1
|
Le YP, Saito K, Parajuli B, Sakai K, Kubota Y, Miyakawa M, Shinozaki Y, Shigetomi E, Koizumi S. Severity of Peripheral Infection Differentially Affects Brain Functions in Mice via Microglia-Dependent and -Independent Mechanisms. Int J Mol Sci 2023; 24:17597. [PMID: 38139424 PMCID: PMC10743593 DOI: 10.3390/ijms242417597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Peripheral infection induces inflammation in peripheral tissues and the brain, impacting brain function. Glial cells are key players in this process. However, the effects of peripheral infection on glial activation and brain function remain unknown. Here, we showed that varying degrees of peripheral infection had different effects on the regulation of brain functions by microglia-dependent and -independent mechanisms. Acute mild infection (one-day LPS challenge: 1LPS) exacerbated middle cerebral artery occlusion (MCAO) injury, and severe infection (four-day LPS challenge: 4LPS) for one week suppressed it. MCAO injury was assessed by triphenyltetrazolium chloride staining. We observed early activation of microglia in the 1LPS and 4LPS groups. Depleting microglia with a colony-stimulating factor-1 receptor (CSF1R) antagonist had no effect on 1LPS-induced brain injury exacerbation but abolished 4LPS-induced protection, indicating microglial independence and dependence, respectively. Microglia-independent exacerbation caused by 1LPS involved peripheral immune cells including macrophages. RNA sequencing analysis of 4LPS-treated microglia revealed increased factors related to anti-inflammatory and neuronal tissue repair, suggesting their association with the protective effect. In conclusion, varying degrees of peripheral inflammation had contradictory effects (exacerbation vs. protection) on MCAO, which may be attributed to microglial dependence. Our findings highlight the significant impact of peripheral infection on brain function, particularly in relation to glial cells.
Collapse
Affiliation(s)
- Yen-Phung Le
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| | - Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| | - Bijay Parajuli
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| | - Kent Sakai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| | - Yuto Kubota
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| | - Miho Miyakawa
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo 409-3898, Japan; (Y.-P.L.); (K.S.); (B.P.); (K.S.); (Y.K.); (M.M.); (Y.S.); (E.S.)
- GLIA Center, University of Yamanashi, Chuo 409-3898, Japan
| |
Collapse
|
2
|
Han H, Yang Y, Jiao Y, Qi H, Han Z, Wang L, Dong L, Tian J, Vanhaesebroeck B, Li X, Liu J, Ma G, Lei H. Leverage of nuclease-deficient CasX for preventing pathological angiogenesis. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:738-748. [PMID: 37662968 PMCID: PMC10469388 DOI: 10.1016/j.omtn.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/03/2023] [Indexed: 09/05/2023]
Abstract
Gene editing with a CRISPR/Cas system is a novel potential strategy for treating human diseases. Pharmacological inhibition of phosphoinositide 3-kinase (PI3K) δ suppresses retinal angiogenesis in a mouse model of oxygen-induced retinopathy. Here we show that an innovative system of adeno-associated virus (AAV)-mediated CRISPR/nuclease-deficient (d)CasX fused with the Krueppel-associated box (KRAB) domain is leveraged to block (81.2% ± 6.5%) in vitro expression of p110δ, the catalytic subunit of PI3Kδ, encoded by Pik3cd. This CRISPR/dCasX-KRAB (4, 269 bp) system is small enough to be fit into a single AAV vector. We then document that recombinant AAV serotype (rAAV)1 efficiently transduces vascular endothelial cells from pathologic retinal vessels, which show high expression of p110δ; furthermore, we demonstrate that blockade of retinal p110δ expression by intravitreally injected rAAV1-CRISPR/dCasX-KRAB targeting the Pik3cd promoter prevents (32.1% ± 5.3%) retinal p110δ expression as well as pathological retinal angiogenesis in a mouse model of oxygen-induced retinopathy. These data establish a strong foundation for treating pathological angiogenesis by AAV-mediated CRISPR interference with p110δ expression.
Collapse
Affiliation(s)
- Haote Han
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, People’s Republic of China
| | - Yanhui Yang
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, the School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, People’s Republic of China
| | - Yunjuan Jiao
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha 410013, People’s Republic of China
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Hui Qi
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, People’s Republic of China
| | - Zhuo Han
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, People’s Republic of China
| | - Luping Wang
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, People’s Republic of China
| | - Lijun Dong
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, People’s Republic of China
| | - Jingkui Tian
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, People’s Republic of China
| | | | - Xiaopeng Li
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Junwen Liu
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha 410013, People’s Republic of China
| | - Gaoen Ma
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Hetian Lei
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, People’s Republic of China
| |
Collapse
|
3
|
Fan Y, Li J, Fang B. A Tale of Two: When Neural Stem Cells Encounter Hypoxia. Cell Mol Neurobiol 2023; 43:1799-1816. [PMID: 36308642 PMCID: PMC11412202 DOI: 10.1007/s10571-022-01293-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/02/2022] [Indexed: 11/12/2022]
Abstract
Normoxia is defined as an oxygen concentration of 20.9%, as in room air, whereas hypoxia refers to any oxygen concentration less than this. Any physiological oxygen deficiency or tissue oxygen deficiency relative to demand is called hypoxia. Neural stem cells (NSCs) are multipotent stem cells that can differentiate into multiple cell lines such as neurons, oligodendrocytes, and astrocytes. Under hypoxic conditions, the apoptosis rate of NSCs increases remarkably in vitro or in vivo. However, some hypoxia promotes the proliferation and differentiation of NSCs. The difference is related to the oxygen concentration, the duration of hypoxia, the hypoxia tolerance threshold of the NSCs, and the tissue source of the NSCs. The main mechanism of hypoxia-induced proliferation and differentiation involves an increase in cyclin and erythropoietin concentrations, and hypoxia-inducible factors play a key role. Multiple molecular pathways are activated during hypoxia, including Notch, Wnt/β-catenin, PI3K/Akt, and altered microRNA expression. In addition, we review the protective effect of exogenous NSCs transplantation on ischemic or anoxic organs, the therapeutic potential of hypoxic preconditioning on exogenous NSCs and clinical application of NSCs.
Collapse
Affiliation(s)
- Yiting Fan
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Jinshi Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Zeng Q, Huang P, Wang Z, Wei L, Lin K. Remote ischemic conditioning in the treatment of acute cerebral infarction: A case control study. Heliyon 2023; 9:e18181. [PMID: 37496897 PMCID: PMC10367274 DOI: 10.1016/j.heliyon.2023.e18181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/01/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023] Open
Abstract
OBJECTIVE This paired case-control study aimed to evaluate the efficacy and safety of remote ischemic conditioning (RIC) in patients with acute cerebral infarction (CI) and explore potential serological markers of RIC. METHODS Patients with acute CI (<72 h) were matched 1:1 according to age, sex, and CI conditions and were divided into the RIC group and the control group. The RIC group received RIC intervention for 7 days on top of routine treatment, while the control group received a sham RIC. The curative effects and adverse reactions were observed. RESULT A total of 66 patients (mean age 60.00 ± 11.37 years; mean time of acute CI onset 32.91 ± 17.94 h) completed the study. The National Institute of Health stroke scale score on day 7, modified Rankin Scale scores on day 7 and day 90 were significantly lower than the baseline in the RIC group (P < 0.001, P = 0.003, P = 0.004, respectively) but not in the control group (P = 0.056, P = 0.169, P = 0.058, respectively). RIC was well-tolerated, and no adverse events were reported. Both plasma hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor increased in the RIC group from day 0 to day 7, while they decreased in the control group. The changes in plasma HIF-1α in the RIC group were statistically different from those in the control group (P = 0.006). CONCLUSION Early and short-term RIC treatment was well-tolerated and effective in improving the prognosis in acute CI. HIF-1α can be recognized as a biomarker for evaluating the efficacy of RIC treatment.
Collapse
Affiliation(s)
- Qiong Zeng
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Peiqi Huang
- Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Ziteng Wang
- Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Liling Wei
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Kun Lin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| |
Collapse
|
5
|
Späth MR, Hoyer-Allo KJR, Seufert L, Höhne M, Lucas C, Bock T, Isermann L, Brodesser S, Lackmann JW, Kiefer K, Koehler FC, Bohl K, Ignarski M, Schiller P, Johnsen M, Kubacki T, Grundmann F, Benzing T, Trifunovic A, Krüger M, Schermer B, Burst V, Müller RU. Organ Protection by Caloric Restriction Depends on Activation of the De Novo NAD+ Synthesis Pathway. J Am Soc Nephrol 2023; 34:772-792. [PMID: 36758124 PMCID: PMC10125653 DOI: 10.1681/asn.0000000000000087] [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: 03/20/2022] [Accepted: 01/10/2023] [Indexed: 02/11/2023] Open
Abstract
SIGNIFICANCE STATEMENT AKI is a major clinical complication leading to high mortality, but intensive research over the past decades has not led to targeted preventive or therapeutic measures. In rodent models, caloric restriction (CR) and transient hypoxia significantly prevent AKI and a recent comparative transcriptome analysis of murine kidneys identified kynureninase (KYNU) as a shared downstream target. The present work shows that KYNU strongly contributes to CR-mediated protection as a key player in the de novo nicotinamide adenine dinucleotide biosynthesis pathway. Importantly, the link between CR and NAD+ biosynthesis could be recapitulated in a human cohort. BACKGROUND Clinical practice lacks strategies to treat AKI. Interestingly, preconditioning by hypoxia and caloric restriction (CR) is highly protective in rodent AKI models. However, the underlying molecular mechanisms of this process are unknown. METHODS Kynureninase (KYNU) knockout mice were generated by Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and comparative transcriptome, proteome and metabolite analyses of murine kidneys pre- and post-ischemia-reperfusion injury in the context of CR or ad libitum diet were performed. In addition, acetyl-lysin enrichment and mass spectrometry were used to assess protein acetylation. RESULTS We identified KYNU as a downstream target of CR and show that KYNU strongly contributes to the protective effect of CR. The KYNU-dependent de novo nicotinamide adenine dinucleotide (NAD+) biosynthesis pathway is necessary for CR-associated maintenance of NAD+ levels. This finding is associated with reduced protein acetylation in CR-treated animals, specifically affecting enzymes in energy metabolism. Importantly, the effect of CR on de novo NAD+ biosynthesis pathway metabolites can be recapitulated in humans. CONCLUSIONS CR induces the de novo NAD+ synthesis pathway in the context of IRI and is essential for its full nephroprotective potential. Differential protein acetylation may be the molecular mechanism underlying the relationship of NAD+, CR, and nephroprotection.
Collapse
Affiliation(s)
- Martin R. Späth
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - K. Johanna R. Hoyer-Allo
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lisa Seufert
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Martin Höhne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christina Lucas
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Theresa Bock
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Genetics, University of Cologne, Cologne, Germany
| | - Lea Isermann
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Medical Faculty, Institute for Mitochondrial Diseases and Aging, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Susanne Brodesser
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jan-Wilm Lackmann
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Katharina Kiefer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Felix C. Koehler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Katrin Bohl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Ignarski
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Petra Schiller
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Marc Johnsen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Torsten Kubacki
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Franziska Grundmann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Aleksandra Trifunovic
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Medical Faculty, Institute for Mitochondrial Diseases and Aging, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Marcus Krüger
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Volker Burst
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Emergency Department, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
6
|
Singh A, Chen R. The Duration of Oxygen and Glucose Deprivation (OGD) Determines the Effects of Subsequent Reperfusion on Rat Pheochromocytoma (PC12) Cells and Primary Cortical Neurons. Int J Mol Sci 2023; 24:7106. [PMID: 37108268 PMCID: PMC10138834 DOI: 10.3390/ijms24087106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Reperfusion is the fundamental treatment for ischaemic stroke; however, many ischaemic stroke patients cannot undergo reperfusion treatment. Furthermore, reperfusion can cause ischaemic reperfusion injuries. This study aimed to determine the effects of reperfusion in an in vitro ischaemic stroke model-oxygen and glucose deprivation (OGD) (0.3% O2)-with rat pheochromocytoma (PC12) cells and cortical neurons. In PC12 cells, OGD resulted in a time-dependent increase in cytotoxicity and apoptosis, and reduction in MTT activity from 2 h onwards. Reperfusion following shorter periods (4 and 6 h) of OGD recovered apoptotic PC12 cells, whereas after 12 h, OGD increased LDH release. In primary neurons, 6 h OGD led to significant increase in cytotoxicity, reduction in MTT activity and dendritic MAP2 staining. Reperfusion following 6 h OGD increased the cytotoxicity. HIF-1a was stabilised by 4 and 6 h OGD in PC12 cells and 2 h OGD onwards in primary neurons. A panel of hypoxic genes were upregulated by the OGD treatments depending on the duration. In conclusion, the duration of OGD determines the mitochondrial activity, cell viability, HIF-1a stabilization, and hypoxic gene expression in both cell types. Reperfusion following OGD of short duration is neuroprotective, whereas OGD of long duration is cytotoxic.
Collapse
Affiliation(s)
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| |
Collapse
|
7
|
Hazra R, Hubert H, Little-Ihrig L, Ghosh S, Ofori-Acquah S, Hu X, Novelli EM. Insulin-like Growth Factor-1 Prevents Hypoxia/Reoxygenation-Induced White Matter Injury in Sickle Cell Mice. Biomedicines 2023; 11:692. [PMID: 36979670 PMCID: PMC10045140 DOI: 10.3390/biomedicines11030692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Occlusion of cerebral blood vessels causes acute cerebral hypoxia-an important trigger of ischemic white matter injury and stroke in sickle cell disease (SCD). While chronic hypoxia triggers compensatory neuroprotection via insulin-like growth factor-1 (IGF-1) and hypoxia inducible factor-1α (HIF-1α), severe bouts of acute hypoxia and subsequent restoration of blood flow (hypoxia/reoxygenation, H/R) overwhelm compensatory mechanisms and cause neuroaxonal damage-identified as white matter lesions-in the brain. The neuroprotective role of IGF-1 in the pathogenesis of white matter injury in SCD has not been investigated; however, it is known that systemic IGF-1 is reduced in individuals with SCD. We hypothesized that IGF-1 supplementation may prevent H/R-induced white matter injury in SCD. Transgenic sickle mice homozygous for human hemoglobin S and exposed to H/R developed white matter injury identified by elevated expression of non-phosphorylated neurofilament H (SMI32) with a concomitant decrease in myelin basic protein (MBP) resulting in an increased SMI32/MBP ratio. H/R-challenge also lowered plasma and brain IGF-1 expression. Human recombinant IGF-1 prophylaxis significantly induced HIF-1α and averted H/R-induced white matter injury in the sickle mice compared to vehicle-treated mice. The expression of the IGF-1 binding proteins IGFBP-1 and IGFBP-3 was elevated in the IGF-1-treated brain tissue indicating their potential role in mediating neuroprotective HIF-1α signaling. This study provides proof-of-concept for IGF-1-mediated neuroprotection in SCD.
Collapse
Affiliation(s)
- Rimi Hazra
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Holland Hubert
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Lynda Little-Ihrig
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Samit Ghosh
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Solomon Ofori-Acquah
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Xiaoming Hu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15240, USA
| | - Enrico M Novelli
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
8
|
Ghori A, Prinz V, Nieminen-Kehlä M, Bayerl SH, Kremenetskaia I, Riecke J, Krechel H, Broggini T, Scherschinski L, Licht T, Keshet E, Vajkoczy P. Vascular Endothelial Growth Factor Augments the Tolerance Towards Cerebral Stroke by Enhancing Neurovascular Repair Mechanism. Transl Stroke Res 2022; 13:774-791. [PMID: 35175562 PMCID: PMC9391249 DOI: 10.1007/s12975-022-00991-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/05/2021] [Accepted: 01/26/2022] [Indexed: 11/30/2022]
Abstract
The breakdown of the blood-brain barrier (BBB) is a critical event in the development of secondary brain injury after stroke. Among the cellular hallmarks in the acute phase after stroke are a downregulation of tight-junction molecules and the loss of microvascular pericyte coverage and endothelial sealing. Thus, a rapid repair of blood vessel integrity and re-stabilization of the BBB is considered an important strategy to reduce secondary brain damage. However, the mechanisms underlying BBB disruption remain poorly understood. Especially, the role of VEGF in this context remains inconclusive. With the conditional and reversible VEGF expression systems, we studied the time windows of deleterious and beneficial VEGF actions on blood vessel integrity in mice. Using genetic systems for gain of function and loss of function experiments, we activated and inhibited VEGF signaling prior and simultaneously to ischemic stroke onset. In both scenarios, VEGF seems to play a vital role in containing the stroke-induced damage after cerebral ischemia. We report that the transgenic overexpression of VEGF (GOF) prior to the stroke stabilizes the vasculature and prevents blood-brain barrier disruption in young and aged animals after stroke. Whereas inhibition of signals for endogenous VEGF (LOF) prior to stroke results in bigger infarction with massive brain swelling and enhanced BBB permeability, furthermore, activating or blocking VEGF signaling after ischemic stroke onset had comparable effects on BBB repair and cerebral edema. VEGF can function as an anti-permeability factor, and a VEGF-based therapy in the context of stroke prevention and recovery has an enormous potential.
Collapse
Affiliation(s)
- Adnan Ghori
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| | - Vincent Prinz
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| | | | - Simon. H. Bayerl
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| | - Jana Riecke
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| | - Hanna Krechel
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| | - Thomas Broggini
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| | - Lea Scherschinski
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| | - Tamar Licht
- Department of Developmental Biology and Cancer Research, Hebrew University Hadassah Medical School, 91120 Jerusalem, Israel
| | - Eli Keshet
- Department of Developmental Biology and Cancer Research, Hebrew University Hadassah Medical School, 91120 Jerusalem, Israel
| | - Peter Vajkoczy
- Department of Neurosurgery, Universitätsmedizin Charité, 10117 Berlin, Germany
| |
Collapse
|
9
|
Koizumi S, Hirayama Y. Ischemic Tolerance Induced by Glial Cells. Neurochem Res 2022; 47:2522-2528. [PMID: 35920970 PMCID: PMC9463280 DOI: 10.1007/s11064-022-03704-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/09/2022] [Accepted: 07/16/2022] [Indexed: 11/05/2022]
Abstract
Ischemic tolerance is a phenomenon in which resistance to subsequent invasive ischemia is acquired by a preceding noninvasive ischemic application, and is observed in many organs, including the brain, the organ most vulnerable to ischemic insult. To date, much research has been conducted on cerebral ischemic tolerance as a cell-autonomous action of neurons. In this article, we review the essential roles of microglia and astrocytes in the acquisition of ischemic tolerance through neuron-non-autonomous mechanisms, where the two types of glial cells function in a concerted manner to induce ischemic tolerance.
Collapse
Affiliation(s)
- Schuichi Koizumi
- Department of Neuropharmacology, Yamanashi, Japan. .,Yamanashi GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 409-3898, Yamanashi, Japan.
| | - Yuri Hirayama
- Department of Neuropharmacology, Yamanashi, Japan.,Department of Pharmacology, Graduate School of Medicine, Chiba University, 260-8670, Chiba, Japan
| |
Collapse
|
10
|
Katsel P, Fam P, Tan W, Khan S, Gama-Sosa M, De Gasperi R, Roussos P, Robinson A, Cooper I, Schnaider-Beeri M, Haroutunian V. Engagement of vascular early response genes typifies mild cognitive impairment. Alzheimers Dement 2022; 18:1357-1369. [PMID: 34758195 PMCID: PMC10878080 DOI: 10.1002/alz.12481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 07/07/2021] [Accepted: 08/11/2021] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Molecular responses in the brains of persons with mild cognitive impairment (MCI), the earliest transitional state between normal aging and early Alzheimer's disease (AD), are poorly understood. METHODS We examined AD-related neuropathology and transcriptome changes in the neocortex of individuals with MCI relative to controls and temporal responses to the mild hypoxia in mouse brains. RESULTS Subsets of vascular early response to hypoxia genes were upregulated in MCI prior to the buildup of AD neuropathology. Early activation of pro-angiogenic hypoxia-inducible factor signaling in response to mild hypoxia was detected in mouse brains similar to those that were altered in MCI. Protracted responses to hypoxia were characterized by activation of phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt)-the mammalian target of rapamycin (mTOR) pathways in brain microvessel isolates. DISCUSSION These findings suggest that cerebrovascular remodeling is an important antecedent to the development of dementia and a component of the homeostatic response to reduced oxygen tension in aging prior to the onset of AD.
Collapse
Affiliation(s)
- Pavel Katsel
- Department of Psychiatry, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
| | - Peter Fam
- Department of Neuroscience, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
| | - Weilun Tan
- Department of Psychiatry, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
| | - Sonia Khan
- Department of Psychiatry, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
| | - Miguel Gama-Sosa
- Department of Psychiatry, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
| | - Rita De Gasperi
- Department of Psychiatry, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
| | - Panos Roussos
- Department of Psychiatry, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
- Genetics and Genomic Sciences, The Icahn School of Medicine
at Mount Sinai, New York, New York, USA
- Pamela Sklar Division of Psychiatric Genomics and Friedman
Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, New York,
USA
| | - Ari Robinson
- The Joseph Sagol Neuroscience Center Tel-Hashomer,
Ramat-Gan, Israel
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center Tel-Hashomer,
Ramat-Gan, Israel
| | - Michal Schnaider-Beeri
- Department of Psychiatry, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
- The Joseph Sagol Neuroscience Center Tel-Hashomer,
Ramat-Gan, Israel
| | - Vahram Haroutunian
- Department of Psychiatry, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
- Department of Neuroscience, The Icahn School of Medicine at
Mount Sinai, New York, New York, USA
- Mental Illness Research, Education and Clinical Center
(MIRECC), James J Peters VA Medical Center, Bronx, New York, USA
| |
Collapse
|
11
|
Neuroprotective Efficacy of Betulinic Acid Hydroxamate, a B55α/PP2A Activator, in Acute Hypoxia-Ischemia-Induced Brain Damage in Newborn Rats. Transl Stroke Res 2022; 14:397-408. [PMID: 35419730 DOI: 10.1007/s12975-022-01017-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/12/2022] [Accepted: 03/29/2022] [Indexed: 10/18/2022]
Abstract
There is an increasing evidence of the neuroprotective effects of hypoxia inducing factor prolyl-hydroxylase inhibitors (HIF-PHDi) after hypoxic-ischemic (HI) brain damage (HIBD). We studied the neuroprotective effects of betulinic hydroxamate (BAH), a novel B55α/PP2A activator that dephosphorylates and inhibits PHD2 activity, in a rat model of neonatal HIBD. Seven-day-old (P7) Wistar rats were exposed to hypoxia after left carotid artery electrocoagulation and then received vehicle (HI + VEH) or BAH 3 mg/kg i.p. 30 min post-insult. Brain damage was assessed by magnetic resonance imaging (MRI) and neurobehavioral studies testing motor and cognitive performance at P14 and P37, as well as immunohistochemical studies (TUNEL and myelin basic protein (MBP) signal) at P37. Mechanisms of damage were assessed at P14 determining excitotoxicity (glutamate/N-acetylaspartate ratio by H+-magnetic resonance spectroscopy), oxidative stress (protein nitrosylation by Oxyblot), and inflammation (cytokine and chemokine concentration). BAH reduced brain damage volume and cell death, preventing the development of motor and working memory deficits. BAH showed a robust protective effect on myelination, restoring MBP expression at P37. BAH modulated excitotoxicity, oxidative stress, and inflammation. Most neuroprotective effects were still present despite BAH administration was delayed for 12 h, whereas beneficial effects on motor strength at P14 and on cell death and myelination at P37 were preserved even when BAH administration was delayed for 24 h. In conclusion, BAH appears as an effective neuroprotective treatment for neonatal HIBD in a manner associated with the modulation of excitotoxicity, oxidative stress, and inflammation, showing a broad therapeutic window.
Collapse
|
12
|
Catalan Serra P, Soler X. Obstructive Sleep apnea and cardiovascular events in Elderly Patients. Expert Rev Respir Med 2022; 16:197-210. [PMID: 35041560 DOI: 10.1080/17476348.2022.2030225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION In recent decades, life expectancy has increased considerably. The cardiovascular effects of Obstructive Sleep Apnea (OSA) in the elderly lead to patient disability and high resource consumption. Intermittent nocturnal hypoxia leads to hemodynamic stress and adrenergic activation, which promotes cardiovascular disease. However, chronic intermittent hypoxia may protect elderly patients from cardiovascular events (CVE) due to biological adaptation. AREAS COVERED OSA patients are at increased risk of cardiovascular events. The severity of OSA increases cardiovascular risk, and this association also exists in the elderly. This article reviews the association between OSA, CPAP treatment, and CVE, particularly stroke and coronary heart disease (CHD), in the elderly. MEDLINE and the Cochrane Collaboration databases were searched from inception to July 2021. EXPERT COMMENTARY Although a positive association between OSA and the incidence of cardiovascular disease in the elderly has been established, the role of sleep apnea in certain cardiovascular events remains controversial. Most authors agree that untreated OSA is a risk factor for stroke or worse stroke prognosis. However, the association between OSA and CHD is usually less pronounced than between OSA and stroke, especially in the elderly.
Collapse
Affiliation(s)
| | - Xavier Soler
- Department of Pulmonary, Critical Care, and Sleep Medicine. University of California, San Diego, California
| |
Collapse
|
13
|
Intracellular Signaling. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
14
|
Guan Y, Liu J, Gu Y, Ji X. Effects of Hypoxia on Cerebral Microvascular Angiogenesis: Benefits or Damages? Aging Dis 2022; 14:370-385. [PMID: 37008044 PMCID: PMC10017152 DOI: 10.14336/ad.2022.0902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/02/2022] [Indexed: 11/18/2022] Open
Abstract
Cerebrovascular microcirculation is essential for maintaining the physiological functions of the brain. The brain can be protected from stress injury by remodeling the microcirculation network. Angiogenesis is a type of cerebral vascular remodeling. It is an effective approach to improve the blood flow of the cerebral microcirculation, which is necessary for preventing and treating various neurological disorders. Hypoxia is one of the most important regulators of angiogenesis, affecting the sprouting, proliferation, and maturation stages of angiogenesis. Moreover, hypoxia negatively affects cerebral vascular tissue by impairing the structural and functional integrity of the blood-brain barrier and vascular-nerve decoupling. Therefore, hypoxia has a dual effect on blood vessels and is affected by confounding factors including oxygen concentration, hypoxia duration, and hypoxia frequency and extent. Establishing an optimal model that promotes cerebral microvasculogenesis without causing vascular injury is essential. In this review, we first elaborate on the effects of hypoxia on blood vessels from two different perspectives: (1) the promotion of angiogenesis and (2) cerebral microcirculation damage. We further discuss the factors influencing the dual role of hypoxia and emphasize the benefits of moderate hypoxic irritation and its potential application as an easy, safe, and effective treatment for multiple nervous system disorders.
Collapse
Affiliation(s)
- Yuying Guan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yakun Gu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Correspondence should be addressed to: Dr. Prof. Xunming Ji; Beijing Institute of Brain Disorders, Capital Medical University, 10 Xi Tou Tiao, You Anmen, Beijing 100069, China. E-mail: .
| |
Collapse
|
15
|
Shen XY, Gao ZK, Han Y, Yuan M, Guo YS, Bi X. Activation and Role of Astrocytes in Ischemic Stroke. Front Cell Neurosci 2021; 15:755955. [PMID: 34867201 PMCID: PMC8635513 DOI: 10.3389/fncel.2021.755955] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke refers to the disorder of blood supply of local brain tissue caused by various reasons. It has high morbidity and mortality worldwide. Astrocytes are the most abundant glial cells in the central nervous system (CNS). They are responsible for the homeostasis, nutrition, and protection of the CNS and play an essential role in many nervous system diseases’ physiological and pathological processes. After stroke injury, astrocytes are activated and play a protective role through the heterogeneous and gradual changes of their gene expression, morphology, proliferation, and function, that is, reactive astrocytes. However, the position of reactive astrocytes has always been a controversial topic. Many studies have shown that reactive astrocytes are a double-edged sword with both beneficial and harmful effects. It is worth noting that their different spatial and temporal expression determines astrocytes’ various functions. Here, we comprehensively review the different roles and mechanisms of astrocytes after ischemic stroke. In addition, the intracellular mechanism of astrocyte activation has also been involved. More importantly, due to the complex cascade reaction and action mechanism after ischemic stroke, the role of astrocytes is still difficult to define. Still, there is no doubt that astrocytes are one of the critical factors mediating the deterioration or improvement of ischemic stroke.
Collapse
Affiliation(s)
- Xin-Ya Shen
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen-Kun Gao
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Mei Yuan
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yi-Sha Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
16
|
Hafez S, Eid Z, Alabasi S, Darwiche Y, Channaoui S, Hess DC. Mechanisms of Preconditioning Exercise-Induced Neurovascular Protection in Stroke. J Stroke 2021; 23:312-326. [PMID: 34649377 PMCID: PMC8521252 DOI: 10.5853/jos.2020.03006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Ischemic stroke is a leading cause of death and disability. Tissue plasminogen activator is the only U.S. Food and Drug Administration approved thrombolytic therapy for ischemic stroke patients till date. However, its use is limited due to increased risk of bleeding and narrow therapeutic window. Most of the preclinically tested pharmacological agents failed to be translated to the clinic. This drives the need for alternative therapeutic approaches that not only provide enhanced neuroprotection, but also reduce the risk of stroke. Physical exercise is a sort of preconditioning that provides the body with brief ischemic episodes that can protect the body from subsequent severe ischemic attacks like stroke. Physical exercise is known to improve cardiovascular health. However, its role in providing neuroprotection in stroke is not clear. Clinical observational studies showed a correlation between regular physical exercise and reduced risk and severity of ischemic stroke and better outcomes after stroke. However, the underlying mechanisms through which prestroke exercise can reduce the stroke injury and improve the outcomes are not completely understood. The purpose of this review is to: demonstrate the impact of exercise on stroke outcomes and show the potential role of exercise in stroke prevention and recovery; uncover the underlying mechanisms through which exercise reduces the neurovascular injury and improves stroke outcomes aiming to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Sherif Hafez
- Department of Pharmaceutical Sciences, College of Pharmacy Mercer University, Atlanta, GA, USA.,Neurology Department, Augusta University, Augusta, GA, USA
| | - Zeina Eid
- College of Pharmacy Larkin University, Miami, FL, USA
| | - Sara Alabasi
- College of Pharmacy Larkin University, Miami, FL, USA
| | | | | | - David C Hess
- Neurology Department, Augusta University, Augusta, GA, USA
| |
Collapse
|
17
|
Tetorou K, Sisa C, Iqbal A, Dhillon K, Hristova M. Current Therapies for Neonatal Hypoxic-Ischaemic and Infection-Sensitised Hypoxic-Ischaemic Brain Damage. Front Synaptic Neurosci 2021; 13:709301. [PMID: 34504417 PMCID: PMC8421799 DOI: 10.3389/fnsyn.2021.709301] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Neonatal hypoxic-ischaemic brain damage is a leading cause of child mortality and morbidity, including cerebral palsy, epilepsy, and cognitive disabilities. The majority of neonatal hypoxic-ischaemic cases arise as a result of impaired cerebral perfusion to the foetus attributed to uterine, placental, or umbilical cord compromise prior to or during delivery. Bacterial infection is a factor contributing to the damage and is recorded in more than half of preterm births. Exposure to infection exacerbates neuronal hypoxic-ischaemic damage thus leading to a phenomenon called infection-sensitised hypoxic-ischaemic brain injury. Models of neonatal hypoxia-ischaemia (HI) have been developed in different animals. Both human and animal studies show that the developmental stage and the severity of the HI insult affect the selective regional vulnerability of the brain to damage, as well as the subsequent clinical manifestations. Therapeutic hypothermia (TH) is the only clinically approved treatment for neonatal HI. However, the number of HI infants needed to treat with TH for one to be saved from death or disability at age of 18-22 months, is approximately 6-7, which highlights the need for additional or alternative treatments to replace TH or increase its efficiency. In this review we discuss the mechanisms of HI injury to the immature brain and the new experimental treatments studied for neonatal HI and infection-sensitised neonatal HI.
Collapse
Affiliation(s)
| | | | | | | | - Mariya Hristova
- Perinatal Brain Repair Group, Department of Maternal and Fetal Medicine, UCL Institute for Women’s Health, London, United Kingdom
| |
Collapse
|
18
|
Altered neurodevelopmental DNA methylation status after fetal growth restriction with brain-sparing. J Dev Orig Health Dis 2021; 13:378-389. [PMID: 34325767 DOI: 10.1017/s2040174421000374] [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: 12/19/2022]
Abstract
It is under debate how preferential perfusion of the brain (brain-sparing) in fetal growth restriction (FGR) relates to long-term neurodevelopmental outcome. Epigenetic modification of neurotrophic genes by altered fetal oxygenation may be involved. To explore this theory, we performed a follow-up study of 21 FGR children, in whom we prospectively measured the prenatal cerebroplacental ratio (CPR) with Doppler sonography. At 4 years of age, we tested their neurodevelopmental outcome using the Wechsler Preschool and Primary Scale of Intelligence, the Child Behavior Checklist, and the Behavior Rating Inventory of Executive Function. In addition, we collected their buccal DNA to determine the methylation status at predefined genetic regions within the genes hypoxia-inducible factor-1 alpha (HIF1A), vascular endothelial growth factor A (VEGFA), erythropoietin (EPO), EPO-receptor (EPOR), brain-derived neurotrophic factor (BDNF), and neurotrophic tyrosine kinase, receptor, type 2 (NTRK2) by pyrosequencing. We found that FGR children with fetal brain-sparing (CPR <1, n = 8) demonstrated a trend (0.05 < p < 0.1) toward hypermethylation of HIF1A and VEGFA at their hypoxia-response element (HRE) compared with FGR children without fetal brain-sparing. Moreover, in cases with fetal brain-sparing, we observed statistically significant hypermethylation at a binding site for cyclic adenosine monophophate response element binding protein (CREB) of BDNF promoter exon 4 and hypomethylation at an HRE located within the NTRK2 promoter (both p <0.05). Hypermethylation of VEGFA was associated with a poorer Performance Intelligence Quotient, while hypermethylation of BDNF was associated with better inhibitory self-control (both p <0.05). These results led us to formulate the hypothesis that early oxygen-dependent epigenetic alterations due to hemodynamic alterations in FGR may be associated with altered neurodevelopmental outcome in later life. We recommend further studies to test this hypothesis.
Collapse
|
19
|
Zhang X, Xiao W, Zhang Q, Xia D, Gao P, Su J, Yang H, Gao X, Ni W, Lei Y, Gu Y. Progression in Moyamoya Disease: Clinical Feature, Neuroimaging Evaluation and Treatment. Curr Neuropharmacol 2021; 20:292-308. [PMID: 34279201 PMCID: PMC9413783 DOI: 10.2174/1570159x19666210716114016] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/08/2021] [Accepted: 07/09/2021] [Indexed: 11/22/2022] Open
Abstract
Moyamoya disease (MMD) is a chronic cerebrovascular disease characterized by progressive stenosis of the arteries of the circle of Willis, with the formation of collateral vascular network at the base of the brain. Its clinical manifestations are complicated. Numerous studies have attempted to clarify the clinical features of MMD, including its epidemiology, genetic characteristics, and pathophysiology. With the development of neuroimaging techniques, various neuroimaging modalities with different advantages have deepened the understanding of MMD in terms of structural, functional, spatial, and temporal dimensions. At present, the main treatment for MMD focuses on neurological protection, cerebral blood flow reconstruction, and neurological rehabilitation, such as pharmacological treatment, surgical revascularization, and cognitive rehabilitation. In this review, we discuss recent progress in understanding the clinical features, in the neuroimaging evaluation and treatment of MMD.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Weiping Xiao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Qing Zhang
- Department of Nursing, Huashan Hospital North, Fudan University, China
| | - Ding Xia
- Department of Radiology, Huashan Hospital North, Fudan University, China
| | - Peng Gao
- Department of Radiology, Huashan Hospital North, Fudan University, China
| | - Jiabin Su
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Heng Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Xinjie Gao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Wei Ni
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Yu Lei
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Yuxiang Gu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, China
| |
Collapse
|
20
|
Liu J, Gu Y, Guo M, Ji X. Neuroprotective effects and mechanisms of ischemic/hypoxic preconditioning on neurological diseases. CNS Neurosci Ther 2021; 27:869-882. [PMID: 34237192 PMCID: PMC8265941 DOI: 10.1111/cns.13642] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
As the organ with the highest demand for oxygen, the brain has a poor tolerance to ischemia and hypoxia. Despite severe ischemia/hypoxia induces the occurrence and development of various central nervous system (CNS) diseases, sublethal insult may induce strong protection against subsequent fatal injuries by improving tolerance. Searching for potential measures to improve brain ischemic/hypoxic is of great significance for treatment of ischemia/hypoxia related CNS diseases. Ischemic/hypoxic preconditioning (I/HPC) refers to the approach to give the body a short period of mild ischemic/hypoxic stimulus which can significantly improve the body's tolerance to subsequent more severe ischemia/hypoxia event. It has been extensively studied and been considered as an effective therapeutic strategy in CNS diseases. Its protective mechanisms involved multiple processes, such as activation of hypoxia signaling pathways, anti-inflammation, antioxidant stress, and autophagy induction, etc. As a strategy to induce endogenous neuroprotection, I/HPC has attracted extensive attention and become one of the research frontiers and hotspots in the field of neurotherapy. In this review, we discuss the basic and clinical research progress of I/HPC on CNS diseases, and summarize its mechanisms. Furthermore, we highlight the limitations and challenges of their translation from basic research to clinical application.
Collapse
Affiliation(s)
- Jia Liu
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yakun Gu
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Mengyuan Guo
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China.,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Burtscher J, Mallet RT, Burtscher M, Millet GP. Hypoxia and brain aging: Neurodegeneration or neuroprotection? Ageing Res Rev 2021; 68:101343. [PMID: 33862277 DOI: 10.1016/j.arr.2021.101343] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022]
Abstract
The absolute reliance of the mammalian brain on oxygen to generate ATP renders it acutely vulnerable to hypoxia, whether at high altitude or in clinical settings of anemia or pulmonary disease. Hypoxia is pivotal to the pathogeneses of myriad neurological disorders, including Alzheimer's, Parkinson's and other age-related neurodegenerative diseases. Conversely, reduced environmental oxygen, e.g. sojourns or residing at high altitudes, may impart favorable effects on aging and mortality. Moreover, controlled hypoxia exposure may represent a treatment strategy for age-related neurological disorders. This review discusses evidence of hypoxia's beneficial vs. detrimental impacts on the aging brain and the molecular mechanisms that mediate these divergent effects. It draws upon an extensive literature search on the effects of hypoxia/altitude on brain aging, and detailed analysis of all identified studies directly comparing brain responses to hypoxia in young vs. aged humans or rodents. Special attention is directed toward the risks vs. benefits of hypoxia exposure to the elderly, and potential therapeutic applications of hypoxia for neurodegenerative diseases. Finally, important questions for future research are discussed.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland; Institute of Sport Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland.
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland
| |
Collapse
|
22
|
Ryou MG, Chen X, Cai M, Wang H, Jung ME, Metzger DB, Mallet RT, Shi X. Intermittent Hypoxia Training Prevents Deficient Learning-Memory Behavior in Mice Modeling Alzheimer's Disease: A Pilot Study. Front Aging Neurosci 2021; 13:674688. [PMID: 34276338 PMCID: PMC8282412 DOI: 10.3389/fnagi.2021.674688] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
In mouse models of Alzheimer's disease (AD), normobaric intermittent hypoxia training (IHT) can preserve neurobehavioral function when applied before deficits develop, but IHT's effectiveness after onset of amyloid-β (Aβ) accumulation is unclear. This study tested the hypothesis that IHT improves learning-memory behavior, diminishes Aβ accumulation in cerebral cortex and hippocampus, and enhances cerebrocortical contents of the neuroprotective trophic factors erythropoietin and brain-derived neurotrophic factor (BDNF) in mice manifesting AD traits. Twelve-month-old female 3xTg-AD mice were assigned to untreated 3xTg-AD (n = 6), AD+IHT (n = 6), and AD+sham-IHT (n = 6) groups; 8 untreated wild-type (WT) mice also were studied. AD+IHT mice alternately breathed 10% O2 for 6 min and room air for 4 min, 10 cycles/day for 21 days; AD+sham-IHT mice breathed room air. Spatial learning-memory was assessed by Morris water maze. Cerebrocortical and hippocampal Aβ40 and Aβ42 contents were determined by ELISA, and cerebrocortical erythropoietin and BDNF were analyzed by immunoblotting and ELISA. The significance of time (12 vs. 12 months + 21 days) and treatment (IHT vs. sham-IHT) was evaluated by two-factor ANOVA. The change in swimming distance to find the water maze platform after 21 d IHT (-1.6 ± 1.8 m) differed from that after sham-IHT (+5.8 ± 2.6 m). Cerebrocortical and hippocampal Aβ42 contents were greater in 3xTg-AD than WT mice, but neither time nor treatment significantly affected Aβ40 or Aβ42 contents in the 3xTg-AD mice. Cerebrocortical erythropoietin and BDNF contents increased appreciably after IHT as compared to untreated 3xTg-AD and AD+sham-IHT mice. In conclusion, moderate, normobaric IHT prevented spatial learning-memory decline and restored cerebrocortical erythropoietin and BDNF contents despite ongoing Aβ accumulation in 3xTg-AD mice.
Collapse
Affiliation(s)
- Myoung-Gwi Ryou
- Department of Medical Laboratory Science and Public Health, Tarleton State University, Texas A&M University System, Stephenville, TX, United States
| | - Xiaoan Chen
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- College of Sports Science, Jishou University, Jishou, China
| | - Ming Cai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hong Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Marianna E. Jung
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Daniel B. Metzger
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Robert T. Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Xiangrong Shi
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
23
|
Ingensiep C, Schaffrath K, Denecke B, Walter P, Johnen S. A multielectrode array-based hypoxia model for the analysis of electrical activity in murine retinae. J Neurosci Res 2021; 99:2172-2187. [PMID: 34110645 DOI: 10.1002/jnr.24899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/14/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022]
Abstract
Several eye diseases, for example, retinal artery occlusion, diabetic retinopathy, and glaucoma, are associated with retinal hypoxia. The lack of oxygen in the retina, especially in retinal ganglion cells (RGCs), causes cell damage up to cell degeneration and leads to blindness. Using multielectrode array recordings, an ex vivo hypoxia acute model was established to analyze the electrical activity of murine wild-type retinae under hypoxic stress conditions. Hypoxia was induced by exchanging the perfusion with oxygen-saturated medium by nitrogen-saturated medium. Hypoxic periods of 0 min (control) up to 60 min were tested on the retinae of adult female C57BL/6J mice. The electrical RGC activity vanished during hypoxia, but conditionally returned after the reestablishment of conventional test conditions. With increasing duration of hypoxia, the returning RGC activity decreased. After a hypoxic period of 30 min and a subsequent recovery time of 30 min, 59.43 ± 11.35% of the initially active channels showed a restored RGC activity. The survival rate of retinal cells after hypoxic stress was analyzed by a live/dead staining assay using two-photon laser scanning microscopy. For detailed information about molecular changes caused by hypoxia, a microarray gene expression analysis was performed. Furthermore, the effect of 2-aminoethanesulfonic acid (taurine, 1 mM) on retinae under hypoxic stress was tested. Treatment with taurine resulted in an increase in the RGC response rate after hypoxia and also increased the survival rate of retinal cells under hypoxic stress, confirming its potential as promising candidate for neuroprotective therapies of eye diseases.
Collapse
Affiliation(s)
- Claudia Ingensiep
- Department of Ophthalmology, University Hospital RWTH Aachen, Aachen, Germany
| | - Kim Schaffrath
- Department of Ophthalmology, University Hospital RWTH Aachen, Aachen, Germany
| | - Bernd Denecke
- Genomics Facility, Interdisciplinary Center for Clinical Research (IZKF), University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Walter
- Department of Ophthalmology, University Hospital RWTH Aachen, Aachen, Germany
| | - Sandra Johnen
- Department of Ophthalmology, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
24
|
Mallet RT, Burtscher J, Richalet JP, Millet GP, Burtscher M. Impact of High Altitude on Cardiovascular Health: Current Perspectives. Vasc Health Risk Manag 2021; 17:317-335. [PMID: 34135590 PMCID: PMC8197622 DOI: 10.2147/vhrm.s294121] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Globally, about 400 million people reside at terrestrial altitudes above 1500 m, and more than 100 million lowlanders visit mountainous areas above 2500 m annually. The interactions between the low barometric pressure and partial pressure of O2, climate, individual genetic, lifestyle and socio-economic factors, as well as adaptation and acclimatization processes at high elevations are extremely complex. It is challenging to decipher the effects of these myriad factors on the cardiovascular health in high altitude residents, and even more so in those ascending to high altitudes with or without preexisting diseases. This review aims to interpret epidemiological observations in high-altitude populations; present and discuss cardiovascular responses to acute and subacute high-altitude exposure in general and more specifically in people with preexisting cardiovascular diseases; the relations between cardiovascular pathologies and neurodegenerative diseases at altitude; the effects of high-altitude exercise; and the putative cardioprotective mechanisms of hypobaric hypoxia.
Collapse
Affiliation(s)
- Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
- Institute of Sport Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
| | - Jean-Paul Richalet
- Laboratoire Hypoxie & Poumon, UMR Inserm U1272, Université Sorbonne Paris Nord 13, Bobigny Cedex, F-93017, France
| | - Gregoire P Millet
- Department of Biomedical Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
- Institute of Sport Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, A-6020, Austria
- Austrian Society for Alpine and High-Altitude Medicine, Mieming, Austria
| |
Collapse
|
25
|
Hypoxia Inducible Factor-1α Attenuates Ischemic Brain Damage by Modulating Inflammatory Response and Glial Activity. Cells 2021; 10:cells10061359. [PMID: 34205911 PMCID: PMC8229365 DOI: 10.3390/cells10061359] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia-inducible factor 1 can sufficiently control the progress of neurological symptoms after ischemic stroke owing to their actions associated with its downstream genes. In this study, we evaluated the role of HIF-1α in attenuating brain damage after endothelin-1 injection. Focal cerebral ischemia in mice were induced by endothelin-1 microinjection. Hypoxia-inducible factor 1 activator, dimethyloxalylglycine (DMOG), and HIF-1α inhibitor, acriflavine (ACF), were used to evaluate the hypoxia-inducible factor 1 activity during cerebral ischemia. The expression levels of HIF-1α, glial fibrillary acidic protein (GFAP), interleukin-10 (IL-10), inducible nitric oxide synthase (iNOS), phosphorylated I-kappa-B-alpha/total I-kappa-B-alpha (p-IκBα/IκBα) and nuclear factor kappa B (NF-kB) were assessed. Besides, mRNA levels of IL-10, tumor necrosis factor- alpha (TNF-α), and NF-kB were also analyzed. Results showed a noticeable increase in hypoxia-inducible factor 1 and IL-10 levels in the DMOG group with a decline in iNOS, TNF-α, and NF-kB levels, implying the anti-inflammatory role of hypoxia-inducible factor 1 activator following stroke. These findings were further corroborated by GFAP immunostaining that showed astrocytic activation to be inhibited 12 days post-ischemia, as well as histological and TEM analyses that demonstrated hypoxia-inducible factor 1 induction to alleviate neuronal soma damage and cell death. Based on our study, HIF-1α could be a potential therapeutic target for ischemic stroke.
Collapse
|
26
|
Lee J, Walter MF, Korach KS, Noguchi CT. Erythropoietin reduces fat mass in female mice lacking estrogen receptor alpha. Mol Metab 2020; 45:101142. [PMID: 33309599 PMCID: PMC7809438 DOI: 10.1016/j.molmet.2020.101142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/23/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
Objective Erythropoietin (EPO), the cytokine required for erythropoiesis, contributes to metabolic regulation of fat mass and glycemic control. EPO treatment in mice on high-fat diets (HFD) improved glucose tolerance and decreased body weight gain via reduced fat mass in males and ovariectomized females. The decreased fat accumulation with EPO treatment during HFD in ovariectomized females was abrogated with estradiol supplementation, providing evidence for estrogen-related gender-specific EPO action in metabolic regulation. In this study, we examined the cross-talk between estrogen mediated through estrogen receptor α (ERα) and EPO for the regulation of glucose metabolism and fat mass accumulation. Methods Wild-type (WT) mice and mouse models with ERα knockout (ERα−/−) and targeted deletion of ERα in adipose tissue (ERαadipoKO) were used to examine EPO treatment during high-fat diet feeding and after diet-induced obesity. Results ERα−/− mice on HFD exhibited increased fat mass and glucose intolerance. EPO treatment on HFD reduced fat accumulation in male WT and ERα−/− mice and female ERα−/− mice but not female WT mice. EPO reduced HFD increase in adipocyte size in WT mice but not in mice with deletion of ERα independent of EPO-stimulated reduction in fat mass. EPO treatment also improved glucose and insulin tolerance significantly greater in female ERα−/− mice and female ERαadipoKO compared with WT controls. Increased metabolic activity by EPO was associated with browning of white adipocytes as shown by reductions in white fat-associated genes and induction of brown fat-specific uncoupling protein 1 (UCP1). Conclusions This study clearly identified the role of estrogen signaling in modifying EPO regulation of glucose metabolism and the sex-differential EPO effect on fat mass regulation. Cross-talk between EPO and estrogen was implicated for metabolic homeostasis and regulation of body mass in female mice. Erythropoietin regulates fat mass in male but not female mice on high-fat diets. Female estrogen receptor alpha deletion restores erythropoietin fat mass regulation. Estrogen receptor alpha deletion increases erythropoietin regulation of glucose tolerance. Erythropoietin reduced white fat-associated genes and increased uncoupling protein 1. Erythropoietin and estrogen cross-talk is implicated for metabolic homeostasis.
Collapse
Affiliation(s)
- Jeeyoung Lee
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mary F Walter
- Clinical Laboratory Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth S Korach
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Constance Tom Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
27
|
Ghaleb AM, Ramadan MZ, Badwelan A, Mansour L, Al-Tamimi J, Aljaloud KS. Determining the time needed for workers to acclimatize to hypoxia. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2020; 64:1995-2005. [PMID: 32794023 DOI: 10.1007/s00484-020-01989-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 06/11/2023]
Abstract
This study aimed to determine the influence of intermittent hypoxia and the days required for a worker to be acclimatized in high-altitude countries. We conducted an experimental study. Ten nonsmoking male students were randomly recruited from King Saud University. Fourteen days of exposure to intermittent normobaric hypoxia (15%) was the independent variable. Heart rate (HR), respiratory frequency (RF), minute ventilation (VE), respiratory exchange ratio (RER), tidal volume (VT), oxygen uptake (VO2),VO2/kg, VO2/HR, VE/VO2, and VE/VCO2 were the dependent variables. Our results showed that 12 days of exposure to intermittent hypoxia were sufficient for workers to acclimatize to hypoxia based on their respiratory responses (i.e., HR, RF, VE). This type of acclimatization session is very important for workers who are suddenly required to work in such an environment, because prolonged exposure to high altitude without acclimatization leads to cell death due to a lack of oxygen, and this, in turn, puts workers' lives at risk.
Collapse
Affiliation(s)
- Atef M Ghaleb
- Department of Industrial Engineering, College of Engineering, King Saud University, Riyadh, Saudi Arabia.
| | - Mohamed Z Ramadan
- Department of Industrial Engineering, College of Engineering, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Badwelan
- Department of Industrial Engineering, College of Engineering, King Saud University, Riyadh, Saudi Arabia
| | - Lamjed Mansour
- Department of Zoology , College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Jameel Al-Tamimi
- Department of Zoology , College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Khalid Saad Aljaloud
- Department of Exercise Physiology, College of Sport Sciences & Physical Activity, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Jarero-Basulto JJ, Rivera-Cervantes MC, Gasca-Martínez D, García-Sierra F, Gasca-Martínez Y, Beas-Zárate C. Current Evidence on the Protective Effects of Recombinant Human Erythropoietin and Its Molecular Variants against Pathological Hallmarks of Alzheimer's Disease. Pharmaceuticals (Basel) 2020; 13:ph13120424. [PMID: 33255969 PMCID: PMC7760199 DOI: 10.3390/ph13120424] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Substantial evidence in the literature demonstrates the pleiotropic effects of the administration of recombinant human erythropoietin (rhEPO) and its molecular variants in different tissues and organs, including the brain. Some of these reports suggest that the chemical properties of this molecule by itself or in combination with other agents (e.g., growth factors) could provide the necessary pharmacological characteristics to be considered a potential protective agent in neurological disorders such as Alzheimer’s disease (AD). AD is a degenerative disorder of the brain, characterized by an aberrant accumulation of amyloid β (Aβ) and hyperphosphorylated tau (tau-p) proteins in the extracellular and intracellular space, respectively, leading to inflammation, oxidative stress, excitotoxicity, and other neuronal alterations that compromise cell viability, causing neurodegeneration in the hippocampus and the cerebral cortex. Unfortunately, to date, it lacks an effective therapeutic strategy for its treatment. Therefore, in this review, we analyze the evidence regarding the effects of exogenous EPOs (rhEPO and its molecular variants) in several in vivo and in vitro Aβ and tau-p models of AD-type neurodegeneration, to be considered as an alternative protective treatment to this condition. Particularly, we focus on analyzing the differential effect of molecular variants of rhEPO when changes in doses, route of administration, duration of treatment or application times, are evaluated for the improved cellular alterations generated in this disease. This narrative review shows the evidence of the effectiveness of the exogenous EPOs as potential therapeutic molecules, focused on the mechanisms that establish cellular damage and clinical manifestation in the AD.
Collapse
Affiliation(s)
- José J. Jarero-Basulto
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, Zapopan 45220, Mexico
- Correspondence: (J.J.J.-B.); (M.C.R.-C.); Tel.: +52-33-37771150 ((J.J.J.-B. & M.C.R.-C.)
| | - Martha C. Rivera-Cervantes
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, Zapopan 45220, Mexico
- Correspondence: (J.J.J.-B.); (M.C.R.-C.); Tel.: +52-33-37771150 ((J.J.J.-B. & M.C.R.-C.)
| | - Deisy Gasca-Martínez
- Behavioral Analysis Unit, Neurobiology Institute, Campus UNAM-Juriquilla, Querétaro 76230, Mexico;
| | - Francisco García-Sierra
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Ciudad de Mexico 07360, Mexico;
| | - Yadira Gasca-Martínez
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, Zapopan 45220, Mexico; (Y.G.-M.); (C.B.-Z.)
| | - Carlos Beas-Zárate
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, Zapopan 45220, Mexico; (Y.G.-M.); (C.B.-Z.)
| |
Collapse
|
29
|
Evaluating the use of hypoxia sensitive markers for body fluid stain age prediction. Sci Justice 2020; 60:547-554. [PMID: 33077038 DOI: 10.1016/j.scijus.2020.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 09/02/2020] [Accepted: 09/05/2020] [Indexed: 01/30/2023]
Abstract
To augment DNA profiling and body fluid identification techniques efforts are being made to increase the amount of information available from a crime scene stain, which includes efforts to identify externally visible characteristics through phenotypic analysis. A key question surrounding crime scene stains is the length of time between deposition of the stain and its subsequent recovery, in that is the stain recovered related to the incident in question or from a previously deposited stain number of weeks earlier? The inability to answer this fundamental question has a detrimental effect upon the successful completion of a criminal investigation. Once a body fluid leaves the body, the oxygen concentration in the environment changes; therefore, it may be that this change could cause a change in the expression of hypoxia-sensitive biomarkers. Here, a range of bloodstains, liquid saliva and liquid semen samples were collected at 0 days, 7 days, 14 days, 21 days and 28 days of degrading at room temperature (19-22 °C), before undergoing total RNA extraction and cDNA synthesis. Blood was recovered from filter paper with 3 mm2, with saliva and semen being left in their tubes and swabbed at the appropriate times. All samples then underwent quantitative PCR targeting Vascular Endothelial Growth Factor A (VEGFA) and Hypoxia-Inducible Factor 1 Alpha (HIF1A), with B-Actin (ACTB) as a reference gene. A range of linear and quadratic correlation values was obtained from the qPCR data and used to develop a predictive model with a mean absolute deviation (MAD) of 4.2, 2.1, and 5 days for blood, saliva, and semen respectively. Blind testing indicated that a stain age prediction model based upon VEGFA with ACTB as a reference gene could be used on samples up to four weeks old with a margin of error ranging from 2 days through to 5 days. While a sizeable potential time frame exists using this model; this represents a significant step towards the target of having an accurate stain age prediction model.
Collapse
|
30
|
Hao Y, Xin M, Feng L, Wang X, Wang X, Ma D, Feng J. Review Cerebral Ischemic Tolerance and Preconditioning: Methods, Mechanisms, Clinical Applications, and Challenges. Front Neurol 2020; 11:812. [PMID: 33071923 PMCID: PMC7530891 DOI: 10.3389/fneur.2020.00812] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke is one of the leading causes of morbidity and mortality worldwide, and it is increasing in prevalence. The limited therapeutic window and potential severe side effects prevent the widespread clinical application of the venous injection of thrombolytic tissue plasminogen activator and thrombectomy, which are regarded as the only approved treatments for acute ischemic stroke. Triggered by various types of mild stressors or stimuli, ischemic preconditioning (IPreC) induces adaptive endogenous tolerance to ischemia/reperfusion (I/R) injury by activating a multitude cascade of biomolecules, for example, proteins, enzymes, receptors, transcription factors, and others, which eventually lead to transcriptional regulation and epigenetic and genomic reprogramming. During the past 30 years, IPreC has been widely studied to confirm its neuroprotection against subsequent I/R injury, mainly including local ischemic preconditioning (LIPreC), remote ischemic preconditioning (RIPreC), and cross preconditioning. Although LIPreC has a strong neuroprotective effect, the clinical application of IPreC for subsequent cerebral ischemia is difficult. There are two main reasons for the above result: Cerebral ischemia is unpredictable, and LIPreC is also capable of inducing unexpected injury with only minor differences to durations or intensity. RIPreC and pharmacological preconditioning, an easy-to-use and non-invasive therapy, can be performed in a variety of clinical settings and appear to be more suitable for the clinical management of ischemic stroke. Hoping to advance our understanding of IPreC, this review mainly focuses on recent advances in IPreC in stroke management, its challenges, and the potential study directions.
Collapse
Affiliation(s)
| | | | | | | | | | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Chou PL, Chen KH, Chang TC, Chien CT. Repetitively hypoxic preconditioning attenuates ischemia/reperfusion-induced liver dysfunction through upregulation of hypoxia-induced factor-1 alpha-dependent mitochondrial Bcl-xl in rat. CHINESE J PHYSIOL 2020; 63:68-76. [PMID: 32341232 DOI: 10.4103/cjp.cjp_74_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Repetitive hypoxic preconditioning (HP) enforces protective effects to subsequently severe hypoxic/ischemic stress. We hypothesized that HP may provide protection against ischemia/reperfusion (I/R) injury in rat livers via hypoxia-induced factor-1 alpha (HIF-1α)/reactive oxygen species (ROS)-dependent defensive mechanisms. Female Wistar rats were exposed to hypoxia (15 h/day) in a hypobaric hypoxic chamber (5500 m) for HP induction, whereas the others were kept in sea level. These rats were subjected to 45 min of hepatic ischemia by portal vein occlusion followed by 6 h of reperfusion. We evaluated HIF-1α in nuclear extracts, MnSOD, CuZnSOD, catalase, Bad/Bcl-xL/caspase 3/poly-(ADP-ribose)-polymerase (PARP), mitochondrial Bcl-xL, and cytosolic cytochrome C expression with Western blot and nitroblue tetrazolium/3-nitrotyrosine stain. Kupffer cell infiltration and terminal deoxynucleotidyl transferase-mediated nick-end labeling method apoptosis were determined by immunocytochemistry. The ROS value from liver surface and bile was detected by an ultrasensitive chemiluminescence-amplification method. Hepatic function was assessed with plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. HP increased nuclear translocation of HIF-1α and enhanced Bcl-xL, MnSOD, CuZnSOD, and catalase protein expression in a time-dependent manner. The response of HP enhanced hepatic HIF-1α, and Bcl-xL expression was abrogated by a HIF-1α inhibitor YC-1. Hepatic I/R increased ROS levels, myeloperoxidase activity, Kupffer cell infiltration, ALT and AST levels associated with the enhancement of cytosolic Bad translocation to mitochondria, release of cytochrome C to cytosol, and activation of caspase 3/PARP-mediated apoptosis. HP significantly ameliorated hepatic I/R-enhanced oxidative stress, apoptosis, and mitochondrial and hepatic dysfunction. In summary, HP enhances HIF-1α/ROS-dependent cascades to upregulate mitochondrial Bcl-xL protein expression and to confer protection against I/R injury in the livers.
Collapse
Affiliation(s)
- Pei-Lei Chou
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Kuo-Hsin Chen
- Department of Surgery, Division of General Surgery, Far-Eastern Memorial Hospital; Department of Electrical Engineering, Yuan Ze University, Taoyuan City, Taiwan
| | - Tzu-Ching Chang
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chiang-Ting Chien
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
32
|
Wang D, Wang L, Bai L, Du Y, Liu L, Chen X. Effects of Inhibition of miR-155-5p in Neural Stem Cell Subarachnoid Transplant on Rats with Cerebral Infarction. Hum Gene Ther Methods 2020; 30:184-193. [PMID: 31618139 DOI: 10.1089/hgtb.2019.118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cerebral infarction is a leading cause of death, which calls for effective prevention and treatment. Transplant of neural stem cells (NSCs) is a potential therapeutic treatment to cerebral infarction although its efficacy still needs to be improved. Overexpression of hypoxia-inducible factor 1α (HIF-1α) has been shown to enhance the protective effects of stem cell transplant on cerebral infarction. The expression of HIF-1α is predicted to be regulated by miR-155-5p. Therefore, we regulated the expression of miR-155-5p in NSCs and evaluated the effects of miR-155-5p-regulated NSC transplant on cerebral infarction. We inhibited miR-155-5p expression in NSCs by overexpressing miR-155-5p inhibitor. HIF-1α expression, cell viability, and the expression of apoptosis markers were examined. We established the middle cerebral artery occlusion (MCAO) rat model, and the infarct volume, neurobehavioral outcomes, inflammation, and oxidative stress were evaluated after NSC transplant. miR-155-5p directly targeted HIF-1α and negatively regulated its expression. Inhibition of miR-155-5p enhanced cell viability and prevented cell apoptosis. Transplant of miR-155-5p-inhibited NSCs significantly decreased infarct volume and improved neurobehavioral outcomes of MCAO rats. Transplant of miR-155-5p-inhibited NSCs significantly inhibited inflammation and oxidative stress. Inhibition of miR-155-5p in NSCs resulted in enhanced protection against cerebral infarction after NSC transplant.
Collapse
Affiliation(s)
- Dong Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Liang Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lin Bai
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuanyuan Du
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Luji Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang Chen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
33
|
Hurst J, Mueller-Buehl AM, Hofmann L, Kuehn S, Herms F, Schnichels S, Joachim SC. iNOS-inhibitor driven neuroprotection in a porcine retina organ culture model. J Cell Mol Med 2020; 24:4312-4323. [PMID: 32130787 PMCID: PMC7171393 DOI: 10.1111/jcmm.15091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/24/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
Nitrite oxide plays an important role in the pathogenesis of various retinal diseases, especially when hypoxic processes are involved. This degeneration can be simulated by incubating porcine retinal explants with CoCl2. Here, the therapeutic potential of iNOS‐inhibitor 1400W was evaluated. Degeneration through CoCl2 and treatment with the 1400W were applied simultaneously to porcine retinae explants. Three groups were compared: control, CoCl2, and CoCl2 + iNOS‐inhibitor (1400W). At days 4 and 8, retinal ganglion cells (RGCs), bipolar, and amacrine cells were analysed. Furthermore, the influence on the glia cells and different stress markers were evaluated. Treatment with CoCl2 resulted in a significant loss of RGCs already after 4 days, which was counteracted by the iNOS‐inhibitor. Expression of HIF‐1α and its downstream targets confirmed the effective treatment with 1400W. After 8 days, the CoCl2 group displayed a significant loss in amacrine cells and also a drastic reduction in bipolar cells was observed, which was prevented by 1400W. The decrease in microglia could not be prevented by the inhibitor. CoCl2 induces strong degeneration in porcine retinae by mimicking hypoxia, damaging certain retinal cell types. Treatment with the iNOS‐inhibitor counteracted these effects to some extent, by preventing loss of retinal ganglion and bipolar cells. Hence, this inhibitor seems to be a very promising treatment for retinal diseases.
Collapse
Affiliation(s)
- José Hurst
- Centre for Ophthalmology Tübingen, University Eye Hospital, Tübingen, Germany
| | - Ana Maria Mueller-Buehl
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Lisa Hofmann
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sandra Kuehn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Fenja Herms
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sven Schnichels
- Centre for Ophthalmology Tübingen, University Eye Hospital, Tübingen, Germany
| | | |
Collapse
|
34
|
Wu X, Wang C, Wang J, Zhu M, Yao Y, Liu J. Hypoxia preconditioning protects neuronal cells against traumatic brain injury through stimulation of glucose transport mediated by HIF-1α/GLUTs signaling pathway in rat. Neurosurg Rev 2020; 44:411-422. [PMID: 31897883 PMCID: PMC7851104 DOI: 10.1007/s10143-019-01228-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/04/2019] [Accepted: 12/18/2019] [Indexed: 11/25/2022]
Abstract
Hypoxia preconditioning (HPC), a well-established preconditioning model, has been shown to protect the brain against severe hypoxia or ischemia caused by traumatic brain injury (TBI), but the mechanism has not been well elucidated. Anaerobic glycolysis is the major way for neurons to produce energy under cerebral ischemia and hypoxia after TBI, and it requires large amounts of glucose. We hypothesized that glucose transport, as a rate-limiting step of glucose metabolism, may play key roles in the neuroprotective effects of HPC on cerebral cortex tissue against TBI. The aim of this study was to investigate the effect of HPC on glucose transport activity of rat cerebral cortex tissue after TBI through examining the gene expression of two major glucose transporters (GLUT1 and GLUT3) and their upstream target gene hypoxia-inducible factor-1α (HIF-1α). Sprague-Dawley rats were treated with HPC (50.47 kPa, 3 h/d, 3d). Twenty-four hours after the last treatment, the rats were injured using the Feeney free falling model. Cortex tissues of injured rats were removed at 1 h, 4 h, 8 h, 12 h, 1 day, 3 days, 7 d, and 14 days post-injury for histological analysis. Compared with TBI alone, HPC before TBI resulted in the expression of HIF-1α, GLUT1, and GLUT3 to increase at 1 h; they were markedly increased at 4 h, 8 h, 12 h, 1 day, and 3 days and decreased thereafter (p < 0.05). HPC before TBI could improve neuronal survival in rats by examining NeuN staining and observing reduced apoptosis by examining TUNEL staining. The result showed that HPC before TBI could increase the expression of GLUT1 and GLUT3. And through double immunofluorescence staining for GLUT3 and NeuN, the results strongly suggest that HPC improved glucose transport activity of neurons in rats with TBI. In summary, our results further support that HPC can improve hypoxia tolerance and attenuate neuronal loss of cerebral cortex in rats after TBI. The mechanism is mainly related to the increase of glucose transport activity through inducing GLUT1 and GLUT3 expression through upregulating HIF-1α expression.
Collapse
Affiliation(s)
- Xiaogang Wu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Chunlin Wang
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Jinbiao Wang
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Meijie Zhu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Yinsheng Yao
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Jiachuan Liu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China.
| |
Collapse
|
35
|
Jin ML, Zou ZH, Tao T, Li J, Xu J, Luo KJ, Liu Z. Effect of the Recombinant Adenovirus-Mediated HIF-1 Alpha on the Expression of VEGF in the Hypoxic Brain Microvascular Endothelial Cells of Rats. Neuropsychiatr Dis Treat 2020; 16:397-406. [PMID: 32103959 PMCID: PMC7012637 DOI: 10.2147/ndt.s238616] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To investigate the effect of recombinant adenovirus-mediated HIF-1 alpha (HIF-1α) on the expression of vascular endothelial growth factor (VEGFA) and HIF-1α in hypoxic brain microvascular endothelial cells (BMEC) in rats. METHODS Primary cultured rat BMEC in vitro were treated without or with either recombinant adenovirus-mediated hypoxia-inducible factor-1 alpha (AdHIF-1α) or recombinant adenovirus empty vector (Ad) in the presence of CoCl2 (simulating hypoxia conditions), or were grown under normoxia conditions. The expression of VEGFA and HIF-1α was analyzed at 12h, 24h, 48h and 72h incubation time, respectively. We also accessed a GEO dataset of stroke to analyze in vivo the alteration of HIF-1α and VEGFA expression, and the correlations between HIF-1α, VEGFA and CD31 mRNA levels in vascular vessels after stroke. RESULTS VEGFA and HIF-1α expression were significantly higher in at each time point in the AdHIF-1α than other groups (p<0.05), whereas the Ad group and hypoxia group, showed no statistically significant difference (p>0.05). Moreover, VEGFA and HIF-1α levels were significantly higher in BMEC under hypoxia conditions than normoxia conditions (p <0.05). Both HIF-1α and VEGFA expression significantly increased after stroke in vivo with 1.30 and 1.57 fold-change in log2, respectively. There were significantly positive associations between HIF-1α, VEGFA and CD31 mRNA levels in vivo after stroke. CONCLUSION Hypoxia-induced HIF-1α and VEGFA expression in vascular vessels, and recombinant AdHIF-1α could up-regulate VEGFA, and enhance HIF-1ααlevels in BMEC in vitro, which may play an important role in the recovery of stroke.
Collapse
Affiliation(s)
- Ming-Lu Jin
- Department of Rehabilitation Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, People's Republic of China.,Department of Neurology, Qijiang Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing 404100, People's Republic of China.,Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, People's Republic of China
| | - Zhe-Hua Zou
- Department of Rehabilitation Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, People's Republic of China.,Department of General Practice, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, People's Republic of China
| | - Tao Tao
- Department of Rehabilitation Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, People's Republic of China
| | - Jun Li
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, People's Republic of China.,Department of Neurology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, People's Republic of China
| | - Jian Xu
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, People's Republic of China
| | - Kai-Jian Luo
- Guizhou Cancer Hospital, Guiyang, Guizhou 550000, People's Republic of China
| | - Zhi Liu
- Department of Pharmacy, The Affiliated Hospital Guizhou Medical University, Guiyang, Guizhou 550001, People's Republic of China
| |
Collapse
|
36
|
Wang H, Shi X, Schenck H, Hall JR, Ross SE, Kline GP, Chen S, Mallet RT, Chen P. Intermittent Hypoxia Training for Treating Mild Cognitive Impairment: A Pilot Study. Am J Alzheimers Dis Other Demen 2020; 35:1533317519896725. [PMID: 31902230 PMCID: PMC10624018 DOI: 10.1177/1533317519896725] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although intermittent hypoxia training (IHT) has proven effective against various clinical disorders, its impact on mild cognitive impairment (MCI) is unknown. This pilot study examined IHT's safety and therapeutic efficacy in elderly patients with amnestic MCI (aMCI). Seven patients with aMCI (age 69 ± 3 years) alternately breathed 10% O2 and room-air, each 5 minutes, for 8 cycles/session, 3 sessions/wk for 8 weeks. The patients' resting arterial pressures fell by 5 to 7 mm Hg (P < .05) and cerebral tissue oxygenation increased (P < .05) following IHT. Intermittent hypoxia training enhanced hypoxemia-induced cerebral vasodilation (P < .05) and improved mini-mental state examination and digit span scores from 25.7 ± 0.4 to 27.7 ± 0.6 (P = .038) and from 24.7 ± 1.2 to 26.1 ± 1.3 (P = .047), respectively. California verbal learning test score tended to increase (P = .102), but trail making test-B and controlled oral word association test scores were unchanged. Adaptation to moderate IHT may enhance cerebral oxygenation and hypoxia-induced cerebrovasodilation while improving short-term memory and attention in elderly patients with aMCI.
Collapse
Affiliation(s)
- Hong Wang
- Departments of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
- Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xiangrong Shi
- Departments of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Hannah Schenck
- Departments of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James R. Hall
- Departments of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sarah E. Ross
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Geoffrey P. Kline
- Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Shande Chen
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Robert T. Mallet
- Departments of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Peijie Chen
- Shanghai University of Sport, Shanghai, China
| |
Collapse
|
37
|
Asaghiar F, Williams G. Evaluating the use of hypoxia sensitive markers for body fluid stain age prediction. FORENSIC SCIENCE INTERNATIONAL GENETICS SUPPLEMENT SERIES 2019. [DOI: 10.1016/j.fsigss.2019.10.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
38
|
Kreisman NR, Wooliscroft LB, Campbell CF, Dotiwala AK, Cox ML, Denson AC, Betancourt AM, Tomchuck SL. Preconditioning hippocampal slices with hypothermia promotes rapid tolerance to hypoxic depolarization and swelling: Mediation by erythropoietin. Brain Res 2019; 1726:146517. [PMID: 31634451 DOI: 10.1016/j.brainres.2019.146517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/06/2019] [Accepted: 10/17/2019] [Indexed: 12/26/2022]
Abstract
We suggested previously that hippocampal slices were protected from hypoxic depolarization and swelling by preincubating them at room temperature (Kreisman et al., 2000). We postulated that hypothermic preconditioning induced tolerance in our slices, which protected against hypoxic depolarization and swelling. Control hippocampal slices were incubated at 34-35 °C for two hours and the response to 10 min of severe hypoxia was compared to slices which were preconditioned for two hours at room temperature (22-23 °C) prior to warming to 34-35 °C. Recordings of the extracellular DC potential provided an index of tissue depolarization and changes in tissue light transmittance provided an index of swelling. Hypothermic preconditioning significantly reduced hypoxia-induced swelling, particularly in CA3 and the dentate inner blade. Since erythropoietin (EPO) had been shown to mediate hypoxic preconditioning, we tested whether EPO also mediated hypothermic preconditioning in our slices. Recombinant rat EPO (1-10 micromolar) mitigated hypoxia-induced swelling and depolarization in dentate inner blade of unconditioned slices in a dose-dependent manner. We also blocked the protective effects of hypothermic preconditioning on hypoxic depolarization and swelling in the inner blade of the dentate gyrus by administering soluble EPO receptor in the bath and treating slices with wortmannin to block phosphorylation of PI3 kinase, a critical step in the activation of the downstream neuroprotectant, Akt. These results suggest that EPO mediates tolerance to hypoxic depolarization and swelling induced by hypothermic preconditioning. They also emphasize that various preincubation protocols used in experiments with hippocampal slices may differentially affect basal electrophysiological and metabolic properties of those slices.
Collapse
Affiliation(s)
- Norman R Kreisman
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States; Tulane Brain Institute, New Orleans, LA 70118, United States.
| | | | - Carolyn F Campbell
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States; Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Ary K Dotiwala
- Tulane Brain Institute, New Orleans, LA 70118, United States
| | - Michael L Cox
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Aaron C Denson
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Aline M Betancourt
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Suzanne L Tomchuck
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| |
Collapse
|
39
|
Wang D, Wang L, Bai L, Du Y, Liu L, Chen X. Effects of inhibition of miR-155-5p in neural stem cell subarachnoid transplant on rats with cerebral infarction. Hum Gene Ther Methods 2019. [DOI: 10.1089/hum.2019.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Dong Wang
- Second Hospital of Hebei Medical University, 71213, the Second Hospital of Hebei Medical University, Shijiazhuang, China, 050000
| | - Liang Wang
- Second Hospital of Hebei Medical University, 71213, Shijiazhuang, Hebei, China
| | - Lin Bai
- Second Hospital of Hebei Medical University, 71213, Shijiazhuang, Hebei, China
| | - Yuanyuan Du
- Second Hospital of Hebei Medical University, 71213, Shijiazhuang, Hebei, China
| | - Luji Liu
- Second Hospital of Hebei Medical University, 71213, Shijiazhuang, Hebei, China
| | - Xiang Chen
- Second Hospital of Hebei Medical University, 71213, Shijiazhuang, Hebei, China
| |
Collapse
|
40
|
Wang H, Niu F, Fan W, Shi J, Zhang J, Li B. Modulating effects of preconditioning exercise in the expression of ET-1 and BNP via HIF-1α in ischemically injured brain. Metab Brain Dis 2019; 34:1299-1311. [PMID: 31222402 DOI: 10.1007/s11011-019-00450-z] [Citation(s) in RCA: 12] [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: 12/20/2018] [Accepted: 06/10/2019] [Indexed: 12/12/2022]
Abstract
It is well-known that in ischemia-induced hypoxia, hypoxia-inducible factor -1α (HIF-1α) is critical in triggering expression of its downstream target genes to produce several products, such as erythropoietin (EPO), vascular endothelial growth factor (VEGF), nitric oxide synthesis (NOS), glucose transportor-1 (GLUT-1), insulin-like growth factor (IGF), which further promote erythropoiesis, angiogenesis, vasodilation and capitalization of glucose to overcome hypoxia. Meanwhile, as the factors with opposite effects on blood vessels, endothelin-1 (ET-1) and brain natriuretic peptide (BNP) also stand out strikingly in ischemic pathophysiology. To this day, several preconditioning manners have been used to induce tolerance to ischemia. During our research, exercise preconditioning was applied and it was demonstrated that HIF-1α triggered expression of ET-1 and BNP, which confirmed their downstream target genes for HIF-1α. And ET-1 may influcence expression of BNP to some degree but not the only factor which regulates BNP expression. Therefore, our findings suggest exercise preconditioning may provide protection to the ischemic brain tissue via HIF-1α which in turn increases expression of BNP to cause vasodilation in cooperation with some other factors, such as VEGF and EPO, to increase the blood flow in the ischemic area and then relieve the injuries induced by ischemia.
Collapse
Affiliation(s)
- Huijie Wang
- Department of Histology& Embryology, Basic Medicine School, Dali University, Yunnan, China
| | - Feng Niu
- Department of Rehabilitation, Jinshan Hospital, Fudan University, Shanghai, China
| | - Wei Fan
- Central Laboratory, Jinshan Hospital, Fudan University, No.1508, Longhang Road, Jinshan district, Shanghai, 201508, China
| | - Jimin Shi
- Central Laboratory, Jinshan Hospital, Fudan University, No.1508, Longhang Road, Jinshan district, Shanghai, 201508, China
| | - Jihong Zhang
- Central Laboratory, Jinshan Hospital, Fudan University, No.1508, Longhang Road, Jinshan district, Shanghai, 201508, China
| | - Bing Li
- Central Laboratory, Jinshan Hospital, Fudan University, No.1508, Longhang Road, Jinshan district, Shanghai, 201508, China.
| |
Collapse
|
41
|
Tayyari F, Khuu L, Sivak JM, Flanagan JG, Singer S, Brent MH, Hudson C. Retinal blood oxygen saturation and aqueous humour biomarkers in early diabetic retinopathy. Acta Ophthalmol 2019; 97:e673-e679. [PMID: 30690929 DOI: 10.1111/aos.14016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE The aim of this study was to assess the relationship between retinal blood oxygen saturation (SO2 ) and specific aqueous humour (AH) concentrations of proangiogenic biomarkers in diabetic patients with nonproliferative diabetic retinopathy (NPDR) and to compare them with those of matched control subjects. METHODS The sample comprised 14 participants with mild-to-moderate NPDR (69.1 ± 6.6 years) and 17 age-matched healthy controls (69.7 ± 6.3 years); all participants were previously scheduled for routine cataract extraction with intraocular lens implantation. Multiplex cytokine analyses of specific biomarkers, including vascular endothelial growth factor A (VEGF-A), angiopoietin2 (Ang2), epidermal growth factor (EGF), hepatocyte growth factor (HGF) and interleukin-8 (IL-8) were performed by BioPlex 200 system. Six non-invasive hyperspectral retinal images were acquired. RESULTS Mean SO2 was significantly higher in both arterioles (94.4 ± 1.9 versus 93.0 ± 1.6) and venules (64.4 ± 5.6 versus 55.9 ± 4.8) of NPDR than in the healthy controls (p < 0.001). AH levels of HGF (p = 0.018), Ang2 (p = 0.005) and IL-8 (p = 0.034) were significantly higher, and EGF (p = 0.030) was significantly lower in NPDR subjects. The study demonstrated a correlation between venular retinal blood oxygen saturation and proangiogenic factors HGF (r = 0.558, p = 0.038), Ang2 (r = 0.556, p = 0.039) and EGF (r = -0.554, p = 0.040), but did not find any correlation for IL-8 (r = 0.330, p = 0.249) even though this biomarker was significantly higher in the diabetic group. CONCLUSION To our knowledge, the present study is the first report considering the association between SO2 and AH concentrations of protein biomarkers in diabetic retinopathy. The biomarkers of interest have been shown to participate in cell death, which may explain higher oxygen saturation in NPDR.
Collapse
Affiliation(s)
- Faryan Tayyari
- Retina Research Group School of Optometry and Vision Science University of Waterloo Waterloo Ontario Canada
- Department of Ophthalmology and Vision Sciences University of Toronto Toronto Ontario Canada
| | - Lee‐Anne Khuu
- Department of Ophthalmology and Vision Sciences University of Toronto Toronto Ontario Canada
| | - Jeremy M. Sivak
- Department of Ophthalmology and Vision Sciences University of Toronto Toronto Ontario Canada
| | - John G. Flanagan
- Retina Research Group School of Optometry and Vision Science University of Waterloo Waterloo Ontario Canada
- Department of Ophthalmology and Vision Sciences University of Toronto Toronto Ontario Canada
| | - Shaun Singer
- Department of Ophthalmology and Vision Sciences University of Toronto Toronto Ontario Canada
| | - Michael H. Brent
- Department of Ophthalmology and Vision Sciences University of Toronto Toronto Ontario Canada
| | - Christopher Hudson
- Retina Research Group School of Optometry and Vision Science University of Waterloo Waterloo Ontario Canada
- Department of Ophthalmology and Vision Sciences University of Toronto Toronto Ontario Canada
| |
Collapse
|
42
|
Sarieva KV, Lyanguzov AY, Galkina OV, Vetrovoy OV. The Effect of Severe Hypoxia on HIF1- and Nrf2-Mediated Mechanisms of Antioxidant Defense in the Rat Neocortex. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419020107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Acun A, Zorlutuna P. CRISPR/Cas9 Edited Induced Pluripotent Stem Cell-Based Vascular Tissues to Model Aging and Disease-Dependent Impairment. Tissue Eng Part A 2019; 25:759-772. [PMID: 30704346 DOI: 10.1089/ten.tea.2018.0271] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPACT STATEMENT Modeling human disease as precisely as possible is of upmost importance in understanding the underlying pathology and discovering effective therapies. Therefore, disease models that are highly controlled and composed of human-origin cells that present the disease phenotype are crucial. The human induced pluripotent stem cell (hiPSC)-based tissue model we present in this study is an important example of human-origin tissue model with controlled gene expression. Through CRISPR/Cas9 editing of hypoxia inducible factor 1α in hiPSCs, we developed tissue models that show the age and disease-dependent endothelial deterioration. This model holds promise for various biomedical applications as more realistic disease phenotypes can be created using fully human-origin platforms.
Collapse
Affiliation(s)
- Aylin Acun
- 1 Bioengineering Graduate Program and University of Notre Dame, Notre Dame, Indiana
| | - Pinar Zorlutuna
- 1 Bioengineering Graduate Program and University of Notre Dame, Notre Dame, Indiana.,2 Aerospace and Mechanical Engineering Department, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
44
|
Das KK, Yendigeri SM, Patil BS, Bagoji IB, Reddy RC, Bagali S, Biradar MS, Saha S. Subchronic hypoxia pretreatment on brain pathophysiology in unilateral common carotid artery occluded albino rats. Indian J Pharmacol 2018; 50:185-191. [PMID: 30505054 PMCID: PMC6234715 DOI: 10.4103/ijp.ijp_312_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE: This study was aimed to assess the effect of unilateral common carotid artery occlusion on brain pathophysiology in rats pretreated with subchronic hypoxia. MATERIALS AND METHODS: Rats (200 ± 20 g) were randomized into three groups: Group 1 served as sham, Group 2 were normoxic (21% O2 and 79% N2), and Group 3 were hypoxia preconditioned (10% O2 and 90% N2) for 21 days before left common carotid artery occlusion (LCCAO). The LCCAO was done for 75 min followed by reperfusion for 12 h. Neurological scores were recorded. Serum malondialdehyde (MDA) and nitric oxide (NO) levels at pre- and 12 h post-LCCAO were measured. Brain histopathological assessments were also done. RESULTS: Higher neurological deficits scores in Group 2 as compared to Group 3 rats were noticed. Serum MDA and NO levels at 12 h post-LCCAO in Group 2 rats showed significant elevation as compared to preocclusion levels. Group 3 rats did not show such elevations. On histopathology of left and right cerebral hemispheres of Group 1 (sham) did not show any specific changes. In Group 2 rats, the right cerebral hemisphere (nonoccluded) showed no areas of ischemia-induced brain changes, but in the left side (occlusive), there were features of ischemic brain damage including cerebral edema. In the case of Group 3 rats, there were less ischemic damages in the left occluded side as compared to the left side of the Group 2 rats. CONCLUSION: This study clearly demonstrates that subchronic hypoxia pretreatment can reduce ischemic brain injury by unilateral common carotid artery occlusion in rats.
Collapse
Affiliation(s)
- Kusal K Das
- Department of Physiology, Laboratory of Vascular Physiology and Medicine, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Bijapur, Karnataka, India
| | - Saeed M Yendigeri
- Department of Pathology, Al Ameen Medical College, Bijapur, Karnataka, India
| | - Bheemshetty S Patil
- Department of Anatomy, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Bijapur, Karnataka, India
| | - Ishwar B Bagoji
- Department of Anatomy, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Bijapur, Karnataka, India
| | - R Chandramouli Reddy
- Department of Physiology, Laboratory of Vascular Physiology and Medicine, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Bijapur, Karnataka, India
| | - Shrilaxmi Bagali
- Department of Physiology, Laboratory of Vascular Physiology and Medicine, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Bijapur, Karnataka, India
| | - M S Biradar
- Department of Medicine, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Bijapur, Karnataka, India
| | - Sikha Saha
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
45
|
Späth MR, Bartram MP, Palacio-Escat N, Hoyer KJR, Debes C, Demir F, Schroeter CB, Mandel AM, Grundmann F, Ciarimboli G, Beyer A, Kizhakkedathu JN, Brodesser S, Göbel H, Becker JU, Benzing T, Schermer B, Höhne M, Burst V, Saez-Rodriguez J, Huesgen PF, Müller RU, Rinschen MM. The proteome microenvironment determines the protective effect of preconditioning in cisplatin-induced acute kidney injury. Kidney Int 2018; 95:333-349. [PMID: 30522767 DOI: 10.1016/j.kint.2018.08.037] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 07/24/2018] [Accepted: 08/16/2018] [Indexed: 01/18/2023]
Abstract
Acute kidney injury (AKI) leads to significant morbidity and mortality; unfortunately, strategies to prevent or treat AKI are lacking. In recent years, several preconditioning protocols have been shown to be effective in inducing organ protection in rodent models. Here, we characterized two of these interventions-caloric restriction and hypoxic preconditioning-in a mouse model of cisplatin-induced AKI and investigated the underlying mechanisms by acquisition of multi-layered omic data (transcriptome, proteome, N-degradome) and functional parameters in the same animals. Both preconditioning protocols markedly ameliorated cisplatin-induced loss of kidney function, and caloric restriction also induced lipid synthesis. Bioinformatic analysis revealed mRNA-independent proteome alterations affecting the extracellular space, mitochondria, and transporters. Interestingly, our analyses revealed a strong dissociation of protein and RNA expression after cisplatin treatment that showed a strong correlation with the degree of damage. N-degradomic analysis revealed that most posttranscriptional changes were determined by arginine-specific proteolytic processing. This included a characteristic cisplatin-activated complement signature that was prevented by preconditioning. Amyloid and acute-phase proteins within the cortical parenchyma showed a similar response. Extensive analysis of disease-associated molecular patterns suggested that transcription-independent deposition of amyloid P-component serum protein may be a key component in the microenvironmental contribution to kidney damage. This proof-of-principle study provides new insights into the pathogenesis of cisplatin-induced AKI and the molecular mechanisms underlying organ protection by correlating phenotypic and multi-layered omics data.
Collapse
Affiliation(s)
- Martin R Späth
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Malte P Bartram
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Nicolàs Palacio-Escat
- COMBINE-Joint Research Center for Computational Biomedicine RWTH Aachen University, Aachen, Germany
| | - K Johanna R Hoyer
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Cedric Debes
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Fatih Demir
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
| | - Christina B Schroeter
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Amrei M Mandel
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Franziska Grundmann
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Giuliano Ciarimboli
- Department of Experimental Nephrology, University Hospital of Münster, Münster, Germany
| | - Andreas Beyer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Jayachandran N Kizhakkedathu
- Department of Pathology, Centre for Blood Research, The University of British Columbia, British Columbia, Vancouver, Canada; Laboratory Medicine, Department of Chemistry, The University of British Columbia, British Columbia, Vancouver, Canada
| | - Susanne Brodesser
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Heike Göbel
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Jan U Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Martin Höhne
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Volker Burst
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Julio Saez-Rodriguez
- COMBINE-Joint Research Center for Computational Biomedicine RWTH Aachen University, Aachen, Germany; Faculty of Medicine Bioquant, Institute for Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | - Pitter F Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
| | - Roman-Ulrich Müller
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany.
| | - Markus M Rinschen
- Department II of Internal Medicine, University of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany.
| |
Collapse
|
46
|
Yang X, Zi XH. LncRNA SNHG1 alleviates OGD induced injury in BMEC via miR-338/HIF-1α axis. Brain Res 2018; 1714:174-181. [PMID: 30414401 DOI: 10.1016/j.brainres.2018.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Brain microvascular endothelial cell (BMEC) is an important therapeutic target for the inhibition of brain vascular dysfunction in ischemic stroke. Expression of long non-coding RNA SNHG1 is reportedly upregulated in BMEC after OGD. The present study aims to investigate the potential roles of SNHG1 in OGD-induced injury in BMEC. METHODS Mice primary brain microvascular endothelial cells (BMEC) were cultured under "normal" or "oxygen/glucose-deprived" (OGD) conditions. The expression of SNHG1 and miR-338 after OGD were examined by qPCR. shRNA against SNHG1 was used to knockdown SNHG1 in BMEC. MiR-338-3p mimic and inhibitor were used to change the expression of miR-338 in BMEC. The relationship between SNHG1 and miR-338, and the relationship between miR-338 and HIF-1α were clarified using RNA pull-down and luciferase reporter gene assays, respectively. RESULTS SNHG1 and miR-338 were upregulated in OGD induced BMEC. SNHG1 silence aggravated OGD-induced cell apoptosis by down-regulating Bcl-2, HIF-1α and VEGF-A, and upregulating caspase 3 activity and Bax. MiR-338 was upregulated in SNHG1-silenced BMEC. RNA pull-down assays showed that SNHG1 could be directly bound by miR-338. In addition, miR-338 overexpression reduced cell viability in OGD while miR-338 inhibition protected BMEC against OGD-induced injury. Furthermore, luciferase reporter assay showed that HIF-1α was a direct target of miR-338. CONCLUSIONS SNHG1 exerted protective effects against OGD induced injury via sponging miR-338, thus upregulating HIF-1α/VEGF-A in BMEC.
Collapse
Affiliation(s)
- Xia Yang
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Xiao-Hong Zi
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| |
Collapse
|
47
|
Stone J, Mitrofanis J, Johnstone DM, Falsini B, Bisti S, Adam P, Nuevo AB, George-Weinstein M, Mason R, Eells J. Acquired Resilience: An Evolved System of Tissue Protection in Mammals. Dose Response 2018; 16:1559325818803428. [PMID: 30627064 PMCID: PMC6311597 DOI: 10.1177/1559325818803428] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/22/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022] Open
Abstract
This review brings together observations on the stress-induced regulation of resilience mechanisms in body tissues. It is argued that the stresses that induce tissue resilience in mammals arise from everyday sources: sunlight, food, lack of food, hypoxia and physical stresses. At low levels, these stresses induce an organised protective response in probably all tissues; and, at some higher level, cause tissue destruction. This pattern of response to stress is well known to toxicologists, who have termed it hormesis. The phenotypes of resilience are diverse and reports of stress-induced resilience are to be found in journals of neuroscience, sports medicine, cancer, healthy ageing, dementia, parkinsonism, ophthalmology and more. This diversity makes the proposing of a general concept of induced resilience a significant task, which this review attempts. We suggest that a system of stress-induced tissue resilience has evolved to enhance the survival of animals. By analogy with acquired immunity, we term this system 'acquired resilience'. Evidence is reviewed that acquired resilience, like acquired immunity, fades with age. This fading is, we suggest, a major component of ageing. Understanding of acquired resilience may, we argue, open pathways for the maintenance of good health in the later decades of human life.
Collapse
Affiliation(s)
- Jonathan Stone
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - John Mitrofanis
- Discipline of Anatomy and Histology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Daniel M. Johnstone
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Benedetto Falsini
- Facolta’ di Medicina e Chirurgia, Fondazione Policlinico A. Gemelli, Universita’ Cattolica del S. Cuore, Rome, Italy
| | - Silvia Bisti
- Department of Biotechnical and Applied Clinical Sciences, Università degli Studi dell’Aquila, IIT Istituto Italiano di Tecnologia Genova and INBB Istituto Nazionale Biosistemi e Biostrutture, Rome, Italy
| | - Paul Adam
- School of Biological, Earth and Environmental Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Arturo Bravo Nuevo
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Mindy George-Weinstein
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Rebecca Mason
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Janis Eells
- College of Health Sciences, University of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
48
|
Lanigan S, Corcoran AE, Wall A, Mukandala G, O'Connor JJ. Acute hypoxic exposure and prolyl-hydroxylase inhibition improves synaptic transmission recovery time from a subsequent hypoxic insult in rat hippocampus. Brain Res 2018; 1701:212-218. [PMID: 30244114 DOI: 10.1016/j.brainres.2018.09.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 01/18/2023]
Abstract
In the CNS short episodes of acute hypoxia can result in a decrease in synaptic transmission which may be fully reversible upon re-oxygenation. Stabilization of hypoxia-inducible factor (HIF) by inhibition of prolyl hydroxylase domain (PHD) enzymes has been shown to regulate the cellular response to hypoxia and confer neuroprotection both in vivo and in vitro. Hypoxic preconditioning has become a novel therapeutic target to induce neuroprotection during hypoxic insults. However, there is little understanding of the effects of repeated hypoxic insults or pharmacological PHD inhibition on synaptic signaling. In this study we have assessed the effects of hypoxic exposure and PHD inhibition on synaptic transmission in the rat CA1 hippocampus. Field excitatory postsynaptic potentials (fEPSPs) were elicited by stimulation of the Schaffer collateral pathway. 30 min hypoxia (gas mixture 95% N2/5% CO2) resulted in a significant and fully reversible decrease in fEPSP slope associated with decreases in partial pressures of tissue oxygen. 15-30 min of hypoxia was sufficient to induce stabilization of HIF in hippocampal slices. Exposure to a second hypoxic insult after 60 min resulted in a similar depression of fEPSP slope but with a significantly greater rate of recovery of the fEPSP. Prior single treatment of slices with the PHD inhibitor, dimethyloxalylglycine (DMOG) also resulted in a significantly greater rate of recovery of fEPSP post hypoxia. These results suggest that hypoxia and 'pseudohypoxia' preconditioning may improve the rate of recovery of hippocampal neurons to a subsequent acute hypoxia.
Collapse
Affiliation(s)
- Sinead Lanigan
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Alan E Corcoran
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Audrey Wall
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gatambwa Mukandala
- College of Natural and Applied Sciences, University of Dar-Es-Salaam (UDSM), P.O Box 35064, Dar-Es-Salaam, Tanzania
| | - John J O'Connor
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
49
|
Geiseler SJ, Morland C. The Janus Face of VEGF in Stroke. Int J Mol Sci 2018; 19:ijms19051362. [PMID: 29734653 PMCID: PMC5983623 DOI: 10.3390/ijms19051362] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 12/12/2022] Open
Abstract
The family of vascular endothelial growth factors (VEGFs) are known for their regulation of vascularization. In the brain, VEGFs are important regulators of angiogenesis, neuroprotection and neurogenesis. Dysregulation of VEGFs is involved in a large number of neurodegenerative diseases and acute neurological insults, including stroke. Stroke is the main cause of acquired disabilities, and normally results from an occlusion of a cerebral artery or a hemorrhage, both leading to focal ischemia. Neurons in the ischemic core rapidly undergo necrosis. Cells in the penumbra are exposed to ischemia, but may be rescued if adequate perfusion is restored in time. The neuroprotective and angiogenic effects of VEGFs would theoretically make VEGFs ideal candidates for drug therapy in stroke. However, contradictory to what one might expect, endogenously upregulated levels of VEGF as well as the administration of exogenous VEGF is detrimental in acute stroke. This is probably due to VEGF-mediated blood–brain-barrier breakdown and vascular leakage, leading to edema and increased intracranial pressure as well as neuroinflammation. The key to understanding this Janus face of VEGF function in stroke may lie in the timing; the harmful effect of VEGFs on vessel integrity is transient, as both VEGF preconditioning and increased VEGF after the acute phase has a neuroprotective effect. The present review discusses the multifaceted action of VEGFs in stroke prevention and therapy.
Collapse
Affiliation(s)
- Samuel J Geiseler
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0371 Oslo, Norway.
| | - Cecilie Morland
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0371 Oslo, Norway.
- Institute for Behavioral Sciences, Faculty of Health Sciences, OsloMet-Oslo Metropolitan University, 0166 Oslo, Norway.
| |
Collapse
|
50
|
Tan H, Lu H, Chen Q, Tong X, Jiang W, Yan H. The Effects of Intermittent Whole-Body Hypoxic Preconditioning on Patients with Carotid Artery Stenosis. World Neurosurg 2018; 113:e471-e479. [DOI: 10.1016/j.wneu.2018.02.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 10/18/2022]
|