1
|
Park JH, Shin MJ, Youn GS, Yeo HJ, Yeo EJ, Kwon HJ, Lee LR, Kim NY, Kwon SY, Kim SM, Cho YJ, Lee SH, Jung HY, Kim DW, Eum WS, Choi SY. PEP-1-PIN1 Promotes Hippocampal Neuronal Cell Survival by Inhibiting Cellular ROS and MAPK Phosphorylation. Biomedicines 2024; 12:2352. [PMID: 39457664 PMCID: PMC11504513 DOI: 10.3390/biomedicines12102352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Background: The peptidyl-prolyl isomerase (PIN1) plays a vital role in cellular processes, including intracellular signaling and apoptosis. While oxidative stress is considered one of the primary mechanisms of pathogenesis in brain ischemic injury, the precise function of PIN1 in this disease remains to be elucidated. Objective: We constructed a cell-permeable PEP-1-PIN1 fusion protein and investigated PIN1's function in HT-22 hippocampal cells as well as in a brain ischemic injury gerbil model. Methods: Transduction of PEP-1-PIN1 into HT-22 cells and signaling pathways were determined by Western blot analysis. Intracellular reactive oxygen species (ROS) production and DNA damage was confirmed by DCF-DA and TUNEL staining. Cell viability was determined by MTT assay. Protective effects of PEP-1-PIN1 against ischemic injury were examined using immunohistochemistry. Results: PEP-1-PIN1, when transduced into HT-22 hippocampal cells, inhibited cell death in H2O2-treated cells and markedly reduced DNA fragmentation and ROS production. This fusion protein also reduced phosphorylation of mitogen-activated protein kinase (MAPK) and modulated expression levels of apoptosis-signaling proteins in HT-22 cells. Furthermore, PEP-1-PIN1 was distributed in gerbil hippocampus neuronal cells after passing through the blood-brain barrier (BBB) and significantly protected against neuronal cell death and also decreased activation of microglia and astrocytes in an ischemic injury gerbil model. Conclusions: These results indicate that PEP-1-PIN1 can inhibit ischemic brain injury by reducing cellular ROS levels and regulating MAPK and apoptosis-signaling pathways, suggesting that PIN1 plays a protective role in H2O2-treated HT-22 cells and ischemic injury gerbil model.
Collapse
Affiliation(s)
- Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Gi Soo Youn
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Hyun Jung Kwon
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Lee Re Lee
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Na Yeon Kim
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Su Yeon Kwon
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Su Min Kim
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University Medical Center, Chuncheon 24253, Republic of Korea;
| | - Sung Ho Lee
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
- Genesen Inc., Teheran-ro, Gangnam-gu, Seoul 06181, Republic of Korea
| | - Hyo Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Republic of Korea;
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea; (J.H.P.); (M.J.S.); (G.S.Y.); (H.J.Y.); (E.J.Y.); (H.J.K.); (L.R.L.); (N.Y.K.); (S.Y.K.); (S.M.K.); (S.H.L.)
| |
Collapse
|
2
|
Yeo EJ, Shin MJ, Youn GS, Park JH, Yeo HJ, Kwon HJ, Lee LR, Kim NY, Kwon SY, Kim SM, Lee J, Lee KW, Lee CH, Cho YJ, Kwon OS, Kim DW, Jung HY, Eum WS, Choi SY. Tat-RAN attenuates brain ischemic injury in hippocampal HT-22 cells and ischemia animal model. Neurochem Int 2023; 167:105538. [PMID: 37207854 DOI: 10.1016/j.neuint.2023.105538] [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: 02/16/2023] [Revised: 04/30/2023] [Accepted: 05/07/2023] [Indexed: 05/21/2023]
Abstract
Oxidative stress plays a key role in the pathogenesis of neuronal injury, including ischemia. Ras-related nuclear protein (RAN), a member of the Ras superfamily, involves in a variety of biological roles, such as cell division, proliferation, and signal transduction. Although RAN reveals antioxidant effect, its precise neuroprotective mechanisms are still unclear. Therefore, we investigated the effects of RAN on HT-22 cell which were exposed to H2O2-induced oxidative stress and ischemia animal model by using the cell permeable Tat-RAN fusion protein. We showed that Tat-RAN transduced into HT-22 cells, and markedly inhibited cell death, DNA fragmentation, and reactive oxygen species (ROS) generation under oxidative stress. This fusion protein also controlled cellular signaling pathways, including mitogen-activated protein kinases (MAPKs), NF-κB, and apoptosis (Caspase-3, p53, Bax and Bcl-2). In the cerebral forebrain ischemia animal model, Tat-RAN significantly inhibited both neuronal cell death, and astrocyte and microglia activation. These results indicate that RAN significantly protects against hippocampal neuronal cell death, suggesting Tat-RAN will help to develop the therapies for neuronal brain diseases including ischemic injury.
Collapse
Affiliation(s)
- Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Gi Soo Youn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Hyun Jung Kwon
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Lee Re Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Na Yeon Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Su Yeon Kwon
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Su Min Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Jaehak Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Chan Hee Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University Medical Center, Chuncheon, 24253, South Korea
| | - Oh-Shin Kwon
- School of Life Sciences, College of Natural Sciences Kyungpook National University, Taegu, 41566, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Hyo Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, South Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
3
|
Kim W, Kwon HJ, Jung HY, Hahn KR, Yoon YS, Hwang IK, Choi SY, Kim DW. Neuroprotective Effects of Purpurin Against Ischemic Damage via MAPKs, Bax, and Oxidative Stress Cascades in the Gerbil Hippocampus. Mol Neurobiol 2022; 59:2580-2592. [PMID: 35094304 PMCID: PMC9016019 DOI: 10.1007/s12035-021-02642-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/07/2021] [Indexed: 11/24/2022]
Abstract
Purpurin has various effects, including anti-inflammatory effects, and can efficiently cross the blood-brain barrier. In the present study, we investigated the effects of purpurin on oxidative stress in HT22 cells and mild brain damage in the gerbil hippocampal CA1 region induced by transient forebrain ischemia. Oxidative stress induced by H2O2 was significantly ameliorated by treatment with purpurin, based on changes in cell death, DNA fragmentation, formation of reactive oxygen species, and pro-apoptotic (Bax)/anti-apoptotic (Bcl-2) protein levels. In addition, treatment with purpurin significantly reduced the phosphorylation of c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase 1/2 (ERK), and p38 signaling in HT22 cells. Transient forebrain ischemia in gerbils led to a significant increase in locomotor activity 1 day after ischemia and significant decrease in number of surviving cells in the CA1 region 4 days after ischemia. Administration of purpurin reduced the travel distance 1 day after ischemia and abrogates the neuronal death in the hippocampal CA1 region 4 days after ischemia based on immunohistochemical and histochemical staining for NeuN and Fluoro-Jade C, respectively. Purpurin treatment significantly decreased the activation of microglia and astrocytes as well as the increases of nuclear factor kappa-light-chain-enhancer of activated B cells p65 in the hippocampal CA1 region 4 days after ischemia and ameliorated the ischemia-induced transient increases of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in the hippocampus 6 h after ischemia. In addition, purpurin significantly alleviated the ischemia-induced phosphorylation of JNK, ERK, and p38 in the hippocampus 1 day after ischemia. Furthermore, purpurin treatment significantly mitigated the increases of Bax in the hippocampus 1 day after ischemia and the lipid peroxidation based on malondialdehyde and hydroperoxides levels 2 days after ischemia. These results suggest that purpurin can be one of the potential candidates to reduce neuronal damage and inflammatory responses after oxidative stress in HT22 cells or ischemic damage in gerbils.
Collapse
Affiliation(s)
- Woosuk Kim
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
- Department of Anatomy, College of Veterinary Medicine, and Veterinary Science Research Institute, Konkuk University, Seoul, 05030, South Korea
| | - Hyun Jung Kwon
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, South Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Soo Young Choi
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea.
| |
Collapse
|
4
|
Park JH, Kim DW, Shin MJ, Park J, Han KH, Lee KW, Park JK, Choi YJ, Yeo HJ, Yeo EJ, Sohn EJ, Kim HC, Shin EJ, Cho SW, Kim DS, Cho YJ, Eum WS, Choi SY. Tat-indoleamine 2,3-dioxygenase 1 elicits neuroprotective effects on ischemic injury. BMB Rep 2020. [PMID: 32684242 PMCID: PMC7704220 DOI: 10.5483/bmbrep.2020.53.11.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
It is well known that oxidative stress participates in neuronal cell death caused production of reactive oxygen species (ROS). The increased ROS is a major contributor to the development of ischemic injury. Indoleamine 2,3-dioxygenase 1 (IDO-1) is involved in the kynurenine pathway in tryptophan metabolism and plays a role as an anti-oxidant. However, whether IDO-1 would inhibit hippocampal cell death is poorly known. Therefore, we explored the effects of cell permeable Tat-IDO-1 protein against oxidative stress-induced HT-22 cells and in a cerebral ischemia/reperfusion injury model. Transduced Tat-IDO-1 reduced cell death, ROS production, and DNA fragmentation and inhibited mitogen-activated protein kinases (MAPKs) activation in H2O2 exposed HT-22 cells. In the cerebral ischemia/reperfusion injury model, Tat-IDO-1 transduced into the brain and passing by means of the blood-brain barrier (BBB) significantly prevented hippocampal neuronal cell death. These results suggest that Tat-IDO-1 may present an alternative strategy to improve from the ischemic injury.
Collapse
Affiliation(s)
- Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jong Kook Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Eun Jeong Sohn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Duk-Soo Kim
- Department of Anatomy and BK21 Plus Center, College of Medicine, Soonchunhyang University, Cheonan 31538, Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University Medical Center, Chuncheon 24253, Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
5
|
Tat-Biliverdin Reductase A Exerts a Protective Role in Oxidative Stress-Induced Hippocampal Neuronal Cell Damage by Regulating the Apoptosis and MAPK Signaling. Int J Mol Sci 2020; 21:ijms21082672. [PMID: 32290442 PMCID: PMC7215548 DOI: 10.3390/ijms21082672] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) is major risk factor in neuronal diseases including ischemia. Although biliverdin reductase A (BLVRA) plays a pivotal role in cell survival via its antioxidant function, its role in hippocampal neuronal (HT-22) cells and animal ischemic injury is not clearly understood yet. In this study, the effects of transducible fusion protein Tat-BLVRA on H2O2-induced HT-22 cell death and in an animal ischemia model were investigated. Transduced Tat-BLVRA markedly inhibited cell death, DNA fragmentation, and generation of ROS. Transduced Tat-BLVRA inhibited the apoptosis and mitogen activated protein kinase (MAPK) signaling pathway and it passed through the blood-brain barrier (BBB) and significantly prevented hippocampal cell death in an ischemic model. These results suggest that Tat-BLVRA provides a possibility as a therapeutic molecule for ischemia.
Collapse
|
6
|
Yeo HJ, Shin MJ, Yeo EJ, Choi YJ, Kim DW, Kim DS, Eum WS, Choi SY. Tat-CIAPIN1 inhibits hippocampal neuronal cell damage through the MAPK and apoptotic signaling pathways. Free Radic Biol Med 2019; 135:68-78. [PMID: 30818058 DOI: 10.1016/j.freeradbiomed.2019.02.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 01/26/2023]
Abstract
Cytokine-induced apoptosis inhibitor 1 (CIAPIN1) protein is widely expressed in the brain and it is known that this protein is involved in cell survival including dopaminergic neuronal cells. Oxidative stress is known as one of the major causes of degenerative diseases including ischemia. In this study, we investigated the effect of CIAPIN1 protein on hippocampal neuronal (HT-22) cell damage induced by hydrogen peroxide (H2O2) and in an animal model of ischemia using Tat-CIAPIN1 fusion protein which can transduce into cells. Tat-CIAPIN1 protein transduced into HT-22 cells and significantly inhibited cell death, DNA fragmentation, and reactive oxygen species (ROS) generation. Also, Tat-CIAPIN1 protein enhances cell survival via the regulation of Akt, MAPK, NF-κB and apoptotic signaling pathways in the H2O2 treated cells. In an ischemic animal model, Tat-CIAPIN1 protein transduced into the brain and protected neuronal cell death of hippocampal CA1 region induced by ischemic insult. In conclusion, we demonstrated that Tat-CIAPIN1 protein has protective effects against hippocampal neuronal cell damage induced by ischemic injury, suggesting that Tat-CIAPIN1 protein may provide a potential therapeutic agent for ischemia.
Collapse
Affiliation(s)
- Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan-Si, 31538, South Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
7
|
Kim SM, Hwang IK, Yoo DY, Eum WS, Kim DW, Shin MJ, Ahn EH, Jo HS, Ryu EJ, Yong JI, Cho SW, Kwon OS, Lee KW, Cho YS, Han KH, Park J, Choi SY. Tat-antioxidant 1 protects against stress-induced hippocampal HT-22 cells death and attenuate ischaemic insult in animal model. J Cell Mol Med 2015; 19:1333-45. [PMID: 25781353 PMCID: PMC4459847 DOI: 10.1111/jcmm.12513] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/14/2014] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress-induced reactive oxygen species (ROS) are responsible for various neuronal diseases. Antioxidant 1 (Atox1) regulates copper homoeostasis and promotes cellular antioxidant defence against toxins generated by ROS. The roles of Atox1 protein in ischaemia, however, remain unclear. In this study, we generated a protein transduction domain fused Tat-Atox1 and examined the roles of Tat-Atox1 in oxidative stress-induced hippocampal HT-22 cell death and an ischaemic injury animal model. Tat-Atox1 effectively transduced into HT-22 cells and it protected cells against the effects of hydrogen peroxide (H2O2)-induced toxicity including increasing of ROS levels and DNA fragmentation. At the same time, Tat-Atox1 regulated cellular survival signalling such as p53, Bad/Bcl-2, Akt and mitogen-activate protein kinases (MAPKs). In the animal ischaemia model, transduced Tat-Atox1 protected against neuronal cell death in the hippocampal CA1 region. In addition, Tat-Atox1 significantly decreased the activation of astrocytes and microglia as well as lipid peroxidation in the CA1 region after ischaemic insult. Taken together, these results indicate that transduced Tat-Atox1 protects against oxidative stress-induced HT-22 cell death and against neuronal damage in animal ischaemia model. Therefore, we suggest that Tat-Atox1 has potential as a therapeutic agent for the treatment of oxidative stress-induced ischaemic damage.
Collapse
Affiliation(s)
- So Mi Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangnung-Wonju National University, Kangneung, Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Eun Hee Ahn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Hyo Sang Jo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Eun Ji Ryu
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Ji In Yong
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Korea
| | - Oh-Shin Kwon
- Department of Biochemistry, School of Life Sciences & Biotechnology, Kyungpook National University, Taegu, Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Yoon Shin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| |
Collapse
|
8
|
Yang XY, Liu QN, Zhang L, Jiang SQ, Gong PL. Neuroprotective Effect of Dauricine after Transient Middle Cerebral Artery Occlusion in Rats: Involvement of Bcl-2 Family Proteins. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 38:307-18. [DOI: 10.1142/s0192415x10007865] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Previous studies have shown that the bisbenzyl isoquinoline alkaloid dauricine can protect the brain against ischemic damage. We investigated here whether dauricine could inhibit neuronal apoptosis and modulate Bcl-2 family protein levels in a rat model of transient focal cerebral ischemia. Male Sprague-Dawley rats underwent a 60 min temporary occlusion of the middle cerebral artery (MCAO). Two doses of dauricine (5 and 10 mg/kg as low and high dose respectively) were administered intraperitoneally at 1 hour after MCAO. After neurological deficits were assessed at 3 hours and 24 hours of reperfusion, rats were killed and brain samples were collected. Apoptotic changes were evaluated by TUNEL method. The immunohistochemistry and Western blot were used to assess the protein expressions of Bcl-2 and Bax. RT-PCR was used to determine Bcl-2 and Bax mRNA expressions. Dauricine (5 and 10 mg/kg) treatment improved neurological deficits, diminished DNA fragmentation, increased Bcl-2 expression and reduced Bax expression in the penumbra. The infarct-reducing effects of dauricine may be due, in part, to the inhibition of apoptotic cell death via modulation Bcl-2 family protein in the penumbra.
Collapse
Affiliation(s)
- Xiao-Yan Yang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang-Ni Liu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zhang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Qin Jiang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei-Li Gong
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Wappler E, Gál A, Skopál J, Nagy Z. Single, high-dose 17β-estradiol therapy has anti-apoptotic effect and induces cerebral plasticity following transient forebrain ischemia in gerbils (Short communication). ACTA ACUST UNITED AC 2011; 98:189-94. [DOI: 10.1556/aphysiol.98.2011.2.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
10
|
Yamagata K, Ichinose S, Miyashita A, Tagami M. Protective effects of ebselen, a seleno-organic antioxidant on neurodegeneration induced by hypoxia and reperfusion in stroke-prone spontaneously hypertensive rat. Neuroscience 2008; 153:428-35. [DOI: 10.1016/j.neuroscience.2008.02.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 02/04/2008] [Accepted: 02/18/2008] [Indexed: 12/17/2022]
|
11
|
Nakka VP, Gusain A, Mehta SL, Raghubir R. Molecular mechanisms of apoptosis in cerebral ischemia: multiple neuroprotective opportunities. Mol Neurobiol 2008; 37:7-38. [PMID: 18066503 DOI: 10.1007/s12035-007-8013-9] [Citation(s) in RCA: 216] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Accepted: 11/05/2007] [Indexed: 12/18/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury triggers multiple and distinct but overlapping cell signaling pathways, which may lead to cell survival or cell damage. There is overwhelming evidence to suggest that besides necrosis, apoptosis do contributes significantly to the cell death subsequent to I/R injury. Both extrinsic and intrinsic apoptotic pathways play a vital role, and upon initiation, these pathways recruit downstream apoptotic molecules to execute cell death. Caspases and Bcl-2 family members appear to be crucial in regulating multiple apoptotic cell death pathways initiated during I/R. Similarly, inhibitor of apoptosis family of proteins (IAPs), mitogen-activated protein kinases, and newly identified apoptogenic molecules, like second mitochondrial-activated factor/direct IAP-binding protein with low pI (Smac/Diablo), omi/high-temperature requirement serine protease A2 (Omi/HtrA2), X-linked mammalian inhibitor of apoptosis protein-associated factor 1, and apoptosis-inducing factor, have emerged as potent regulators of cellular apoptotic/antiapoptotic machinery. All instances of cell survival/death mechanisms triggered during I/R are multifaceted and interlinked, which ultimately decide the fate of brain cells. Moreover, apoptotic cross-talk between major subcellular organelles suggests that therapeutic strategies should be optimally directed at multiple targets/mechanisms for better therapeutic outcome. Based on the current knowledge, this review briefly focuses I/R injury-induced multiple mechanisms of apoptosis, involving key apoptotic regulators and their emerging roles in orchestrating cell death programme. In addition, we have also highlighted the role of autophagy in modulating cell survival/death during cerebral ischemia. Furthermore, an attempt has been made to provide an encouraging outlook on emerging therapeutic approaches for cerebral ischemia.
Collapse
Affiliation(s)
- Venkata Prasuja Nakka
- Division of Pharmacology, Central Drug Research Institute, Chatter Manzil Palace, POB-173, Lucknow, 226001, India
| | | | | | | |
Collapse
|
12
|
Müller GJ, Lassmann H, Johansen FF. Anti-apoptotic signaling and failure of apoptosis in the ischemic rat hippocampus. Neurobiol Dis 2007; 25:582-93. [PMID: 17207631 DOI: 10.1016/j.nbd.2006.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2006] [Revised: 10/03/2006] [Accepted: 11/03/2006] [Indexed: 11/18/2022] Open
Abstract
Several anti-apoptotic proteins are induced in CA1 neurons after transient forebrain ischemia (TFI), but fail to protect the majority of these cells from demise. Correlating cell death morphologies (apoptosis-like and necrosis-like death) with immunohistochemistry (IHC), we investigated whether anti-apoptosis contributes to survival, compromises apoptosis effector functions and/or delays death in CA1 neurons 1-7 days after TFI. As surrogate markers for bioenergetic failure, the IHC of respiratory chain complex (RCC) subunits was investigated. Dentate granule cell (DGC) apoptosis following colchicine injection severed as a reference for classical apoptosis. Heat shock protein 70 (Hsp70), neuronal apoptosis inhibitory protein (NAIP) and manganese superoxide dismutase (MnSOD) were upregulated in the majority of intact CA1 neurons paralleling the occurrence of CA1 neuronal death (days 3-7) as well as in a proportion of apoptosis-(<50%) and necrosis-like (<30%) CA1 neurons. Colchicine did not provoke an anti-apoptotic response in DGC at all. In addition, more than 70% of apoptosis- and necrosis-like CA1 neurons had completely lost their RCC subunits suggesting bioenergetic failure; by contrast, following colchicine injection, 88% of all apoptotic DGC presented RCC subunits. Thus, anti-apoptotic proteins may, in a subset of ischemic CA1 neurons, prevent cell death, while in others, affected by pronounced energy failure, they may cause secondary necrosis.
Collapse
Affiliation(s)
- Georg Johannes Müller
- Molecular Neuropathology Group, University of Copenhagen, 11, Frederik V's vej, 2100-Copenhagen-O, Denmark
| | | | | |
Collapse
|
13
|
Portier BP, Taglialatela G. Bcl-2 localized at the nuclear compartment induces apoptosis after transient overexpression. J Biol Chem 2006; 281:40493-502. [PMID: 17090549 DOI: 10.1074/jbc.m606181200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Bcl-2 is the best characterized member of a large family of proteins that regulate apoptosis. Although it is established that Bcl-2 localized at the mitochondria functions as an anti-apoptotic protein, the function of Bcl-2 at the nucleus remains unclear. Recently we showed that nuclear compartment-associated Bcl-2 inhibits transcription factor activation. Based on this observation, we hypothesized that presence of Bcl-2 at the nucleus may induce rather than protect cells from apoptosis. Here we investigated the putative apoptotic role of nuclear compartment-associated Bcl-2. Additionally, we examined the role of the Bcl-2 BH4 domain in mediating binding to FKBP38, the Bcl-2 mitochondrial chaperone. Our results demonstrate a novel, pro-apoptotic function for nuclear Bcl-2 and identify the Bcl-2 BH4 domain as a key regulator in mediating Bcl-2/FKBP38 binding. These results indicate that Bcl-2 has a dual role as both a protector and a killer and that the ability to switch roles depends on Bcl-2 subcellular localization.
Collapse
Affiliation(s)
- Bryce Patrick Portier
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-1043, USA
| | | |
Collapse
|
14
|
Gerber J, Brück W, Stadelmann C, Bunkowski S, Lassmann H, Nau R. Expression of death-related proteins in dentate granule cells in human bacterial meningitis. Brain Pathol 2006; 11:422-31. [PMID: 11556687 PMCID: PMC8098385 DOI: 10.1111/j.1750-3639.2001.tb00410.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Neuronal apoptosis in the dentate gyrus has been observed in animal models of bacterial meningitis and in humans dying in the course of the disease. To evaluate the mechanisms of neuronal cell death, hippocampal sections of 20 patients dying from bacterial meningitis were investigated by immunohistochemistry using antibodies against the proform of caspase-3 and the active enzyme, bcl-2, bax and p53. In the dentate granule cell layer, the median density of neurons with an apoptotic morphology was 7.6/mm2 (0-15.6/mm2). The median density of immunoreactive neurons was 2.3/mm2 (procaspase-3), 0.9/mm2 (activated caspase-3), 1.8/mm2 (bcl-2), 1.1/mm2 (bax) and 0.4/mm2 (p53). 80% of neurons immunoreactive for active caspase-3 had an apoptotic morphology, whereas only 10% of all procaspase-3 stained neurons showed signs of apoptosis. Apoptotic cell death is present in humans dying in the course of bacterial meningitis in the dentate gyrus of the Formatio hippocampi. Neuronal expression of caspase-3, bcl-2 and bax suggests an involvement of these proteins in neuronal death.
Collapse
Affiliation(s)
- Joachim Gerber
- Department of Neurology, Georg‐August‐Universität Göttingen, Germany
| | - Wolfgang Brück
- Department of Neuropathology, Charité, Humboldt‐Universität, Berlin, Germany
| | | | | | - Hans Lassmann
- Institute of Brain Research, Department of Neuroimmunology, University of Vienna, Austria
| | - Roland Nau
- Department of Neurology, Georg‐August‐Universität Göttingen, Germany
| |
Collapse
|
15
|
Uchino H, Morota S, Takahashi T, Ikeda Y, Kudo Y, Ishii N, Siesjö BK, Shibasaki F. A novel neuroprotective compound FR901459 with dual inhibition of calcineurin and cyclophilins. ACTA NEUROCHIRURGICA. SUPPLEMENT 2006; 96:157-62. [PMID: 16671446 DOI: 10.1007/3-211-30714-1_35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Brain ischemia leads to severe damage in the form of delayed neuronal cell death. In our study, we show that the marked neuroprotection of the new immunosuppressant FR901495 in forebrain ischemia is due not only to inhibition of calcineurin, but also to protection against mitochondrial damage caused by mitochondrial permeability transition pore formation through cyclophilin D, one of the prolyl cis/trans isomerase family members. These findings shed light on the clinical application and development of new drugs for the treatment of ischemic damage in the brain as well as in the heart and liver.
Collapse
Affiliation(s)
- H Uchino
- Department of Anesthesiology, Hachioji Medical Center, Tokyo Medical University, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Lindenboim L, Kringel S, Braun T, Borner C, Stein R. Bak but not Bax is essential for Bcl-xS-induced apoptosis. Cell Death Differ 2005; 12:713-23. [PMID: 15861188 DOI: 10.1038/sj.cdd.4401638] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Bcl-x(S), a proapoptotic member of the Bcl-2 protein family, is localized in the mitochondria and induces apoptosis in a caspase- and BH3-dependent manner by a mechanism involving cytochrome c release. The way in which Bcl-x(S) induces caspase activation and cytochrome c release, as well as the relationship between Bcl-x(S) and other proapoptotic members of the Bcl-2 family, is not known. Here we used embryonic fibroblasts derived from mice deficient in the multidomain proapoptotic members of the Bcl-2 family (Bax and Bak) and the apoptotic components of the apoptosome (Apaf-1 and caspase-9) to unravel the cascade of events by which Bcl-x(S) promotes apoptosis. Our results show that Bak but not Bax is essential for Bcl-x(S)-induced apoptosis. Bcl-x(S) induced activation of Bak, which in turn promoted apoptosis by apoptosome-dependent and -independent pathways. These findings provide the first evidence that a proapoptotic Bcl-2 family protein induces apoptosis exclusively via Bak.
Collapse
Affiliation(s)
- L Lindenboim
- Department of Neurobiochemistry, George S Wise Faculty of Life Sciences, Tel Aviv University, 69978 Ramat Aviv, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
17
|
Vis JC, Schipper E, de Boer-van Huizen RT, Verbeek MM, de Waal RMW, Wesseling P, ten Donkelaar HJ, Kremer B. Expression pattern of apoptosis-related markers in Huntington's disease. Acta Neuropathol 2005; 109:321-8. [PMID: 15668790 DOI: 10.1007/s00401-004-0957-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Revised: 10/27/2004] [Accepted: 11/02/2004] [Indexed: 11/26/2022]
Abstract
Inappropriate apoptosis has been implicated in the mechanism of neuronal death in Huntington's disease (HD). In this study, we report the expression of apoptotic markers in HD caudate nucleus (grades 1-4) and compare this with controls without neurological disease. Terminal transferase-mediated biotinylated-UTP nick end-labeling (TUNEL)-positive cells were detected in both control and HD brains. However, typical apoptotic cells were present only in HD, especially in grade 3 and 4 specimens. Expression of the pro-apoptotic protein Bax was increased in HD brains compared to controls, demonstrating a cytoplasmic expression pattern in predominantly shrunken and dark neurons, which were most frequently seen in grades 2 and 3. Control brains displayed weak perinuclear expression of the anti-apoptotic protein Bcl-2, whereas in HD brains Bcl-2 immunoreactivity was markedly enhanced, especially in severely affected grade 4 brains, and was observed in both healthy neurons and dark neurons. Caspase-3, an executioner protease, was only found in four HD brains of different grades and was not expressed in controls. A strong neuronal and glial expression of poly(ADP-ribose) polymerase (PARP)-immunoreactivity was observed in HD brains. These data strongly suggest the involvement of apoptosis in HD. The exact apoptotic pathway occurring in HD neurodegeneration remains yet unclear. However, the presence of late apoptotic events, such as enhanced PARP expression and many TUNEL-positive cells accompanied with weak caspase-3 immunoreactivity in severely affected HD brains, suggests that caspase-mediated neuronal death only plays a minor role in HD.
Collapse
Affiliation(s)
- José C Vis
- Department of Neurology 321, University Medical Centre Nijmegen, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Lindenboim L, Schlipf S, Kaufmann T, Borner C, Stein R. Bcl-xS induces an NGF-inhibitable cytochrome c release. Exp Cell Res 2004; 297:392-403. [PMID: 15212942 DOI: 10.1016/j.yexcr.2004.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2003] [Revised: 01/13/2004] [Indexed: 10/26/2022]
Abstract
Bcl-x(S), a pro-apoptotic member of the Bcl-2 protein family, is localized in the mitochondrial outer membrane and induces caspase-dependent and nerve growth factor (NGF)-inhibitable apoptosis in PC12 cells. The mechanism of action of Bcl-x(S) and how NGF inhibits this death are not fully understood. It is still unknown whether Bcl-x(S) induces mitochondrial cytochrome c release, and which apoptotic step NGF inhibits. We show that Bcl-x(S) induces cytochrome c release and caspase-3 activation in several cell types, and that in PC12 cells, these events are inhibited by NGF treatment. The survival effect of NGF was inhibited by inhibitors of protein kinase C (PKC), phosphatidylinositol-3-kinase (PI 3-kinase), and the mitogen-activated protein kinase kinase (MEK) inhibitors GF109203X, LY294002, and U0126. These findings show that cytochrome c release and caspase-3 activation participate in Bcl-x(S)-induced apoptosis, and that NGF inhibits Bcl-x(S)-induced apoptosis at the mitochondrial level via the PKC, PI 3-kinase, and MEK signaling pathways.
Collapse
Affiliation(s)
- Liora Lindenboim
- Department of Neurobiochemistry, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | | | | | | | | |
Collapse
|
19
|
Ferrer I, Friguls B, Dalfó E, Justicia C, Planas AM. Caspase-dependent and caspase-independent signalling of apoptosis in the penumbra following middle cerebral artery occlusion in the adult rat. Neuropathol Appl Neurobiol 2003; 29:472-81. [PMID: 14507339 DOI: 10.1046/j.1365-2990.2003.00485.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Transient focal ischaemia by middle cerebral artery occlusion (MCAO) may produce cell death, but the mechanisms leading to cell death differ in the infarct core and in the penumbra, the immediate zone surrounding the infarct core. In the present study, transient focal ischaemia to adult rats was produced by intraluminal occlusion of the middle cerebral artery for 1 h followed by 0 h (n=6), 1 h (n=10), 4 h (n=8), 6 h (n=2) and 12 h (n=3) of reperfusion. The present model of ischaemia causes a large cortico-striatal infarct extending through the mediolateral cortex and dorsolateral striatum at 12 h. The expression and subcellular distribution of several proteins involved in apoptosis have been examined in the penumbra and in the infarct core by using combined methods of immunohistochemistry, cell subfractionation and Western blotting. Transient focal ischaemia by MCAO results in activation of complex signal pathways for cell death in the penumbra. Increased expression of Bcl-2 and Bax, but not of Bcl-x, occurs in the penumbra at the time when Bax translocates from the cytosol to the mitochondria, cytochrome c is released to the cytoplasm and active caspase-3 is expressed. Bax translocation, cytochrome c release and active caspase-3 are observed at 4 h, but not at 1 h, following reperfusion, and together indicate activation of the caspase-dependent pathway of apoptosis in the penumbra. In contrast, reduced Bax expression but not Bax translocation and cytochrome c release occurs in the infarct core, thus suggesting apoptosis signals restricted to the penumbra. In addition, increased expression of an apoptosis-inducing factor in the cytoplasm and nuclei of selected cells shows, for the first time, activation of the caspase-independent mitochondrial pathway in the penumbra following transient focal ischaemia and reperfusion.
Collapse
Affiliation(s)
- I Ferrer
- Institut de Neuropatologia, Servei d'Anatomia Patològica, Hospital Bellvitge Hospitalet de Llobregat, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
20
|
Wu C, Fujihara H, Yao J, Qi S, Li H, Shimoji K, Baba H. Different expression patterns of Bcl-2, Bcl-xl, and Bax proteins after sublethal forebrain ischemia in C57Black/Crj6 mouse striatum. Stroke 2003; 34:1803-8. [PMID: 12791942 DOI: 10.1161/01.str.0000077255.15597.69] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Ischemic injury in neurons can be strongly reduced by a preceding sublethal ischemic episode, of which the mechanism is poorly understood. Although changes in the expression of apoptosis-related proteins (Bcl-2, Bcl-xl, and Bax) have been considered to be crucially important in ischemic injury, the roles these proteins play in ischemic preconditioning induced by sublethal forebrain ischemia have not been elucidated. Therefore, we investigated the transcription and expression of Bcl-2, Bcl-xl, and Bax in striatum of mice subjected to sublethal forebrain ischemia and lethal ischemia, with or without ischemic preconditioning. METHODS Sublethal forebrain ischemia was induced in C57Black/Crj6 (C57BL/6) mice by 6 minutes of bilateral common carotid artery occlusion. The transcription and expression of Bcl-2 family genes were detected by reverse transcription-polymerase chain reaction, Western blot, and immunofluorescent staining. RESULTS No detectable neuronal loss was induced in striatum by 6 minutes of bilateral common carotid artery occlusion. Transcription and expression of Bcl-2 and Bcl-xl were increased after sublethal forebrain ischemia, which attenuated the DNA fragmentation induced by lethal ischemia. The transcription and expression of Bax remained unchanged. CONCLUSIONS Upregulation of Bcl-2 and Bcl-xl but not Bax may have a role in protective ischemic preconditioning. This result indicates a potential strategy for further ischemic neuronal injury therapies.
Collapse
Affiliation(s)
- Chaoran Wu
- Department of Anesthesiology, Niigata University School of Medicine, Japan.
| | | | | | | | | | | | | |
Collapse
|
21
|
Tahara K, Tsuchimoto D, Tominaga Y, Asoh S, Ohta S, Kitagawa M, Horie H, Kadoya T, Nakabeppu Y. DeltaFosB, but not FosB, induces delayed apoptosis independent of cell proliferation in the Rat1a embryo cell line. Cell Death Differ 2003; 10:496-507. [PMID: 12728248 DOI: 10.1038/sj.cdd.4401173] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The fates of Rat1a cells expressing FosB and DeltaFosB as fusion proteins (ER-FosB, ER-DeltaFosB) with the ligand binding domain of human estrogen receptor were examined. The binding of estrogen to the fusion proteins resulted in their nuclear translocation and triggered cell proliferation, and thereafter delayed cell death was observed only in cells expressing ER-DeltaFosB. The proliferation of Rat1a cells, but not cell death triggered by ER-DeltaFosB, was completely abolished by butyrolactone I, an inhibitor of cycline-dependent kinases, and was partly suppressed by antisense oligonucleotides against galectin-1, whose expression is induced after estrogen administration. The cell death was accompanied by the activation of caspase-3 and -9, the fragmentation of the nuclear genome and cytochrome c release from the mitochondria, and was suppressed by zDEVD-fmk and zLEHD-fmk but not zIETD-fmk. The cell death was not suppressed by exogenous His-PTD-Bcl-x(L) at all, suggesting involvement of a Bcl-x(L)-resistant pathway for cytochrome c release.
Collapse
Affiliation(s)
- K Tahara
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi Higashi-ku, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Xu H, Steven Richardson J, Li XM. Dose-related effects of chronic antidepressants on neuroprotective proteins BDNF, Bcl-2 and Cu/Zn-SOD in rat hippocampus. Neuropsychopharmacology 2003; 28:53-62. [PMID: 12496940 DOI: 10.1038/sj.npp.1300009] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It has been proposed that antidepressants have neuroprotective effects on hippocampal neurons. To further test this hypothesis, brain-derived neurotrophic factor (BDNF), B cell lymphoma protein-2 (Bcl-2), and copper-zinc superoxide dismutase (Cu/Zn-SOD) were examined immunohistochemically in hippocampal neurons of Sprague-Dawley rats following daily treatment with 5 or 10 mg/kg of amitriptyline or venlafaxine for 21 days. At 5 mg/kg, both amitriptyline and venlafaxine increased the intensity of BDNF immunostaining in hippocampal pyramidal neurons, and the intensity of Bcl-2 immunostaining in hippocampal mossy fibers, but did not alter the Cu/Zn-SOD immunoreactivity. The high dose of venlafaxine, however, decreased the intensity of BDNF immunostaining in all subareas of the hippocampus and increased the intensity of Cu/Zn-SOD immunostaining in the dentate granular cell layer. The high dose of amitriptyline increased the intensity of Cu/Zn-SOD immunostaining, but did not affect the immunoreactivity of Bcl-2 or BDNF. These findings suggest that the chronic administration of amitriptyline or venlafaxine at 5 mg/kg, but not 10 mg/kg, may be neuroprotective to hippocampal neurons. These dose-related effects of antidepressant drugs on hippocampal neurons may have relevance to disparate findings in the field.
Collapse
Affiliation(s)
- Haiyun Xu
- Neuropsychiatric Research Institute, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatooon, Canada
| | | | | |
Collapse
|
23
|
Lee MY, Kim SY, Shin SL, Choi YS, Lee JH, Tsujimoto Y, Lee JH. Reactive astrocytes express bis, a bcl-2-binding protein, after transient forebrain ischemia. Exp Neurol 2002; 175:338-46. [PMID: 12061864 DOI: 10.1006/exnr.2002.7903] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bis (also called Bag-3), identified as a novel Bcl-2-interacting protein, has been shown to enhance anti-cell death activity of Bcl-2. Because ischemia/reperfusion induces expression of Bcl-2, we examined the changes in the pattern of Bis expression in the adult rat hippocampus after transient forebrain ischemia. Western blot analysis with protein extracts from the hippocampus showed that, compared with controls, levels of Bis were markedly increased seven days after ischemia. An immunohistochemical study showed that the expression of Bis increased preferentially in the CA1 and the dentate hilar regions, and peaked at 3-7 days after reperfusion. The temporal and spatial patterns of expression for both Bis and glial fibrillary acidic protein (GFAP) were very similar, and double immunofluorescence histochemistry showed that Bis was expressed in reactive astrocytes, which express GFAP. Immunolabeling of adjacent sections with anti-Bcl-2 and anti-Hsp70 antibodies revealed that the pattern of Bis expression closely correlates with that of Bcl-2, but clearly differs from that of Hsp70. Coexpression of Bis and Bcl-2 in reactive astrocytes was confirmed by double immunofluorescence histochemistry. Our results demonstrate that reactive astrocytes transiently up-regulate Bis after ischemia/reperfusion in the adult rat hippocampus. However, the precise role of Bis in the astrocytic response to ischemia/reperfusion in relation to Bcl-2 remains to be determined.
Collapse
Affiliation(s)
- Mun-Yong Lee
- Department of Anatomy, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | | | |
Collapse
|
24
|
Chuang JI, Chen ST. Differential expression of Bcl-2 and APP immunoreactivity after intrastriatal injection of MPP+ in the rat. Neurochem Int 2002; 40:169-79. [PMID: 11738483 DOI: 10.1016/s0197-0186(01)00075-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
While there is growing evidence that Bcl-2 proto-oncogene and beta-amyloid precursor proteins (APP) are neuroprotective in function, our recent studies have demonstrated that Bcl-2 and APP may be co-expressed and co-regulated in retinal neurons or glia under normal or experimental conditions. Whether Bcl-2 and APP are functionally coupled in other neuronal systems is not clear. This issue was investigated further in the present experiments by examining the expression pattern of two molecules after unilateral intrastriatal injection of 1-methyl-4-phenyl-pyridinium (MPP(+)), a neurotoxic metabolite that selectively damages dopaminergic neurons. One hour to 2 months after MPP(+) injection into rat striatum, a significant increase in Bcl-2 expression was observed in distinct populations of neurons, astrocyte-like and OX-42-positive cells not only in traumatic regions but also in remote areas including the ipsilateral cortex and substantia nigra (SN). No detectable change was observed in the striatum, cortex or SN on the contralateral side of the brain. The immunoreactive pattern and time-dependent APP increase was similar to that of Bcl-2 in the severely injured striatum and cortex. However, an up-regulation of Bcl-2 expression, but not APP, appears in dopaminergic neurons in the ipsilateral SN pars compacta where there was retrograde degeneration. In contrast, APP immunoreactivity was decreased in the hippocampus following intrastriatal injury, whereas, no alteration in Bcl-2 expression was detected. The differential changes in Bcl-2 and APP expression in nigral neurons and some other brain tissues suggest that these proteins may not be co-regulated by a common mechanism, at least in certain neuronal pathways.
Collapse
Affiliation(s)
- Jih Ing Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, ROC
| | | |
Collapse
|
25
|
Zhang Z, Sobel RA, Cheng D, Steinberg GK, Yenari MA. Mild hypothermia increases Bcl-2 protein expression following global cerebral ischemia. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 95:75-85. [PMID: 11687278 DOI: 10.1016/s0169-328x(01)00247-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Mild hypothermia protects the brain against experimental ischemia, but the reasons are not well known. We examined whether the protective effects of mild hypothermia could be correlated with alterations in expression of Bcl-2, an anti-apoptotic protein in a rat model of transient global ischemia. Following 10 min of forebrain ischemia, hippocampal neurons were examined 72 h later for survival, expression of Bcl-2 family proteins and apoptosis. Intraischemic mild hypothermia was applied for 3 h (33 degrees C, isch-33) or normal body temperature was maintained (37 degrees C, isch-37). Survival of CA1 neurons was significantly improved in the isch-33 group compared to the isch-37 group (90 vs. 53% survival; P<0.01). The proportion of Bcl-2-positive cells among surviving CA1 neurons in the isch-33 group was increased compared to that of sham and isch-37 groups (P<0.01). Bax expression in CA1 was no different between sham and isch-33 groups, but was significantly decreased in isch-37 (P<0.05). TUNEL staining was positive in many isch-37 CA1 neurons, but absent in isch-33. Utilizing electron microscopy, more cells meeting criteria for apoptosis were observed in the isch-37 than isch-33. These data suggest that mild hypothermia attenuates apoptotic death, and that this protection may be related to increases in Bcl-2.
Collapse
Affiliation(s)
- Z Zhang
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Rd. MSLS Building, P304, Stanford, CA 94305-5487, USA
| | | | | | | | | |
Collapse
|
26
|
Friguls B, Justicia C, Pallàs M, Planas AM. Focal cerebral ischemia causes two temporal waves of Akt activation. Neuroreport 2001; 12:3381-4. [PMID: 11711890 DOI: 10.1097/00001756-200110290-00046] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We studied whether pro-survival Akt was activated after transient focal cerebral ischemia and whether it inhibited pro-apoptotic Bad. Phosphorylation of Akt (serine-473) was enhanced in cortex after 1-hour ischemia, and also after 1h and 6 h of reperfusion, but it returned back to that in controls by 24 h. After this first wave of Akt activation, a second increase was observed between 4 and 7 days. In striatum, only the late Akt activation was seen. In contrast to Akt, no Bad phosphorylation (serine-136) was detected after ischemia. Therefore, injury spontaneously activated Akt, but this did not suppress Bad signalling. It is proposed that further pharmacological activation of Akt shortly after ischemia might promote cell survival, whereas Akt activation at longer time points is involved with glial reactivity.
Collapse
Affiliation(s)
- B Friguls
- Departament de Farmacologia i Toxicologia, Institut d'Investigacions Biomèdiques de Barcelona, CSIC-IDIBAPS, Rosselló, 161, planta 6, 08036 Barcelona, Spain
| | | | | | | |
Collapse
|
27
|
DeVries AC, Joh HD, Bernard O, Hattori K, Hurn PD, Traystman RJ, Alkayed NJ. Social stress exacerbates stroke outcome by suppressing Bcl-2 expression. Proc Natl Acad Sci U S A 2001; 98:11824-8. [PMID: 11553785 PMCID: PMC58815 DOI: 10.1073/pnas.201215298] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2001] [Indexed: 12/18/2022] Open
Abstract
The relationship between stressful life events and the onset of disease is well documented. However, the role of psychological stress as a risk factor for life-threatening cerebrovascular insults such as stroke remains unspecified, but could explain individual variation in stroke outcome. To discover the mechanisms through which psychological stress may alter stroke outcome, we modeled the effects of chronic social intimidation and stress on ischemia-induced bcl-2 expression and early neuronal cell loss resulting from cerebral artery occlusion in mice (C57BL/6). The bcl-2 protooncogene promotes cell survival and protects against apoptosis and cellular necrosis in numerous neurodegenerative disorders, including stroke. In our study, male mice were chronically exposed to aggressive social stimuli before induction of a controlled, mild ischemic insult. Stressed mice expressed approximately 70% less bcl-2 mRNA than unstressed mice after ischemia. In addition, social stress greatly exacerbated infarct in wild-type mice but not in transgenic mice that constitutively express increased neuronal bcl-2. Despite similar postischemic concentrations of corticosterone, the major stress hormone in mice, high corticosterone concentrations were significantly correlated with larger infarcts in wild-type mice but not bcl-2 transgenic mice. Thus, enhanced bcl-2 expression offsets the potentially deleterious consequences of high postischemic plasma corticosterone concentrations. Taken together, these data demonstrate that stressful prestroke social milieu strongly compromises an endogenous molecular mechanism of neuroprotection in injured brain and offer a new behavioral target for stroke therapy.
Collapse
Affiliation(s)
- A C DeVries
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Kondratyev A, Sahibzada N, Gale K. Electroconvulsive shock exposure prevents neuronal apoptosis after kainic acid-evoked status epilepticus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 91:1-13. [PMID: 11457487 DOI: 10.1016/s0169-328x(01)00099-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In the aftermath of prolonged continuous seizure activity (status epilepticus, SE), neuronal cell death occurs in the brain regions through which the seizure propagates. The vulnerability to adrenalectomy-induced apoptotic neuronal death was recently reported to be reduced by prior exposure to repeated daily noninjurious electroconvulsive shock (ECS). The present studies identified apoptosis and apoptosis-associated gene products in the neurodegenerative response to experimentally controlled periods (1 or 2 h) of SE in the rat, and determined whether exposure to ECS can interrupt these apoptotic responses mechanisms. Internucleosomal DNA fragmentation and the presence of apoptotic-like neurons (as assessed by in situ double labeling technique) was detected in hippocampus and rhinal cortex at 24 h after SE. Under these conditions, levels of both mRNA and protein encoded by the 'death promoting' bcl-XS gene were increased in the same brain areas. Pretreatment of animals for 7 days with low intensity (minimal) ECS conferred resistance to SE-evoked neurodegeneration, as assessed histopathologically by silver staining. Associated with this neuroprotective action was a reduction in the incidence of apoptosis-like neuronal morphology and DNA fragmentation, and a prevention of the increase in Bcl-XS protein and mRNA in hippocampus and rhinal cortex. These data suggest that pre-exposure to controlled, brief noninjurious seizures decreases vulnerability to programmed neuronal cell death, that this neuroprotective action occurs upstream from Bcl-XS, and that increases in bcl-XS gene expression may serve as a sensitive indicator of neurodegeneration following SE.
Collapse
Affiliation(s)
- A Kondratyev
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC 20007, USA.
| | | | | |
Collapse
|
29
|
Glasgow JN, Qiu J, Rassin D, Grafe M, Wood T, Perez-Pol JR. Transcriptional regulation of the BCL-X gene by NF-kappaB is an element of hypoxic responses in the rat brain. Neurochem Res 2001; 26:647-59. [PMID: 11519724 DOI: 10.1023/a:1010987220034] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Signal transduction pathways that mediate neuronal commitment to apoptosis involve the nuclear factor kappa B (NF-kappaB) transcription factor. The bcl-x gene is a member of the bcl-2 family of genes that regulate apoptosis, and gives rise to two proteins, Bcl-XL and Bcl-XS, via alternative mRNA splicing. BCl-XL protein, like Bcl-2, is a dominant inhibitor of apoptotic cell death, whereas Bcl-XS promotes apoptosis. While there is high expression of Bcl-XL in the developing and adult brain, few transcriptional control elements have been identified in the bcl-x promoter. There are two functional nuclear factor-kappa B (NF-kappaB) DNA binding sites clustered upstream of the brain-specific transcription start site in the upstream promoter region of murine bcl-x. Recombinant NF-kappaB proteins bind to these sites. Also NF-kappaB overexpression, coupled with bcl-x promoter/reporter assays using a series of murine bcl-x promoter and deletion mutants, has identified the downstream 1.1kb of the bcl-x promoter as necessary for basal promoter activity and induction by NF-kappaB in support of the hypothesis that NF-kappaB can act to enhance BCl-XL expression via highly selective interactions with the bcl-x promoter, where NF-kappaB binding and promoter activation are dependent on specific DNA binding site sequences and NF-kappaB protein dimer composition. Hypoxia induces apoptosis in the hippocampus where the NF-kappaB dimers c-Rel/p50 and p50/pS0 bind to the bcl-x promoter NF-kappaB site.
Collapse
Affiliation(s)
- J N Glasgow
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch, Galveston 77555-0652, USA
| | | | | | | | | | | |
Collapse
|
30
|
Chuang JI, Chen ST, Chang YH, Jen LS. Alteration of Bcl-2 expression in the nigrostriatal system after kainate injection with or without melatonin co-treatment. J Chem Neuroanat 2001; 21:215-23. [PMID: 11382533 DOI: 10.1016/s0891-0618(01)00109-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In order to understand further the role of the anti-apoptotic Bcl-2 proto-oncogene protein in excitotoxin-induced brain injury and possible interaction between Bcl-2 and the antioxidant melatonin, the expression of Bcl-2 in various brain parts was studied after intrastriatal injection of kainate (KA, 2.5 nmol) with or without co-treatment of melatonin (10 mg/kg, intraperitoneally (i.p.)). Three days after unilateral injection of KA to the striatum in the rat, a dramatic direct cytotoxic effect was observed, as indicated an expression of Bcl-2 immunoreactivity in TUNEL- and OX-42-positive cells in the KA-injected striatum and traumatized cortical region. A less severe detrimental effect was also observed in the ipsilateral substantia nigra and peritraumatic cortex, as reflected by an upregulation of Bcl-2-immunostained neurons. Surprisingly, a reduction in Bcl-2-immunoreactive neurons that was accompanied by a less severe loss of tyrosine hydroxylase-immunoreactive neurons in the nigrostriatal pathway was observed after co-treatment with melatonin. Western blot analysis confirmed that Bcl-2 expression is elevated in striatum and cortex on the lesioned side, and that its expression was attenuated substantially after systemic administration of melatonin. The results showing an upregulation of Bcl-2 in nigral neurons and reactive microglia after KA lesion are consistent with the view that Bcl-2 is protective in function in the central nervous system.
Collapse
Affiliation(s)
- J I Chuang
- Department of Physiology, National Cheng Kung University, Taiwan 701, Taiwan, ROC
| | | | | | | |
Collapse
|
31
|
Vis JC, Verbeek MM, de Waal RM, ten Donkelaar HJ, Kremer B. The mitochondrial toxin 3-nitropropionic acid induces differential expression patterns of apoptosis-related markers in rat striatum. Neuropathol Appl Neurobiol 2001; 27:68-76. [PMID: 11299004 DOI: 10.1046/j.0305-1846.2001.00305.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The mitochondrial toxin 3-nitropropionic acid (3-NP) causes selective striatal lesions in rats and serves as an experimental model for the neurodegenerative disorder Huntington's disease (HD). Apoptotic cell death has been implicated for the neuronal degeneration that occurs in HD brains. The present study was designed to investigate whether the 3-NP-induced cell death in rats involves apoptosis and an altered expression of Bcl-2 family proteins. Systemic administration of 3-NP via subcutaneous Alzet pumps resulted in lesions of variable severity with neuronal loss and gliosis in the striatum. Using the terminal transferase-mediated biotinylated-UTP nick end-labelling (TUNEL) of DNA, TUNEL-positive cells exhibiting typical apoptotic morphology were detected only in the striatum of rats with a severe lesion. Furthermore, the neuronal expression of the pro-apoptotic protein Bax was strongly increased in the core of the severe lesion. Expression of the anti-apoptotic marker Bcl-2 was unchanged in this location, but was enhanced in the margins of the lesions. A moderately increased expression of both Bax and Bcl-2 was observed in dark neurones in the mild lesion and in the subtle lesion. The presence of nuclear DNA fragmentation, strong granular Bax expression and an increased Bax/Bcl-2 ratio in the centre of severe lesions suggests the occurrence of apoptotic cell death following 3-NP administration. In contrast, the dark compromised neurones observed in 3-NP-treated animals revealed an equally enhanced expression of both Bax and Bcl-2, but lacked TUNEL-labelling, and are therefore not apoptotic.
Collapse
Affiliation(s)
- J C Vis
- Departments of Neurology and Pathology, University Hospital Nijmegen, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
32
|
Abstract
Brain ischemia triggers a complex cascade of molecular events that unfolds over hours to days. Identified mechanisms of postischemic neuronal injury include altered Ca(2+) homeostasis, free radical formation, mitochondrial dysfunction, protease activation, altered gene expression, and inflammation. Although many of these events are well characterized, our understanding of how they are integrated into the causal pathways of postischemic neuronal death remains incomplete. The primary goal of this review is to provide an overview of molecular injury mechanisms currently believed to be involved in postischemic neuronal death specifically highlighting their time course and potential interactions.
Collapse
Affiliation(s)
- R W Neumar
- Department of Emergency Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19107-4283, USA.
| |
Collapse
|
33
|
Abstract
In vivo models of cerebral hypoxia-ischemia have shown that neuronal death may occur via necrosis or apoptosis. Necrosis is, in general, a rapidly occurring form of cell death that has been attributed, in part, to alterations in ionic homeostasis. In contrast, apoptosis is a delayed form of cell death that occurs as the result of activation of a genetic program. In the past decade, we have learned considerably about the mechanisms underlying apoptotic neuronal death following cerebral hypoxia-ischemia. With this growth in knowledge, we are coming to the realization that apoptosis and necrosis, although morphologically distinct, are likely part of a continuum of cell death with similar operative mechanisms. For example, following hypoxia-ischemia, excitatory amino acid release and alterations in ionic homeostasis contribute to both necrotic and apoptotic neuronal death. However, apoptosis is distinguished from necrosis in that gene activation is the predominant mechanism regulating cell survival. Following hypoxic-ischemic episodes in the brain, genes that promote as well as inhibit apoptosis are activated. It is the balance in the expression of pro- and anti-apoptotic genes that likely determines the fate of neurons exposed to hypoxia. The balance in expression of pro- and anti-apoptotic genes may also account for the regional differences in vulnerability to hypoxic insults. In this review, we will examine the known mechanisms underlying apoptosis in neurons exposed to hypoxia and hypoxia-ischemia.
Collapse
Affiliation(s)
- K J Banasiak
- Department of Pediatrics, Section of Critical Care, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | |
Collapse
|
34
|
Banasiak KJ, Cronin T, Haddad GG. bcl-2 prolongs neuronal survival during hypoxia-induced apoptosis. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1999; 72:214-25. [PMID: 10529480 DOI: 10.1016/s0169-328x(99)00189-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In vivo models of hypoxic-ischemic brain injury have shown altered expression of a number of genes that are important in regulating neuronal survival. However, it is not clear as to whether hypoxia alone can alter the expression of genes regulating neuronal survival. We hypothesized that (1) hypoxia alone alters the expression of bcl-2 in neurons, (2) the severity and duration of hypoxia influence bcl-2 expression, and (3) the alteration of bcl-2 expression has an important role in regulating neuronal survival during hypoxia. Embryonic rat neocortical neurons cultured for 7-10 days were exposed to 0.1, 1, or 3% oxygen for various durations and were removed for analyses at 24-h intervals. Under all hypoxic conditions, neurons exhibited morphologic changes, as assessed by electron microscopy and Annexin V staining, consistent with apoptosis. Immunoblot and immunofluorescence analyses revealed an increase in neuronal bcl-2 protein during hypoxic exposure. Quantitative immunofluorescence analyses of bcl-2 immunostained neurons indicated that expression of bcl-2 was altered by the duration and severity of hypoxia. Attenuation of bcl-2 expression by antisense oligonucleotides decreased the proportion of surviving neurons by approximately 50% after 48 h of exposure to 0.1% oxygen. We conclude that observed increase in bcl-2, in part, plays an important role in neuronal survival during exposure to hypoxia.
Collapse
Affiliation(s)
- K J Banasiak
- Department of Pediatrics, Sections of Critical Care and Applied Physiology, LCI 305, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | | | | |
Collapse
|
35
|
López E, Pozas E, Rivera R, Ferrer I. Bcl-2, Bax and Bcl-x expression following kainic acid administration at convulsant doses in the rat. Neuroscience 1999; 91:1461-70. [PMID: 10391451 DOI: 10.1016/s0306-4522(98)00704-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Neuronal death was produced in the CA1 and CA3 areas of the hippocampus, amygdala, and piriform and entorhinal cortices after intraperitioneal administration of kainic acid at convulsant doses to adult rats. To assess the involvement of members of the Bcl-2 family in cell death or survival, immunohistochemistry, western and northern blotting to Bcl-2, Bcl-x and Bax, and in situ hybridization to Bax were examined at different time-points after kainic acid treatment. Members of the Bcl-2 family were expressed in the cytoplasm of pyramidal neurons in the hippocampus, and in a subset of neurons of the piriform and the entorhinal cortices, amygdala and neocortex in the normal adult brain. Dying neurons in the pyramidal cell layer of CA1 and CA3 areas, entorhinal and piriform cortices, and amygdala also expressed Bcl-2, Bax and Bcl-x following excitotoxicity, although many dying cells did not. In addition, a number of cells in the affected areas showed Bax immunoreactivity in their nuclei at 24-48 h following kainic acid administration, thus indicating Bax nuclear translocation in a subset of dying cells. Western blots disclosed no modifications in the intensity of the bands corresponding to Bcl-2, Bcl-x and Bax, between control and kainic acid-treated rats. No modifications in the intensity of the bcl-2 messenger RNA band on northern blots was observed in kainic acid-treated rats. However, a progressive increase in the intensity of the bax messenger RNA band was found in kainic acid-treated rats at 6 h, 12 h and 24 h following kainic acid administration. Interestingly, a slight increase in Bax immunoreactivity was observed in the cytoplasm of neurons of the dentate gyrus at 24-48 h, a feature which matches the increase of bax messenger RNA in the same area, as shown by in situ hybridization at 12-24 h following kainic acid injection. The present results suggest that cell death or survival does not correlate with modifications of Bcl-2, Bax and Bcl-x protein, and messenger RNA expression, but rather that kainic acid excitotoxicity is associated with Bax translocation to the nucleus in a subset of dying cells.
Collapse
Affiliation(s)
- E López
- Hospital Prínceps d'Espanya, Departament de Biologia Cellular i Anatomia Patològica, Facultat de Medecina, Universitat de Barcelona, Hospitalet de Llobregat, Spain
| | | | | | | |
Collapse
|