H. Y. Wu
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan, Republic of China
| | - Faith C. H. Li
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan, Republic of China
| | - Kuang-Yu Dai
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan, Republic of China
| | - Enya Y. H. Sun
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan, Republic of China
| | - Samuel H. H. Chan
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan, Republic of China
- * E-mail: (SHHC); (AYWC)
| | - Alice Y. W. Chang
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan, Republic of China
- * E-mail: (SHHC); (AYWC)
| |
Collapse
|
31
|
Abstract
String vessels are thin connective tissue strands, remnants of capillaries, with no endothelial cells; they do not carry blood flow. They occur in numerous species, particularly in the central nervous system, but can occur in any tissue where capillaries have died. String vessels are often associated with pathologies such as Alzheimer's disease, ischemia, and irradiation, but are also found in normal human brains from preterm babies to the aged. They provide a record of the original blood vessel location, but gradually disappear after months or years. There have been numerous studies of string vessels (acellular capillaries) in the retina, because retinal vessels can be seen in great detail in whole mounts after trypsin digestion. Capillary regression occurs by apoptosis, synchronously along capillary segments, with macrophages engulfing apoptotic endothelial cells. Macrophages may cause the apoptosis, or the regression may be triggered by loss of the endothelial cell survival factor VEGF. VEGF expression is induced by hypoxia and promotes capillary growth. Cessation of blood flow eliminates the shear stress that helps maintain endothelial cell survival. Capillaries can re-grow by proliferation and migration of endothelial cells into empty basement membrane tubes, which provide a structural scaffold, replete with signaling molecules. This is a problem in tumor control, but useful for recovery from capillary loss. There is an age-related waning of VEGF expression in response to hypoxia. This causes an age-related decline in cerebral angiogenesis and results in neuronal loss. It may also contribute to the proposed age-related loss of brain reserve.
Collapse
Affiliation(s)
- William R Brown
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
32
|
Abstract
The glycemic index (GI) indicates how fast blood glucose is raised after consuming a carbohydrate-containing food. Human metabolic studies indicate that GI is related to patho-physiological responses after meals. Compared with a low-GI meal, a high-GI meal is characterized with hyperglycemia during the early postprandial stage (0-2h) and a compensatory hyperlipidemia associated with counter-regulatory hormone responses during late postprandial stage (4-6h). Over the past three decades, several human health disorders have been related to GI. The strongest relationship suggests that consuming low-GI foods prevents diabetic complications. Diabetic retinopathy (DR) is a complication of diabetes. In this aspect, GI appears to be useful as a practical guideline to help diabetic people choose foods. Abundant epidemiological evidence also indicates positive associations between GI and risk for type 2 diabetes, cardiovascular disease, and more recently, age-related macular degeneration (AMD) in people without diabetes. Although data from randomized controlled intervention trials are scanty, these observations are strongly supported by evolving molecular mechanisms which explain the pathogenesis of hyperglycemia. This wide range of evidence implies that dietary hyperglycemia is etiologically related to human aging and diseases, including DR and AMD. In this context, these diseases can be considered as metabolic retinal diseases. Molecular theories that explain hyperglycemic pathogenesis involve a mitochondria-associated pathway and four glycolysis-associated pathways, including advanced glycation end products formation, protein kinase C activation, polyol pathway, and hexosamine pathway. While the four glycolysis-associated pathways appear to be universal for both normoxic and hypoxic conditions, the mitochondria-associated mechanism appears to be most relevant to the hyperglycemic, normoxic pathogenesis. For diseases that affect tissues with highly active metabolism and that frequently face challenge from low oxygen tension, such as retina in which metabolism is determined by both glucose and oxygen homeostases, these theories appear to be insufficient. Several lines of evidence indicate that the retina is particularly vulnerable when hypoxia coincides with hyperglycemia. We propose a novel hyperglycemic, hypoxia-inducible factor (HIF) pathway, to complement the current theories regarding hyperglycemic pathogenesis. HIF is a transcription complex that responds to decrease oxygen in the cellular environment. In addition to playing a significant role in the regulation of glucose metabolism, under hyperglycemia HIF has been shown to increase the expression of HIF-inducible genes, such as vascular endothelial growth factor (VEGF) leading to angiogenesis. To this extent, we suggest that HIF can also be described as a hyperglycemia-inducible factor. In summary, while management of dietary GI appears to be an effective intervention for the prevention of metabolic diseases, specifically AMD and DR, more interventional data is needed to evaluate the efficacy of GI management. There is an urgent need to develop reliable biomarkers of exposure, surrogate endpoints, as well as susceptibility for GI. These insights would also be helpful in deciphering the detailed hyperglycemia-related biochemical mechanisms for the development of new therapeutic agents.
Collapse
|
33
|
Anderson J, Sandhir R, Hamilton ES, Berman NEJ. Impaired expression of neuroprotective molecules in the HIF-1alpha pathway following traumatic brain injury in aged mice. J Neurotrauma 2009; 26:1557-66. [PMID: 19203226 DOI: 10.1089/neu.2008.0765] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Elderly traumatic brain injury (TBI) patients have higher rates of mortality and worse functional outcome than non-elderly TBI patients. The mechanisms involved in poor outcomes in the elderly are not well understood. Hypoxia-inducible factor-1 alpha (HIF-1alpha) is a basic helix-loop-helix transcription factor that modulates expression of key genes involved in neuroprotection. In this study, we studied the expression of HIF-1alpha and its target survival genes, heme oxygenase-1 (HO-1), vascular endothelial growth factor (VEGF), and erythropoietin (EPO) in the brains of adult versus aged mice following controlled cortical impact (CCI) injury. Adult (5-6 months) and aged (23-24 months) C57Bl/6 mice were injured using a CCI device. At 72 h post-injury, mice were sacrificed and the injured cortex was used for mRNA and protein analysis using real-time reverse transcription--polymerase chain reaction (RT-PCR) and Western blotting protocols. Following injury, HIF-1alpha, HO-1, and VEGF showed upregulation in both the young and aged mice, but in the aged animals the increase in HIF-1alpha and VEGF in response to injury was much lower than in the adult injured animals. EPO was upregulated in the adult injured brain, but not in the aged injured brain. These results support the hypothesis that reduced expression of genes in the HIF-1alpha neuroprotective pathway in aging may contribute to poor prognosis in the elderly following TBI.
Collapse
Affiliation(s)
- Joshua Anderson
- Steve Palermo Nerve Regeneration Laboratory, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | | | | |
Collapse
|
34
|
Cataldi A, Zingariello M, Rapino M, Zara S, Daniele F, Di Giulio C, Antonucci A. Effect of hypoxia and aging on PKC delta-mediated SC-35 phosphorylation in rat myocardial tissue. Anat Rec (Hoboken) 2009; 292:1135-42. [PMID: 19645017 DOI: 10.1002/ar.20936] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Nuclear speckles, which are sites of pre-mRNA splicing and/or assembly components, are diffusely distributed throughout the nucleoplasm. They are composed of splicing factors (SFs), including SC-35, which are nuclear proteins that remove introns (noncoding sequences in the genes) from precursor mRNA molecules, to form mature RNA, which will be transported to the cytoplasm, site of protein synthesis and activation. In light of such evidences, here we report that hypoxia modulates in vivo SC-35 SF phosphorylation via protein kinase C (PKC) delta in young rat heart. Trichrome Mallory staining and TUNEL analysis along with immunohistochemistry and Western blotting have been performed on left ventricles excised from young and old rats exposed to intermittent hypoxia. Although young hypoxic myocardial cells appear smaller than normoxic cells, connective and endothelial components increase, SC-35 phosphorylation is particularly evident in the endothelium and paralleled by an increased expression of vascular endothelial growth factor (VEGF). In addition, SC-35 and PKC delta coimmunoprecipitation occurs, suggesting that SC-35 phosphorylation could be PKC delta-mediated and that hypoxic young heart needs to counteract the damage through a process of neoangiogenesis involving such SF. Even though the levels of SC-35 and PKC delta are high, the similar response disclosed by normoxic and hypoxic old rat hearts (both showing a fibrotic organization, similar endothelial components and VEGF levels) could be due to the existence of an impaired oxygen sensing mechanism and thus to a low rate of angiogenesis.
Collapse
Affiliation(s)
- Amelia Cataldi
- Cattedra di Anatomia Umana, Facoltà di Farmacia, Università G. d'Annunzio, Chieti-Pescara, Chieti, Italy.
| | | | | | | | | | | | | |
Collapse
|
35
|
Regulatory role of HIF-1alpha in the pathogenesis of age-related macular degeneration (AMD). Ageing Res Rev 2009; 8:349-58. [PMID: 19589398 DOI: 10.1016/j.arr.2009.06.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 06/26/2009] [Accepted: 06/29/2009] [Indexed: 01/10/2023]
Abstract
Age-related macular degeneration (AMD) is a leading cause of irreversible blindness in the elderly throughout the world. AMD is attributed to a complex interaction of genetic and environmental factors. It is characterized by degeneration involving the retinal photoreceptors, retinal pigment epithelium (RPE), and Bruch's membrane, as well as alterations in choroidal capillaries. Aging and age-associated degenerative diseases, such as AMD, are intimately associated with decreased levels of tissue oxygenation and hypoxia that may induce accumulation of detrimental RPE-associated deposits, inflammation and neovascularization processes in retina. Hypoxia-inducible factor (HIF) is the master regulator for hypoxia-induced cellular adaptation that is involved in NF-kappaB signaling and the autophagic protein clearance system. In this review, we discuss role of HIF in AMD pathology and as a possible therapeutic target.
Collapse
|
36
|
Desferroxamine infusion increases cerebral blood flow: a potential association with hypoxia-inducible factor-1. Clin Sci (Lond) 2009; 116:771-9. [PMID: 19014354 DOI: 10.1042/cs20080320] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Finding an effective means to improve cerebral perfusion during hypoxic/ischaemic stress is essential for neuroprotection. Studies in animal models of stroke have shown that desferroxamine activates HIF-1 (hypoxia-inducible factor-1), reduces brain damage and promotes functional recovery. The present study was designed to investigate the effects of desferroxamine infusion on the cerebral circulation in humans. Fifteen volunteers were enrolled in a randomized double-blind placebo-controlled crossover study. We measured cerebral blood flow velocity by transcranial Doppler ultrasonography in the middle cerebral artery, arterial blood pressure, end-tidal CO(2), as well as HIF-1 protein and serum lactate dehydrogenase concentrations in response to 8 h of desferroxamine compared with placebo infusion. Cerebrovascular resistance was calculated from the ratio of steady-state beat-to-beat values for blood pressure to blood flow velocity. We found that desferroxamine infusion was associated with a significant cerebral vasodilation. Moreover, decreased cerebrovascular resistance was temporally correlated with an increased HIF-1 protein concentration as well as HIF-1 transcriptional activation, as measured by serum lactate dehydrogenase concentration. The findings of the present study provide preliminary data suggesting that activators of HIF-1, such as desferroxamine, may protect neurons against ischaemic injury by dilating cerebral vessels and enhancing cerebral perfusion.
Collapse
|
37
|
Kitase Y, Yokozeki M, Fujihara S, Izawa T, Kuroda S, Tanimoto K, Moriyama K, Tanaka E. Analysis of gene expression profiles in human periodontal ligament cells under hypoxia: the protective effect of CC chemokine ligand 2 to oxygen shortage. Arch Oral Biol 2009; 54:618-24. [PMID: 19406381 DOI: 10.1016/j.archoralbio.2009.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 02/13/2009] [Accepted: 03/27/2009] [Indexed: 01/29/2023]
Abstract
Periodontal ligament (PDL) cells appear to play important functional roles in response to mechanical stress. We hypothesized that hypoxia caused by a deformation of blood vessels and the following ischaemia may play a crucial role in differential gene expression in PDL cells affected by mechanical stress. Gene induction in cultured human PDL cells by hypoxia was analyzed using cDNA array, followed by RT-PCR analysis. Eleven hypoxia-responsive genes were found differentially expressed under low-oxygen conditions in PDL cells. Among them, CCR2, CC chemokine ligand 2 (CCL2) receptor was studied in more detail since little information is available on the role of chemokines in adaptive responses of PDL cells under hypoxia. Here we investigate whether CCR2 mediates the signalling to maintain the homeostasis of PDL cells. We found that cell death of PDL cells was induced under hypoxia with down-regulation of CCL2 mRNA expression. However, the exogenous CCL2 prevented PDL cell death under oxygen shortage with the increment of cellular inhibitor of apoptosis (cIAP) mRNA expression. The present study demonstrated substantial effects of hypoxia on gene expression of CCL2 and CCR2 in PDL cells, indicating that mechanical loading accompanied with mild hypoxia allows PDL cells to elicit adaptive responses with up-regulation of CCR2.
Collapse
Affiliation(s)
- Yukiko Kitase
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8504, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Post-translational modification of proteins during intermittent hypoxia. Respir Physiol Neurobiol 2009; 164:272-6. [PMID: 18602876 DOI: 10.1016/j.resp.2008.05.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 05/22/2008] [Accepted: 05/24/2008] [Indexed: 12/14/2022]
Abstract
Post-translational modification (PTM) is one of the mechanisms by which protein function is regulated by chronic hypoxia. This article presents an overview of recent findings on PTM of proteins induced by chronic intermittent hypoxia (CIH) which is experienced by humans with sleep disordered breathing resulting in autonomic abnormalities. The analysis of PTM of proteins involves electrophoretic separation of tissue or cellular proteins followed by immunolabeling using antibodies specific to native and post-translationally modified forms. Recent results demonstrate that CIH, depending on the pattern, duration and severity of hypoxia, alters the state of phosphorylation of a subset of proteins associated with transcriptional factor activation, signaling pathways and neurotransmitter synthesis via activation of appropriate enzymatic machinery that catalyzes specific phosphorylation reactions. Investigation pertaining to PTMs associated with CIH is at its infant stage and future application of high throughput proteomics techniques are necessary to unravel other important PTMs associated with various critical metabolic and signaling pathways that are activated by intermittent hypoxia.
Collapse
|
39
|
Abstract
Chronic intermittent or episodic hypoxia, as occurs during a number of disease states, can have devastating effects, and prolonged exposure to this hypoxia can result in cell injury or cell death. Indeed, intermittent hypoxia activates a number of signaling pathways that are involved in oxygen sensing, oxidative stress, metabolism, catecholamine biosynthesis, and immune responsiveness. The cumulative effect of these processes over time can undermine cell integrity and lead to a decline in function. Furthermore, the ability to respond adequately to various stressors is hampered, and this is traditionally defined as premature aging or senescence. This review highlights recent advances in our understanding of the cellular and molecular mechanisms that are involved in the response to intermittent hypoxia and the potential interplay among various pathways that may accelerate the aging process.
Collapse
Affiliation(s)
- Robert M Douglas
- Department of Pediatrics, University of California San Diego, and Rady Children's Hospital-San Diego, San Diego, La Jolla, CA, USA.
| | | |
Collapse
|
40
|
Anderson J, Sandhir R, Hamilton ES, Berman NE. Impaired Expression of Neuroprotective Molecules in the HIF-1-α Pathway following Traumatic Brain Injury in Aged Mice. J Neurotrauma 2009. [DOI: 10.1089/neu.2008-0765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
41
|
Shi H. Hypoxia inducible factor 1 as a therapeutic target in ischemic stroke. Curr Med Chem 2009; 16:4593-600. [PMID: 19903149 PMCID: PMC2819104 DOI: 10.2174/092986709789760779] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2009] [Accepted: 11/13/2009] [Indexed: 11/22/2022]
Abstract
In stroke research, a significant focus is to develop therapeutic strategies that prevent neuronal death and improve recovery. Yet, few successful therapeutic strategies have emerged. Hypoxia-inducible factor 1 (HIF-1) is a key regulator in hypoxia. It has been suggested to be an important player in neurological outcomes following ischemic stroke due to the functions of its downstream genes. These include genes that promote glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Many lines of evidence have shown that HIF-1 is induced in ischemic brains. Importantly, it seems that HIF-1 is primarily induced in the salvageable tissue of an ischemic brain, penumbra. However, the effect of HIF-1 on neuronal tissue injuries is still debatable based on evidence from in vitro and preclinical studies. Furthermore, it is of importance to understand the mechanism of HIF-1 degradation after its induction in ischemic brain. This review provides a present understanding of the mechanism of HIF-1 induction in ischemic neurons and the potential effect of HIF-1 on ischemic brain tissue. The author also elaborates on potential therapeutic approaches through understanding of the induction mechanism and of the potential role of HIF-1 in ischemic stroke.
Collapse
Affiliation(s)
- Honglian Shi
- Department of Pharmacology and Toxicology, University of Kansas, School of Pharmacy, 1251 Wescoe Hall Drive, Malott Hall 5044, Lawrence, KS 66045, USA.
| |
Collapse
|
42
|
Shatilo VB, Korkushko OV, Ischuk VA, Downey HF, Serebrovskaya TV. Effects of intermittent hypoxia training on exercise performance, hemodynamics, and ventilation in healthy senior men. High Alt Med Biol 2008; 9:43-52. [PMID: 18331220 DOI: 10.1089/ham.2007.1053] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The efficacy and safety of intermittent hypoxia training (IHT) were investigated in healthy, 60- to 74-yr-old men. Fourteen men (Gr 1) who routinely exercised daily for 20 to 30 min were compared with 21 (Gr 2) who avoided exercise. Their submaximal work-load power values before the IHT training were 94 +/- 3.7 and 66 +/- 3.1, respectively. Before and after 10 days of IHT, the ventilatory response to sustained hypoxia (SH; 12% O(2) for 10 min), work capacity (bicycle ergometer), and forearm cutaneous perfusion (laser Doppler) were determined. During SH, no negative electrocardiogram (ECG) changes were observed in either group, and the ventilatory response to SH was unaltered by IHT. In Gr 1, IHT (normobaric rebreathing for 5 min, final Sa(O(2)) = 85% to 86%, followed by 5 min normoxia, 4/day) produced no changes in hemodynamic indixes and work capacity. In Gr 2, IHT decreased blood pressure (BP) by 7.9 +/- 3.1 mmHg (p < 0.05) and increased submaximal work by 11.3% (p < 0.05) and anaerobic threshold by 12.7% (p < 0.05). The increase in HR and BP caused by a 55 W-work load was reduced by 5% and 6.5%, respectively (p < 0.05). Cutaneous perfusion increased by 0.06 +/- 0.04 mL/min/100 g in Gr 1 and by 0.11 +/- 0.04 mL/min/100 g in Gr 2 (p < 0.05). Hyperemia recovery time increased significantly by 15.3 +/- 4.6 sec in Gr 1 and by 25.2 +/- 11.2 sec in Gr 2. Thus, healthy senior men well tolerate IHT as performed in this investigation. In untrained, healthy senior men, IHT had greater positive effects on hemodynamics, microvascular endothelial function, and work capacity.
Collapse
|
43
|
Bielefeldt-Ohmann H, Tolnay AE, Reisenhauer C, Hansen T, Smirnova N, Van Campen H. Transplacental Infection with Non-Cytopathic Bovine Viral Diarrhoea Virus Types 1b and 2: Viral Spread and Molecular Neuropathology. J Comp Pathol 2008; 138:72-85. [DOI: 10.1016/j.jcpa.2007.10.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 10/27/2007] [Indexed: 11/24/2022]
|
44
|
Abstract
Hypoxic acclimatization includes increased brain capillary density. Adaptive angiogenesis, which occurs over a 3-week period, is mediated by upregulation of vascular endothelial growth factor induced by hypoxia-inducible factor-1 in concert with the capillary remodeling molecule angiopoietin-2, which is upregulated through cyclooxygenase-2 production of prostaglandin E2. The process is apparently orchestrated by pericytes, which regulate the microvascular milieu and coordinate the interactions within the neurovascular unit. The return to normoxia is accompanied by microvascular regression and decreasing numbers of capillaries to prehypoxic densities. Regression is the result of endothelial cell apoptosis, suggesting the existence of physiologic mechanisms for adjusting capillary density to balance oxygen availability and oxygen consumption. The capacity for adaptation is diminished in older rats because of the attenuation of the hypoxia-inducible factor-1 response.
Collapse
Affiliation(s)
- Paula Dore-Duffy
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | |
Collapse
|
45
|
Qing M, Görlach A, Schumacher K, Wöltje M, Vazquez-Jimenez JF, Hess J, Seghaye MC. The hypoxia-inducible factor HIF-1 promotes intramyocardial expression of VEGF in infants with congenital cardiac defects. Basic Res Cardiol 2007. [DOI: 10.1007/s00395-007-0654-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
46
|
Qing M, Quing M, Görlach A, Schumacher K, Wöltje M, Vazquez-Jimenez JF, Hess J, Seghaye MC. The hypoxia-inducible factor HIF-1 promotes intramyocardial expression of VEGF in infants with congenital cardiac defects. Basic Res Cardiol 2007; 102:224-32. [PMID: 17268888 DOI: 10.1007/s00395-007-0639-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2006] [Revised: 11/07/2006] [Accepted: 12/06/2006] [Indexed: 10/23/2022]
Abstract
OBJECTIVES The response to hypoxia is primarily mediated by the transcription factor hypoxia-inducible factor-1 (HIF-1) which leads to the induction of a variety of adaptive gene products including vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS). This study was designed to test the hypothesis that HIF-1 and its target genes would be upregulated in the ventricular myocardium of infants with cyanotic congenital cardiac defects. METHODS 14 infants with cyanotic (n = 7) or acyanotic cardiac defects (n = 7) were investigated. Samples from the right ventricular myocardium taken immediately after aortic clamping were studied for protein expression and DNA-binding activity. RESULTS Protein levels of HIF-1alpha were significantly elevated in patients with cyanotic compared to acyanotic congenital heart disease and inversely correlated with the degree of hypoxemia. This response was accompanied by significantly enhanced HIF-1 DNA binding activity. Furthermore, protein levels of VEGF and eNOS were significantly higher in the myocardium of cyanotic than of acyanotic infants. To test the potential involvement of upstream regulatory pathways, activation of MAP kinases was determined. Intramyocardial levels of phosphorylated p38 MAP kinase, but not of ERK1/2 were significantly higher in infants with cyanotic compared to those with acyanotic congenital heart disease and inversely correlated to hypoxemia. CONCLUSIONS These findings show that chronic hypoxemia is associated with the induction and stabilization of the transcription factor HIF-1 as well as its target genes VEGF and eNOS in the myocardium of infants with cyanotic cardiac defects. Thus, stabilization of HIF-1 and induction of the adaptive hypoxia response could particularly participate in myocardial remodeling in children with congenital cardiac defects and chronic hypoxemia.
Collapse
Affiliation(s)
- Ma Qing
- Dept. of Pediatric Cardiology, Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Di Giulio C, Rapino M, Zingariello M, Antonucci A, Cataldi A. PKC alpha-mediated CREB activation is oxygen and age-dependent in rat myocardial tissue. Histochem Cell Biol 2006; 127:327-33. [PMID: 17120015 DOI: 10.1007/s00418-006-0245-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2006] [Indexed: 02/02/2023]
Abstract
Both hypoxia and aging affect the morphology and the function of rat myocardial tissue. Moreover the heart tries to counteract the impaired function by activating specific signalling cascades. Here we report the involvement of CREB protein in "in vivo" response to hypoxic challenge and during aging in rat hearts. CREB is activated in parallel to HIF-1alpha nuclear translocation in the young after hypoxia exposure followed by reoxygenation, while this kind of response is not so dramatic in the old, neither in terms of CREB activation, neither in terms of HIF-1alpha expression and translocation, suggesting in the old the existence of an impaired oxygen-sensing mechanism or an adaptation of the cells to hypoxia. Moreover in the young a PKC alpha/Erk pathway seems to be involved in the activation of HIF-1alpha along with CREB, suggesting an attempt of the young to counteract the damage evoked by hypoxia, while in the old a PKC alpha/p38 MAPK/CREB pathway could determine the occurrence of both aging and aged cell hypoxia response.
Collapse
Affiliation(s)
- Camillo Di Giulio
- Dipartimento di Scienze Mediche di Base ed Applicate, Università G. d'Annunzio, Chieti-Pescara, Chieti, Italy
| | | | | | | | | |
Collapse
|
48
|
Kirkham FJ, Datta AK. Hypoxic adaptation during development: relation to pattern of neurological presentation and cognitive disability. Dev Sci 2006; 9:411-27. [PMID: 16764614 PMCID: PMC1931424 DOI: 10.1111/j.1467-7687.2006.00507.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Children with acute hypoxic-ischaemic events (e.g. stroke) and chronic neurological conditions associated with hypoxia frequently present to paediatric neurologists. Failure to adapt to hypoxia may be a common pathophysiological pathway linking a number of other conditions of childhood with cognitive deficit. There is evidence that congenital cardiac disease, asthma and sleep disordered breathing, for example, are associated with cognitive deficit, but little is known about the mechanism and whether there is any structural change. This review describes what is known about how the brain reacts and adapts to hypoxia, focusing on epilepsy and sickle cell disease (SCD). We prospectively recorded overnight oxyhaemoglobin saturation (SpO2) in 18 children with intractable epilepsy, six of whom were currently or recently in minor status (MS). Children with MS were more likely to have an abnormal sleep study defined as either mean baseline SpO2 <94% or >4 dips of >4% in SpO2/hour (p = .04). In our series of prospectively followed patients with SCD who subsequently developed acute neurological symptoms and signs, mean overnight SpO2 was lower in those with cerebrovascular disease on magnetic resonance angiography (Mann-Whitney, p = .01). Acute, intermittent and chronic hypoxia may have detrimental effects on the brain, the clinical manifestations perhaps depending on rapidity of presentation and prior exposure.
Collapse
Affiliation(s)
- Fenella J Kirkham
- Department of Child Health, Southampton University Hospitals NHS Trust, Southampton, UK.
| | | |
Collapse
|