1
|
Stepien BK, Wielockx B. From Vessels to Neurons-The Role of Hypoxia Pathway Proteins in Embryonic Neurogenesis. Cells 2024; 13:621. [PMID: 38607059 PMCID: PMC11012138 DOI: 10.3390/cells13070621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Embryonic neurogenesis can be defined as a period of prenatal development during which divisions of neural stem and progenitor cells give rise to neurons. In the central nervous system of most mammals, including humans, the majority of neocortical neurogenesis occurs before birth. It is a highly spatiotemporally organized process whose perturbations lead to cortical malformations and dysfunctions underlying neurological and psychiatric pathologies, and in which oxygen availability plays a critical role. In case of deprived oxygen conditions, known as hypoxia, the hypoxia-inducible factor (HIF) signaling pathway is activated, resulting in the selective expression of a group of genes that regulate homeostatic adaptations, including cell differentiation and survival, metabolism and angiogenesis. While a physiological degree of hypoxia is essential for proper brain development, imbalanced oxygen levels can adversely affect this process, as observed in common obstetrical pathologies such as prematurity. This review comprehensively explores and discusses the current body of knowledge regarding the role of hypoxia and the HIF pathway in embryonic neurogenesis of the mammalian cortex. Additionally, it highlights existing gaps in our understanding, presents unanswered questions, and provides avenues for future research.
Collapse
Affiliation(s)
- Barbara K. Stepien
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Experimental Centre, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
2
|
Embryotoxicity of Polystyrene Microspheres of Different Sizes to the Marine Medaka Oryzias melastigma (McClelland, 1839). WATER 2022. [DOI: 10.3390/w14121831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Polystyrene microplastics (PS-MPs) are potentially harmful to marine organisms, especially during the early developmental stages, although the underlying mechanism remains unclear. The present study evaluated the growth and morphological characteristics of marine medaka Oryzias melastigma (McClelland, 1839) embryos exposed to PS-MP. PS-MPs of three different sizes (0.05, 0.5, and 6.0 μm with a concentration of 106 particles/L) were subjected to waterborne exposure for 19 d. The hatching time and rate of embryos exposed to 0.5 and 6.0 μm PS-MPs were significantly lower than those of the control, while no significant difference was observed in the 0.05 μm treatment. No significant differences were observed in the mortality rate of the embryos, embryo diameter, and relevant gene expression levels, including il6, il8, il-1β, jak, stat-3, nf-κb, hif-1α, epo, cyp1a1, ahr, sod, cat, and gpx, but with the exception of vtg. Fluorescent PS-MPs were found on the embryo surfaces when the embryos were exposed to 0.5 and 6.0 μm PS-MPs, but no signals were detected inside embryos using confocal microscopy. Therefore, the results indicate that PS-MPs having a diameter of 6.0 μm can only attach to the surface or villus of embryos and not enter the embryos through the membrane pores, whereas PS-MPs with diameters of 0.05 and 0.5 μm cannot enter the embryos.
Collapse
|
3
|
Shen J, Yang C, Zhang MS, Chin DWC, Chan FF, Law CT, Wang G, Cheng CLH, Chen M, Wan RTC, Wu M, Kuang Z, Sharma R, Lee TKW, Ng IOL, Wong CCL, Wong CM. Histone chaperone FACT complex coordinates with HIF to mediate an expeditious transcription program to adapt to poorly oxygenated cancers. Cell Rep 2022; 38:110304. [PMID: 35108543 DOI: 10.1016/j.celrep.2022.110304] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 11/16/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer cells adapt to hypoxia through HIFs (hypoxia-inducible factors), which initiate the transcription of numerous genes for cancer cell survival in the hypoxia microenvironment. In this study, we find that the FACT (facilitates chromatin transcription) complex works cooperatively with HIFs to facilitate the expeditious expression of HIF targets for hypoxia adaptation. Knockout (KO) of the FACT complex abolishes HIF-mediated transcription by impeding transcription elongation in hypoxic cancer cells. Interestingly, the FACT complex is post-translationally regulated by PHD/VHL-mediated hydroxylation and proteasomal degradation, in similar fashion to HIF-1/2α. Metabolic tracing confirms that FACT KO suppresses glycolytic flux and impairs lactate extrusion, leading to intracellular acidification and apoptosis in cancer cells. Therapeutically, hepatic artery ligation and anti-angiogenic inhibitors adversely induce intratumoral hypoxia, while co-treatment with FACT inhibitor curaxin remarkably hinders the growth of hypoxic tumors. In summary, our findings suggest that the FACT complex is a critical component of hypoxia adaptation and a therapeutic target for hypoxic tumors.
Collapse
Affiliation(s)
- Jialing Shen
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Chunxue Yang
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Misty Shuo Zhang
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Don Wai-Ching Chin
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - For-Fan Chan
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Cheuk-Ting Law
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Gengchao Wang
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Carol Lai-Hung Cheng
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Mengnuo Chen
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Rebecca Ting-Chi Wan
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Mengjie Wu
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Zhijian Kuang
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Rakesh Sharma
- Proteomic and Metabolic Core Facility, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Terence Kin Wah Lee
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Irene Oi-Lin Ng
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Carmen Chak-Lui Wong
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| | - Chun-Ming Wong
- State Key Laboratory of Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
4
|
Wang B, Zeng H, Liu J, Sun M. Effects of Prenatal Hypoxia on Nervous System Development and Related Diseases. Front Neurosci 2021; 15:755554. [PMID: 34759794 PMCID: PMC8573102 DOI: 10.3389/fnins.2021.755554] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
The fetal origins of adult disease (FOAD) hypothesis, which was proposed by David Barker in the United Kingdom in the late 1980s, posited that adult chronic diseases originated from various adverse stimuli in early fetal development. FOAD is associated with a wide range of adult chronic diseases, including cardiovascular disease, cancer, type 2 diabetes and neurological disorders such as schizophrenia, depression, anxiety, and autism. Intrauterine hypoxia/prenatal hypoxia is one of the most common complications of obstetrics and could lead to alterations in brain structure and function; therefore, it is strongly associated with neurological disorders such as cognitive impairment and anxiety. However, how fetal hypoxia results in neurological disorders remains unclear. According to the existing literature, we have summarized the causes of prenatal hypoxia, the effects of prenatal hypoxia on brain development and behavioral phenotypes, and the possible molecular mechanisms.
Collapse
Affiliation(s)
- Bin Wang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongtao Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingliu Liu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
5
|
Robertson CE, Wilsterman K. Developmental and reproductive physiology of small mammals at high altitude: challenges and evolutionary innovations. ACTA ACUST UNITED AC 2020; 223:223/24/jeb215350. [PMID: 33443053 DOI: 10.1242/jeb.215350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High-altitude environments, characterized by low oxygen levels and low ambient temperatures, have been repeatedly colonized by small altricial mammals. These species inhabit mountainous regions year-round, enduring chronic cold and hypoxia. The adaptations that allow small mammals to thrive at altitude have been well studied in non-reproducing adults; however, our knowledge of adaptations specific to earlier life stages and reproductive females is extremely limited. In lowland natives, chronic hypoxia during gestation affects maternal physiology and placental function, ultimately limiting fetal growth. During post-natal development, hypoxia and cold further limit growth both directly by acting on neonatal physiology and indirectly via impacts on maternal milk production and care. Although lowland natives can survive brief sojourns to even extreme high altitude as adults, reproductive success in these environments is very low, and lowland young rarely survive to sexual maturity in chronic cold and hypoxia. Here, we review the limits to maternal and offspring physiology - both pre-natal and post-natal - that highland-adapted species have overcome, with a focus on recent studies on high-altitude populations of the North American deer mouse (Peromyscus maniculatus). We conclude that a combination of maternal and developmental adaptations were likely to have been critical steps in the evolutionary history of high-altitude native mammals.
Collapse
Affiliation(s)
| | - Kathryn Wilsterman
- Division of Biological Sciences, University of Montana, Missoula, MT 59802, USA
| |
Collapse
|
6
|
Richbourg HA, Hu DP, Xu Y, Barczak AJ, Marcucio RS. miR-199 family contributes to regulation of sonic hedgehog expression during craniofacial development. Dev Dyn 2020; 249:1062-1076. [PMID: 32391617 DOI: 10.1002/dvdy.191] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The frontonasal ectodermal zone (FEZ) is a signaling center that regulates patterned development of the upper jaw, and Sonic hedgehog (SHH) mediates FEZ activity. Induction of SHH expression in the FEZ results from SHH-dependent signals from the brain and neural crest cells. Given the role of miRNAs in modulating gene expression, we investigated the extent to which miRNAs regulate SHH expression and FEZ signaling. RESULTS In the FEZ, the miR-199 family appears to be regulated by SHH-dependent signals from the brain; expression of this family increased from HH18 to HH22, and upon activation of SHH signaling in the brain. However, the miR-199 family is more broadly expressed in the mesenchyme of the frontonasal process and adjacent neuroepithelium. Downregulating the miR-199 genes expanded SHH expression in the FEZ, resulting in wider faces, while upregulating miR-199 genes resulted in decreased SHH expression and narrow faces. Hypoxia inducible factor 1 alpha (HIF1A) and mitogen-activated protein kinase kinase kinase 4 (MAP3K4) appear to be potential targets of miR-199b. Reduction of MAP3K4 altered beak development but increased apoptosis, while reducing HIF1A reduced expression of SHH in the FEZ and produced malformations independent of apoptosis. CONCLUSIONS Our results demonstrate that this miRNA family appears to participate in regulating SHH expression in the FEZ; however, specific molecular mechanisms remain unknown.
Collapse
Affiliation(s)
- Heather A Richbourg
- Department of Orthopaedic Surgery, San Francisco General Hospital, University of California, San Francisco, San Francisco, California, USA
| | - Diane P Hu
- Department of Orthopaedic Surgery, San Francisco General Hospital, University of California, San Francisco, San Francisco, California, USA
| | - Yanhua Xu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Zhejiang University Life Sciences Institute, Hangzhou, China
| | - Andrea J Barczak
- Functional Genomics Core, University of California, San Francisco, San Francisco, California, USA
| | - Ralph S Marcucio
- Department of Orthopaedic Surgery, San Francisco General Hospital, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
7
|
Nalivaeva NN, Turner AJ, Zhuravin IA. Role of Prenatal Hypoxia in Brain Development, Cognitive Functions, and Neurodegeneration. Front Neurosci 2018; 12:825. [PMID: 30510498 PMCID: PMC6254649 DOI: 10.3389/fnins.2018.00825] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/22/2018] [Indexed: 12/15/2022] Open
Abstract
This review focuses on the role of prenatal hypoxia in the development of brain functions in the postnatal period and subsequent increased risk of neurodegenerative disorders in later life. Accumulating evidence suggests that prenatal hypoxia in critical periods of brain formation results in significant changes in development of cognitive functions at various stages of postnatal life which correlate with morphological changes in brain structures involved in learning and memory. Prenatal hypoxia also leads to a decrease in brain adaptive potential and plasticity due to the disturbance in the process of formation of new contacts between cells and propagation of neuronal stimuli, especially in the cortex and hippocampus. On the other hand, prenatal hypoxia has a significant impact on expression and processing of a variety of genes involved in normal brain function and their epigenetic regulation. This results in changes in the patterns of mRNA and protein expression and their post-translational modifications, including protein misfolding and clearance. Among proteins affected by prenatal hypoxia are a key enzyme of the cholinergic system-acetylcholinesterase, and the amyloid precursor protein (APP), both of which have important roles in brain function. Disruption of their expression and metabolism caused by prenatal hypoxia can also result, apart from early cognitive dysfunctions, in development of neurodegeneration in later life. Another group of enzymes affected by prenatal hypoxia are peptidases involved in catabolism of neuropeptides, including amyloid-β peptide (Aβ). The decrease in the activity of neprilysin and other amyloid-degrading enzymes observed after prenatal hypoxia could result over the years in an Aβ clearance deficit and accumulation of its toxic species which cause neuronal cell death and development of neurodegeneration. Applying various approaches to restore expression of neuronal genes disrupted by prenatal hypoxia during postnatal development opens an avenue for therapeutic compensation of cognitive dysfunctions and prevention of Aβ accumulation in the aging brain and the model of prenatal hypoxia in rodents can be used as a reliable tool for assessment of their efficacy.
Collapse
Affiliation(s)
- Natalia N. Nalivaeva
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Anthony J. Turner
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Igor A. Zhuravin
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Research Centre, Saint-Petersburg State Pediatric Medical University, St. Petersburg, Russia
| |
Collapse
|
8
|
Efremov YR, Proskurina AS, Potter EA, Dolgova EV, Efremova OV, Taranov OS, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. Cancer Stem Cells: Emergent Nature of Tumor Emergency. Front Genet 2018; 9:544. [PMID: 30505319 PMCID: PMC6250818 DOI: 10.3389/fgene.2018.00544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
A functional analysis of 167 genes overexpressed in Krebs-2 tumor initiating cells was performed. In the first part of the study, the genes were analyzed for their belonging to one or more of the three groups, which represent the three major phenotypic manifestation of malignancy of cancer cells, namely (1) proliferative self-sufficiency, (2) invasive growth and metastasis, and (3) multiple drug resistance. 96 genes out of 167 were identified as possible contributors to at least one of these fundamental properties. It was also found that substantial part of these genes are also known as genes responsible for formation and/or maintenance of the stemness of normal pluri-/multipotent stem cells. These results suggest that the malignancy is simply the ability to maintain the stem cell specific genes expression profile, and, as a consequence, the stemness itself regardless of the controlling effect of stem niches. In the second part of the study, three stress factors combined into the single concept of "generalized cellular stress," which are assumed to activate the expression of these genes, were defined. In addition, possible mechanisms for such activation were identified. The data obtained suggest the existence of a mechanism for the de novo formation of a pluripotent/stem phenotype in the subpopulation of "committed" tumor cells.
Collapse
Affiliation(s)
- Yaroslav R Efremov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Ekaterina A Potter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenia V Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oksana V Efremova
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Oleg S Taranov
- The State Research Center of Virology and Biotechnology Vector, Koltsovo, Russia
| | - Aleksandr A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
9
|
Kurabayashi A, Tanaka C, Matsumoto W, Naganuma S, Furihata M, Inoue K, Kakinuma Y. Murine remote preconditioning increases glucose uptake and suppresses gluconeogenesis in hepatocytes via a brain-liver neurocircuit, leading to counteracting glucose intolerance. Diabetes Res Clin Pract 2018. [PMID: 29526685 DOI: 10.1016/j.diabres.2018.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS Our previous study revealed that cyclic hindlimb ischaemia-reperfusion (IR) activates cardiac acetylcholine (ACh) synthesis through the cholinergic nervous system and cell-derived ACh accelerates glucose uptake. However, the mechanisms regulating glucose metabolism in vivo remain unknown. We investigated the effects and mechanisms of IR in mice under pathophysiological conditions. METHODS Using IR-subjected male C57BL/6J mice, the effects of IR on blood sugar (BS), glucose uptake, central parasympathetic nervous system (PNS) activity, hepatic gluconeogenic enzyme expression and those of ACh on hepatocellular glucose uptake were assessed. RESULTS IR decreased BS levels by 20% and increased c-fos immunoreactivity in the center of the PNS (the solitary tract and the dorsal motor vagal nucleus). IR specifically downregulated hepatic gluconeogenic enzyme expression and activities (glucose-6-phosphatase and phosphoenolpyruvate carboxykinase) and accelerated hepatic glucose uptake. Transection of a hepatic vagus nerve branch decreased this uptake and reversed BS decrease. Suppressed gluconeogenic enzyme expression was reversed by intra-cerebroventricular administration of a choline acetyltransferase inhibitor. Moreover, IR significantly attenuated hyperglycaemia in murine model of type I and II diabetes mellitus. CONCLUSIONS IR provides another insight into a therapeutic modality for diabetes mellitus due to regulating gluconeogenesis and glucose-uptake and advocates an adjunctive mode rectifying disturbed glucose metabolism.
Collapse
Affiliation(s)
| | - Chiharu Tanaka
- Department of Pathology, Kochi Medical School, Kochi 783-8505, Japan
| | - Waka Matsumoto
- Department of Pathology, Kochi Medical School, Kochi 783-8505, Japan
| | - Seiji Naganuma
- Department of Pathology, Kochi Medical School, Kochi 783-8505, Japan
| | - Mutsuo Furihata
- Department of Pathology, Kochi Medical School, Kochi 783-8505, Japan
| | - Keiji Inoue
- Department of Urology, Kochi Medical School, Kochi 783-8505, Japan
| | - Yoshihiko Kakinuma
- Department of Physiology, Nippon Medical School Graduate School of Medicine, Tokyo 113-8602, Japan.
| |
Collapse
|
10
|
Zheng Y, Wang XM. Expression Changes in Lactate and Glucose Metabolism and Associated Transporters in Basal Ganglia following Hypoxic-Ischemic Reperfusion Injury in Piglets. AJNR Am J Neuroradiol 2018; 39:569-576. [PMID: 29326137 DOI: 10.3174/ajnr.a5505] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/30/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND PURPOSE The neonatal brain has active energy metabolism, and glucose oxidation is the major energy source of brain tissue. Lactate is produced by astrocytes and released to neurons. In the central nervous system, lactate is transported between neurons and astrocytes via the astrocyte-neuron lactate shuttle. The aim of this study was to investigate the regulatory mechanisms of energy metabolism in neurons and astrocytes in the basal ganglia of a neonatal hypoxic-ischemic brain injury piglet model. MATERIALS AND METHODS A total of 35 healthy piglets (3-5 days of age; 1.0-1.5 kg) were assigned to a control group (n = 5) or a hypoxic-ischemic model group (n = 30). The hypoxic-ischemic model group was further divided into 6 groups according to the 1H-MR spectroscopy and PET/CT scan times after hypoxia-ischemia (0-2, 2-6, 6-12, 12-24, 24-48, and 48-72 hours; n = 5/group). 1H-MR spectroscopy data were processed with LCModel software. Maximum standard uptake values refer to the maximum standard uptake values for glucose (or FDG). The maximum standard uptake values of the basal ganglia-to-occipital cortex ratio were analyzed. The expression levels of glucose transporters and monocarboxylate transporters were detected by immunohistochemical analysis. RESULTS Lactate levels decreased after an initial increase, with the maximal level occurring around 2-6 hours following hypoxia-ischemia. After hypoxia-ischemia, the maximum standard uptake values of the basal ganglia and basal ganglia/occipital cortex initially increased then decreased, with the maximum occurring at approximately 6-12 hours. The lactate and glucose uptake (basal ganglia/occipital cortex maximum standard uptake values) levels were positively correlated. The expression levels of glucose transporter-1 and glucose transporter-3 were positively correlated with the basal ganglia/occipital cortex. The expression levels of monocarboxylic acid transporter-2 and monocarboxylic acid transporter-4 were positively correlated with lactate content. CONCLUSIONS The results indicate that lactate and glucose transporters have a synergistic effect on the energy metabolism of neurons and astrocytes following hypoxic-ischemic reperfusion brain injury.
Collapse
Affiliation(s)
- Y Zheng
- From the Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - X-M Wang
- From the Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
11
|
Digital gene expression analysis of Takifugu rubripes brain after acute hypoxia exposure using next-generation sequencing. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2017; 24:12-18. [DOI: 10.1016/j.cbd.2017.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/07/2017] [Accepted: 05/27/2017] [Indexed: 01/21/2023]
|
12
|
Sousa LMMDC, Silva RDS, Fonseca VUD, Leandro RM, Di Vincenzo TS, Alves-Wagner AB, Machado UF, Papa PDC. Is the canine corpus luteum an insulin-sensitive tissue? J Endocrinol 2016; 231:223-233. [PMID: 27679426 DOI: 10.1530/joe-16-0173] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 09/27/2016] [Indexed: 01/08/2023]
Abstract
This study aimed to determine in the canine corpus luteum throughout the dioestrus (1) the influence of insulin on glucose uptake; (2) the regulation of genes potentially involved; and (3) the influence of hypoxia on glucose transporter expression and steroidogenesis, after treatment with cobalt chloride (CoCl2). Glucose uptake by luteal cells increased 2.7 folds (P < 0.05) in response to insulin; a phenomenon related to increased expression of glucose transporter (GLUT) 4 and phosphorylation of protein kinase B (AKT). The gene expression of insulin receptor and SLC2A4 (codifier of GLUT4) genes after insulin stimulation increased on day 20 post ovulation (p.o.) and declined on day 40 p.o. (P < 0.05). Regarding potentially involved molecular mechanisms, the nuclear factor kappa B gene RELA was upregulated on days 30/40 p.o., when SLC2A4 mRNA was low, and the interleukin 6 (IL6) gene was upregulated in the first half of dioestrus, when SLC2A4 mRNA was high. CoCl2 in luteal cell cultures increased the hypoxia-inducible factor HIF1A/HIF1A and the SLC2A4/GLUT4 expression, and decreased progesterone (P4) production and hydroxyl-delta-5-steroid dehydrogenase 3 beta (HSD3B) mRNA expression (P < 0.05). This study shows that the canine luteal cells are responsive to insulin, which stimulates glucose uptake in AKT/GLUT4-mediated pathway; that may be related to local activity of RELA and IL6. Besides, the study reveals that luteal cells under hypoxia activate HIF1A-modulating luteal function and insulin-stimulated glucose uptake. These data indicate that insulin regulates luteal cells' glucose disposal, participating in the maintenance and functionality of the corpus luteum.
Collapse
Affiliation(s)
| | - Renata Dos Santos Silva
- Department of SurgerySchool of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Vanessa Uemura da Fonseca
- Department of SurgerySchool of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Rafael Magdanelo Leandro
- Department of SurgerySchool of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Thiago Senna Di Vincenzo
- Department of SurgerySchool of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Bárbara Alves-Wagner
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ubiratan Fabres Machado
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paula de Carvalho Papa
- Department of SurgerySchool of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Varshney P, Dey CS. P21-activated kinase 2 (PAK2) regulates glucose uptake and insulin sensitivity in neuronal cells. Mol Cell Endocrinol 2016; 429:50-61. [PMID: 27040307 DOI: 10.1016/j.mce.2016.03.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 12/15/2022]
Abstract
P21-activated kinases (PAKs) are recently reported as important players of insulin signaling and glucose homeostasis in tissues like muscle, pancreas and liver. However, their role in neuronal insulin signaling is still unknown. Present study reports the involvement of PAK2 in neuronal insulin signaling, glucose uptake and insulin resistance. Irrespective of insulin sensitivity, insulin stimulation decreased PAK2 activity. PAK2 downregulation displayed marked enhancement of GLUT4 translocation with increase in glucose uptake whereas PAK2 over-expression showed its reduction. Treatment with Akti-1/2 and wortmannin suggested that Akt and PI3K are mediators of insulin effect on PAK2 and glucose uptake. Rac1 inhibition demonstrated decreased PAK2 activity while inhibition of PP2A resulted in increased PAK2 activity, with corresponding changes in glucose uptake. Taken together, present study demonstrates an inhibitory role of insulin signaling (via PI3K-Akt) and PP2A on PAK2 activity and establishes PAK2 as a Rac1-dependent negative regulator of neuronal glucose uptake and insulin sensitivity.
Collapse
Affiliation(s)
- Pallavi Varshney
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India.
| |
Collapse
|
14
|
Hypoxic Adaptation in the Nervous System: Promise for Novel Therapeutics for Acute and Chronic Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 903:221-43. [PMID: 27343100 DOI: 10.1007/978-1-4899-7678-9_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Homeostasis is the process by which cells adapt to stress and prevent or repair injury. Unique programs have evolved to sense and activate these homeostatic mechanisms and as such, homeostatic sensors may be potent therapeutic targets. The hypoxic response mediated by hypoxia inducible factor (HIF) downstream of oxygen sensing by HIF prolyl 4-hydroxylases (PHDs) has been well-studied, revealing cell-type specific regulation of HIF stability, activity, and transcriptional targets. HIF's paradoxical roles in nervous system development, physiology, and pathology arise from its complex roles in hypoxic adaptation and normoxic biology. Understanding how to engage the hypoxic response so as to recapitulate the protective mechanism of ischemic preconditioning is a high priority. Indeed, small molecules that activate the hypoxic response provide broad neuroprotection in several clinically relevant injury models. Screens for PHD inhibitors have identified novel therapeutics for neuroprotection that are ready to proceed to clinical trials for ischemic stroke. Better understanding the mechanisms of how to engage hypoxic adaption without altering development or physiology may identify additional novel therapeutic targets for diverse acute and chronic neuropathologies.
Collapse
|
15
|
Stroev SA, Tyul’kova EI, Vataeva LA, Miettinen MT. Modifications of the expression of thioredoxins and superoxide dismutases in the rat hippocampus that were induced by prenatal hypoxia are preserved in mature animals. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415030101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Docanto MM, Ham S, Corbould A, Brown KA. Obesity-Associated Inflammatory Cytokines and Prostaglandin E2 Stimulate Glucose Transporter mRNA Expression and Glucose Uptake in Primary Human Adipose Stromal Cells. J Interferon Cytokine Res 2015; 35:600-5. [PMID: 25839190 DOI: 10.1089/jir.2014.0194] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Obesity is associated with chronic low-grade inflammation. This occurs largely as a result of the infiltration of immune cells within the obese adipose, which produce a number of inflammatory factors, including interleukin-6 (IL-6), IL-1β, tumor necrosis factor-α (TNFα), and prostaglandin E(2) (PGE(2)). These factors have previously been shown to affect insulin-mediated glucose uptake in differentiated adipocytes. However, the insulin-independent effect of inflammation on adipocyte precursors, the adipose stromal cells, has not been explored. This study therefore aimed to examine the effect of obesity-associated inflammatory factors on the expression of insulin-independent glucose transporters (GLUT1 and GLUT3) and on the uptake of glucose within adipose stromal cells. Primary human subcutaneous adipose stromal cells were isolated from abdominoplasty, and the effect of inflammatory cytokines (IL-6, IL-1β, and TNFα) and PGE(2) on GLUT mRNA expression and glucose transport was assessed using real-time polymerase chain reaction and radiolabeled deoxyglucose uptake assays, respectively. Results demonstrate that all four inflammatory mediators caused a dose-dependent increase in GLUT1 mRNA expression and glucose uptake. GLUT3 mRNA expression was also upregulated by IL-6 (0.5 ng/mL), TNFα (0.1 and 10 ng/mL), and PGE(2) (0.1 μM). Overall, these results demonstrate that obesity-associated inflammation increases insulin-independent glucose transporter expression and glucose uptake in undifferentiated adipose stromal cells.
Collapse
Affiliation(s)
- Maria M Docanto
- 1 Metabolism & Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research , Victoria, Australia
| | - Seungmin Ham
- 1 Metabolism & Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research , Victoria, Australia
| | - Anne Corbould
- 1 Metabolism & Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research , Victoria, Australia .,2 Department of Physiology, Monash University , Clayton, Victoria, Australia
| | - Kristy A Brown
- 1 Metabolism & Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research , Victoria, Australia .,2 Department of Physiology, Monash University , Clayton, Victoria, Australia .,3 Department of Molecular and Translational Sciences, Monash University , Clayton, Victoria, Australia
| |
Collapse
|
17
|
Moraes PA, Yonamine CY, Pinto Junior DC, Esteves JVD, Machado UF, Mori RC. Insulin acutely triggers transcription of Slc2a4 gene: participation of the AT-rich, E-box and NFKB-binding sites. Life Sci 2014; 114:36-44. [PMID: 25123536 DOI: 10.1016/j.lfs.2014.07.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 11/16/2022]
Abstract
AIMS The insulin-sensitive glucose transporter protein GLUT4 (solute carrier family 2 member 4 (Slc2a4) gene) plays a key role in glycemic homeostasis. Decreased GLUT4 expression is a current feature in insulin resistant conditions such as diabetes, and the restoration of GLUT4 content improves glycemic control. This study investigated the effect of insulin upon Slc2a4/GLUT4 expression, focusing on the AT-rich element, E-box and nuclear factor NF-kappa-B (NFKB) site. MAIN METHODS Rat soleus muscles were incubated during 180 min with insulin, added or not with wortmannin (phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit gamma isoform (PI3K)-inhibitor), ML9 (serine/threonine protein kinase (AKT) inhibitor) and tumor necrosis factor (TNF, GLUT4 repressor), and processed for analysis of GLUT4 protein (Western blotting); Slc2a4, myocyte enhancer factor 2a/d (Mef2a/d), hypoxia inducible factor 1a (Hif1a), myogenic differentiation 1 (Myod1) and nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (Nfkb1) messenger ribonucleic acids (mRNAs) (polymerase chain reaction (PCR)); and AT-rich- (myocyte-specific enhancer factor 2 (MEF2)-binding site), E-box- (hypoxia inducible factor 1 alpha (HIF1A)- and myoblast determination protein 1 (MYOD1)-binding site), and NFKB-binding activity (electrophoretic mobility assay). KEY FINDINGS Insulin increased Slc2a4 mRNA expression (140%) and nuclear proteins binding to AT-rich and E-box elements (~90%), all effects were prevented by wortmannin and ML9. Insulin also increased Mef2a/d and Myod1 mRNA expression, suggesting the participation of these transcriptional factors in the Slc2a4 enhancing effect. Conversely, insulin decreased Nfkb1 mRNA expression and protein binding to the NFKB-site (~50%). Furthermore, TNF-induced inhibition of GLUT4 expression (~40%) was prevented by insulin in an NFKB-binding repressing mechanism. GLUT4 protein paralleled the Slc2a4 mRNA regulations. SIGNIFICANCE Insulin enhances the Slc2a4/GLUT4 expression in the skeletal muscle by activating AT-rich and E-box elements, in a PI3K/AKT-dependent mechanism, and repressing NFKB-site activity as well. These results unravel how post-prandial increase of insulin may guarantee GLUT4 expression, and how the insulin signaling impairment can participate in insulin resistance-induced repression of GLUT4.
Collapse
Affiliation(s)
- Paulo Alexandre Moraes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Caio Yogi Yonamine
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Danilo Correa Pinto Junior
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - João Victor DelConti Esteves
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Ubiratan Fabres Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Rosana Cristina Mori
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
18
|
Treatment with thyroxine restores myelination and clinical recovery after intraventricular hemorrhage. J Neurosci 2013; 33:17232-46. [PMID: 24174657 DOI: 10.1523/jneurosci.2713-13.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Intraventricular hemorrhage (IVH) remains a major cause of white matter injury in preterm infants with no viable therapeutic strategy to restore myelination. Maturation of oligodendrocytes and myelination is influenced by thyroid hormone (TH) signaling, which is mediated by TH receptor α (TRα) and TRβ. In the brain, cellular levels of TH are regulated by deiodinases, with deiodinase-2 mediating TH activation and deiodinase-3 TH inactivation. Therefore, we hypothesized that IVH would decrease TH signaling via changes in the expression of deiodinases and/or TRs, and normalization of TH signaling would enhance maturation of oligodendrocytes and myelination in preterm infants with IVH. These hypotheses were tested using both autopsy materials from human preterm infants and a rabbit model of IVH. We found that deiodinase-2 levels were reduced, whereas deiodinase-3 levels were increased in brain samples of both humans and rabbits with IVH compared with controls without IVH. TRα expression was also increased in human infants with IVH. Importantly, treatment with TH accelerated the proliferation and maturation of oligodendrocytes, increased transcription of Olig2 and Sox10 genes, augmented myelination, and restored neurological function in pups with IVH. Consistent with these findings, the density of myelinating oligodendrocytes was almost doubled in TH-treated human preterm infants compared with controls. Thus, in infants with IVH the combined elevation in deiodinase-3 and reduction in deiodinase-2 decreases TH signaling that can be worsened by an increase in unliganded TRα. Given that TH promotes neurological recovery in IVH, TH treatment might improve the neurodevelopmental outcome of preterm infants with IVH.
Collapse
|
19
|
Mechanisms regulating GLUT4 transcription in skeletal muscle cells are highly conserved across vertebrates. PLoS One 2013; 8:e80628. [PMID: 24260440 PMCID: PMC3832493 DOI: 10.1371/journal.pone.0080628] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/03/2013] [Indexed: 12/30/2022] Open
Abstract
The glucose transporter 4 (GLUT4) plays a key role in glucose uptake in insulin target tissues. This transporter has been extensively studied in many species in terms of its function, expression and cellular traffic and complex mechanisms are involved in its regulation at many different levels. However, studies investigating the transcription of the GLUT4 gene and its regulation are scarce. In this study, we have identified the GLUT4 gene in a teleost fish, the Fugu (Takifugu rubripes), and have cloned and characterized a functional promoter of this gene for the first time in a non-mammalian vertebrate. In silico analysis of the Fugu GLUT4 promoter identified potential binding sites for transcription factors such as SP1, C/EBP, MEF2, KLF, SREBP-1c and GC-boxes, as well as a CpG island, but failed to identify a TATA box. In vitro analysis revealed three transcription start sites, with the main residing 307 bp upstream of the ATG codon. Deletion analysis determined that the core promoter was located between nucleotides -132/+94. By transfecting a variety of 5´deletion constructs into L6 muscle cells we have determined that Fugu GLUT4 promoter transcription is regulated by insulin, PG-J2, a PPARγ agonist, and electrical pulse stimulation. Furthermore, our results suggest the implication of motifs such as PPARγ/RXR and HIF-1α in the regulation of Fugu GLUT4 promoter activity by PPARγ and contractile activity, respectively. These data suggest that the characteristics and regulation of the GLUT4 promoter have been remarkably conserved during the evolution from fish to mammals, further evidencing the important role of GLUT4 in metabolic regulation in vertebrates.
Collapse
|
20
|
Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3481-3498. [PMID: 23830918 DOI: 10.1016/j.bbamcr.2013.06.026] [Citation(s) in RCA: 760] [Impact Index Per Article: 69.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/21/2013] [Accepted: 06/22/2013] [Indexed: 02/07/2023]
Abstract
Anoikis is a programmed cell death induced upon cell detachment from extracellular matrix, behaving as a critical mechanism in preventing adherent-independent cell growth and attachment to an inappropriate matrix, thus avoiding colonizing of distant organs. As anchorage-independent growth and epithelial-mesenchymal transition, two features associated with anoikis resistance, are vital steps during cancer progression and metastatic colonization, the ability of cancer cells to resist anoikis has now attracted main attention from the scientific community. Cancer cells develop anoikis resistance due to several mechanisms, including change in integrins' repertoire allowing them to grow in different niches, activation of a plethora of inside-out pro-survival signals as over-activation of receptors due to sustained autocrine loops, oncogene activation, growth factor receptor overexpression, or mutation/upregulation of key enzymes involved in integrin or growth factor receptor signaling. In addition, tumor microenvironment has also been acknowledged to contribute to anoikis resistance of bystander cancer cells, by modulating matrix stiffness, enhancing oxidative stress, producing pro-survival soluble factors, triggering epithelial-mesenchymal transition and self-renewal ability, as well as leading to metabolic deregulations of cancer cells. All these events help cancer cells to inhibit the apoptosis machinery and sustain pro-survival signals after detachment, counteracting anoikis and constituting promising targets for anti-metastatic pharmacological therapy. This article is part of a Special Section entitled: Cell Death Pathways.
Collapse
Affiliation(s)
- Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; Tuscany Tumor Institute and "Center for Research, Transfer and High Education, DENOTHE", 50134 Florence, Italy.
| |
Collapse
|
21
|
Kakinuma Y, Tsuda M, Okazaki K, Akiyama T, Arikawa M, Noguchi T, Sato T. Heart-specific overexpression of choline acetyltransferase gene protects murine heart against ischemia through hypoxia-inducible factor-1α-related defense mechanisms. J Am Heart Assoc 2013; 2:e004887. [PMID: 23525439 PMCID: PMC3603257 DOI: 10.1161/jaha.112.004887] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background Murine and human ventricular cardiomyocytes rich in acetylcholine (Ach) receptors are poorly innervated by the vagus, compared with whole ventricular innervation by the adrenergic nerve. However, vagal nerve stimulation produces a favorable outcome even in the murine heart, despite relatively low ventricular cholinergic nerve density. Such a mismatch and missing link suggest the existence of a nonneuronal cholinergic system in ventricular myocardium. Methods and Results To examine the role of the nonneuronal cardiac cholinergic system, we generated choline acetyltransferase (ChAT)–expressing cells and heart‐specific ChAT transgenic (ChAT‐tg) mice. Compared with cardiomyocytes of wild‐type (WT) mice, those of the ChAT‐tg mice had high levels of ACh and hypoxia‐inducible factor (HIF)‐1α protein and augmented glucose uptake. These phenotypes were also reproduced by ChAT‐overexpressing cells, which utilized oxygen less. Before myocardial infarction (MI), the WT and ChAT‐tg mice showed similar hemodynamics; after MI, however, the ChAT‐tg mice had better survival than did the WT mice. In the ChAT‐tg hearts, accelerated angiogenesis at the ischemic area, and accentuated glucose utilization prevented post‐MI remodeling. The ChAT‐tg heart was more resistant to ischemia–reperfusion injury than was the WT heart. Conclusions These results suggest that the activated cardiac ACh‐HIF‐1α cascade improves survival after MI. We conclude that de novo synthesis of ACh in cardiomyocytes is a pivotal mechanism for self‐defense against ischemia.
Collapse
Affiliation(s)
- Yoshihiko Kakinuma
- Department of Cardiovascular Control, Kochi Medical School, Nankoku, Japan.
| | | | | | | | | | | | | |
Collapse
|
22
|
Mattmiller SA, Corl CM, Gandy JC, Loor JJ, Sordillo LM. Glucose transporter and hypoxia-associated gene expression in the mammary gland of transition dairy cattle. J Dairy Sci 2011; 94:2912-22. [PMID: 21605761 DOI: 10.3168/jds.2010-3936] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 02/02/2011] [Indexed: 11/19/2022]
Abstract
Glucose is an important energy substrate, especially needed by dairy cows postpartum to support the onset of lactation. The prioritization and regulation of glucose uptake is accomplished, in part, by changes in expression of cellular glucose transport molecules (GLUT) within the mammary gland. The objectives of this study were to (1) evaluate the expression and cell-type specific localization of GLUT and hypoxia-associated genes that may regulate GLUT expression over the transition period and through lactation in bovine mammary tissue and (2) determine functionality of GLUT on primary bovine mammary endothelial cells (BMEC). Mammary tissue biopsies were taken from cows at 15 d before calving and again at 1, 15, 30, 60, 120, and 240 d post-parturition for quantitative real-time PCR analysis of GLUT and hypoxia-associated genes. Additional mammary tissue samples were used to localize GLUT within the cells of the lobulo-alveolar system via fluorescence microscopy. Cultures of primary bovine mammary endothelial cells were used to confirm the functionality of GLUT with a fluorescent glucose analog uptake assay. Significant increases in GLUT1 gene expression were observed during early lactation, whereas both GLUT3 and GLUT4 gene expression increased during late lactation. The gene expression for 2 receptors of vascular endothelial growth factor increased significantly during early lactation and remained increased throughout lactation when compared with gene expression during the transition period. All GLUT were detected on cultured BMEC and were capable of internalizing glucose through GLUT-mediated mechanisms. These data suggest mammary vascular tissues express GLUT during lactation and BMEC express functional glucose transporters. A better understanding of glucose uptake at the endothelial level may prove to be critical to improve glucose absorption from the blood for utilization by mammary epithelial cells.
Collapse
Affiliation(s)
- S A Mattmiller
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing 48824, USA
| | | | | | | | | |
Collapse
|
23
|
Stroev SA, Tyul’kova EI, Vataeva LA, Samoilov MO, Pelto-Huikko MT. Effects of prenatal hypoxia on expression of thioredoxin-1 in the rat hippocampus at different stages of postnatal ontogeny. NEUROCHEM J+ 2011. [DOI: 10.1134/s1819712411030111] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Gillies RM, Robinson SP, McPhail LD, Carter ND, Murray JF. Immunohistochemical assessment of intrinsic and extrinsic markers of hypoxia in reproductive tissue: differential expression of HIF1α and HIF2α in rat oviduct and endometrium. J Mol Histol 2011; 42:341-54. [PMID: 21732047 PMCID: PMC3136703 DOI: 10.1007/s10735-011-9338-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 06/19/2011] [Indexed: 12/11/2022]
Abstract
Hypoxia is thought to be critical in regulating physiological processes within the female reproductive system, including ovulation, composition of the fluid in the oviductal/uterine lumens and ovarian follicle development. This study examined the localisation of exogenous (pimonidazole) and endogenous [hypoxia inducible factor 1α and 2α (HIF1α, -2α), glucose transporter type 1 (GLUT1) and carbonic anhydrase 9 (CAIX)] hypoxia-related antigens within the oviduct and uterus of the rat reproductive tract. The extent to which each endogenous antigen co-compartmentalised with pimonidazole was also assessed. Female Wistar Furth rats (n = 10) were injected intraperitoneally with pimonidazole (60 mg/kg) 1 h prior to death. Reproductive tissues were removed immediately following death and fixed in 4% paraformaldehyde before being embedded in paraffin. Serial sections were cut (6-7 μm thick) and antigens of interest identified using standard immunohistochemical procedures. The mucosal epithelia of the ampulla, isthmus and uterus were immunopositive for pimonidazole in most sections. Co-compartmentalisation of pimonidazole with HIF1α was only expressed in the mucosa of the uterus whilst co-compartmentalisation with HIF2α was observed in the mucosa of the ampulla, isthmus and uterus. Both GLUT1 and CAIX were co-compartmentalised with pimonidazole in mucosa of the isthmus and uterus. This study confirms that mucosal regions of the rat oviduct and uterus frequently experience severe hypoxia and there are compartment specific variations in expression of endogenous hypoxia-related antigens, including the HIF isoforms. The latter observation may relate to target gene specificity of HIF isoforms or perhaps HIF2α's responsiveness to non-hypoxic stimuli such as hypoglycaemia independently of HIF1α.
Collapse
Affiliation(s)
- Robert M Gillies
- School of Life Sciences, University of Westminster, 115 New Cavendish St., London, UK
| | | | | | | | | |
Collapse
|
25
|
Gómez-Ruiz A, Milagro FI, Campión J, Martínez JA, de Miguel C. High-fat diet feeding alters metabolic response to fasting/non fasting conditions. Effect on caveolin expression and insulin signalling. Lipids Health Dis 2011; 10:55. [PMID: 21489269 PMCID: PMC3083369 DOI: 10.1186/1476-511x-10-55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 04/13/2011] [Indexed: 12/14/2022] Open
Abstract
Background The effect of food intake on caveolin expression in relation to insulin signalling was studied in skeletal muscle and adipocytes from retroperitoneal (RP) and subcutaneous (SC) adipose tissue, comparing fasted (F) to not fasted (NF) rats that had been fed a control or high-fat (HF) diet for 72 days. Methods Serum glucose was analysed enzymatically and insulin and leptin by ELISA. Caveolins and insulin signalling intermediaries (IR, IRS-1 and 2 and GLUT4) were determined by RT-PCR and western blotting. Caveolin and IR phosphorylation was measured by immunoprecipitation. Data were analysed with Mann-Whitney U test. Results High-fat fed animals showed metabolic alterations and developed obesity and insulin resistance. In skeletal muscle, food intake (NF) induced activation of IR and increased expression of IRS-2 in control animals with normal metabolic response. HF animals became overweight, hyperglycaemic, hyperinsulinemic, hyperleptinemic and showed insulin resistance. In skeletal muscle of these animals, food intake (NF) also induced IRS-2 expression together with IR, although this was not active. Caveolin 3 expression in this tissue was increased by food intake (NF) in animals fed either diet. In RP adipocytes of control animals, food intake (NF) decreased IR and IRS-2 expression but increased that of GLUT4. A similar but less intense response was found in SC adipocytes. Food intake (NF) did not change caveolin expression in RP adipocytes with either diet, but in SC adipocytes of HF animals a reduction was observed. Food intake (NF) decreased caveolin-1 phosphorylation in RP but increased it in SC adipocytes of control animals, whereas it increased caveolin-2 phosphorylation in both types of adipocytes independently of the diet. Conclusions Animals fed a control-diet show a normal response to food intake (NF), with activation of the insulin signalling pathway but without appreciable changes in caveolin expression, except a small increase of caveolin-3 in muscle. Animals fed a high-fat diet develop metabolic changes that result in insulin signalling impairment. In these animals, caveolin expression in muscle and adipocytes seems to be regulated independently of insulin signalling.
Collapse
Affiliation(s)
- Ana Gómez-Ruiz
- Department of Biochemistry and Molecular Biology, University of Navarra, Pamplona, Spain
| | | | | | | | | |
Collapse
|
26
|
Kumar P, Taha A, Kale RK, Cowsik SM, Baquer NZ. Physiological and biochemical effects of 17β estradiol in aging female rat brain. Exp Gerontol 2011; 46:597-605. [PMID: 21377519 DOI: 10.1016/j.exger.2011.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2010] [Revised: 01/13/2011] [Accepted: 02/22/2011] [Indexed: 01/10/2023]
Abstract
Aging in females and males is considered as the end of natural protection against age related diseases like osteoporosis, coronary heart disease, diabetes, Alzheimer's disease and Parkinson's disease. These changes increase during menopausal condition in females when the level of estradiol is decreased. The objective of this study was to observe the changes in activities of monoamine oxidase, glucose transporter-4 levels, membrane fluidity, lipid peroxidation levels and lipofuscin accumulation occurring in brains of female rats of 3 months (young), 12 months (adult) and 24 months (old) age groups, and to see whether these changes are restored to normal levels after exogenous administration of estradiol (0.1 μg/g body weight for 1 month). The results obtained in the present work revealed that normal aging was associated with significant increases in the activity of monoamine oxidase, lipid peroxidation levels and lipofuscin accumulation in the brains of aging female rats, and a decrease in glucose transporter-4 level and membrane fluidity. Our data showed that estradiol treatment significantly decreased monoamine oxidase activity, lipid peroxidation and lipofuscin accumulation in brain regions of aging rats, and a reversal of glucose transporter-4 levels and membrane fluidity was achieved, therefore it can be concluded from the present findings that estradiol's beneficial effects seemed to arise from its antilipofuscin, antioxidant and antilipidperoxidative effects, implying an overall anti-aging action. The results of this study will be useful for pharmacological modification of the aging process and applying new strategies for control of age related disorders.
Collapse
Affiliation(s)
- Pardeep Kumar
- School of Life Sciences, Jawaharlal Nehru University, 110067, New Delhi, India
| | | | | | | | | |
Collapse
|
27
|
Osada-Oka M, Hashiba Y, Akiba S, Imaoka S, Sato T. Glucose is necessary for stabilization of hypoxia-inducible factor-1α under hypoxia: Contribution of the pentose phosphate pathway to this stabilization. FEBS Lett 2010; 584:3073-9. [DOI: 10.1016/j.febslet.2010.05.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 04/10/2010] [Accepted: 05/22/2010] [Indexed: 01/22/2023]
|
28
|
Gómez-Ruiz A, Milagro FI, Campión J, Martínez JA, de Miguel C. Caveolin expression and activation in retroperitoneal and subcutaneous adipocytes: Influence of a high-fat diet. J Cell Physiol 2010; 225:206-13. [DOI: 10.1002/jcp.22241] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
29
|
Tong W, Chen W, Ostrowski RP, Ma Q, Souvenir R, Zhang L, Zhang JH, Tang J. Maternal hypoxia increases the activity of MMPs and decreases the expression of TIMPs in the brain of neonatal rats. Dev Neurobiol 2010; 70:182-94. [PMID: 20017119 DOI: 10.1002/dneu.20770] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A recent study has shown that increased activity of matrix metalloproteinases-2 and metalloproteinases-9 (MMP-2 and MMP-9) has detrimental effect on the brain after neonatal hypoxia. The present study determined the effect of maternal hypoxia on neuronal survivability and the activity of MMP-2 and MMP-9, as well as the expression of tissue inhibitors of metalloproteinase 1 and 2 (TIMP-1 and TIMP-2) in the brain of neonatal rats. Pregnant rats were exposed to 10.5% oxygen for 6 days from the gestation day 15 to day 21. Pups were sacrificed at day 0, 4, 7, 14, and 21 after birth. Body weight and brain weight of the pups were measured at each time point. The activity of MMP-2 and MMP-9 and the protein abundance of TIMP-1 and TIMP-2 were determined by zymography and Western blotting, respectively. The tissue distribution of MMPs was examined by immunofluorescence staining. The neuronal death was detected by Nissl staining. Maternal hypoxia caused significant decreases in body and brain size, increased activity of MMP-2 at day 0, and increased MMP-9 at day 0 and 4. The increased activity of the MMPs was accompanied by an overall tendency towards a reduced expression of TIMPs at all ages with the significance observed for TIMPs at day 0, 4, and 7. Immunofluorescence analysis showed an increased expression of MMP-2, MMP-9 in the hippocampus at day 0 and 4. Nissl staining revealed significant cell death in the hippocampus at day 0, 4, and 7. Functional tests showed worse neurobehavioral outcomes in the hypoxic animals.
Collapse
Affiliation(s)
- Wenni Tong
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California 92350, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Lima GA, Anhê GF, Giannocco G, Nunes MT, Correa-Giannella ML, Machado UF. Contractile activity per se induces transcriptional activation of SLC2A4 gene in soleus muscle: involvement of MEF2D, HIF-1a, and TRalpha transcriptional factors. Am J Physiol Endocrinol Metab 2009; 296:E132-8. [PMID: 18957617 DOI: 10.1152/ajpendo.90548.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Skeletal muscle is a target tissue for approaches that can improve insulin sensitivity in insulin-resistant states. In muscles, glucose uptake is performed by the GLUT-4 protein, which is encoded by the SLC2A4 gene. SLC2A4 gene expression increases in response to conditions that improve insulin sensitivity, including chronic exercise. However, since chronic exercise improves insulin sensitivity, the increased SLC2A4 gene expression could not be clearly attributed to the muscle contractile activity per se and/or to the improved insulin sensitivity. The present study was designed to investigate the role of contractile activity per se in the regulation of SLC2A4 gene expression as well as in the participation of the transcriptional factors myocyte enhancer factor 2D (MEF2D), hypoxia inducible factor 1a (HIF-1a), and thyroid hormone receptor-alpha (TRalpha). The performed in vitro protocol excluded the interference of metabolic, hormonal, and neural effects. The results showed that, in response to 10 min of electrically induced contraction of soleus muscle, an early 40% increase in GLUT-4 mRNA (30 min) occurred, with a subsequent 65% increase (120 min) in GLUT-4 protein content. EMSA and supershift assays revealed that the stimulus rapidly increased the binding activity of MEF2D, HIF-1a, and TRalpha into the SLC2A4 gene promoter. Furthermore, chromatin immunoprecipitation assay confirmed, in native nucleosome, that contraction induced an approximate fourfold (P < 0.01) increase in MEF2D and HIF-1a-binding activity. In conclusion, muscle contraction per se enhances SLC2A4 gene expression and that involves MEF2D, HIF-1a, and TRalpha transcription factor activation. This finding reinforces the importance of physical activity to improve glycemic homeostasis independently of other additional insulin sensitizer approaches.
Collapse
Affiliation(s)
- Guilherme Alves Lima
- Dept. of Physiology and Biophysics, Institute of Biomedical Sciences, Univ. of Sao Paulo, Av. Prof. Lineu Prestes, 1524, 05508-900 Sao Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
31
|
Chen W, Ostrowski RP, Obenaus A, Zhang JH. Prodeath or prosurvival: two facets of hypoxia inducible factor-1 in perinatal brain injury. Exp Neurol 2008; 216:7-15. [PMID: 19041643 DOI: 10.1016/j.expneurol.2008.10.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 09/12/2008] [Accepted: 10/24/2008] [Indexed: 01/13/2023]
Abstract
Hypoxia, which occurs in the brain when oxygen availability drops below the normal level, is a major cause of perinatal hypoxic-ischemic injury (HII). The transcriptional factor hypoxia inducible factor-1 (HIF-1) is a key regulator in the pathophysiological response to the stress of hypoxia. Genes regulated by HIF-1 are involved in energy metabolism, erythropoiesis, angiogenesis, vasodilatation, cell survival and apoptosis. Compared with the adult brain, the neonatal brain is different in physiological structure, function, cellular composition and signaling pathway related gene activation and response after hypoxia. The purpose of this review is to determine if developmental susceptibility of the brain after hypoxic/ischemic injury is related to HIF-1alpha, which also plays a pivotal role in the normal brain development. HIF-1alpha regulates both prosurvival and prodeath responses in the neonatal brain and various mechanisms underlie the apparent contradictory effects, including duration of ischemic injury and severity, cell-types, and/or dependent on the nature of the stimulus after HII. Studies report an excessive induction of HIF-1 in the immature brain, which suggests that a cell death promoting role of HIF may prevail. Inhibition of HIF-1alpha and targeted activation of its prosurvival genes appear as a favorable therapeutic strategy. However, a better understanding of multifaceted HIF-1 function during brain development is required to explore potential targets for further therapeutic interventions in the neonate.
Collapse
Affiliation(s)
- Wanqiu Chen
- Department of Physiology, Loma Linda University, Loma Linda, CA 92354, USA
| | | | | | | |
Collapse
|
32
|
Rintala E, Wiebe MG, Tamminen A, Ruohonen L, Penttilä M. Transcription of hexose transporters of Saccharomyces cerevisiae is affected by change in oxygen provision. BMC Microbiol 2008; 8:53. [PMID: 18373847 PMCID: PMC2324102 DOI: 10.1186/1471-2180-8-53] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 03/28/2008] [Indexed: 12/03/2022] Open
Abstract
Background The gene family of hexose transporters in Saccharomyces cerevisiae consists of 20 members; 18 genes encoding transporters (HXT1-HXT17, GAL2) and two genes encoding sensors (SNF3, RGT2). The effect of oxygen provision on the expression of these genes was studied in glucose-limited chemostat cultivations (D = 0.10 h-1, pH 5, 30°C). Transcript levels were measured from cells grown in five steady state oxygen levels (0, 0.5, 1, 2.8 and 20.9% O2), and from cells under conditions in which oxygen was introduced to anaerobic cultures or removed from cultures receiving oxygen. Results The expression pattern of the HXT gene family was distinct in cells grown under aerobic, hypoxic and anaerobic conditions. The transcription of HXT2, HXT4 and HXT5 was low when the oxygen concentration in the cultures was low, both under steady state and non-steady state conditions, whereas the expression of HXT6, HXT13 and HXT15/16 was higher in hypoxic than in fully aerobic or anaerobic conditions. None of the HXT genes showed higher transcript levels in strictly anaerobic conditions. Expression of HXT9, HXT14 and GAL2 was not detected under the culture conditions studied. Conclusion When oxygen becomes limiting in a glucose-limited chemostat cultivation, the glucose uptake rate per cell increases. However, the expression of none of the hexose transporter encoding genes was increased in anaerobic conditions. It thus seems that the decrease in the moderately low affinity uptake and consequently the relative increase of high affinity uptake may itself allow the higher specific glucose consumption rate to occur in anaerobic compared to aerobic conditions.
Collapse
Affiliation(s)
- Eija Rintala
- VTT, Technical Research Centre of Finland, PO Box 1000, FI-02044 VTT, Finland.
| | | | | | | | | |
Collapse
|
33
|
Peyronnet J, Roux JC, Mamet J, Perrin D, Lachuer J, Pequignot JM, Dalmaz Y. Developmental plasticity of the carotid chemoafferent pathway in rats that are hypoxic during the prenatal period. Eur J Neurosci 2008; 26:2865-72. [PMID: 18001283 DOI: 10.1111/j.1460-9568.2007.05884.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The chemoreflex pathway undergoes postnatal maturation, and the perinatal environment plays a critical role in shaping respiratory control system. We investigated the role of prenatal hypoxia on the maturation of the chemoreflex neural circuits regulating ventilation in rat. Effects of hypoxia (10% O2) from the 5th to the 20th day of gestation were studied on male offspring at birth and on postnatal days 3, 7, 21 and 68. Maturation of the respiratory control system was assessed by in vivo tyrosine hydroxylase (TH) activity measurement in peripheral chemoreceptors (carotid bodies, petrosal ganglia), and in brainstem catecholaminergic cell groups (A2C2c and A1C1 areas in the medulla, A5 and A6 areas in the pons). Resting ventilation and ventilatory response to hypoxia were evaluated as functional sequelae. In peripheral structures, prenatal hypoxia reduced TH activity within the first postnatal week and enhanced it later. In contrast, in central areas, prenatal hypoxia upregulated TH activity within the first postnatal week and downregulated it later. The in vivo TH activity impairment is therefore tissue specific, with an opposite effect on the peripheral and central neural circuits. A shift of the effect of prenatal hypoxia occurred between 1 and 3 weeks, indicating a postnatal temporal effect of prenatal hypoxia. An important period in the development of the chemoafferent pathway occurred between the first and the third postnatal week. Functionally, prenatal hypoxia impaired resting ventilation and ventilatory response to hypoxia. The alterations of the catecholaminergic components of the chemoafferent pathway resulting from prenatal hypoxia might contribute to impair postnatal respiratory behaviour.
Collapse
Affiliation(s)
- J Peyronnet
- Université Lyon 1, UMR CNRS 5123, Physiologie intégrative Cellulaire et Moléculaire, Villeurbanne, F-69622, Lyon, France.
| | | | | | | | | | | | | |
Collapse
|
34
|
Chen XQ, Wang SJ, Du JZ, Chen XC. Diversities in hepatic HIF-1, IGF-I/IGFBP-1, LDH/ICD, and their mRNA expressions induced by CoCl(2) in Qinghai-Tibetan plateau mammals and sea level mice. Am J Physiol Regul Integr Comp Physiol 2006; 292:R516-26. [PMID: 16990490 DOI: 10.1152/ajpregu.00397.2006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ochotona curzoniae and Microtus oeconomus are the native mammals living on the Qinghai-Tibetan-Plateau of China. The molecular mechanisms of their acclimatization to the Plateau-hypoxia remain unclear. Expressions of hepatic hypoxia-inducible factor (HIF)-1alpha, insulin-like growth factor-I (IGF-I)/IGF binding protein (BP)-1(IGFBP-1; including genes), and key metabolic enzymatic genes [lactate dehydrogenase (LDH)-A/isocitrate dehydrogenase (ICD)] are compared in Qinghai-Tibetan-Plateau mammals and sea-level mice after injection of CoCl(2) (20, 40, or 60 mg/kg) and normobaric hypoxia (16.0% O(2), 10.8% O(2), and 8.0% O(2)) for 6 h, tested by histochemistry, Western blot analysis, ELISA, and RT-PCR. Major results are CoCl(2) markedly increased 1) HIF-1alpha only in mice, 2) hepatic and circulatory IGF-I in M. oeconomus, 3) hepatic IGFBP-1 in mice and O. curzoniae, and 4) LDH-A but reduced ICD mRNA in mice (CoCl(2) 20 mg/kg) but were unchanged in the Tibetan mammals. Normobaric hypoxia markedly 1) increased HIF-1alpha and LDH-A mRNA in mice and M. oeconomus (8.0% O(2)) not in O. curzoniae, and 2) reduced ICD mRNA in mice and M. oeconomus (8.0% O(2)) not in O. curzoniae. Results suggest that 1) HIF-1alpha responsiveness to hypoxia is distinct in lowland mice and plateau mammals, reflecting a diverse tolerance of the three species to hypoxia; 2) CoCl(2) induces diversities in HIF-1, IGF-I/IGFBP-1 protein or genes in mice, M. oeconomus, and O. curzoniae. In contrast, HIF-1 mediates IGFBP-1 transcription only in mice and in M. oeconomus (subjected to severe hypoxia); 3) differences in IGF-I/IGFBP-1 expressions induced by CoCl(2) reflect significant diversities in hormone regulation and cell protection from damage; and 4) activation of anaerobic glycolysis and reduction of Krebs cycle represents strategies of lowland-animals vs. the stable metabolic homeostasis of plateau-acclimatized mammals.
Collapse
Affiliation(s)
- Xue-Qun Chen
- Division of Neurobiology and Physiology, College of Life Sciences, Zhejiang University, Yuquan Campus, Hangzhou 310027, China.
| | | | | | | |
Collapse
|
35
|
Rojas DA, Perez-Munizaga DA, Centanin L, Antonelli M, Wappner P, Allende ML, Reyes AE. Cloning of hif-1alpha and hif-2alpha and mRNA expression pattern during development in zebrafish. Gene Expr Patterns 2006; 7:339-45. [PMID: 16997637 DOI: 10.1016/j.modgep.2006.08.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2006] [Revised: 08/04/2006] [Accepted: 08/08/2006] [Indexed: 10/24/2022]
Abstract
Hypoxia-inducible factors (HIFs) regulate gene expression in response to hypoxia and in vertebrates they are known to participate in several developmental processes, including angiogenesis, vasculogenesis, heart and central nervous system development. Over the last decade, major progress in unraveling the molecular mechanisms that mediate regulation of HIF proteins by oxygen tension has been reported, but our knowledge on their developmental regulation during embryogenesis in model organisms is limited. Expression of hif-1alpha and hif-2alpha genes has been characterized during normal mouse development and they were found to be expressed from stages E7.5, later in E9.5 and E15.5 in several different tissues such as the brain, heart and blood vessels. However, there is no detailed temporal information on their expression at other embryonic stages, even though orthologous genes have been described in several different vertebrate species. In this study, we describe the cloning and detailed expression pattern of zebrafish hif-1alpha and hif-2alpha genes. Sequence analysis revealed that zebrafish Hif proteins are highly homologous to other vertebrate orthologues. Zebrafish hif-1alpha and hif-2alpha are both expressed throughout development in discrete territories in a dynamic pattern. Interestingly, in the notochord the expression of hif-1alpha is switched off, while hif-2alpha transcription is turned on, signifying that the two genes might have partially overlapping, although non-redundant functions in development. This is the first time that a detailed comparison of the expression of hif-1alpha and hif-2alpha is directly assessed in a vertebrate model system throughout development.
Collapse
Affiliation(s)
- Diego A Rojas
- Facultad de Ciencias de la Salud, Universidad Diego Portales, Av. Ejército Libertador 141, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
36
|
Kang MJ, Kim HJ, Kim HK, Lee JY, Kim DH, Jung KJ, Kim KW, Baik HS, Yoo MA, Yu BP, Chung HY. The effect of age and calorie restriction on HIF-1-responsive genes in aged liver. Biogerontology 2006; 6:27-37. [PMID: 15834661 DOI: 10.1007/s10522-004-7381-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Accepted: 09/20/2004] [Indexed: 12/22/2022]
Abstract
Hypoxia inducible factor-1 (HIF-1) regulates transactivation of several genes in response to hypoxia condition. We explore hepatic HIF-1 responsive gene regulation during aging and the age-related changes of the HIF-1 related gene activation in young and old rats. Results indicate that the aging process induces the activation of HIF-1alpha, which is accompanied by increased HIF-1 DNA binding. This increased binding activity is accompanied by the increase of HIF-1-dependent genes, heme oxygenase-1 (HO-1), vascular endothelial growth factor (VEGF), erythropoietin (EPO), and inducible nitric oxide synthase (iNOS), which all showed remarkable up-regulation during aging process. In contrast, the increased HIF-1 related gene expression was effectively blunted by the anti-oxidative action of calorie restriction in aged rat liver. We propose that age-related HIF-1 binding activity may well be influenced by the increased pro-oxidative conditions of aged animals, which up-regulate HIF-1-dependent gene expression.
Collapse
Affiliation(s)
- Min Ju Kang
- College of Pharmacy, Aging Tissue Bank, Pusan National University, Busan, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Tuncer MC, Hatipoglu ES, Ozturk H, Kervancioglu P, Buyukbayram H. The Effects of L-Arginine on Neurological Function, Histopathology, and Expression of Hypoxia-Inducible Factor-1 Alpha following Spinal Cord Ischemia in Rats. Eur Surg Res 2006; 37:323-9. [PMID: 16465055 DOI: 10.1159/000090331] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Accepted: 10/27/2005] [Indexed: 11/19/2022]
Abstract
The aim of this study was to investigate the effects of L-arginine (L-Arg) on neurological function, histopathology, and expression of hypoxia-inducible factor-1 alpha (HIF-1alpha) following spinal cord ischemia in rats, and the interaction between therapy with the nitric oxide donor L-Arg and up-regulation of the expression of HIF-1alpha. Thirty Wistar rats weighing between 200 and 250 g were divided into three groups, each containing 10 rats: group 1, sham operation; group 2, untreated ischemia-reperfusion (I-R); group 3, I-R plus L-Arg treatment. Spinal cord ischemia was applied for 20 min. There were no significant differences in mean arterial pressures, temperatures, and blood gas levels among the groups. In group 2, malondialdehyde values were significantly increased compared with groups 1 and 3. The rats with aortic occlusion in group 2 had paraplegia or paraparesis. In group 3, all animals were neurologically intact. In group 3, spinal motor neurons did not decrease significantly, and little proliferation of microglia was observed compared with those in group 2. In group 2, spinal motor neurons in ventral gray matter decreased significantly compared with those in groups 1 and 3. HIF-1alpha-positive immunostaining was mildly detected in group 2 animals. The expression of immunoreactive cells was intensely increased in spinal cord tissue from I-R/L-Arg rats. In conclusion, our findings suggest that HIF-1alpha-positive immunostaining may be critical factors in the pathophysiology of inflammatory spinal cord injury induced by I-R. Nitric oxide may play an important role in the immunohistochemical expression of these molecules, and the neuroprotective benefit of L-Arg may be attributed to preventing neural cell necrosis.
Collapse
Affiliation(s)
- M Cudi Tuncer
- Department of Anatomy, Dicle University, Medical School, Diyarbakir, Turkey.
| | | | | | | | | |
Collapse
|
38
|
Training in the Morris Water Maze of Female and Male Rats Exposed to Hypoxia at Various Periods of Prenatal Development. J EVOL BIOCHEM PHYS+ 2005. [DOI: 10.1007/s10893-006-0007-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
39
|
Hall JR, Richards RC, MacCormack TJ, Ewart KV, Driedzic WR. Cloning of GLUT3 cDNA from Atlantic cod (Gadus morhua) and expression of GLUT1 and GLUT3 in response to hypoxia. ACTA ACUST UNITED AC 2005; 1730:245-52. [PMID: 16081168 DOI: 10.1016/j.bbaexp.2005.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 07/04/2005] [Accepted: 07/07/2005] [Indexed: 10/25/2022]
Abstract
A putative facilitative glucose transporter, GLUT3, cDNA was cloned from Atlantic cod. It is ubiquitously expressed, with substantial levels in kidney. The 519 aa protein has the highest sequence identity (66.3%) to grass carp GLUT3. Atlantic cod were exposed to a hypoxic challenge (45% DO2) for 24 h and the effects on GLUT1 and GLUT3 expression assessed. GLUT1 expression in gill is upregulated; however, in spleen, there is a significant decrease in both GLUT1 and GLUT3 expression. The increase in GLUT1 mRNA is considered to be associated with an increased energy demand on gill, whereas, the decrease in gene expression in spleen potentially reflects a general decrease in rates of transcription.
Collapse
Affiliation(s)
- Jennifer R Hall
- Ocean Sciences Centre, Memorial University of Newfoundland, St. John's, Newfoundland, Canada A1C 5S7
| | | | | | | | | |
Collapse
|
40
|
Silva JLT, Giannocco G, Furuya DT, Lima GA, Moraes PAC, Nachef S, Bordin S, Britto LRG, Nunes MT, Machado UF. NF-kappaB, MEF2A, MEF2D and HIF1-a involvement on insulin- and contraction-induced regulation of GLUT4 gene expression in soleus muscle. Mol Cell Endocrinol 2005; 240:82-93. [PMID: 16024167 DOI: 10.1016/j.mce.2005.05.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Revised: 05/13/2005] [Accepted: 05/20/2005] [Indexed: 02/07/2023]
Abstract
The GLUT4 gene transcriptional activity has a profound impact on the insulin-mediated glucose disposal and it is, therefore, important to understand the mechanisms underlying it. Insulin and exercise modulate GLUT4 expression in vivo, but the net control and involved mechanisms of each one have not been established yet. This paper sought to discriminate, in soleus muscle, the effects of insulin and muscle contraction on GLUT4 gene expression, and the involvement of transcriptional factors: myocite enhancer factor 2 (MEF2 A/C/D), hypoxia inducible factor 1-a (HIF1-a) and nuclear factor-kappa B (NF-kappaB). The GLUT4 mRNA was reduced by fasting (40%), and increased by in vitro incubation with insulin (25%) or insulin plus glucose (40%), which was accompanied by opposite regulations of NF-kappaB mRNA. Differently, in vitro, muscle contraction led to a rapid increase (35-80%) in GLUT4, MEF2A, MEF2D and HIF1-a mRNAs. Additionally, electrophoretic mobility shift assay confirmed changes in the binding activity of nuclear proteins to consensus NF-kappaB, GLUT4-Ebox and GLUT4-AT-rich element probes, parallel to the mRNA changes of their respective transcriptional factors NF-kappaB, HIF1-a and MEF2s. Concluding, insulin- and contraction-induced regulation of GLUT4 expression involves distinct transcriptional factors.
Collapse
Affiliation(s)
- Jose L T Silva
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Xu JF, Chen XQ, Du JZ, Wang TY. CRF receptor type 1 mediates continual hypoxia-induced CRF peptide and CRF mRNA expression increase in hypothalamic PVN of rats. Peptides 2005; 26:639-46. [PMID: 15752579 DOI: 10.1016/j.peptides.2004.11.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Revised: 11/15/2004] [Accepted: 11/17/2004] [Indexed: 11/19/2022]
Abstract
We demonstrated previously that hypoxia activated CRF and CRF mRNA in PVN, and CRF receptor 1 (CRFR1) mRNA in rat pituitary. The aim of the study is to test whether the hypoxia-activated CRF and CRF mRNA is associated with triggering CRFR1. Rats were exposed to hypobaric hypoxia at altitude of 2 and 5 km. CRF and CRF mRNA were assayed by immunostaining and in situ hybridization. CRFR1 mRNA was assayed by RT-PCR. Results showed that 5 km continual hypoxia increased CRF and CRF mRNA in PVN, CRFR1 mRNA in pituitary, and plasma corticosterone. The hypoxia-increased CRF, CRF mRNA, CRFR1 mRNA, and corticosterone were blocked by CRFR1 antagonist (CP-154,526), suggesting that CRFR1 in PVN and pituitary are responsible for the hypoxia-increased CRF and CRF mRNA in PVN.
Collapse
Affiliation(s)
- Jian-Fen Xu
- Division of Neurobiology and Physiology, College of Life Sciences, Zhejiang University (Yuquan Campus), Hangzhou 310027, PR China
| | | | | | | |
Collapse
|
42
|
Zhang F, Vannucci SJ, Philp NJ, Simpson IA. Monocarboxylate transporter expression in the spontaneous hypertensive rat: effect of stroke. J Neurosci Res 2005; 79:139-45. [PMID: 15578721 DOI: 10.1002/jnr.20312] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The expression of the monocarboxylate transporters (MCT) 1, 2, and 4 have been studied in the brains of spontaneous hypertensive (SH) rats after an ischemic insult induced by a permanent occlusion of the left middle cerebral artery (MCAO). Profound temporal changes in MCT1 expression were observed in various regions of the ipsilateral hemisphere over the period of 1 hr to 5 days after MCAO. Initially, a very rapid and transient increase in MCT1 mRNA was observed in neurons in the second layer of the cortex and in the piriform cortex at 1-3 hr. A slower but sustained increase in MCT1 mRNA expression was observed in astrocytes in the peri-infarct region beginning at 6 hr after MCAO and persisting over a period of 120 hr coinciding with their activation, migration, and involvement in scar formation. An increase in MCT1 expression in endothelial cells was seen over the same period. These increases in MCT1 expression in astrocytes and endothelial cells were accompanied by a corresponding increase in MCT1 protein. Finally at 120 hr post-MCAO, increases in MCT1, MCT2, and MCT4 expression were observed in cells within the infarct and bordering the scar, the identity of which remains to be determined. Consistent with cell death, the levels of MCT1, MCT2, and MCT4 mRNA decreased with cell death within the infarcted area but unlike MCT1, no increases in either MCT2 or 4 were observed within the remaining ipsilateral hemisphere. These studies reveal that the pattern of regulation of MCT1 expression after MCAO is similar to that observed previously for glucose transporter 1 (GLUT1) and suggest that the regulation of MCT1 mRNA expression is mediated by the hypoxia-inducible transcription factor HIF1.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
43
|
Sipe CW, Gruber EJ, Saha MS. Short upstream region drives dynamic expression of hypoxia-inducible factor 1alpha during Xenopus development. Dev Dyn 2005; 230:229-38. [PMID: 15162502 DOI: 10.1002/dvdy.20049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Hypoxia-inducible factor 1alpha (HIF-1alpha) plays a central role in regulating oxygen-dependent gene expression and is involved in a range of pathways implicated in cellular survival, proliferation, and development. While the posttranslational regulation of HIF-1alpha is well characterized, the relative importance of its control at the transcriptional level during development remains less clear. Although the mouse and human promoter regions have been analyzed in vitro, to date, there has been no in vivo analysis of any vertebrate HIF-1alpha promoter. To investigate the transcriptional regulation of HIF-1alpha during development of the amphibian Xenopus laevis, we have described the gene's expression pattern and isolated the xHIF-1alpha upstream regulatory regions. We show xHIF-1alpha mRNA to be constitutively expressed at low levels throughout embryogenesis, but with significant up-regulation during gastrula stages, and subsequently, in specific regions of the central nervous system and axial tissues. Our functional analysis using a series of truncated xHIF-1alpha promoter constructs demonstrates that a 173-bp region of the proximal promoter, which is 100% conserved among five allelic variants, is sufficient to drive correct expression in transgenic embryos. Although these results are corroborated by a parallel set of in vitro transfection experiments in a Xenopus cell line, some key differences suggest the importance of using transgenic methods in conjunction with in vitro assays.
Collapse
MESH Headings
- Alleles
- Animals
- Animals, Genetically Modified
- Base Sequence
- Cell Hypoxia/genetics
- Cell Hypoxia/physiology
- Cloning, Molecular
- Conserved Sequence/genetics
- Embryo, Nonmammalian/cytology
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Gene Expression Regulation, Developmental
- Genes, Reporter/genetics
- Green Fluorescent Proteins/analysis
- Green Fluorescent Proteins/genetics
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit
- Molecular Sequence Data
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Sequence Analysis, DNA
- Transcription Factors/genetics
- Transcription, Genetic/genetics
- Xenopus laevis/embryology
- Xenopus laevis/genetics
- Xenopus laevis/metabolism
Collapse
Affiliation(s)
- Conor W Sipe
- Department of Biology, College of William and Mary, Williamsburg, Virginia 23187, USA
| | | | | |
Collapse
|
44
|
Zhao WQ, Chen H, Quon MJ, Alkon DL. Insulin and the insulin receptor in experimental models of learning and memory. Eur J Pharmacol 2004; 490:71-81. [PMID: 15094074 DOI: 10.1016/j.ejphar.2004.02.045] [Citation(s) in RCA: 343] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2004] [Indexed: 01/25/2023]
Abstract
Insulin is best known for its action on peripheral insulin target tissues such as the adipocyte, muscle and liver to regulate glucose homeostasis. In the central nervous system (CNS), insulin and the insulin receptor are found in specific brain regions where they show evidence of participation in a variety of region-specific functions through mechanisms that are different from its direct glucose regulation in the periphery. While the insulin/insulin receptor associated with the hypothalamus plays important roles in regulation of the body energy homeostasis, the hippocampus- and cerebral cortex-distributed insulin/insulin receptor has been shown to be involved in brain cognitive functions. Emerging evidence has suggested that insulin signaling plays a role in synaptic plasticity by modulating activities of excitatory and inhibitory receptors such as glutamate and GABA receptors, and by triggering signal transduction cascades leading to alteration of gene expression that is required for long-term memory consolidation. Furthermore, deterioration of insulin receptor signaling appears to be associated with aging-related brain degeneration such as the Alzheimer's dementia and cognitive impairment in aged subjects suffering type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Wei-Qin Zhao
- Blanchette Rockefeller Neurosciences Institute, 3rd floor, Academic and Research Building, 9601 Medical Center Drive, Rockville, MD 20850, USA.
| | | | | | | |
Collapse
|
45
|
Shao R, Zhang FP, Tian F, Anders Friberg P, Wang X, Sjöland H, Billig H. Increase of SUMO-1 expression in response to hypoxia: direct interaction with HIF-1alpha in adult mouse brain and heart in vivo. FEBS Lett 2004; 569:293-300. [PMID: 15225651 DOI: 10.1016/j.febslet.2004.05.079] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Revised: 05/25/2004] [Accepted: 05/30/2004] [Indexed: 11/28/2022]
Abstract
The present study investigates the regulation of small ubiquitin-related modifier-1 (SUMO-1) expression in response to hypoxia in adult mouse brain and heart. We observed a significant increase in SUMO-1 mRNAs and proteins after hypoxic stimulation in vivo. Because SUMO-1 interacts with various transcription factors, including hypoxia-inducible factor-1beta (HIF-1beta) in vitro, we not only demonstrated that the HIF-1alpha expression is increased by hypoxia in brain and heart, but also provided evidence that SUMO-1 co-localizes in vivo with HIF-1alpha in response to hypoxia by demonstrating the co-expression of these two proteins in neurons and cardiomyocytes. The specific interaction between SUMO-1 and HIF-1alpha was additionally demonstrated with co-immunoprecipitation. These results indicate that the increased levels of SUMO-1 participate in the modulation of HIF-1alpha function through sumoylation in brain and heart.
Collapse
Affiliation(s)
- Ruijin Shao
- Division of Endocrinology, Department of Physiology and Pharmacology, Göteborg University, SE-40530 Göteborg, Sweden
| | | | | | | | | | | | | |
Collapse
|
46
|
Perrin D, Mamet J, Scarna H, Roux JC, Bérod A, Dalmaz Y. Long-term prenatal hypoxia alters maturation of brain catecholaminergic systems and motor behavior in rats. Synapse 2004; 54:92-101. [PMID: 15352134 DOI: 10.1002/syn.20065] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In order to determine the influence of long-term prenatal hypoxia on the maturation of the brain catecholaminergic structures involved in motor and cognitive functions, pregnant rats were subjected to hypoxia (10% O2) from the 5th to 20th day of gestation. The in vivo activity of tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis, was assessed, by accumulation of L-DOPA after i.p. administration of NSD-1015, in the motor cortex areas, the hippocampus, and the striatum at birth and at the 3rd, 7th, 14th, 21st, and 68th postnatal days. The motor reactivity to novelty and the circadian motor activity were measured at the 21st and 68th postnatal days. Exposure to prenatal hypoxia strongly altered the developmental pattern of in vivo TH activity in restricted noradrenergic terminals of the brain. In the 21-day-old prenatal hypoxic rats, the TH activity was reduced by 80% in the motor cortex areas and by 43% in the hippocampus, compared to control rats, while no differences could be detected in the striatum. Compared to control rats, the prenatal hypoxic pups exhibited a higher motor reactivity to novelty and a nocturnal motor hypoactivity at the 21st postnatal day. The neurochemical and behavioral alterations were no longer observed at the 68th postnatal day. The altered in vivo TH activity in the young rats might be part of the neural mechanisms contributing to the motor behavioral impairments induced by prenatal hypoxia. Long-term prenatal hypoxia could be linked to the development of psychopathologies that can be detected in infancy.
Collapse
Affiliation(s)
- David Perrin
- Laboratoire de Physiologie Intégrative, Cellulaire et Moléculaire UMR CNRS 5123, Université Claude Bernard, Lyon, France.
| | | | | | | | | | | |
Collapse
|
47
|
Lampl M, Jeanty P. Exposure to maternal diabetes is associated with altered fetal growth patterns: A hypothesis regarding metabolic allocation to growth under hyperglycemic-hypoxemic conditions. Am J Hum Biol 2004; 16:237-63. [PMID: 15101051 DOI: 10.1002/ajhb.20015] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The prevalence of diabetes is rising worldwide, including women who grew poorly in early life, presenting intergenerational health problems for their offspring. It is well documented that fetuses exposed to maternal diabetes during pregnancy experience both macrosomia and poor growth outcomes in birth size. Less is known about the in utero growth patterns that precede these risk factor expressions. Fetal growth patterns and the effects of clinical class and glycemic control were investigated in 37 diabetic pregnant women and their fetuses and compared to 29 nondiabetic, nonsmoking maternal/fetal pairs who were participants in a biweekly longitudinal ultrasound study with measurements of the head, limb, and trunk dimensions. White clinical class of the diabetic women was recorded (A2-FR) and glycosylated hemoglobin levels taken at the time of measurement assessed glycemic control (median 6.9%, interquartile range 5.6-9.2%). No significant difference in fetal weight was found by exposure. The exposed sample had greater abdominal circumferences from 21 weeks (P < or = 0.05) and shorter legs, but greater upper arm and thigh circumferences accompanied increasing glycemia in the second trimester. In the third trimester, exposed fetuses had a smaller slope for the occipital frontal diameter (P = 0.00) and were brachycephalic. They experienced a proximal/distal growth gradient in limb proportionality with higher humerus / femur ratios (P = 0.04) and arms relatively long by comparison with legs (P = 0.02). HbA1c levels above 7.5% accompanied shorter femur length for thigh circumference after 30 gestational weeks of age. Significant effects of diabetic clinical class and glycemic control were identified in growth rate timing. These growth patterns suggest that hypoxemic and hyperglycemic signals cross-talk with their target receptors in a developmentally regulated, hierarchical sequence. The increase in fetal fat often documented with diabetic pregnancy may reflect altered growth at the level of cell differentiation and proximate mechanisms controlling body composition. These data suggest that the maternal-fetal interchange circuit, designed to share and capture resources on the fetal side, may not have had a long evolutionary history of overabundance as a selective force, and modern health problems drive postnatal sequelae that become exacerbated by increasing longevity.
Collapse
Affiliation(s)
- Michelle Lampl
- Department of Anthropology, Emory University, Atlanta, Georgia 30324, USA.
| | | |
Collapse
|
48
|
Leloup C, Magnan C, Alquier T, Mistry S, Offer G, Arnaud E, Kassis N, Ktorza A, Pénicaud L. Intrauterine hyperglycemia increases insulin binding sites but not glucose transporter expression in discrete brain areas in term rat fetuses. Pediatr Res 2004; 56:263-7. [PMID: 15181191 DOI: 10.1203/01.pdr.0000132853.35660.27] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Diabetic pregnancy results in several metabolic and hormonal disorders, both in the embryo and the fetus of different species, including humans. Insulin is a potent modulator of brain development and is suggested to promote the differentiation and maturation of hypothalamic or related extrahypothalamic structures, which are directly involved in neural inputs to the pancreas. Because these structures are known to be specifically responsive both to insulin and glucose, we examined the effects of 48-h hyperglycemic clamps in unrestrained pregnant rats on insulin binding and glucose transporter expression in hypothalamic and extrahypothalamic-related areas of their fetal offspring. The main result was an increase in insulin binding in the ventromedial hypothalamic nucleus (VMH), the arcuate nucleus (AN), and the lateral hypothalamus (LH), and in the nucleus of the tractus solitarius (NTS) for extrahypothalamic areas (+30% in the VMH, +37% in the AN, +25.8% in the LH, and +37.3% in the NTS). The deleterious effect of brain hyperinsulinism during the late gestational stage does not seem to act through glucose transporter (GLUT) expression, inasmuch as no relationship between GLUT level and hyperinsulinism in brain areas could be observed. The specific increase in insulin binding in areas involved in the nervous control of metabolism could be a factor in the increased glucose intolerance and impairment of insulin secretion that was previously observed in the adult rats from hyperglycemic mothers.
Collapse
Affiliation(s)
- Corinne Leloup
- CNRS UMR 5018-UPS, CHU de Rangueil, 1 Ave Jean Poulhès, 31 403 Toulouse cedex, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Baud O, Daire J, Dalmaz Y, Fontaine RH, Krueger RC, Sebag G, Evrard P, Gressens P, Verney C. Gestational hypoxia induces white matter damage in neonatal rats: a new model of periventricular leukomalacia. Brain Pathol 2004; 14:1-10. [PMID: 14997932 PMCID: PMC8095946 DOI: 10.1111/j.1750-3639.2004.tb00492.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In the premature infant, periventricular leukomalacia, usually related to hypoxicischemic white matter damage, is the main cause of neurological impairment. We hypothesized that protracted prenatal hypoxia might induce white matter damage during the perinatal period. Pregnant Sprague-Dawley rats were placed in a chamber supplied with hypoxic gas (10% O2-90% N2) from embryonic day 5 (E5) to E20. Neonatal rat brains were investigated by histology, immunocytochemistry, western blotting, in situ hybridization, DNA fragmentation analysis, and in vivo magnetic resonance imaging (MRI). Body weight of pups subjected to prenatal hypoxia was 10 to 30% lower from P0 to P14 than in controls. Specific white matter cysts were detected between P0 and P7 in pups subjected to prenatal hypoxia, in addition to abnormal extra-cellular matrix, increased lipid peroxidation, white matter cell death detected by TUNEL, and increased activated macrophage counts in white matter. Subsequently, gliotic scars and delayed myelination primarily involving immature oligodendrocytes were seen. In vivo MRI with T1, T2, and diffusion sequences disclosed similar findings immediately after birth, showing strong correlations with histological abnormalities. We speculate that protracted prenatal hypoxia in rat induces white matter damage occurring through local inflammatory response and oxidative stress linked to re-oxygenation during the perinatal period.
Collapse
Affiliation(s)
- Olivier Baud
- Laboratoire de Neurobiologie du Développement, INSERM E9935, Service de Néonatologie et service de Neurologie Hôpital Robert, Debré, Paris, Farnce
| | - Jean‐Luc Daire
- Service d'imagerie médicale, Hôpital Robert Debré, Paris, France
| | - Yvette Dalmaz
- Laboratoire de Physiologie des Régulations Energétiques, Cellulaires et Moléculaires UMR CNRS 5123, Lyon, France
| | - Romain H Fontaine
- Laboratoire de Neurobiologie du Développement, INSERM E9935, Service de Néonatologie et service de Neurologie Hôpital Robert, Debré, Paris, Farnce
| | - Richard C. Krueger
- Division of Neonatology, Cedars Sinai Medical Center, Los Angeles, Calif
| | - Guy Sebag
- Service d'imagerie médicale, Hôpital Robert Debré, Paris, France
| | - Philippe Evrard
- Laboratoire de Neurobiologie du Développement, INSERM E9935, Service de Néonatologie et service de Neurologie Hôpital Robert, Debré, Paris, Farnce
| | - Pierre Gressens
- Laboratoire de Neurobiologie du Développement, INSERM E9935, Service de Néonatologie et service de Neurologie Hôpital Robert, Debré, Paris, Farnce
| | - Catherine Verney
- Laboratoire de Neurobiologie du Développement, INSERM E9935, Service de Néonatologie et service de Neurologie Hôpital Robert, Debré, Paris, Farnce
| |
Collapse
|
50
|
Makino Y, Nakamura H, Ikeda E, Ohnuma K, Yamauchi K, Yabe Y, Poellinger L, Okada Y, Morimoto C, Tanaka H. Hypoxia-inducible factor regulates survival of antigen receptor-driven T cells. THE JOURNAL OF IMMUNOLOGY 2004; 171:6534-40. [PMID: 14662854 DOI: 10.4049/jimmunol.171.12.6534] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peripheral T lymphocytes undergo activation by antigenic stimulation and function in hypoxic areas of inflammation. We demonstrated in CD3-positive human T cells accumulating in inflammatory tissue expression of the hypoxia-inducible factor-1alpha (HIF-1alpha), indicating a role of hypoxia-mediated signals in regulation of T cell function. Surprisingly, accumulation of HIF-1alpha in human T cells required not only hypoxia but also TCR/CD3-mediated activation. Moreover, hypoxia repressed activation-induced cell death (AICD) by TCR/CD3 stimulation, resulting in an increased survival of the cells. Microarray analysis suggested the involvement of HIF-1 target gene product adrenomedullin (AM) in this process. Indeed, AM receptor antagonist abrogated hypoxia-mediated repression of AICD. Moreover, synthetic AM peptides repressed AICD even in normoxia. Taken together, we propose that hypoxia is a critical determinant of survival of the activated T cells via the HIF-1alpha-AM cascade, defining a previously unknown mode of regulation of peripheral immunity.
Collapse
Affiliation(s)
- Yuichi Makino
- Division of Clinical Immunology, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|