1
|
Fujii R, Nambu Y, Sawant Shirikant N, Furube E, Morita M, Yoshimura R, Miyata S. Neuronal regeneration in the area postrema of adult mouse medulla oblongata following glutamate-induced neuronal elimination. Neuroscience 2024; 563:188-201. [PMID: 39521321 DOI: 10.1016/j.neuroscience.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Neural stem cells and/or progenitor cells (NSCs/NPCs) in the subventricular and subgranular zones of the adult mammal forebrain generate new neurons and are involved in partial repair after injury. Recently, NSCs/NPCs were identified in the area postrema (AP) of the medulla oblongata of the hindbrain. In this study, we used the properties of fenestrate capillaries to observe specific neuronal elimination in the AP of adult mice and investigated subsequent neuronal regeneration by neurogenesis. Subcutaneous administration of monosodium glutamate (MSG) induced prominent Fos expression in HuC/D+ neurons in the AP 2 h after administration. MSG administration caused a marked decrease in HuC/D+ neuronal density by neuronal death 3 to 21 days after administration, which recovered to the control level 35 days later. After MSG administration, the density of TUNEL+ dying neurons and phagocytic microglia surrounding or engulfing neurons increased. Within 7 days of MSG administration, the number of BrdU+ Sox2+ and BrdU+ Math1+ cells increased markedly, and at least the BrdU+ Math1+ cells similarly increased for the next following 7 days. A remarkable number of HuC/D+ neurons with BrdU+ nuclei were observed 35 days after MSG administration. This study reveals that neurogenesis occurs in the AP of adult mice, recovering and maintaining normal neuronal density after neuronal death.
Collapse
Affiliation(s)
- Rena Fujii
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Yuri Nambu
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Nitin Sawant Shirikant
- Department of Biology, Graduate School of Science, Kobe University, Kobe 657-8501, Japan
| | - Eriko Furube
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan; Department of Anatomy, Asahikawa Medical University School of Medicine, Midorigaoka, Asahikawa, Hokkaido 078-8510, Japan
| | - Mitsuhiro Morita
- Department of Biology, Graduate School of Science, Kobe University, Kobe 657-8501, Japan
| | - Ryoichi Yoshimura
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| |
Collapse
|
2
|
Shadman J, Panahpour H, Alipour MR, Salimi A, Shahabi P, Azar SS. Investigating the therapeutic effects of nimodipine on vasogenic cerebral edema and blood-brain barrier impairment in an ischemic stroke rat model. Neuropharmacology 2024; 257:110054. [PMID: 38950691 DOI: 10.1016/j.neuropharm.2024.110054] [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: 03/24/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Vasogenic brain edema, a potentially life-threatening consequence following an acute ischemic stroke, is a major clinical problem. This research aims to explore the therapeutic benefits of nimodipine, a calcium channel blocker, in mitigating vasogenic cerebral edema and preserving blood-brain barrier (BBB) function in an ischemic stroke rat model. In this research, animals underwent the induction of ischemic stroke via a 60-min blockage of the middle cerebral artery and treated with a nonhypotensive dose of nimodipine (1 mg/kg/day) for a duration of five days. The wet/dry method was employed to identify cerebral edema, and the Evans blue dye extravasation technique was used to assess the permeability of the BBB. Furthermore, immunofluorescence staining was utilized to assess the protein expression levels of matrix metalloproteinase-9 (MMP-9) and intercellular adhesion molecule-1 (ICAM-1). The study also examined mitochondrial function by evaluating mitochondrial swelling, succinate dehydrogenase (SDH) activity, the collapse of mitochondrial membrane potential (MMP), and the generation of reactive oxygen species (ROS). Post-stroke administration of nimodipine led to a significant decrease in cerebral edema and maintained the integrity of the BBB. The protective effects observed were associated with a reduction in cell apoptosis as well as decreased expression of MMP-9 and ICAM-1. Furthermore, nimodipine was observed to reduce mitochondrial swelling and ROS levels while simultaneously restoring MMP and SDH activity. These results suggest that nimodipine may reduce cerebral edema and BBB breakdown caused by ischemia/reperfusion. This effect is potentially mediated through the reduction of MMP-9 and ICAM-1 levels and the enhancement of mitochondrial function.
Collapse
Affiliation(s)
- Javad Shadman
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamdollah Panahpour
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | | | - Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saied Salimpour Azar
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
3
|
Krzyzowska M, Patrycy M, Chodkowski M, Janicka M, Kowalczyk A, Skulska K, Thörn K, Eriksson K. Fas/FasL-Mediated Apoptosis and Inflammation Contribute to Recovery from HSV-2-Mediated Spinal Cord Infection. Viruses 2024; 16:1363. [PMID: 39339840 PMCID: PMC11436029 DOI: 10.3390/v16091363] [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: 07/21/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Herpes simplex virus type 2 (HSV-2) is a sexually transmitted pathogen that causes a persistent infection in sensory ganglia. The infection manifests itself as genital herpes but in rare cases it can cause meningitis. In this study, we used a murine model of HSV-2 meningitis to show that Fas and FasL are induced within the CNS upon HSV-2 infection, both on resident microglia and astrocytes and on infiltrating monocytes and lymphocytes. Mice lacking Fas or FasL had a more severe disease development with significantly higher morbidity, mortality, and an overall higher CNS viral load. In parallel, these Fas/FasL-deficient mice showed a severely impaired infection-induced CNS inflammatory response with lower levels of infiltrating CD4+ T-cells, lower levels of Th1 cytokines and chemokines, and a shift in the balance between M1 and M2 microglia/monocytes. In vitro, we confirmed that Fas and FasL is required for the induction of leucocyte apoptosis, but also show that the Fas/FasL pathway is required for adequate cytokine and chemokine production by glial cells. In summary, our data show that the Fas/FasL cell death receptor pathway is an important defense mechanism in the spinal cord as it down-regulates HSV-2-induced inflammation while at the same time promoting adequate anti-viral immune responses against infection.
Collapse
Affiliation(s)
- Malgorzata Krzyzowska
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (M.P.); (M.C.); (M.J.)
| | - Magdalena Patrycy
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (M.P.); (M.C.); (M.J.)
| | - Marcin Chodkowski
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (M.P.); (M.C.); (M.J.)
| | - Martyna Janicka
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (M.P.); (M.C.); (M.J.)
| | - Andrzej Kowalczyk
- PORT Polish Center for Technology Development, 54-066 Wroclaw, Poland; (A.K.); (K.S.)
| | - Katarzyna Skulska
- PORT Polish Center for Technology Development, 54-066 Wroclaw, Poland; (A.K.); (K.S.)
| | - Karolina Thörn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (K.T.); (K.E.)
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (K.T.); (K.E.)
| |
Collapse
|
4
|
Wang G, Li Z, Wang G, Sun Q, Lin P, Wang Q, Zhang H, Wang Y, Zhang T, Cui F, Zhong Z. Advances in Engineered Nanoparticles for the Treatment of Ischemic Stroke by Enhancing Angiogenesis. Int J Nanomedicine 2024; 19:4377-4409. [PMID: 38774029 PMCID: PMC11108071 DOI: 10.2147/ijn.s463333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Gongchen Wang
- Department of Vascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People’s Hospital, Yantai, Shandong, 265600, People’s Republic of China
| | - Peng Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Feiyun Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
5
|
Veeravalli KK. Implications of MMP-12 in the pathophysiology of ischaemic stroke. Stroke Vasc Neurol 2024; 9:97-107. [PMID: 37336584 PMCID: PMC11103161 DOI: 10.1136/svn-2023-002363] [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: 02/03/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
This article focuses on the emerging role of matrix metalloproteinase-12 (MMP-12) in ischaemic stroke (IS). MMP-12 expression in the brain increases dramatically in animal models of IS, and its suppression reduces brain damage and promotes neurological, sensorimotor and cognitive functional outcomes. Thus, MMP-12 could represent a potential target for the management of IS. This article provides an overview of MMP-12 upregulation in the brain following IS, its deleterious role in the post-stroke pathogenesis (blood-brain barrier disruption, inflammation, apoptosis and demyelination), possible molecular interactions and mechanistic insights, its involvement in post-ischaemic functional deficits and recovery as well as the limitations, perspectives, challenges and future directions for further research. Prior to testing any MMP-12-targeted therapy in patients with acute IS, additional research is needed to establish the effectiveness of MMP-12 suppression against IS in older animals and in animals with comorbidities. This article also examines the clinical implications of suppressing MMP-12 alone or in combination with MMP-9 for extending the currently limited tissue plasminogen activator therapy time window. Targeting of MMP-12 is expected to have a profound influence on the therapeutic management of IS in the future.
Collapse
Affiliation(s)
- Krishna Kumar Veeravalli
- Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| |
Collapse
|
6
|
Chung S, Yi Y, Ullah I, Chung K, Park S, Lim J, Kim C, Pyun SH, Kim M, Kim D, Lee M, Rhim T, Lee SK. Systemic Treatment with Fas-Blocking Peptide Attenuates Apoptosis in Brain Ischemia. Int J Mol Sci 2024; 25:661. [PMID: 38203830 PMCID: PMC10780202 DOI: 10.3390/ijms25010661] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Apoptosis plays a crucial role in neuronal injury, with substantial evidence implicating Fas-mediated cell death as a key factor in ischemic strokes. To address this, inhibition of Fas-signaling has emerged as a promising strategy in preventing neuronal cell death and alleviating brain ischemia. However, the challenge of overcoming the blood-brain barrier (BBB) hampers the effective delivery of therapeutic drugs to the central nervous system (CNS). In this study, we employed a 30 amino acid-long leptin peptide to facilitate BBB penetration. By conjugating the leptin peptide with a Fas-blocking peptide (FBP) using polyethylene glycol (PEG), we achieved specific accumulation in the Fas-expressing infarction region of the brain following systemic administration. Notably, administration in leptin receptor-deficient db/db mice demonstrated that leptin facilitated the delivery of FBP peptide. We found that the systemic administration of leptin-PEG-FBP effectively inhibited Fas-mediated apoptosis in the ischemic region, resulting in a significant reduction of neuronal cell death, decreased infarct volumes, and accelerated recovery. Importantly, neither leptin nor PEG-FBP influenced apoptotic signaling in brain ischemia. Here, we demonstrate that the systemic delivery of leptin-PEG-FBP presents a promising and viable strategy for treating cerebral ischemic stroke. Our approach not only highlights the therapeutic potential but also emphasizes the importance of overcoming BBB challenges to advance treatments for neurological disorders.
Collapse
Affiliation(s)
- Sungeun Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Yujong Yi
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
- Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Seongjun Park
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Jaeyeoung Lim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Chaeyeon Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Seon-Hong Pyun
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Minkyung Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Minhyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Taiyoun Rhim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| |
Collapse
|
7
|
Tsai YT, Cheng CY. Electroacupuncture at the Dazhui and Baihui acupoints and different frequencies (10 and 50 Hz) protects against apoptosis by up-regulating ERK1/2-mediated signaling in rats after global cerebral ischemia. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:706-716. [PMID: 38645497 PMCID: PMC11024414 DOI: 10.22038/ijbms.2024.72279.15716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 12/27/2023] [Indexed: 04/23/2024]
Abstract
Objectives This study assessed the effects of electroacupuncture (EA) stimulation at different frequencies at the Dazhui and Baihui acupoints in the subacute phase after transient global cerebral ischemia (GCI). Materials and Methods Rats were subjected to GCI for 25 min, followed by reperfusion for 7 days. EA at acupoints was performed at 10, 30, or 50 Hz, 1 day after reperfusion and then once daily for 6 consecutive days. Results EA at acupoints at 10 and 50 Hz effectively down-regulated apoptosis in the hippocampal cornu ammonis 1(CA1) area and ameliorated memory deficits. Moreover, EA treatment at 10 and 50 Hz markedly increased phospho (p)-extracellular signal-regulated protein kinase 1/2 (ERK1/2), p-ERK1/2/neuronal nuclei (NeuN), p-cAMP response element-binding protein (CREB)/p-ERK1/2, B-cell lymphoma-2 (Bcl-2)/p-CREB, and X-linked inhibitor of apoptosis protein/NeuN expression levels and decreased Bcl-2 homologous antagonist/killer, second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI, cytochrome c, cleaved caspase-3, and apoptosis-inducing factor expression levels. Furthermore, 10-Hz EA treatment effectively increased p-p38 mitogen-activated protein kinase (MAPK), p-p38 MAPK/NeuN, and p-CREB/p-p38 MAPK expression levels. Pretreatment with U0126 (ERK1/2 inhibitor) completely abrogated the effects of 10- and 50-Hz EA treatments on the aforementioned protein expression levels. Similarly, pretreatment with SB203580 (p38 MAPK inhibitor) completely abrogated the effects of 10-Hz treatment on the aforementioned protein expression levels. Conclusion The effects of 10- and 50-Hz EA treatments on mitochondria-related apoptosis can be attributed to the activation of ERK1/2/p38 MAPK/CREB/Bcl-2- and ERK1/2/CREB/Bcl-2-mediated signaling, respectively, in the hippocampal CA1 area at 7 days after transient GCI.
Collapse
Affiliation(s)
- Yueh-Ting Tsai
- School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Traditional Chinese Medicine, Kuang Tien General Hospital, Taichung 43303, Taiwan
| | - Chin-Yi Cheng
- School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Chinese Medicine, Hui-Sheng Hospital, Taichung 42056, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung 42056, Taiwan
| |
Collapse
|
8
|
Davoodi Asl F, Sahraei SS, Kalhor N, Fazaeli H, Sheykhhasan M, Soleimani Moud S, Naserpour L, Sheikholeslami A. Promising effects of exosomes from menstrual blood-derived mesenchymal stem cells on endometriosis. Reprod Biol 2023; 23:100788. [PMID: 37542905 DOI: 10.1016/j.repbio.2023.100788] [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: 05/06/2023] [Revised: 06/26/2023] [Accepted: 07/14/2023] [Indexed: 08/07/2023]
Abstract
Endometriosis as a non-malignant gynecological disease leads to dysregulation of numerous cellular functions including apoptosis, angiogenesis, migration, proliferation, and inflammation. Accumulating evidence has shed light on the importance of endometrial stem cells within the menstrual blood which are involved in the establishment and progression of endometriotic lesions in a retrograde manner. According to the fact that the therapeutic benefits of mesenchymal stem cells are provided through paracrine functions, we used exosomes from menstrual blood-derived stem cells (MenSCs) for treating endometriotic stem cells to inhibit their lesion formation tendency. Menstrual blood samples from healthy and endometriosis women were collected. Isolated MenSCs by the density-gradient centrifugation method were characterized by flow cytometry. Secreted exosomes were isolated from healthy MenSCs (NE-MenSCs) and used to treat endometriotic cells (E-MenSCs). 72 h after treatment, different mechanisms and pathways including inflammation, proliferation, apoptosis, migration, and angiogenesis were analyzed using Real-Time PCR, ELISA, immunocytochemistry, annexin V/PI, and scratching assay. Exosome treatment significantly reduce the expression level of markers related to inflammation, proliferation, migration, and angiogenesis in E-MenSCs which are aberrantly expressed in endometriosis. Moreover, apoptosis was induced in E-MenSCs after treatment which was evaluated in both gene and protein levels. In this study, we give preliminary evidence for the potential of MenSCs-Exo in ameliorating endometriosis. Regarding our results, we suggest that after relevant clinical trial, MenSCs-derived exosomes can be considered as a better treatment option to improve endometriosis compared to common and conventional treatments and show their potential as a cell-free product in endometriosis repair.
Collapse
Affiliation(s)
- Faezeh Davoodi Asl
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Seyedeh Saeideh Sahraei
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Hoda Fazaeli
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Mohsen Sheykhhasan
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Sanaz Soleimani Moud
- Midwifery ward, Infertility treatment center, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Leila Naserpour
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Azar Sheikholeslami
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran.
| |
Collapse
|
9
|
Lee HY, Song SY, Hwang J, Baek A, Baek D, Kim SH, Park JH, Choi S, Pyo S, Cho SR. Very early environmental enrichment protects against apoptosis and improves functional recovery from hypoxic-ischemic brain injury. Front Mol Neurosci 2023; 15:1019173. [PMID: 36824441 PMCID: PMC9942523 DOI: 10.3389/fnmol.2022.1019173] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 12/29/2022] [Indexed: 02/10/2023] Open
Abstract
Appropriate rehabilitation of stroke patients at a very early phase results in favorable outcomes. However, the optimal strategy for very early rehabilitation is at present unclear due to the limited knowledge on the effects of very early initiation of rehabilitation based on voluntary exercise (VE). Environmental enrichment (EE) is a therapeutic paradigm for laboratory animals that involves complex combinations of physical, cognitive, and social stimuli, as well as VE. Few studies delineated the effect of EE on apoptosis in very early stroke in an experimental model. Although a minimal benefit of early rehabilitation in stroke models has been claimed in previous studies, these were based on a forced exercise paradigm. The aim of this study is to determine whether very early exposure to EE can effectively regulate Fas/FasL-mediated apoptosis following hypoxic-ischemic (HI) brain injury and improve neurobehavioral function. C57Bl/6 mice were housed for 2 weeks in either cages with EE or standard cages (SC) 3 h or 72 h after HI brain injury. Very early exposure to EE was associated with greater improvement in motor function and cognitive ability, reduced volume of the infarcted area, decreased mitochondria-mediated apoptosis, and decreased oxidative stress. Very early exposure to EE significantly downregulated Fas/FasL-mediated apoptosis, decreased expression of Fas, Fas-associated death domain, cleaved caspase-8/caspase-8, cleaved caspase-3/caspase-3, as well as Bax and Bcl-2, in the cerebral cortex and the hippocampus. Delayed exposure to EE, on the other hand, failed to inhibit the extrinsic pathway of apoptosis. This study demonstrates that very early exposure to EE is a potentially useful therapeutic translation for stroke rehabilitation through effective inhibition of the extrinsic and intrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Hoo Young Lee
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea,National Traffic Injury Rehabilitation Hospital, Gyeonggi-do, Republic of Korea,Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Suk-Young Song
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jihye Hwang
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea,Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ahreum Baek
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea,Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Dawoon Baek
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea,Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Sung Hoon Kim
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Jung Hyun Park
- Yonsei University College of Medicine, Seoul, Republic of Korea,Department of Rehabilitation Medicine, Rehabilitation Institute of Neuromuscular Disease, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sungchul Choi
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soonil Pyo
- Neuracle Science Co. Ltd., Seoul, Republic of Korea,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Sung-Rae Cho
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea,Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea,Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea,*Correspondence: Sung-Rae Cho, ✉
| |
Collapse
|
10
|
Cranial electrotherapy stimulation alleviates depression-like behavior of post-stroke depression rats by upregulating GPX4-mediated BDNF expression. Behav Brain Res 2023; 437:114117. [PMID: 36116735 DOI: 10.1016/j.bbr.2022.114117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022]
Abstract
To elucidate whether cranial electrotherapy stimulation (CES) improves depression-like behavior of post-stroke depression (PSD) via regulation of glutathione peroxidase 4 (GPX4)-mediated brain-derived neurotrophic factor (BDNF) expression. Middle cerebral artery occlusion (MCAO) and chronic unpredictable mild stress (CUMS) were used to develop a rat PSD model. CES was applied, and RAS-selective lethal 3 (RSL3) was injected into the hippocampus to inhibit GPX4 in PSD rats. The depression behavior was detected by sucrose preference and forced swimming tests. The structure and morphology of the hippocampus were observed and analyzed by histopathological hematoxylin-eosin (HE) staining. The mRNA and protein expressions of GPX4 and BDNF in the hippocampus were detected by qRT-PCR, western blot and immunohistochemical analysis.The degeneration and necrosis of hippocampal neurons, the depression-like behavior were severer and the expression of BDNF in the hippocampus were decreased in PSD rats than those in MCAO and control groups. CES promoted the hippocampal neuron repair, alleviated the depression-like behavior and increased the expression of BDNF in PSD rats. The inhibition of GPX4 by RSL3 exacerbated the depression-like behavior and decreased the expression of BDNF in PSD rats. In addition, we found that RSL3 disrupted the positive effects of CES on the PSD rats. Conclusion: CES improves depression-like behavior of PSD rats through upregulation of GPX4-mediated BDNF expression in the hippocampus.
Collapse
|
11
|
Kadir RRA, Alwjwaj M, Bayraktutan U. MicroRNA: An Emerging Predictive, Diagnostic, Prognostic and Therapeutic Strategy in Ischaemic Stroke. Cell Mol Neurobiol 2022; 42:1301-1319. [PMID: 33368054 PMCID: PMC9142420 DOI: 10.1007/s10571-020-01028-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
Stroke continues to be the third-leading cause of death and disability worldwide. The limited availability of diagnostic tools approved therapeutics and biomarkers that help monitor disease progression or predict future events remain as the major challenges in the field of stroke medicine. Hence, attempts to discover safe and efficacious therapeutics and reliable biomarkers are of paramount importance. MicroRNAs (miRNAs) are a class of non-coding RNAs that play important roles in regulating gene expression. Since miRNAs also play important roles in key mechanisms associated with the pathogenesis of stroke, including energy failure, inflammation and cell death, it is possible that miRNAs may serve as reliable blood-based markers for risk prediction, diagnosis and prognosis of ischaemic stroke. Discovery of better neurological outcome and smaller cerebral infarcts in animal models of ischaemic stroke treated with miRNA agomirs or antagomirs indicate that miRNAs may also play a cerebrovascular protective role after an ischaemic stroke. Nonetheless, further evidences on the optimum time for treatment and route of administration are required before effective translation of these findings into clinical practice. Bearing these in mind, this paper reviews the current literature discussing the involvement of miRNAs in major pathologies associated with ischaemic stroke and evaluates their value as reliable biomarkers and therapeutics for ischaemic stroke.
Collapse
Affiliation(s)
- Rais Reskiawan A Kadir
- Stroke, Division of Clinical Neuroscience, School of Medicine, The University of Nottingham, Clinical Sciences Building, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Mansour Alwjwaj
- Stroke, Division of Clinical Neuroscience, School of Medicine, The University of Nottingham, Clinical Sciences Building, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, School of Medicine, The University of Nottingham, Clinical Sciences Building, Hucknall Road, Nottingham, NG5 1PB, UK.
| |
Collapse
|
12
|
Song L, Mu L, Wang H. MicroRNA-489-3p aggravates neuronal apoptosis and oxidative stress after cerebral ischemia-reperfusion injury. Bioengineered 2022; 13:14047-14056. [PMID: 35730531 PMCID: PMC9342425 DOI: 10.1080/21655979.2022.2062534] [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] [Indexed: 11/26/2022] Open
Abstract
Cerebral ischemia-reperfusion injury (CIRI) mostly occurs in the treatment stage of ischemic diseases and aggravate brain tissue damage. Although studies have demonstrated that miR-489-3p is closely related to CIRI, the effects of miR-489-3p on neural function in CIRI have not been directly studied. The transient middle cerebral artery occlusion (tMCAO) model was established by suture method, and the corresponding plasmids that interfered with the expression of miR-489-3p or Sirtuin1 (SIRT1) were injected into the model mice, and the behavioral changes of the mice were observed. Then the concentration of serum neuronal injury markers and oxidative stress indices were examined. Next, the pathological conditions, neuronal loss and apoptosis of brain tissue were observed by hematoxylin-eosin staining, Nissl staining, and Transferase-mediated deoxyuridine triphosphate-biotin nick end labeling staining. Finally, the hemoglobin content and cerebral edema in the mouse brain were determined. In addition, the expression levels of miR-489-3p and SIRT1 were detected by reverse transcription quantitative polymerase chain reaction or Western blot, and the targeting relationship between miR-489-3p and SIRT1 was verified by bioinformatics analysis and luciferase reporter assay. The experimental results found that in tMCAO mice, miR-489-3p in brain tissue was up-regulated and SIRT1 was down-regulated. Down-regulating miR-489-3p or up-regulating SIRT1 ameliorated behavioral dysfunction, neuronal damage and apoptosis, oxidative stress and brain histopathology. miR-489-3p targeted the regulation of SIRT1 expression, and down-regulating SIRT1 can reverse the protective effect of silenced miR-489-3p on brain injury. Taken together, by targeting SIRT1, elevated miR-489-3p aggravates CIRI-induced neuronal apoptosis and oxidative stress.
Collapse
Affiliation(s)
- LiGuo Song
- Department of Neurosurgery, The First People's Hospital of Mudanjiang, Mudanjiang City, Heilongjiang Province, China
| | - LuYan Mu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Haerbin, Heilongjiang Province, China
| | - HongLiang Wang
- Department of Neurology, The Sixth People's Hospital of Nantong City, Nantong City, Jiangsu Province, China
| |
Collapse
|
13
|
Neuronal Pnn Deficiency Increases Oxidative Stress and Exacerbates Cerebral Ischemia/Reperfusion Injury in Mice. Antioxidants (Basel) 2022; 11:antiox11030466. [PMID: 35326115 PMCID: PMC8944488 DOI: 10.3390/antiox11030466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/10/2022] Open
Abstract
Cerebral stroke remains one of the leading causes of death worldwide. Ischemic stroke caused by the sudden loss of blood flow in brain is the major type of cerebral stroke. In addition to necrotic cell death in the ischemic core region, neuronal apoptosis is usually observed in the ischemic penumbra. Pnn, a multi-functional protein, participates in cellular proliferation, migration, differentiation, apoptosis as well as cell–cell interaction through its abilities in regulating gene transcription and mRNA processing. Our recent studies have demonstrated that Pnn has a cell type-specific distribution manner in neural cells under ischemic injury and plays a protective role in astrocytes against ischemic stress. In this study, we generated an inducible neuron-specific Pnn deficiency mouse model to further investigate the physiological role of Pnn in neurons. To directly examine the role of neuronal Pnn in ischemic stress, four weeks after induction of Pnn deficiency in neurons, middle cerebral artery occlusion (MCAO) was applied to induce cerebral ischemia/reperfusion in mice. In the cerebrum and hippocampus with neuronal Pnn depletion, the expression of SRSF2, a mRNA splicing regulator, was increased, while the expression of SRSF1, a functional antagonist of SRSF2, was reduced. Expression levels of ROS generators (NOX-1 and NOX-2) and antioxidant proteins (GR, HO-1, NQO-1) were upregulated in brain tissue with loss of neuronal Pnn, echoing an increase in oxidized proteins in cortical and hippocampal neurons. Furthermore, the expression of DNA damage marker, p53bp1, was found in the choroid plexus of mice with neuronal Pnn depletion. In mice with MCAO, compared to wild type mice, both increased cerebral infarcted area and elevated expressions of proapoptotic proteins were found in mice with neuronal Pnn depletion. In conclusion, Pnn deficiency increases oxidative stress in neurons and exacerbates cerebral ischemia/reperfusion injury in mice.
Collapse
|
14
|
Wasan H, Singh D, Joshi B, Sharma U, Dinda AK, Reeta KH. Post Stroke Safinamide Treatment Attenuates Neurological Damage by Modulating Autophagy and Apoptosis in Experimental Model of Stroke in Rats. Mol Neurobiol 2021; 58:6121-6135. [PMID: 34453687 DOI: 10.1007/s12035-021-02523-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/04/2021] [Indexed: 11/30/2022]
Abstract
Exploring and repurposing a drug have become a lower risk alternative. Safinamide, approved for Parkinson's disease, has shown neuroprotection in various animal models of neurological disorders. The present study aimed to explore the potential of safinamide in cerebral ischemia/reperfusion (I/R) in rats. Sprague-Dawley rats were used in middle cerebral artery occlusion model of stroke. The effective dose of safinamide was selected based on the results of neurobehavioral parameters and reduction in infarct size assessed 24 h post-reperfusion. For sub-acute study, the treatment with effective dose was extended for 3 days and effects on neurobehavioral parameters, infarct size (TTC staining and MRI), oxidative stress parameters (MDA, GSH, SOD, NOX-2), inflammatory cytokines (TNF-α, IL-1β, IL-10), apoptosis (Bax, Bcl-2, cleaved caspase-3 expression, and TUNEL staining), and autophagy (pAMPK, Beclin-1, LC3-II expression) were studied. The results of dose selection study showed significant reduction (p < 0.05) in infarct size and improvement in neurobehavioral parameters with safinamide (80 mg/kg). In sub-acute study, safinamide showed significant (p < 0.05) improvement in motor coordination and infarct size reduction. Additionally, safinamide treatment significantly normalized altered redox homeostasis and inflammatory cytokine levels. However, no change was observed in expression of NOX-2 in I/R or safinamide treatment group when compared with sham. I/R induced deranged expression of apoptotic markers and increased TUNEL positive cells in cortex were significantly normalized with safinamide treatment. Further, safinamide significantly (p < 0.05) induced the expressions of autophagic proteins (Beclin-1 and LC3-II) in cortex. Overall, the results demonstrated neuroprotective potential of safinamide via anti-oxidant, anti-inflammatory, anti-apoptotic, and autophagy inducing properties. Thus, safinamide can be explored for repurposing in ischemic stroke after further exploration.
Collapse
Affiliation(s)
- Himika Wasan
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Devendra Singh
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Balu Joshi
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Uma Sharma
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| | - A K Dinda
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
15
|
Du J, Li W, Wang B. Long non-coding RNA TUG1 aggravates cerebral ischemia and reperfusion injury by sponging miR-493-3p/miR-410-3p. Open Med (Wars) 2021; 16:919-930. [PMID: 34222667 PMCID: PMC8231466 DOI: 10.1515/med-2021-0253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Background Cerebral ischemia and reperfusion injury (CIRI) affects bodily function by causing irreversible damage to brain cells. The diverse pathophysiological course factors hinder the research work to go deeper. Long noncoding RNA taurine-upregulated gene 1 (TUG1) has been reported to be related to CIRI. This study explored the undefined regulatory pathway of TUG1 in CIRI. Methods Quantitative real-time polymerase chain reaction was applied to test the expression of TUG1, microRNA (miR)-493-3p and miR-410-3p. The viability and apoptosis of oxygen and glucose deprivation/reoxygen (OGD/R) model cells were evaluated by cell counting kit-8 and flow cytometry assay, respectively. The determination of inflammatory factors of interleukin-6, interleukin-1β and tumor necrosis factor-α was presented by enzyme-linked immunosorbent assay. The oxidative stress was performed by measuring the generation of malondialdehyde, reactive oxygen species and the activity of superoxide dismutase. Cytotoxicity was presented by measuring the generation of lactate dehydrogenase. Western blot assay was devoted to assessing the level of apoptosis-related factors (cleaved-caspase-3 and cleaved-caspase-9) and the protein level of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK) pathway-related factors in neuro-2a cells treated by OGD/R. Besides, online database starBase was applied to predict the potential binding sites of TUG1 to miR-493-3p and miR-410-3p, which was further confirmed by the dual-luciferase reporter system. Results The expression of TUG1 was upregulated, while miR-493-3p or miR-410-3p was downregulated in the serum of CIRI and OGD/R model cells. Meanwhile, knockdown of TUG1 eliminated the suppression in proliferation, the promotion in apoptosis, inflammation and oxidative stress, as well as the cytotoxicity in OGD/R model cells. Interestingly, the inhibition of miR-493-3p or miR-410-3p allayed the above effects. In addition, TUG1 harbored miR-493-3p or miR-410-3p and negatively regulated their expression. Finally, the TUG1 activated JNK and p38 MAPK pathways by sponging miR-493-3p/miR-410-3p. Conclusion TUG1 motivated the development of CIRI by sponging miR-493-3p/miR-410-3p to activate JNK and p38 pathways. The novel role of TUG1 in CIRI may contribute to the advancement of CIRI treatment.
Collapse
Affiliation(s)
- Jinlong Du
- Department of Critical Care Medicine, Jingzhou Central Hospital, The Second Clinical Medical College of Yangtze University, Jingzhou, Hubei, 434020, China
| | - Wenjing Li
- Department of Ultrasound, Jingzhou Central Hospital, The Second Clinical Medical College of Yangtze University, Jingzhou, Hubei, 434020, China
| | - Bing Wang
- Department of Critical Care Medicine, Jingzhou Central Hospital, The Second Clinical Medical College of Yangtze University, Jingzhou, Hubei, 434020, China
| |
Collapse
|
16
|
Gu C, Yang J, Luo Y, Ran D, Tan X, Xiang P, Fei H, Lu Y, Guo W, Tu Y, Liu X, Wang H. ZNRF2 attenuates focal cerebral ischemia/reperfusion injury in rats by inhibiting mTORC1-mediated autophagy. Exp Neurol 2021; 342:113759. [PMID: 33992580 DOI: 10.1016/j.expneurol.2021.113759] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/25/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023]
Abstract
Zinc and ring finger 2 (ZNRF2), an E3 ubiquitin ligase, plays a crucial role in many diseases. However, its role in cerebral ischemia/reperfusion injury (CIRI) still remains unknown. In this study, the function and molecular mechanism of ZNRF2 in CIRI in vivo and vitro was studied. ZNRF2 was found to be dramatically downregulated in CIRI. Overexpression of ZNRF2 could significantly reduce the neurological deficit, brain infarct volume and histopathological damage of cortex in middle cerebral artery occlusion/reperfusion. Concomitantly, overexpression of ZNRF2 increased the primary neuronal viability and decreased the neuronal apoptosis induced by oxygen-glucose deprivation and reoxygenation (OGD/R). Mechanistically, overexpression of ZNRF2 inhibited the over-induction of autophagy induced by OGD/R which was abolished by mTORC1 inhibitor rapamycin. It can be concluded that ZNRF2 plays a protective effect in CIRI and the underlying mechanism may be related to the inhibition of mTORC1-mediated autophagy.
Collapse
Affiliation(s)
- Chao Gu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Junqing Yang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Ying Luo
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Dongzhi Ran
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Xiaodan Tan
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Pu Xiang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China; Dianjiang People's Hospital of Chongqing, Dianjiang, Chongqing 408300, China
| | - Huizhi Fei
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Yi Lu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Wenjia Guo
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Yujun Tu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Xia Liu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Hong Wang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
17
|
He J, Zhang X, He W, Xie Y, Chen Y, Yang Y, Chen R. Neuroprotective effects of zonisamide on cerebral ischemia injury via inhibition of neuronal apoptosis. ACTA ACUST UNITED AC 2021; 54:e10498. [PMID: 33656055 PMCID: PMC7917778 DOI: 10.1590/1414-431x202010498] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 11/02/2020] [Indexed: 11/22/2022]
Abstract
It is known that neuronal apoptosis contributes to pathology of cerebral ischemia injury. Zonisamide (ZNS) has shown anti-apoptosis effects in recent studies. The present study investigated whether the anti-apoptotic effect can account for the neuroprotective action of ZNS on cerebral ischemia. Neuronal cells were maintained under oxygen-glucose deprivation conditions to simulate cerebral ischemia and treated with ZNS simultaneously. The apoptosis of the cells and expression of apoptosis-related proteins were investigated by flow cytometry and western blot analysis, respectively. A cerebral ischemia mouse model was created via middle cerebral artery occlusion, and the mice were treated with ZNS. Neurological deficit scores and infarct volumes of the cerebral ischemia mice were measured. The apoptosis status of the neuronal cells was evaluated by TUNEL staining. In vitro, the ZNS treatment inhibited both the apoptosis of the neuronal cells and apoptosis-related protein expression (caspase-3, caspase-8, and calpain-1) induced by the oxygen-glucose deprivation. The anti-apoptosis effect of ZNS could occur through the blocking of reactive oxygen species. Moreover, ZNS treatment significantly ameliorated neurological deficits and reduced infarct volumes in the cerebral ischemia mice model. In this study, ZNS exerted neuroprotective effects by inhibition of apoptosis in neuronal cells in cerebral ischemia. Therefore, ZNS might be a promising therapy for cerebral ischemia.
Collapse
Affiliation(s)
- Junna He
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Weiliang He
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yanzhao Xie
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yanxia Chen
- Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yang Yang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Rong Chen
- Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei, China
| |
Collapse
|
18
|
Kale S, Sarode LP, Kharat A, Ambulkar S, Prakash A, Sakharkar AJ, Ugale RR. Protocatechuic Acid Prevents Early Hour Ischemic Reperfusion Brain Damage by Restoring Imbalance of Neuronal Cell Death and Survival Proteins. J Stroke Cerebrovasc Dis 2021; 30:105507. [PMID: 33285352 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105507] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To investigate the neuroprotective effect of protocatechuic acid (PCA) on cell death/survival protein imbalance in a rat model of middle cerebral artery occlusion and reperfusion. METHODS Focal ischemia was induced by middle cerebral artery occlusion in adult male Wistar rats and confirmed by measuring infarction of brain by 2,3,5-Triphenyltetrazolium chloride (TTC) staining. Rats were treated with vehicle or PCA at 10, 30 or 50 mg/kg dose intraperitoneally and subjected to neurological deficits or beam walk assessment at 24 h of reperfusion. Effective dose of PCA (50 mg/kg) was administered at 1, 2 and 3 h time point of post-ictus ischemia. Cellular damage and nuclear condensation was observed by haematoxylin and eosin (H and E) staining and Hoechst 33342 staining respectively. Additionally, immunohistochemical expression of caspase 3 and cAMP-response element binding protein (CREB) and their mRNA's were observed. RESULTS PCA at 30 and 50 mg/kg significantly improved behavioural performance and reduced infarction. Maximum neuroprotective effect of PCA (50 mg/kg) was found at 1 h (early hours) post-ictus ischemia along with reduction in cellular damage and nuclear condensation. PCA increased CREB protein and it's mRNA, while suppressed caspase-3 protein and mRNA at 1 h of reperfusion injury. CONCLUSION PCA exhibit the potential to prevent early hour (1h) reperfusion injury restoring balance of survival and death protein may offer a cost effective adjuvant therapy in stroke.
Collapse
Affiliation(s)
- Swapnil Kale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur, Maharashtra 440 033, India.
| | - Lopmudra P Sarode
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur, Maharashtra 440 033, India.
| | - Amol Kharat
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur, Maharashtra 440 033, India.
| | - Saurabh Ambulkar
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur, Maharashtra 440 033, India.
| | - Anand Prakash
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar, India.
| | - Amul J Sakharkar
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India.
| | - Rajesh R Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur, Maharashtra 440 033, India.
| |
Collapse
|
19
|
Li Y, Xiang L, Wang C, Song Y, Miao J, Miao M. Protection against acute cerebral ischemia/reperfusion injury by Leonuri Herba Total Alkali via modulation of BDNF-TrKB-PI3K/Akt signaling pathway in rats. Biomed Pharmacother 2021; 133:111021. [PMID: 33227709 DOI: 10.1016/j.biopha.2020.111021] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/09/2020] [Accepted: 11/15/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To observe the brain protective effect of Leonuri Herba Total Alkali (LHA) on cerebral ischemia reperfusion injury in rats, so as to provide basis for clinical research. METHODS Adult male SD rats were randomly assigned into sham group, middle cerebral artery occlusion/reperfusion (MCAO/R) group, and LHA + MCAO/R group (25 mg/kg, 50 mg/kg, and 100 mg/kg). Fourteen days before MCAO/R surgery, the rats in treatment groups were orally administered with LHA in ultrapure water once daily for 14 days, while rats in the sham and MCAO groups were given the same amount of saline in advance. After 1 h of administration on the 14th day, MCAO surgery was subjected. The neurological deficits, brain infarct volume, histopathology, immunofluorescence, inflammation indicators and the gene/protein expressions of BDNF-TrKB-PI3K/Akt signaling pathway in the rat brain tissue were evaluated 24 h after the MCAO/R-injury. RESULTS It was found that rats in LHA pre-administration group showed significantly reduced neurological deficit scores, infarction volume, the serum levels of NSE and S100β. Meanwhile, the content of Evans Blue (EB) in brain tissue from LHA group was decreased, as well as the levels of inflammatory cytokines and their gene levels. Moreover, LHA pre-administration inhibited the expression of CD44, GFAP, FOXO1 and promoted the expression of BDNF and NeuN. In addition, LHA pre-administration could up-regulate the protein expression of TrkB, p-PI3K, p-Akt, Bcl-2, and down-regulate the protein expression of Bax, and increase the level of Bcl-2/Bax. CONCLUSIONS The study demonstrated that LHA pre-administration could regulate the PI3K/Akt pathway by increasing BDNF levels, and play a neuroprotective role in cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yan Li
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Liling Xiang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Can Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Yagang Song
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Jinxin Miao
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China.
| | - Mingsan Miao
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China.
| |
Collapse
|
20
|
Expression and Distribution Pattern of Pnn in Ischemic Cerebral Cortex and Cultured Neural Cells Exposed to Oxygen-Glucose Deprivation. Brain Sci 2020; 10:brainsci10100708. [PMID: 33027948 PMCID: PMC7599571 DOI: 10.3390/brainsci10100708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 10/03/2020] [Indexed: 12/23/2022] Open
Abstract
Pinin (Pnn), a multifunctional protein, participates in embryonic development as well as in cellular apoptosis, proliferation, and migration through regulating mRNA alternative splicing and gene transcription. Previous studies have shown that Pnn plays important roles in neural system development and the expression level of Pnn in astrocytes is altered by ischemic stress and associated with cellular apoptosis. In the present study, we further utilized primary cultured rat neurons and astrocytes with oxygen-glucose deprivation (OGD) and a mouse model with middle cerebral artery occlusion (MCAO)-induced ischemic stroke to examine the effect of ischemic stress on Pnn expression and distribution in different types of neural cells. Under normoxia, Pnn is mainly localized in the nuclear speckle of primary cultured neurons. The expression level of Pnn was increased after the OGD treatment and then decreased in the reoxygenation period. Moreover, the cytoplasmic expression of Pnn was observed in neurons with OGD and reoxygenation (OGD/R). Unlike that in neurons, the Pnn expression in astrocytes was decreased after OGD treatment and then gradually increased during the reoxygenation period. Of interest, the nuclear–cytoplasmic translocation of Pnn was not observed in astrocytes with OGD/R. In the MCAO mouse model, the neuronal expression of Pnn in the peri-ischemic region was reduced by three days post induction of ischemic stroke. However, the Pnn expression in astrocytes was not altered. Moreover, the nuclear speckle distribution of Pnn in neurons was also diminished following ischemic stroke. In conclusion, the Pnn expression and distribution after OGD and during reoxygenation showed distinct manners in neurons and astrocytes, implying that Pnn may play different roles in different types of neural cells in the stress response to ischemic injury.
Collapse
|
21
|
Zhang D, Jin W, Liu H, Liang T, Peng Y, Zhang J, Zhang Y. ENT1 inhibition attenuates apoptosis by activation of cAMP/pCREB/Bcl2 pathway after MCAO in rats. Exp Neurol 2020; 331:113362. [PMID: 32445645 DOI: 10.1016/j.expneurol.2020.113362] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND PURPOSE The present study was designed to investigate the potential role and the mechanism of equilibrative nucleoside transporter 1 (ENT1) on neuronal apoptosis and neurological deficits after middle cerebral artery occlusion (MCAO) in rats. METHODS One hundred and thirty-four male Sprague-Dawley rats were subjected to two hours of MCAO followed by reperfusion. The time course of the expression level of ENT1 and phosphorylation of CREB were detected by western blot and immunofluorescence staining. Another set of animals were administrated with NBTI, the ENT1 inhibitor, by daily intraperitoneal injection starting at 0.5 h post-MCAO, infarction volume and neurological deficits were measured both at 24 h and 72 h post MCAO. We further explored the neuroprotection machenism by using H89, cAMP dependent protein kinase inhibitor, the expression of Bcl-2, Bax, phosphorylated CREB and Cleaved caspase-3 were quantified by Western blot, neuronal apoptosis were analyed by TUNEL staining. RESULTS The endogenous expression of ENT1 were significantly increased and peaked at 12 h after MCAO. High-dose of NBTI (15 mg/kg) reduced brain infarction volume and improved neurologic deficits both at 24 h and 72 h post MCAO. Moreover, NBTI significantly increased the level of CREB phosphorylation and extracellular adenosine concentration, and decreased the neuronal apoptosis 24 h after MCAO. NBTI treatment reduced the expression of Bax and cleaved caspase-3, while up-regulated Bcl-2 compared with vehicle group. These effects were abolished by H89 pretreatment. CONCLUSIONS ENT1 inhibition prevented neuronal apoptosis and improves neurological deficits through cAMP/PKA/CREB/Bcl-2 signaling pathway after MCAO in rats. ENT1 might be an effective target in the treatment strategy for ischemic stroke.
Collapse
Affiliation(s)
- Dongyun Zhang
- Department of rehabilitation medicine, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of neurology, Affiliated Hospital, Zunyi medical University, Zunyi, China
| | - Weidong Jin
- Department of Laboratory Medicine, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongliang Liu
- Department of rehabilitation medicine, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Liang
- Department of neurology, Affiliated Hospital, Zunyi medical University, Zunyi, China
| | - Yan Peng
- Department of neurology, Affiliated Hospital, Zunyi medical University, Zunyi, China
| | - Jun Zhang
- Department of neurology, Affiliated Hospital, Zunyi medical University, Zunyi, China.
| | - Yang Zhang
- Department of Laboratory Medicine, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
22
|
Wang Y, Zhao P, Song Z, Du X, Huo X, Lu J, Liu X, Lv J, Li C, Guo M, Chen Z. Generation of Gene-Knockout Mongolian Gerbils via CRISPR/Cas9 System. Front Bioeng Biotechnol 2020; 8:780. [PMID: 32733872 PMCID: PMC7360674 DOI: 10.3389/fbioe.2020.00780] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/18/2020] [Indexed: 12/27/2022] Open
Abstract
The Mongolian gerbil (Meriones unguiculatus), a well-known "multifunctional" experimental animal, plays a crucial role in the research of hearing, cerebrovascular diseases and Helicobacter pylori infection. Although the whole-genome sequencing of Mongolian gerbils has been recently completed, lack of valid gene-editing systems for gerbils largely limited the further usage of Mongolian gerbils in biomedical research. Here, efficient targeted mutagenesis in Mongolian gerbils was successfully conducted by pronuclear injection with Cas9 protein and single-guide RNAs (sgRNAs) targeting Cystatin C (Cst3) or Apolipoprotein A-II (Apoa2). We found that 22 h after human chorionic gonadotropin (hCG) injection, zygote microinjection was conducted, and the injected zygotes were transferred into the pseudopregnant gerbils, which were induced by injecting equine chorionic gonadotropin (eCG) and hCG at a 70 h interval and being caged with ligated male gerbils. We successfully obtained Cst3 and Apoa2 gene knockout gerbils with the knockout efficiencies of 55 and 30.9%, respectively. No off-target effects were detected in all knockout gerbils and the mutations can be germline-transmitted. The absence of CST3 protein was observed in the tissues of homozygous Cst3 knockout (Cst3-KO) gerbils. Interestingly, we found that disruption of the Cst3 gene led to more severe brain damage and neurological deficits after unilateral carotid artery ligation, thereby indicating that the gene modifications happened at both genetic and functional levels. In conclusion, we successfully generated a CRISPR/Cas9 system based genome editing platform for Mongolian gerbils, which provided a foundation for obtaining other genetically modified gerbil models for biomedical research.
Collapse
Affiliation(s)
- Yan Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Peikun Zhao
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Zidai Song
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Xiaoyan Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Xueyun Huo
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Jing Lu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Xin Liu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Jianyi Lv
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Changlong Li
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Meng Guo
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Zhenwen Chen
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Science, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Zhang H, Song Y, Feng C. Improvement of cerebral ischemia/reperfusion injury by daucosterol palmitate-induced neuronal apoptosis inhibition via PI3K/Akt/mTOR signaling pathway. Metab Brain Dis 2020; 35:1035-1044. [PMID: 32363473 DOI: 10.1007/s11011-020-00575-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/14/2020] [Indexed: 02/07/2023]
Abstract
Traditional Chinese medicine has growing importance in the treatment of ischemia stroke due to its abundance and low drug resistance. In this study, we aim to investigate the therapeutic potential of daucosterol palmitate against ischemia stroke, as well as its neuro-protective mechanism. The dose-response effects of daucosterol palmitate in the protection from brain damage were evaluated in a cerebral ischemia/reperfusion (I/R) rat model. The correlation of neuro-protective effects of daucosterol palmitate with apoptosis inhibition was examined and the possible signaling targets were identified. Our findings revealed that daucosterol palmitate treatment after 2 h' ischemia significantly lowered brain damage, and neuronal cell apoptosis caused by I/R injury in a dose-response mode (20, 40 and 80 mg/kg). Western blot analysis indicated that daucosterol palmitate could reverse the effects of I/R injury on protein expression of PI3K and mTOR, and phosphorylation of Akt. Contrarily, inactivation of PI3K using wortmannin dramatically antagonized the effect of daucosterol palmitate for I/R injury. With these findings, it supports the application potential of daucosterol palmitate in the treatment of ischemia stroke. Besides, the PI3K/Akt/mTOR pathway might be potential cellular targets for daucosterol palmitate.
Collapse
Affiliation(s)
- Huiyuan Zhang
- Department of Neurology, Liaocheng People's Hospital, Huashan road, NO 45, Liaocheng city, 252000, Shandong Province, China
| | - Yamin Song
- Department of Neurology, Liaocheng People's Hospital, Huashan road, NO 45, Liaocheng city, 252000, Shandong Province, China.
| | - Cong Feng
- Department of Surgery, Tumour Hospital of Liaocheng, Liaocheng city, 252000, Shandong, China
| |
Collapse
|
24
|
Zhang AP, Zhang YY, Liu AF, Wang K, Li C, Liu YE, Zhang YQ, Zhou J, Lv J, Jiang WJ. Molecular mechanism of long-term neuroprotective effects of gradual flow restoration on cerebral ischemia reperfusion injury in MCAO rats. J Stroke Cerebrovasc Dis 2020; 29:105041. [PMID: 32807453 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/18/2020] [Accepted: 06/07/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Ischemia-reperfusion injuries (IRIs) can aggravate the condition of some patients with acute occlusion of major intracranial artery (AOMIA) who received endovascular thrombectomy. Here, we provided data confirming the association of Repressor Element-1 Silencing Transcription factor (REST) with the long-term neuroprotective effect of the middle cerebral artery occlusion (MCAO) rats underwent Gradual Flow Restoration (GFR). METHODS Long term neuroprotective effects of GFR intervention were evaluated on MCAO rats model after 3d and 7d reperfusion. The neurological deficit score and TTC staining were performed to evaluate the degree of brain damage in GFR and other interventions at different time. Differentially expressed genes related to cerebral ischemia reperfusion injury (CIRI) were initially screened and identified using GSE32529 microarray analysis. REST protein expression in rat brain cortex infarction was detected by Western blot analysis. RESULTS MCAO rats intervened with GFR exhibited reduced neurological deficit (P < 0.05) and alleviated brain infarction volume (P < 0.01). The REST gene with up-regulated expression and its downstream genes with down-regulated expression were screened by Microarray analysis. The brain cortex infarction in MCAO rats produced high levels of REST expression. The GFR intervention inhibited REST expression, and alleviated brain injury on MCAO rats. CONCLUSION Our results demonstrated that GFR intervention plays a long-term neuroprotective role and reduces brain edema and damage at reperfusion, possibly by inhibiting REST expression.
Collapse
Affiliation(s)
- Ai-Ping Zhang
- Medical College of Soochow University, Suzhou, Jiangsu 215123; The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China; Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China
| | - Ying-Ying Zhang
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China
| | - Ao-Fei Liu
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China
| | - Kai Wang
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China
| | - Chen Li
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China
| | - Yun-E Liu
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China
| | - Yi-Qun Zhang
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China
| | - Ji Zhou
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China
| | - Jin Lv
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China; Central Laboratory of Research Department, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR china.
| | - Wei-Jian Jiang
- Medical College of Soochow University, Suzhou, Jiangsu 215123; The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China; Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The PLA Rocket Force Characteristic Medical Center, Beijing 100088, PR China.
| |
Collapse
|
25
|
Mahale A, Kumar R, Sarode LP, Gakare S, Prakash A, Ugale RR. Dapsone prolong delayed excitotoxic neuronal cell death by interacting with proapoptotic/survival signaling proteins. J Stroke Cerebrovasc Dis 2020; 29:104848. [PMID: 32689584 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Dapsone prevents ischemic injury, inhibits apoptosis and shows functional improvement post-ischemia. However, its effect on proapoptotic or survival proteins in delayed ischemia remains unclear. METHODS Male adult Wistar rats were subjected to middle cerebral artery occlusion (MCAO) for 90 min followed by 24 h of ischemic reperfusion (I/R). Dapsone [9.375 or 12.5 mg/kg, intraperitoneally (IP)] was administered at 3, 6 and 12 h of I/R followed by behavioural assessment, brain infarction, histological alteration and cell viability study. Further, dapsone (25 and 50 µM) was added at 3, 6 and 12 h after L-glutamate (100 µM) in primary cortical culture (DIV 14) and cell viability, cytotoxicity, apoptosis was observed. Proteins expression were observed using immunocytochemistry. All experiments were performed after 24 h of I/R (In-Vivo) and 24 h of recovery post glutamate insult (In-Vitro). RESULTS Reduced brain infarction, improved neurobehavioural functions in addition to reduction in abnormal morphological structures of ischemic brain and improvement in cell viability was observed with treatment of dapsone (12.5 mg/kg) administered upto 6 h. Similarly, dapsone (25, 50 µM) increased cell survival post glutamate insult in cortical culture (In-vitro). Further, dapsone treatment at delayed hours (6 h) reduced apoptotic nuclei and proapoptotic proteins JNK, PTEN, Calpain, Caspase 3 expression along with activation of prosurvival protein BDNF expression post-glutamate insult. CONCLUSION Our results suggest that dapsone has the potential to limit the neuronal damage post-glutamate insult in delayed hours (6 h) through repressing proapoptotic proteins JNK, PTEN, Calpain, Caspase-3 of cerebral ischemia along with activation of pro-survival protein BDNF.
Collapse
Affiliation(s)
- Ashutosh Mahale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur 440033, Maharashtra, India
| | - Rakesh Kumar
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur 440033, Maharashtra, India
| | - Lopmudra P Sarode
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur 440033, Maharashtra, India
| | - Sukanya Gakare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur 440033, Maharashtra, India
| | - Anand Prakash
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar, India.
| | - Rajesh R Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Amravati Road, Nagpur 440033, Maharashtra, India.
| |
Collapse
|
26
|
Lu X, Dong J, Zheng D, Li X, Ding D, Xu H. Reperfusion combined with intraarterial administration of resveratrol-loaded nanoparticles improved cerebral ischemia-reperfusion injury in rats. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102208. [PMID: 32334100 DOI: 10.1016/j.nano.2020.102208] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/13/2020] [Accepted: 03/28/2020] [Indexed: 01/28/2023]
Abstract
Endovascular thrombectomy (EVT) has been recommended as the first line therapy for large artery occlusion (LAO) stroke. However, abrupt recovery of blood flow induces oxidative stress which breaks down the blood-brain barrier (BBB), activates apoptosis and inhibits neurogenesis. Supplement of exogenous antioxidants to relieve the injuries related to oxidative stress is a rational treatment combined to EVT for acute LAO therapy. Resveratrol (RES), an antioxidant, was encapsulated into polymeric nanoparticles (RES-NPs). In transient middle cerebral artery occlusion (tMCAO) rats, intraarterial administration of RES-NPs demonstrated significant protection against cerebral ischemia/reperfusion (I/R) injuries. RES-NPs attenuated the oxidative stress induced by I/R, prevented brain edema, protected neurons from undergoing apoptosis, and contributed to neurogenesis through enhanced expression of brain-derived neurotrophic factor (BDNF). These results suggested that intra-arterial infusion of RES-NPs in conjunction with EVT could be a potential strategy for the LAO stroke therapy.
Collapse
Affiliation(s)
- Xiaowei Lu
- Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Jingde Dong
- Department of Geriatric Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Donghui Zheng
- Department of Nephrology, the Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an City, Huai'an, PR China
| | - Xiaolin Li
- Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, China
| | - Huae Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
27
|
Habib P, Stamm AS, Zeyen T, Noristani R, Slowik A, Beyer C, Wilhelm T, Huber M, Komnig D, Schulz JB, Reich A. EPO regulates neuroprotective Transmembrane BAX Inhibitor-1 Motif-containing (TMBIM) family members GRINA and FAIM2 after cerebral ischemia-reperfusion injury. Exp Neurol 2019; 320:112978. [PMID: 31211943 DOI: 10.1016/j.expneurol.2019.112978] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/03/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Transmembrane BAX Inhibitor-1 Motif-containing (TMBIM) family members exert inhibitory activities in apoptosis and necroptosis. FAIM2 (TMBIM-2) is neuroprotective against murine focal ischemia and is regulated by erythropoietin (EPO). Similar to FAIM2, GRINA (TMBIM-3) is predominantly expressed in the brain. The role of GRINA in transient brain ischemia, its potential synergistic effects with FAIM2 and its regulation by EPO treatment were assessed. METHODS We performed transient (30 min) middle cerebral artery occlusion (tMCAo) followed by 72 h of reperfusion in GRINA-deficient (GRINA-/-), FAIM2-deficient (FAIM2-/-), double-deficient (GRINA-/-FAIM2-/-) and wildtype littermates (WT) mice. We administered EPO or saline 0, 24 and 48 h after tMCAo. We subjected primary murine cortical neurons (pMCN) of all mouse strains to oxygen-glucose deprivation (OGD) after GRINA and/or FAIM2 gene transfection. RESULTS Compared to wildtype controls GRINA deficiency led to a similar increase in infarct volumes as FAIM2 deficiency (p < .01). We observed the highest neurological deficits and largest infarct sizes in double-deficient mice. EPO administration upregulated GRINA and FAIM2 mRNA levels in wildtype littermates. EPO decreased infarct sizes and abrogated neurological impairments in wildtype controls. GRINA and/or FAIM2 deficient mice showed increased expression levels of cleaved-caspase 3 and of pro-apoptotic BAX mRNA. Further, caspase 8 was upregulated in FAIM2-/- and caspase 9 in GRINA-/- mice. Overexpression of GRINA and FAIM2 in wildtype and in double deficient pMCN decreased cell death rate after OGD. CONCLUSIONS GRINA and FAIM2 are highly expressed in the brain and convey EPO-mediated neuroprotection after ischemic stroke involving different caspases.
Collapse
Affiliation(s)
- Pardes Habib
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany.
| | - Ann-Sophie Stamm
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Thomas Zeyen
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Rozina Noristani
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Medical School, RWTH Aachen University, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Medical School, RWTH Aachen University, Aachen, Germany
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Daniel Komnig
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Jörg B Schulz
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany; JARA-BRAIN Institute of Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
28
|
Behdarvandy M, Karimian M, Atlasi MA, Azami Tameh A. Heat shock protein 27 as a neuroprotective biomarker and a suitable target for stem cell therapy and pharmacotherapy in ischemic stroke. Cell Biol Int 2019; 44:356-367. [PMID: 31502740 DOI: 10.1002/cbin.11237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/08/2019] [Indexed: 12/12/2022]
Abstract
Ischemic stroke is a major common cause of death and long-term disability worldwide. Several pathophysiological events including excitotoxicity, oxidative/nitrative stress, inflammation, and apoptosis are involved in ischemic injuries. Recently, the molecular mechanisms involved in cerebral ischemia through a focus on a member of small heat shock proteins family, Hsp27, has been developed. Notably, following exposure to ischemia, Hsp27 expression in the brain could be increased rather than the normal condition and it may play an important role in neuroprotection after ischemic stroke. The neuroprotection effects of Hsp27 may arise from its anti-oxidant, anti-inflammatory, anti-apoptotic, and chaperonic properties. Moreover, some therapeutic strategies such as stem cell therapy and pharmacotherapy have been developed with Hsp27 targeting. In this review, we describe the function and structure of Hsp27 and its possible role in neuroprotection after ischemic stroke. Finally, we present current studies in stroke therapy, which focused on Hsp27 targeting.
Collapse
Affiliation(s)
- Marjan Behdarvandy
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Qotb-e Ravandi Blvd., 8715988141, Kashan, Iran
| | - Mohammad Karimian
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Qotb-e Ravandi Blvd., 8715988141, Kashan, Iran
| | - Mohammad Ali Atlasi
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Qotb-e Ravandi Blvd., 8715988141, Kashan, Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Qotb-e Ravandi Blvd., 8715988141, Kashan, Iran
| |
Collapse
|
29
|
The Neuroprotective Effect of the HDAC2/3 Inhibitor MI192 on the Penumbra After Photothrombotic Stroke in the Mouse Brain. Mol Neurobiol 2019; 57:239-248. [DOI: 10.1007/s12035-019-01773-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/20/2022]
|
30
|
Demyanenko S, Berezhnaya E, Neginskaya M, Rodkin S, Dzreyan V, Pitinova M. Сlass II histone deacetylases in the post-stroke recovery period-expression, cellular, and subcellular localization-promising targets for neuroprotection. J Cell Biochem 2019; 120:19590-19609. [PMID: 31264264 DOI: 10.1002/jcb.29266] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/12/2019] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDAC) inhibitors can protect nerve cells after a stroke, but it is unclear which HDAC isoform is involved in this effect. We studied cellular and intracellular rearrangement of class II HDACs at late periods after photothrombotic infarct (PTI) in the mouse sensorimotor cortex in the tissue surrounding the ischemia core and in the corresponding region of the contralateral hemisphere. We observed a decrease in HDAC4 in cortical neurons and an increase in its nuclear translocation. HDAC6 expression in neurons was also increased. Moreover, HDAC6-positive cells had elevated apoptosis. Tubostatin A (Tub A)-induced decrease in the activity of HDAC6 restored acetylation of α-tubulin during the early poststroke recovery period and reduced apoptosis of nerve cells thus protecting the brain tissue. Selective inhibition of HDAC6 elevated expression of growth-associated protein-43 (GAP43), which remained high up to 14 days after stroke and promoted axogenesis and recovery from the PTI-induced neurological deficit. Selective HDAC6 inhibitor Tub A markedly reduced neuronal death and increased acetylation of α-tubulin and the level of GAP43. Thus, HDAC6 inhibition could be a promising strategy for modulation of brain recovery as it can increase the intensity and reduce the duration of reparation processes in the brain after stroke.
Collapse
Affiliation(s)
- Svetlana Demyanenko
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Elena Berezhnaya
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Maria Neginskaya
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Stanislav Rodkin
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Valentina Dzreyan
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Maria Pitinova
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| |
Collapse
|
31
|
Calcitriol protects the Blood-Brain Barrier integrity against ischemic stroke and reduces vasogenic brain edema via antioxidant and antiapoptotic actions in rats. Brain Res Bull 2019; 150:281-289. [PMID: 31220552 DOI: 10.1016/j.brainresbull.2019.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND Vasogenic brain edema is the most important complication of ischemic stroke that aggravates primary brain injury. Ischemia-Reperfusion (IR)-induced Blood-Brain Barrier (BBB) impairment limits the use of recombinant tissue plasminogen activator (r-tPA) by increasing the possibility of hemorrhagic transformation and contributing to vasogenic edema and neuroinflammation. This study examined the effects of post-ischemic treatment with calcitriol on cerebral infarction, vasogenic edema formation and BBB disruption in a rat model of ischemic stroke. METHODS Male Sprague-Dawley rats were divided into three main groups, including the sham, IR + vehicle and IR + calcitriol groups. Transient focal cerebral ischemia was induced by a 60-min-long occlusion of the left middle cerebral artery. The infarct volume, brain edema, BBB permeability and antioxidant enzyme activities were evaluated 24 h after ischemia. Immunohistochemical analysis was conducted to investigate cell apoptosis and Brain-Derived Neurotrophic Factor (BDNF) protein expression five days after ischemia. RESULTS Compared to the IR + vehicle group, the IR + calcitriol group showed a reduced brain infarction volume, attenuated brain edema formation and improved BBB function. These protective effects were followed by the upregulation of antioxidant enzyme activities in the brain tissue. Additionally, a diminished cell apoptosis and an increased BDNF immunoreactivity were obtained in the IR + calcitriol group. CONCLUSION Calcitriol may reduce brain injury and attenuate vasogenic edema by upregulating antioxidant enzymes activities, reducing cell apoptosis and increasing BDNF protein in the brain tissue in a rat model of ischemic stroke.
Collapse
|
32
|
Mukda S, Tsai CY, Leu S, Yang JL, Chan SHH. Pinin protects astrocytes from cell death after acute ischemic stroke via maintenance of mitochondrial anti-apoptotic and bioenergetics functions. J Biomed Sci 2019; 26:43. [PMID: 31167655 PMCID: PMC6549339 DOI: 10.1186/s12929-019-0538-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/26/2019] [Indexed: 11/27/2022] Open
Abstract
Background Stroke is the second most common cause of deaths worldwide. After an ischemic stroke, the proliferated reactive astrocytes in the peri-infarct areas play a beneficial role in neuronal survival. As such, astrocytes have gradually become a target for neuroprotection in stroke. The present study assessed the hypothesis that Pinin (Pnn), originally identified as a nuclear and desmosome-associated protein and is now known to possess anti-apoptotic capacity, protects astrocytes from cell death after ischemic stroke and delineated the underlying mechanisms. Methods In in vivo experiments, adult male Sprague-Dawley rats (12-week old) were used to induce acute focal cerebral ischemia employing the middle cerebral artery occlusion (MCAO) method. In in vitro experiments, postnatal day 1 (P1) Sprague-Dawley rat pups were used to prepare cultures of primary astrocytes. Oxygen-glucose deprivation (OGD) and re-oxygenation (OGD/R) procedures were employed to mimic the hypoxic-ischemic condition of stroke in those astrocytes. Results We found in the peri-infarct area of the ipsilateral cortex and striatum in Sprague-Dawley rats after transient MCAO an increase in Pnn expression that correlated positively with the time-course of infarction as detected by T2-weighted imaging and triphenyltetrazolium chloride staining, augmented number of reactive astrocytes that double-labelled with Pnn as determined by immunofluorescence, and enhanced cytotoxic edema as revealed by diffusion weighted imaging; but mirrored the decreased cleaved caspase-3 as measured by western blot. In an OGD and OGD/R model using primary cultured astrocytes, treatment with Pnn siRNA doubled the chance of surviving astrocytes to manifest cell death as revealed by flow cytometry, and blunted activated ERK signaling, reduced Bcl-2 expression and augmented cleaved caspase 3 detected by western blot in the normoxia, OGD or OGD/R group. Gene-knockdown of Pnn also impeded the reversal from decline in cell viability, elevation in lactate dehydrogenase leakage and decrease in ATP production in the OGD/R group. Conclusion We conclude that the endogenous Pnn participates in neuroprotection after acute ischemic stroke by preserving the viability of astrocytes that survived the ischemic challenge via maintenance of mitochondrial anti-apoptotic and bioenergetics functions. Electronic supplementary material The online version of this article (10.1186/s12929-019-0538-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, 25/25 Phuttamonthon 4 Road, Salaya, Nakhon Pathom, 73170, Thailand.,Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Rd, Kaohsiung, 83301, Taiwan
| | - Ching-Yi Tsai
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Rd, Kaohsiung, 83301, Taiwan
| | - Steve Leu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Rd, Kaohsiung, 83301, Taiwan
| | - Jenq-Lin Yang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Rd, Kaohsiung, 83301, Taiwan
| | - Samuel H H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Rd, Kaohsiung, 83301, Taiwan.
| |
Collapse
|
33
|
Lorente L, Martín MM, Pérez-Cejas A, González-Rivero AF, Sabatel R, Ramos L, Argueso M, Solé-Violán J, Cáceres JJ, Jiménez A, García-Marín V. Serum Caspase-3 Levels and Early Mortality of Patients with Malignant Middle Cerebral Artery Infarction. Neurocrit Care 2019; 31:486-493. [PMID: 31115825 DOI: 10.1007/s12028-019-00739-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE Circulating caspase-3 levels at 24 h of ischemic stroke were found to be associated with poorer functional neurological outcome in a previous study. The aim of this study was to determine whether there is an association between serum caspase-3 levels and early mortality in patients with malignant middle cerebral artery infarction (MMCAI). METHODS We included patients with MMCAI defined as computer tomography showing ischemic changes in more than 50% of the middle cerebral artery territory and Glasgow Coma Scale ≤ 8. Serum caspase-3 levels at days 1, 4, and 8 of MMCAI were determined. RESULTS Non-surviving MMCAI (n = 34) showed higher serum caspase-3 levels at days 1 (p < 0.001), 4 (p = 0.001), and 8 (p = 0.01) than surviving patients (n = 34). We found that the area under the curve of serum caspase-3 levels for prediction of mortality at 30 days was 88% (95% CI = 78-95%; p < 0.001). Multiple logistic regression showed that serum caspase-3 levels were associated with 30-day mortality (OR = 51.25; 95% CI = 8.30-316.31; p < 0.001). CONCLUSIONS The novel and more important findings of our study were that high serum caspase-3 levels were associated with mortality in MMCAI patients.
Collapse
Affiliation(s)
- Leonardo Lorente
- Intensive Care Unit, Hospital Universitario de Canarias, Ofra s/n, La Laguna, 38320, Santa Cruz de Tenerife, Spain.
| | - María M Martín
- Intensive Care Unit, Hospital Universitario Nuestra Señora de Candelaria, Crta del Rosario s/n, 38010, Santa Cruz de Tenerife, Spain
| | - Antonia Pérez-Cejas
- Laboratory Department, Hospital Universitario de Canarias, Ofra, s/n, La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | - Agustín F González-Rivero
- Laboratory Department, Hospital Universitario de Canarias, Ofra, s/n, La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | - Rafael Sabatel
- Department of Radiology, Hospital Universitario de Canarias, Ofra, s/n, La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | - Luis Ramos
- Intensive Care Unit, Hospital General La Palma, Buenavista de Arriba s/n, Breña Alta, 38713, La Palma, Spain
| | - Mónica Argueso
- Intensive Care Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez nº17-19, 46004, Valencia, Spain
| | - Jordi Solé-Violán
- Intensive Care Unit, Hospital Universitario Dr. Negrín, CIBERES, Barranco de la Ballena s/n, 35010, Las Palmas de Gran Canaria, Spain
| | - Juan J Cáceres
- Intensive Care Unit, Hospital Insular, Plaza Dr. Pasteur s/n, 35016, Las Palmas de Gran Canaria, Spain
| | - Alejandro Jiménez
- Research Unit, Hospital Universitario de Canarias, Ofra s/n, La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | - Victor García-Marín
- Department of Neurosurgery, Hospital Universitario de Canarias, Ofra, s/n, La Laguna, 38320, Santa Cruz de Tenerife, Spain
| |
Collapse
|
34
|
Zhang B, Zhang HX, Shi ST, Bai YL, Zhe X, Zhang SJ, Li YJ. Interleukin-11 treatment protected against cerebral ischemia/reperfusion injury. Biomed Pharmacother 2019; 115:108816. [PMID: 31096144 DOI: 10.1016/j.biopha.2019.108816] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Inflammation and immune responses are crucial factors associated with the onset and progression of stroke. Interleukin-11 (IL-11) is a hematopoietic IL-6 family cytokine that functions as an anti-inflammatory agent against various inflammatory diseases. However, its roles in stroke remain unknown. In this study, we investigated the effects of IL-11 on cerebral ischemia-reperfusion injury in a model of focal cerebral ischemia. METHODS Mice were randomly divided into five groups the vehicle group, the middle cerebral artery occlusion (MCAO) group, the MCAO plus adenosine monophosphate-activated protein kinase (AMPK) inhibitor compound C group, the MCAO plus IL-11 treatment group, and the MCAO plus IL-11 treatment and compound C group. Focal cerebral ischemia was induced by occluding the left middle cerebral artery, and reperfusion was achieved by withdrawing the suture 2 h after ischemia. The protein expression levels of IL-11 were measured using Western blot analysis, and its location was detected using immunohistochemistry and immunofluorescence staining. The infarct volume was examined using 2,3,5-triphenyl tetrazolium chloride (TTC) staining, and the neurobehavioral progression was assessed using the neurological scoring system. The expression of astrocytes and microglia was detected using immunochemistry, and real-time quantitative PCR was used for the gene quantification of inflammatory cytokines. The extent of cerebral ischemia-reperfusion injury was tested using Nissl staining and the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay. The expression of the apoptotic proteins Bax, Bcl-2 and cleaved caspase-3 were detected using Western blot analysis, and the oxidative stress was also measured. RESULTS The expression of IL-11 mRNA and protein significantly decreased after cerebral ischemia. Immunohistochemical staining showed a large amount of IL-11 in the cerebral cortex of the mice in the vehicle group, whereas the immunoreactivity of IL-11 remained weak for 24 h in the MCAO group. Immunofluorescent staining further confirmed that IL-11 was mainly expressed in the neurons. It was suggested that IL-11 (20 μg/kg) treatment ameliorated infarction and reduced neurological scores. In addition, IL-11 proved to reduce neuropathic damage, glial activation, and the expression of proinflammatory cytokines and increase the expression of anti-inflammatory cytokines after cerebral ischemia. IL-11 was also able to alleviate oxidative stress caused by cerebral ischemia, and AMPK inhibition enhanced the alleviation. Moreover, IL-11 was found to inhibit apoptosis caused by cerebral ischemia, which could also be facilitated by AMPK inhibitors. SIGNIFICANCE Our research suggests that IL-11 is decreased during cerebral ischemia-reperfusion injury, but IL-11 treatment can improve neurological function and reduce the cerebral infarct volume, which can trigger stroke in mice. AMPK inhibition can further promote the protective effect of IL-11 in stroke. Overall, we demonstrate that IL-11 is of therapeutic interest in controlling stroke and managing cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Bei Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Hai-Xiong Zhang
- Otolaryngology Department, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Shao-Ting Shi
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Yu-Lan Bai
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Xiao Zhe
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Shi-Jun Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Ya-Jun Li
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China.
| |
Collapse
|
35
|
Nouraee C, Fisher M, Di Napoli M, Salazar P, Farr TD, Jafarli A, Divani AA. A Brief Review of Edema-Adjusted Infarct Volume Measurement Techniques for Rodent Focal Cerebral Ischemia Models with Practical Recommendations. JOURNAL OF VASCULAR AND INTERVENTIONAL NEUROLOGY 2019; 10:38-45. [PMID: 31308870 PMCID: PMC6613487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
BACKGROUND Determining cerebral infarction volume is an important part of preclinical studies to determine the benefit of potential therapies on stroke outcome. A well-known problem in determining the actual infarction volume of rodent models is the presence of edema. Because of this, algorithms must be utilized to obtain the edema-adjusted (E A)-infarct volume. Different methods based on 2,3,5-triphenyltetrazolium hydrochloride (TTC) staining have been published describing algorithms to determine the E A-infarct volume. MATERIALS AND METHODS Simulated models of infarction and corresponding swelling were employed to determine which absolute method of calculation (Lin et al., Reglodi et al., or Belayev et al.) is the most accurate in calculating the absolute E A-infarct volume. RESULTS The Reglodi and Belayev methods were statistically more accurate in measuring E A-infarct volume than Lin's method, p = 0.0078. Though there was no significant difference between Reglodi's and Belayev's methods for the E A-infarction volume calculation, Reglodi's approach was closer to the ground-truth infarct volume while also being simpler and more straightforward to use. CONCLUSION We recommend that Reglodi's method, that is E A-infarct volume = infarct volume × (contralateral hemisphere/ipsilateral hemisphere), to be used in calculating E A-infarct volume in TTC stained rodent brains. Further, factors such as inhomogeneous infarction distribution in a given brain slice can also contribute to the error in volume calculation. Therefore, the average of the infarct area obtained from anterior and posterior views of a given slice should be used to account for the variation. Considering different factors, we have provided a summary recommendation for calculating the infarction volume.
Collapse
Affiliation(s)
- Cyrus Nouraee
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Marc Fisher
- Department of Neurology, Beth Israel Lahey Health, Harvard Medical School, Boston, MA, USA
| | - Mario Di Napoli
- Department of Neurology, San Camillo de’ Lellis District General Hospital, Rieti, Italy
| | | | - Tracy D. Farr
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Alibay Jafarli
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Afshin A. Divani
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
36
|
Sun J, Li X, Liu J, Pan X, Zhao Q. Stigmasterol Exerts Neuro-Protective Effect Against Ischemic/Reperfusion Injury Through Reduction Of Oxidative Stress And Inactivation Of Autophagy. Neuropsychiatr Dis Treat 2019; 15:2991-3001. [PMID: 31695390 PMCID: PMC6805119 DOI: 10.2147/ndt.s220224] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/09/2019] [Indexed: 01/15/2023] Open
Abstract
PURPOSE Stroke remains the primary cause of pain, suffering, and death in patients. One of the major thrusts in stroke therapy is to find an effective prevention strategy. Objectives of this study are to testify the neuro-protection effect of stigmasterol in ischemic/reperfusion injury model. METHODS The dosage-dependent effects (20, 40, and 80 mg/kg) of stigmasterol on physiological behaviors and oxidative stress biomarkers were investigated. Expression and phosphorylation of beclin1, microtubule-associated protein 1 light chain 3 (LC3), adenosine monophosphate-activated protein kinase (AMPK), mTOR, and N-terminal kinase (JNK) were detected. RESULTS The results showed that stigmasterol was able to effectively reduce neurological deficits and infarct damage induced by the ischemic/reperfusion injury, improve histopathology changes, and restore the levels of the endogenous antioxidant defense system in a dose-response mode. Stigmasterol effectively depressed the expression level of beclin1, and the conversion of LC3 I to LC3 II, while promoted the phosphorylation of mTOR, and remarkably inhibited the phosphorylation of AMPK and JNK, as well as the expression of JNK induced by 24 hrs of reperfusion. CONCLUSION These findings reveal that stigmasterol has neuro-protective effect against the ischemic/reperfusion injury, possibly associated with reduction of oxidative stress and inactivation of autophagy via AMPK/mTOR and JNK pathways.
Collapse
Affiliation(s)
- Jiadong Sun
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang City, Shandong Province 262100, People's Republic of China
| | - Xuemei Li
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang City, Shandong Province 262100, People's Republic of China
| | - Junling Liu
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang City, Shandong Province 262100, People's Republic of China
| | - Xin Pan
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang City, Shandong Province 262100, People's Republic of China
| | - Qianqian Zhao
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang City, Shandong Province 262100, People's Republic of China
| |
Collapse
|
37
|
Li L, Liu T, Li X, Liu X, Liu L, Li S, Li Z, Zhou Y, Liu F. Protein chip and bioinformatic analyses of differentially expressed proteins involved in the effect of hydrogen-rich water on myocardial ischemia-reperfusion injury. Int J Med Sci 2019; 16:1254-1259. [PMID: 31588191 PMCID: PMC6775260 DOI: 10.7150/ijms.35984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/30/2019] [Indexed: 12/17/2022] Open
Abstract
Background: The differentially expressed proteins (DEPs) involved in the effect of hydrogen-rich water on myocardial ischemia reperfusion injury (MIRI) and their biological processes and signaling pathway were analyzed. Methods: 20 Wistar rats were randomly and equally divided into a control and a hydrogen-rich group. Hearts were removed and fixed in a Langendorff device. The control group was perfused with K-R solution, and the hydrogen-rich water group was perfused with K-R solution + hydrogen-rich water. Protein was extracted from the ventricular tissues, and GSR-CAA-67 was used to identify the DEPs between two groups. DEPs were analyzed through bioinformatic methods. Results: Compared with the control group, in the treatment group, the expression of 25 proteins was obviously decreased (P<0.05). For the DEPs, 359 biological processes, including the regulation of signaling pathways, immune reaction and formation of cardiovascular endothelial cells, were selected by GO enrichment analysis. Five signaling pathways were selected by KEGG pathway enrichment analysis. Conclusions: 25 proteins that are involved in hydrogen-water reducing MIRI were selected by high-throughput GSR-CAA-67. The biological processes and metabolic pathways involved in the DEPs were summarized, providing theoretical evidence for the clinical application of hydrogen-rich water.
Collapse
Affiliation(s)
- Liangtong Li
- School of Medicine, Hebei University, Baoding, 071000, China.,Central Laboratory of Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Tongtong Liu
- Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Xiangzi Li
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Xuanchen Liu
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Li Liu
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Shaochun Li
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Zhilin Li
- School of Chemistry, Hebei University, Baoding 071000, China
| | - Yujuan Zhou
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Fulin Liu
- Affiliated Hospital of Hebei University, Baoding, 071000, China
| |
Collapse
|
38
|
Liu W, Guo Q, Zhao H. Oxidative stress-elicited YY1 potentiates antioxidative response via enhancement of NRF2-driven transcriptional activity: A potential neuronal defensive mechanism against ischemia/reperfusion cerebral injury. Biomed Pharmacother 2018; 108:698-706. [DOI: 10.1016/j.biopha.2018.09.082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 12/21/2022] Open
|
39
|
Ullah I, Chung K, Oh J, Beloor J, Bae S, Lee SC, Lee M, Kumar P, Lee SK. Intranasal delivery of a Fas-blocking peptide attenuates Fas-mediated apoptosis in brain ischemia. Sci Rep 2018; 8:15041. [PMID: 30301943 PMCID: PMC6178348 DOI: 10.1038/s41598-018-33296-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022] Open
Abstract
Ischemic stroke-induced neuronal cell death results in the permanent disabling of brain function. Apoptotic mechanisms are thought to play a prominent role in neuronal injury and ample evidence implicates Fas signaling in mediating cell death. In this study, we describe the neuroprotective effects of a Fas-blocking peptide (FBP) that by obstructing Fas signaling in cerebral ischemia inhibits apoptosis. Using an intranasal administration route in a rat model of focal cerebral ischemia, we demonstrate that nose-to-brain delivery of FBP after middle cerebral artery occlusion (MCAO) surgery results in the delivery and retention of FBP in Fas-expressing ischemic areas of the brain. A single intranasal administration of 2 mg/kg FBP resulted in significantly reduced neuronal cell death by inhibiting Fas-mediated apoptosis leading to decreased infarct volumes, reduced neurologic deficit scores and recovery from cerebral ischemia. Intranasally delivered FBP might be a promising strategy for the treatment of cerebral ischemic stroke.
Collapse
Affiliation(s)
- Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Jungju Oh
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea
| | - Jagadish Beloor
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Sumin Bae
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea
| | - Sangah Clara Lee
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
- Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| | - Minhyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea
| | - Priti Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA.
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul, Korea.
| |
Collapse
|
40
|
Lorente L, Martín MM, Pérez-Cejas A, Ramos L, Argueso M, Solé-Violán J, Cáceres JJ, Jiménez A, García-Marín V. High serum levels of caspase-cleaved cytokeratin-18 are associated with malignant middle cerebral artery infarction patient mortality. BMC Neurol 2018; 18:32. [PMID: 29573748 PMCID: PMC5866523 DOI: 10.1186/s12883-018-1038-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/15/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND There have been found apoptotic changes in brain tissue samples from humans after cerebral ischemia. Caspase-cleaved cytokeratin (CCCK)-18 could appears in blood during apoptosis. High circulating levels of CCCK-18 have been associated with a poor prognosis in patients with cerebral process, such as traumatic brain injury and spontaneous cerebral hemorrhage. However, they have not been explored in patients with ischemic stroke. Thus, the aim of this study was to determine whether there is an association between serum CCCK-18 levels and mortality in patients with severe malignant middle cerebral artery infarction (MMCAI). METHODS This was an observational, prospective and multicentre study. We included patients with severe MMCAI. We considered MMCAI as severe when Glasgow Coma Scale (GCS) was lower than 9. We measured serum CCCK-18 levels at the diagnosis moment of the severe MMCAI. RESULTS We found that non-surviving severe MMCAI patients (n = 33) showed lower GCS and platelet count, and higher serum CCCK-18 levels than survivor ones (n = 33). We found an area under the curve (AUC) of serum CCCK-18 levels to predict 30-day mortality of 82% (95% CI = 71%-91%; p < 0.001). In the multiple logistic regression analysis was found that serum CCCK-18 levels were associated with 30-day mortality (OR = 1.023; 95% CI = 1.010-1.037; p = 0.001) after to control for platelet count and GCS. CONCLUSIONS To our knowledge, this is the first series reporting data on serum CCCK-18 levels in ischemic stroke patients. The novel findings of our study were that non-surviving severe MMCAI patients had higher serum CCCK-18 levels than surviving patients, and that there is an association between high serum CCCK-18 levels and MMCAI patients mortality.
Collapse
Affiliation(s)
- Leonardo Lorente
- Intensive Care Unit, Hospital Universitario de Canarias, Ofra s/n, La Laguna, -38320, Santa Cruz de Tenerife, Spain.
| | - María M Martín
- Intensive Care Unit, Hospital Universitario Nuestra Señora de Candelaria, Crta del Rosario s/n, -38010, Santa Cruz de Tenerife, Spain
| | - Antonia Pérez-Cejas
- Laboratory Deparment, Hospital Universitario de Canarias, Ofra, s/n., La Laguna -, 38320, Tenerife, Spain
| | - Luis Ramos
- Intensive Care Unit, Hospital General La Palma, Buenavista de Arriba s/n, -38713, Breña Alta, La Palma, Spain
| | - Mónica Argueso
- Intensive Care Unit, Hospital Clínico Universitario de Valencia, Avda Blasco Ibáñez n°17-19, -46004, Valencia, Spain
| | - Jordi Solé-Violán
- Intensive Care Unit, Hospital Universitario Dr. Negrín, CIBERES, Barranco de la Ballena s/n, -35010, Las Palmas de Gran Canaria, Spain
| | - Juan J Cáceres
- Intensive Care Unit, Hospital Insular, Plaza Dr. Pasteur s/n, 35016, Las Palmas de Gran Canaria, Spain
| | - Alejandro Jiménez
- Research Unit, Hospital Universitario de Canarias, Ofra s/n. La Laguna, -38320, Santa Cruz de Tenerife, Spain
| | - Victor García-Marín
- Deparment of Neurosurgery, Hospital Universitario de Canarias, Ofra, s/n. La Laguna, 38320, Santa Cruz de Tenerife, Spain
| |
Collapse
|
41
|
Yamamoto Y, Hosoda K, Imahori T, Tanaka J, Matsuo K, Nakai T, Irino Y, Shinohara M, Sato N, Sasayama T, Tanaka K, Nagashima H, Kohta M, Kohmura E. Pentose phosphate pathway activation via HSP27 phosphorylation by ATM kinase: A putative endogenous antioxidant defense mechanism during cerebral ischemia-reperfusion. Brain Res 2018; 1687:82-94. [PMID: 29510140 DOI: 10.1016/j.brainres.2018.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/24/2018] [Accepted: 03/01/2018] [Indexed: 11/19/2022]
Abstract
Molecular mechanism underlying ischemic stroke remains poorly understood. We previously reported glucose 6-phosphate dehydrogenase (G6PD) activity in pentose phosphate pathway (PPP) is activated via heat shock protein 27 (HSP27) phosphorylation at serine 85 (S85) by ataxia telangiectasia mutated (ATM) kinase during cerebral ischemia. This mechanism seems to be endogenous antioxidative system. To determine whether this system also works during reperfusion, we performed comparative metabolic analysis of reperfusion effect on metabolism in rat cortex using middle cerebral artery occlusion (MCAO). Metabolic profiling using gas-chromatography/mass-spectrometry analysis showed changes in metabolic state that depended on reperfusion time. Enrichment analysis showed PPP was significantly upregulated during ischemia-reperfusion. Significant increases in fructose 6-phosphate and ribulose 5-phosphate after reperfusion also suggested enhancement of PPP. In relation to PPP, ischemia-reperfusion induced an increase of up to 69-fold in HSP27 transcripts after 24-h reperfusion. Immunoblotting showed gradual increase in HSP27 protein and marked increase in HSP27 phosphorylation (S85) that were time-dependent (4.5-fold after 24-h reperfusion). G6PD activity was significantly elevated after 1-h MCAO (20%), reduced after 1-h reperfusion, increased gradually thereafter and significantly elevated after 24-h reperfusion. The NADPH/NAD+ ratio displayed similar increasing pattern. Intracerebroventricular injection of ATM kinase inhibitor (KU-55933) significantly reduced HSP27 phosphorylation and G6PD activity, significantly increased protein carbonyl, and resulted in increase in infarct size (100%) 24-h after reperfusion following 90-min MCAO. Consequently, G6PD activation via HSP27 phosphorylation by ATM kinase may be part of endogenous antioxidant defense neuroprotection mechanism that is activated during ischemia-reperfusion. These findings have important implications for treatment of stroke.
Collapse
Affiliation(s)
- Yusuke Yamamoto
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kohkichi Hosoda
- Department of Neurosurgery, Kobe City Nishi-Kobe Medical Center, 5-7-1, Kojidai, Nishi-ku, Kobe 651-2273, Japan.
| | - Taichiro Imahori
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Jun Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kazuya Matsuo
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tomoaki Nakai
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yasuhiro Irino
- Division of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masakazu Shinohara
- Division of Epidemiology, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Naoko Sato
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takashi Sasayama
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kazuhiro Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Hiroaki Nagashima
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masaaki Kohta
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Eiji Kohmura
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
42
|
Komnig D, Gertz K, Habib P, Nolte KW, Meyer T, Brockmann MA, Endres M, Rathkolb B, Hrabě de Angelis M, Schulz JB, Falkenburger BH, Reich A. Faim2 contributes to neuroprotection by erythropoietin in transient brain ischemia. J Neurochem 2018; 145:258-270. [PMID: 29315561 DOI: 10.1111/jnc.14296] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/22/2017] [Accepted: 12/23/2017] [Indexed: 11/28/2022]
Abstract
Delayed cell death in the penumbra region of acute ischemic stroke occurs through apoptotic mechanisms, making it amenable to therapeutic interventions. Fas/CD95 mediates apoptotic cell death in response to external stimuli. In mature neurons, Fas/CD95 signaling is modulated by Fas-apoptotic inhibitory molecule 2 (Faim2), which reduces cell death in animal models of stroke, meningitis, and Parkinson disease. Erythropoietin (EPO) has been studied as a therapeutic strategy in ischemic stroke. Erythropoietin stimulates the phosphatidylinositol-3 kinase/Akt (PI3K/Akt) pathway, which regulates Faim2 expression. Therefore, up-regulation of Faim2 may contribute to neuroprotection by EPO. Male Faim2-deficient mice (Faim2-/- ) and wild-type littermates (WT) were subjected to 30 min of middle cerebral artery occlusion (MCAo) followed by 72 h of reperfusion. EPO was applied before (30 min) and after (24 and 48 h) MCAo. In WT mice application of EPO at a low dose (5000 U/kg) significantly reduced stroke volume, whereas treatment with high dose (90 000 U/kg) did not. In Faim2-/- animals administration of low-dose EPO did not result in a significant reduction in stroke volume. Faim2 expression as measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR) increased after low-dose EPO but not with high dose. An extensive phenotyping including analysis of cerebral vessel architecture did not reveal confounding differences between the genotypes. In human post-mortem brain Faim2 displayed a differential expression in areas of penumbral ischemia. Faim2 up-regulation may contribute to the neuroprotective effects of low-dose erythropoietin in transient brain ischemia. The dose-dependency may explain mixed effects of erythropoietin observed in clinical stroke trials.
Collapse
Affiliation(s)
- Daniel Komnig
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Karen Gertz
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Pardes Habib
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Kay W Nolte
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Tareq Meyer
- Department of Diagnostic and Interventional Neuroradiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Endres
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany.,Excellence Cluster NeuroCure, Berlin, Germany.,German Center for Neurodegenerative Disease (DZNE), Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Ludwig-Maximilians-Universität München, Gene Center, Institute of Molecular Animal Breeding and Biotechnology, München, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | | | - Jörg B Schulz
- Department of Neurology, RWTH Aachen University, Aachen, Germany.,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Björn H Falkenburger
- Department of Neurology, RWTH Aachen University, Aachen, Germany.,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
43
|
Demyanenko S, Neginskaya M, Berezhnaya E. Expression of Class I Histone Deacetylases in Ipsilateral and Contralateral Hemispheres after the Focal Photothrombotic Infarction in the Mouse Brain. Transl Stroke Res 2017; 9:471-483. [PMID: 29218547 DOI: 10.1007/s12975-017-0595-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/19/2022]
Abstract
Histone acetylation and deacetylation are among the most important epigenetic processes that regulate gene expression. Nonselective inhibitors of histone deacetylases (HDAC) can protect brain cells during ischemia and stroke. However, which HDAC isoform is involved in this effect is unknown. Some isoforms of histone deacetylases (HDACs) protect brain cells after ischemia, whereas others can promote their death. Most studies consider early periods (1-24 h) after stroke, whereas little is known on the involvement of HDACs during recovery after stroke. In this study, cellular and intracellular rearrangement of class I HDACs (HDAC1, HDAC2, HDAC3, HDAC8) was investigated at late periods after photothrombotic infarction (PTI) of the mouse sensorimotor cortex in intact tissue that surrounds the ischemia core, in the corresponding region of the contralateral hemisphere, and in the hippocampus. Each HDAC isoform had a specific pattern of expression and intracellular distribution in neurons and astrocytes at different periods after the ischemia. We did not observe ischemia-induced changes in the subcellular localization of HDACs under study. Three days after the PTI, the expression of HDAC2 was increased in neurons of the damaged hemisphere. The activity of HDAC2 and HDAC8 was elevated 7 days after the ischemia both in neurons and astrocytes of the studied brain structures; the activity of HDAC8 was also increased 14 days after the ischemia. It is notable that the expression of class I HDACs in the intact hemisphere changes in the same way as their expression in the living tissue of the damaged hemisphere. HDAC1 was found both in the nuclei and cytoplasm of the brain cells; HDAC2 was predominantly localized in the nuclei, and HDAC8 was predominantly observed in the cytoplasm. This in addition to the regulation of gene transcription indicates nontranscriptional activity of HDAC1 and HDAC8 during recovery of the brain tissue after the ischemia. HDAC2 and HDAC8 were identified as potential mediators in an early recovery period after stroke, suggesting that selective inhibitors and activators of HDACs can be considered for therapeutic approaches in this period.
Collapse
Affiliation(s)
- Svetlana Demyanenko
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, prospect Stachki 194/1, Rostov-on-Don, 344090, Russia.
| | - Maria Neginskaya
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, prospect Stachki 194/1, Rostov-on-Don, 344090, Russia
| | - Elena Berezhnaya
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, prospect Stachki 194/1, Rostov-on-Don, 344090, Russia
| |
Collapse
|
44
|
Li H, Wu Y, Suo G, Shen F, Zhen Y, Chen X, Lv H. Profiling neuron-autonomous lncRNA changes upon ischemia/reperfusion injury. Biochem Biophys Res Commun 2017; 495:104-109. [PMID: 29101036 DOI: 10.1016/j.bbrc.2017.10.157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/29/2017] [Indexed: 11/18/2022]
Abstract
Extensive changes of neuronal transcriptome occur post ischemic stroke and during the following reperfusion. Although numerous studies focused on transcriptome changes of mRNAs associated with ischemic stroke, little is known about whether and how long non-coding RNAs (lncRNAs), which play critical roles in cellular homeostasis, are involved in this process. In this study, we performed high throughput screening to analyze expression changes of lncRNAs in primarily cultured hippocampal neurons under an oxygen-glucose deprivation/reperfusion (OGD/R) condition at 0 h, 6 h, 12 h, and 18 h, respectively. Knock down of one validated lncRNAs (Tnxa-ps1) promoted neuronal survival by inhibiting apoptosis. Coding non-coding co-expression network analysis revealed that the expression of Tnxa-ps1 was highly correlated with changes of a particular group of genes, many of which are associated with neural protection. Finally, we showed that down-regulation of Tnxa-ps1 reversed the expression changes of four mRNAs post OGD/R, revealing a regulatory effect between Tnxa-ps1 and selected genes. Together, our data revealed possible participation of lncRNAs in the pathophysiology of OGD/R and thereby provided new insights into the studies of potential therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Haiying Li
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, PR China; Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Youjia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Guihai Suo
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Feifei Shen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Yuqin Zhen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Xia Chen
- Basic Medical Research Center, Medical School, Nantong University, Nantong, Jiangsu 226001, PR China.
| | - Haitao Lv
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, PR China.
| |
Collapse
|
45
|
Grell AS, Frederiksen SD, Edvinsson L, Ansar S. Cerebrovascular gene expression in spontaneously hypertensive rats. PLoS One 2017; 12:e0184233. [PMID: 28880918 PMCID: PMC5589213 DOI: 10.1371/journal.pone.0184233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/21/2017] [Indexed: 11/22/2022] Open
Abstract
Hypertension is a hemodynamic disorder and one of the most important and well-established risk factors for vascular diseases such as stroke. Blood vessels exposed to chronic shear stress develop structural changes and remodeling of the vascular wall through many complex mechanisms. However, the molecular mechanisms involved are not fully understood. Hypertension-susceptible genes may provide a novel insight into potential molecular mechanisms of hypertension and secondary complications associated with hypertension. The aim of this exploratory study was to identify gene expression differences in the middle cerebral arteries between 12-week-old male spontaneously hypertensive rats and their normotensive Wistar-Kyoto rats using an Affymetrix whole-transcriptome expression profiling. Quantitative PCR and western blotting were used to verify genes of interest. 169 genes were differentially expressed in the middle cerebral arteries from hypertensive compared to normotensive rats. The gene expression of 72 genes was decreased and the gene expression of 97 genes was increased. The following genes with a fold difference ≥1.40 were verified by quantitative PCR; Postn, Olr1, Fas, Vldlr, Mmp2, Timp1, Serpine1, Mmp11, Cd34, Ptgs1 and Ptgs2. The gene expression of Postn, Olr1, Fas, Vldlr, Mmp2, Timp1 and Serpine1 and the protein expression of LOX1 (also known as OLR1) were significantly increased in the middle cerebral arteries from spontaneously hypertensive rats compared to Wistar-Kyoto rats. In conclusion, the identified genes in the middle cerebral arteries from spontaneously hypertensive rats could be possible mediators of the vascular changes and secondary complications associated with hypertension. This study supports the selection of key genes to investigate in the future research of hypertension-induced end-organ damage.
Collapse
Affiliation(s)
- Anne-Sofie Grell
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet Glostrup, Glostrup, Denmark
- * E-mail:
| | - Simona Denise Frederiksen
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet Glostrup, Glostrup, Denmark
- Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Saema Ansar
- Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
46
|
Imahori T, Hosoda K, Nakai T, Yamamoto Y, Irino Y, Shinohara M, Sato N, Sasayama T, Tanaka K, Nagashima H, Kohta M, Kohmura E. Combined metabolic and transcriptional profiling identifies pentose phosphate pathway activation by HSP27 phosphorylation during cerebral ischemia. Neuroscience 2017; 349:1-16. [PMID: 28257891 DOI: 10.1016/j.neuroscience.2017.02.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 02/10/2017] [Accepted: 02/17/2017] [Indexed: 12/28/2022]
Abstract
The metabolic pathophysiology underlying ischemic stroke remains poorly understood. To gain insight into these mechanisms, we performed a comparative metabolic and transcriptional analysis of the effects of cerebral ischemia on the metabolism of the cerebral cortex using middle cerebral artery occlusion (MCAO) rat model. Metabolic profiling by gas-chromatography/mass-spectrometry analysis showed clear separation between the ischemia and control group. The decreases of fructose 6-phosphate and ribulose 5-phosphate suggested enhancement of the pentose phosphate pathway (PPP) during cerebral ischemia (120-min MCAO) without reperfusion. Transcriptional profiling by microarray hybridization indicated that the Toll-like receptor and mitogen-activated protein kinase (MAPK) signaling pathways were upregulated during cerebral ischemia without reperfusion. In relation to the PPP, upregulation of heat shock protein 27 (HSP27) was observed in the MAPK signaling pathway and was confirmed through real-time polymerase chain reaction. Immunoblotting showed a slight increase in HSP27 protein expression and a marked increase in HSP27 phosphorylation at serine 85 after 60-min and 120-min MCAO without reperfusion. Corresponding upregulation of glucose 6-phosphate dehydrogenase (G6PD) activity and an increase in the NADPH/NAD+ ratio were also observed after 120-min MCAO. Furthermore, intracerebroventricular injection of ataxia telangiectasia mutated (ATM) kinase inhibitor (KU-55933) significantly reduced HSP27 phosphorylation and G6PD upregulation after MCAO, but that of protein kinase D inhibitor (CID755673) did not affect HSP27 phosphorylation. Consequently, G6PD activation via ischemia-induced HSP27 phosphorylation by ATM kinase may be part of an endogenous antioxidant defense neuroprotection mechanism during the earliest stages of ischemia. These findings have important therapeutic implications for the treatment of stroke.
Collapse
Affiliation(s)
- Taichiro Imahori
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kohkichi Hosoda
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | - Tomoaki Nakai
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yusuke Yamamoto
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yasuhiro Irino
- Division of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masakazu Shinohara
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; Division of Epidemiology, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Naoko Sato
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takashi Sasayama
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kazuhiro Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Hiroaki Nagashima
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masaaki Kohta
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Eiji Kohmura
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
47
|
Martín-Aragón Baudel MAS, Poole AV, Darlison MG. Chloride co-transporters as possible therapeutic targets for stroke. J Neurochem 2016; 140:195-209. [PMID: 27861901 DOI: 10.1111/jnc.13901] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/08/2016] [Accepted: 11/08/2016] [Indexed: 02/06/2023]
Abstract
Stroke is one of the major causes of death and disability worldwide. The major type of stroke is an ischaemic one, which is caused by a blockage that interrupts blood flow to the brain. There are currently very few pharmacological strategies to reduce the damage and social burden triggered by this pathology. The harm caused by the interruption of blood flow to the brain unfolds in the subsequent hours and days, so it is critical to identify new therapeutic targets that could reduce neuronal death associated with the spread of the damage. Here, we review some of the key molecular mechanisms involved in the progression of neuronal death, focusing on some new and promising studies. In particular, we focus on the potential of the chloride co-transporter (CCC) family of proteins, mediators of the GABAergic response, both during the early and later stages of stroke, to promote neuroprotection and recovery. Different studies of CCCs during the chronic and recovery phases post-stroke reveal the importance of timing when considering CCCs as potential neuroprotective and/or neuromodulator targets. The molecular regulatory mechanisms of the two main neuronal CCCs, NKCC1 and KCC2, are further discussed as an indirect approach for promoting neuroprotection and neurorehabilitation following an ischaemic insult. Finally, we mention the likely importance of combining different strategies in order to achieve more effective therapies.
Collapse
Affiliation(s)
| | - Amy V Poole
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, UK
| | - Mark G Darlison
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, UK
| |
Collapse
|
48
|
Diaz-Ruiz A, Roldan-Valadez E, Ortiz-Plata A, Mondragón-Lozano R, Heras-Romero Y, Mendez-Armenta M, Osorio-Rico L, Nava-Ruiz C, Ríos C. Dapsone improves functional deficit and diminishes brain damage evaluated by 3-Tesla magnetic resonance image after transient cerebral ischemia and reperfusion in rats. Brain Res 2016; 1646:384-392. [PMID: 27321157 DOI: 10.1016/j.brainres.2016.06.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/13/2016] [Accepted: 06/15/2016] [Indexed: 02/08/2023]
|
49
|
Chelluboina B, Klopfenstein JD, Pinson DM, Wang DZ, Vemuganti R, Veeravalli KK. Matrix Metalloproteinase-12 Induces Blood–Brain Barrier Damage After Focal Cerebral Ischemia. Stroke 2015; 46:3523-31. [DOI: 10.1161/strokeaha.115.011031] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/07/2015] [Indexed: 12/12/2022]
Abstract
Background and Purpose—
Matrix metalloproteinases (MMPs) have a central role in compromising the integrity of the blood–brain barrier (BBB). The role of MMP-12 in brain damage after ischemic stroke remains unknown. The main objective of the current study is to investigate the effect of MMP-12 suppression at an early time point before reperfusion on the BBB damage in rats.
Methods—
Sprague–Dawley rats were subjected to middle cerebral artery occlusion and reperfusion. MMP-12 shRNA–expressing plasmids formulated as nanoparticles were administered at a dose of 1 mg/kg body weight. The involvement of MMP-12 on BBB damage was assessed by performing various techniques, including Evans blue dye extravasation, 2,3,5-triphenyltetrazolium chloride staining, immunoblot, gelatin zymography, and immunofluorescence analysis.
Results—
MMP-12 is upregulated ≈31-, 47-, and 66-fold in rats subjected 1–, 2-, or 4-hour ischemia, respectively, followed by 1-day reperfusion. MMP-12 suppression protected the BBB integrity by inhibiting the degradation of tight-junction proteins. Either intravenous or intra-arterial delivery of MMP-12 shRNA-expressing plasmid significantly reduced the percent Evans blue dye extravasation and infarct size. Furthermore, MMP-12 suppression reduced the endogenous levels of other proteases, such as tissue-type plasminogen activator and MMP-9, which are also known to be the key players involved in BBB damage.
Conclusions—
These results demonstrate the adverse role of MMP-12 in acute brain damage that occurs after ischemic stroke and, thereby, suggesting that MMP-12 suppression could be a promising therapeutic target for cerebral ischemia.
Collapse
Affiliation(s)
- Bharath Chelluboina
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Jeffrey D. Klopfenstein
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - David M. Pinson
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - David Z. Wang
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Raghu Vemuganti
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Krishna Kumar Veeravalli
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| |
Collapse
|
50
|
Zhang Y, Lin P, Jiang H, Xu J, Luo S, Mo J, Li Y, Chen X. Extensive serum biomarker analysis in patients with ST segment elevation myocardial infarction (STEMI). Cytokine 2015; 76:356-362. [PMID: 26153394 DOI: 10.1016/j.cyto.2015.06.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/10/2015] [Accepted: 06/25/2015] [Indexed: 11/16/2022]
Abstract
ST segment elevation myocardial infarction (STEMI) is one of the leading causes of morbidity and mortality and some characteristics of STEMI are poorly understood. The aim of the present study is to detect protein expression profiles in the serum of STEMI patients, and to identify biomarkers for this disease. Cytokine profiles of serum from STEMI patients and healthy controls were analyzed with a semi-quantitative human antibody array for 174 proteins, and the results showed blood serum concentrations of 21 cytokines differed considerably between STEMI patients and healthy subjects. In the next phase, a sandwich ELISA kit individually validated eight biomarker results from 21 of the microarray experiments. Clinical validation demonstrated a significant increase of BNDF, PDGF-AA and MMP-9 in patients with AMI. Meanwhile, BNDF, PDGF-AA and MMP-9 distinguished AMI patients from healthy controls with a mean area under the receiver operating characteristic (ROC) curves of 0.870, 0.885, and 0.81, respectively, with diagnostic cut-off points of 0.688 ng/mL, 297.86 ng/mL and 690.066 ng/mL. Our study indicated that these three cytokines were up-regulated in STEMI samples, and may hold promise for the assessment of STEMI.
Collapse
Affiliation(s)
- Yi Zhang
- The Second Affiliated Hospital of Guangzhou Medical University, 250 Changangdong Rd., Guangzhou, Guangdong 510260, People's Republic of China
| | - Peiyi Lin
- The Second Affiliated Hospital of Guangzhou Medical University, 250 Changangdong Rd., Guangzhou, Guangdong 510260, People's Republic of China
| | - Huilin Jiang
- The Second Affiliated Hospital of Guangzhou Medical University, 250 Changangdong Rd., Guangzhou, Guangdong 510260, People's Republic of China
| | - Jieling Xu
- The Second Affiliated Hospital of Guangzhou Medical University, 250 Changangdong Rd., Guangzhou, Guangdong 510260, People's Republic of China
| | - Shuhong Luo
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University, 1838 N. Guangzhou Avenue, Guangzhou, Guangdong 510515, People's Republic of China
| | - Junrong Mo
- The Second Affiliated Hospital of Guangzhou Medical University, 250 Changangdong Rd., Guangzhou, Guangdong 510260, People's Republic of China
| | - Yunmei Li
- The Second Affiliated Hospital of Guangzhou Medical University, 250 Changangdong Rd., Guangzhou, Guangdong 510260, People's Republic of China
| | - Xiaohui Chen
- The Second Affiliated Hospital of Guangzhou Medical University, 250 Changangdong Rd., Guangzhou, Guangdong 510260, People's Republic of China.
| |
Collapse
|