1
|
Rieder GS, Braga MM, Mussulini BHM, Silva ES, Lazzarotto G, Casali EA, Oliveira DL, Franco JL, Souza DOG, Rocha JBT. Diphenyl Diselenide Attenuates Mitochondrial Damage During Initial Hypoxia and Enhances Resistance to Recurrent Hypoxia. Neurotox Res 2024; 42:13. [PMID: 38332435 DOI: 10.1007/s12640-024-00691-6] [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: 08/09/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024]
Abstract
Hypoxia plays a significant role in the development of various cerebral diseases, many of which are associated with the potential risk of recurrence due to mitochondrial damage. Conventional drug treatments are not always effective for hypoxia-related brain diseases, necessitating the exploration of alternative compounds. In this study, we investigated the potential of diphenyl diselenide [(PhSe)2] to ameliorate locomotor impairments and mitigate brain mitochondrial dysfunction in zebrafish subjected to hypoxia. Additionally, we explored whether these improvements could confer resistance to recurrent hypoxia. Through a screening process, an appropriate dose of (PhSe)2 was determined, and animals exposed to hypoxia received a single intraperitoneal injection of 100 mg/kg of the compound or vehicle. After 1 h from the injection, evaluations were conducted on locomotor deficits, (PhSe)2 content, mitochondrial electron transport system, and mitochondrial viability in the brain. The animals were subsequently exposed to recurrent hypoxia to assess the latency time to hypoxia symptoms. The findings revealed that (PhSe)2 effectively crossed the blood-brain barrier, attenuated locomotor deficits induced by hypoxia, and improved brain mitochondrial respiration by modulating complex III. Furthermore, it enhanced mitochondrial viability in the telencephalon, contributing to greater resistance to recurrent hypoxia. These results demonstrate the beneficial effects of (PhSe)2 on both hypoxia and recurrent hypoxia, with cerebral mitochondria being a critical target of its action. Considering the involvement of brain hypoxia in numerous pathologies, (PhSe)2 should be further tested to determine its effectiveness as a potential treatment for hypoxia-related brain diseases.
Collapse
Affiliation(s)
- Guilherme S Rieder
- Programa de Pós Graduação Em Bioquímica Toxicológica, Departamento de Bioquímica E Biologia Molecular, Centro de Ciências Naturais E Exatas, Universidade Federal de Santa Maria, Avenida Roraima 1000, Santa Maria, RS, 97105-900, Brazil
| | - Marcos M Braga
- Programa de Pós Graduação Em Bioquímica Toxicológica, Departamento de Bioquímica E Biologia Molecular, Centro de Ciências Naturais E Exatas, Universidade Federal de Santa Maria, Avenida Roraima 1000, Santa Maria, RS, 97105-900, Brazil
| | - Ben Hur M Mussulini
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Emerson S Silva
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Gabriela Lazzarotto
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Emerson André Casali
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Diogo L Oliveira
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Jeferson L Franco
- Universidade Federal Do Pampa, Campus São Gabriel, São Gabriel, RS, Brazil
| | - Diogo O G Souza
- Programa de Pós-Graduação Em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - João Batista T Rocha
- Programa de Pós Graduação Em Bioquímica Toxicológica, Departamento de Bioquímica E Biologia Molecular, Centro de Ciências Naturais E Exatas, Universidade Federal de Santa Maria, Avenida Roraima 1000, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
2
|
Bastian C, Zerimech S, Nguyen H, Doherty C, Franke C, Faris A, Quinn J, Baltan S. Aging astrocytes metabolically support aging axon function by proficiently regulating astrocyte-neuron lactate shuttle. Exp Neurol 2022; 357:114173. [PMID: 35863500 PMCID: PMC11218845 DOI: 10.1016/j.expneurol.2022.114173] [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: 02/07/2022] [Revised: 06/23/2022] [Accepted: 07/14/2022] [Indexed: 11/19/2022]
Abstract
The astrocyte-neuron lactate shuttle (ANLS) is an essential metabolic support system that uptakes glucose, stores it as glycogen in astrocytes, and provides glycogen-derived lactate for axonal function. Aging intrinsically increases the vulnerability of white matter (WM) to injury. Therefore, we investigated the regulation of this shuttle to understand vascular-glial metabolic coupling to support axonal function during aging in two different WM tracts. Aging astrocytes displayed larger cell bodies and thicker horizontal processes in contrast to thinner vertically oriented processes of young astrocytes. Aging axons recovered less following aglycemia in mouse optic nerves (MONs) compared to young axons, although providing lactate during aglycemia equally supported young and aging axonal function. Incubating MONs in high glucose to upregulate glycogen stores in astrocytes delayed loss of function during aglycemia and improved recovery in both young and aging axons. Providing lactate during recovery from aglycemia unmasked a metabolic switch from glucose to lactate in aging axons. Young and aging corpus callosum consisting of a mixture of myelinated and unmyelinated axons sustained their function fully when lactate was available during aglycemia and surprisingly showed a greater resilience to aglycemia compared to fully myelinated axons of optic nerve. We conclude that lactate is a universal substrate for axons independent of their myelination content and age.
Collapse
Affiliation(s)
- Chinthasagar Bastian
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 441952, United States of America
| | - Sarah Zerimech
- Anesthesia and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, United States of America
| | - Hung Nguyen
- Anesthesia and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, United States of America
| | - Christine Doherty
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 441952, United States of America
| | - Caroline Franke
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 441952, United States of America
| | - Anna Faris
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 441952, United States of America
| | - John Quinn
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 441952, United States of America
| | - Selva Baltan
- Anesthesia and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, United States of America; Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 441952, United States of America.
| |
Collapse
|
3
|
Ransom BR, Goldberg MP, Arai K, Baltan S. White Matter Pathophysiology. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
4
|
Demyanenko S, Dzreyan V, Sharifulina S. Histone Deacetylases and Their Isoform-Specific Inhibitors in Ischemic Stroke. Biomedicines 2021; 9:biomedicines9101445. [PMID: 34680562 PMCID: PMC8533589 DOI: 10.3390/biomedicines9101445] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 01/01/2023] Open
Abstract
Cerebral ischemia is the second leading cause of death in the world and multimodal stroke therapy is needed. The ischemic stroke generally reduces the gene expression due to suppression of acetylation of histones H3 and H4. Histone deacetylases inhibitors have been shown to be effective in protecting the brain from ischemic damage. Histone deacetylases inhibitors induce neurogenesis and angiogenesis in damaged brain areas promoting functional recovery after cerebral ischemia. However, the role of different histone deacetylases isoforms in the survival and death of brain cells after stroke is still controversial. This review aims to analyze the data on the neuroprotective activity of nonspecific and selective histone deacetylase inhibitors in ischemic stroke.
Collapse
|
5
|
Baltan S, Sandau US, Brunet S, Bastian C, Tripathi A, Nguyen H, Liu H, Saugstad JA, Zarnegarnia Y, Dutta R. Identification of miRNAs That Mediate Protective Functions of Anti-Cancer Drugs During White Matter Ischemic Injury. ASN Neuro 2021; 13:17590914211042220. [PMID: 34619990 PMCID: PMC8642107 DOI: 10.1177/17590914211042220] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We have previously shown that two anti-cancer drugs, CX-4945 and MS-275, protect and preserve white matter (WM) architecture and improve functional recovery in a model of WM ischemic injury. While both compounds promote recovery, CX-4945 is a selective Casein kinase 2 (CK2) inhibitor and MS-275 is a selective Class I histone deacetylase (HDAC) inhibitor. Alterations in microRNAs (miRNAs) mediate some of the protective actions of these drugs. In this study, we aimed to (1) identify miRNAs expressed in mouse optic nerves (MONs); (2) determine which miRNAs are regulated by oxygen glucose deprivation (OGD); and (3) determine the effects of CX-4945 and MS-275 treatment on miRNA expression. RNA isolated from MONs from control and OGD-treated animals with and without CX-4945 or MS-275 treatment were quantified using NanoString nCounter® miRNA expression profiling. Comparative analysis of experimental groups revealed that 12 miRNAs were expressed at high levels in MONs. OGD upregulated five miRNAs (miR-1959, miR-501-3p, miR-146b, miR-201, and miR-335-3p) and downregulated two miRNAs (miR-1937a and miR-1937b) compared to controls. OGD with CX-4945 upregulated miR-1937a and miR-1937b, and downregulated miR-501-3p, miR-200a, miR-1959, and miR-654-3p compared to OGD alone. OGD with MS-275 upregulated miR-2134, miR-2141, miR-2133, miR-34b-5p, miR-153, miR-487b, miR-376b, and downregulated miR-717, miR-190, miR-27a, miR-1959, miR-200a, miR-501-3p, and miR-200c compared to OGD alone. Interestingly, miR-501-3p and miR-1959 were the only miRNAs upregulated by OGD, and downregulated by OGD plus CX-4945 and MS-275. Therefore, we suggest that protective functions of CX-4945 or MS-275 against WM injury maybe mediated, in part, through miRNA expression.
Collapse
Affiliation(s)
- Selva Baltan
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Selva Baltan, Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Mackenzie Hall 2140A, L459, 3181 S.W. Sam Jackson Park Rd., Portland, OR 97239, USA.
| | - Ursula S. Sandau
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Sylvain Brunet
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Chinthasagar Bastian
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Ajai Tripathi
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Hung Nguyen
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Helen Liu
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Julie A. Saugstad
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Yalda Zarnegarnia
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| |
Collapse
|
6
|
Khan H, Tiwari P, Kaur A, Singh TG. Sirtuin Acetylation and Deacetylation: a Complex Paradigm in Neurodegenerative Disease. Mol Neurobiol 2021; 58:3903-3917. [PMID: 33877561 DOI: 10.1007/s12035-021-02387-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Sirtuins are the class III of histone deacetylases that depend on nicotinamide adenine dinucleotide for their activity. Sirtuins can influence the progression of neurodegenerative disorders by switching between deacetylation and acetylation processes. Histone acetylation occurs when acetyl groups are added to lysine residues on the N-terminal part of histone proteins. Deacetylation, on the other hand, results in the removal of acetyl groups. Pharmacological modulation of sirtuin activity has been shown to influence various neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, stroke, and amyotrophic lateral sclerosis. In this review, mechanistic perspective of sirtuins has been discussed in anti-inflammatory, antiapoptotic, and neuroprotective effects in various disorders. We have discussed the structure, neurobiology, and physiology of sirtuins in neurodegenerative disease. Recent preclinical and clinical studies and their outcome have also been elucidated. The aim of this review is to fill in the gaps in our understanding of sirtuins' role in histone acetylation and deacetylation in all neurodegenerative diseases. Here, we emphasized on reviewing all the studies carried out in various labs depicting the role of sirtuin modulators in neuroprotection and highlighted the ideas that can be considered for future perspectives. Taken together, sirtuins may serve as a promising therapeutic target for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Palak Tiwari
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
7
|
Kalotra S, Kaur G. PSA mimetic 5-nonyloxytryptamine protects cerebellar neurons against glutamate induced excitotoxicity: An in vitro perspective. Neurotoxicology 2020; 82:69-81. [PMID: 33197482 DOI: 10.1016/j.neuro.2020.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 12/29/2022]
Abstract
PSA-NCAM is a molecule of therapeutic interest for its key role in promoting neuritogenesis and synaptic plasticity. The current study was aimed to investigate the neuroregenerative potential of 5-nonyloxytryptamine (5-NOT) as PSA mimetic compound against glutamate induced excitotoxicity. 2D and 3D cultures of cerebellar neurons challenged with glutamate were used to ascertain the effect of 5-NOT on neurite outgrowth, migration and expression of neuronal plasticity markers. Glutamate excitotoxicity is one of the major underlying pathological factor responsible for neurodegeneration in various neurological disorders such as trauma, stroke, ischemia, epilepsy and neurodegenerative diseases.5-NOT treatment was observed to promote axonal growth and defasiculation in glutamate challenged neurons as well as promoted the migration of cerebellar neurons in both wound scratched area and cerebellar explant cultures. Further, 5-NOT treatment upregulated the expression of synaptic plasticity and cell survival pathway proteins which showed reduced expression after glutamate induced excitotoxicity. Thus, this preliminary data reveals thatPSA-mimetic,5-NOT may prove to be a potential neuroprotective candidate for neurodegenerative diseases.
Collapse
Affiliation(s)
- Shikha Kalotra
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| |
Collapse
|
8
|
PPAR-γ Is Critical for HDAC3-Mediated Control of Oligodendrocyte Progenitor Cell Proliferation and Differentiation after Focal Demyelination. Mol Neurobiol 2020; 57:4810-4824. [PMID: 32803489 DOI: 10.1007/s12035-020-02060-8] [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: 05/12/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022]
Abstract
Disruption of remyelination contributes to neurodegeneration and cognitive impairment in chronically disabled patients. Valproic acid (VPA) inhibits histone deacetylase (HDAC) function and probably promotes oligodendrocyte progenitor cell (OPC) proliferation and differentiation; however, the relevant molecular mechanisms remain unknown. Here, focal demyelinating lesions (FDLs) were generated in mice by two-point stereotactic injection of lysophosphatidylcholine (LPC) into the corpus callosum. Cognitive functions, sensorimotor abilities and histopathological changes were assessed for up to 28 days post-injury with or without VPA treatment. Primary OPCs were harvested and used to study the effect of VPA on OPC differentiation under inflammatory conditions. VPA dose-dependently attenuated learning and memory deficits and robustly protected white matter after FDL induction, as demonstrated by reductions in SMI-32 and increases in myelin basic protein staining. VPA also promoted OPC proliferation and differentiation and increased subsequent remyelination efficiency by day 28 post-FDL induction. VPA treatment did not affect HDAC1, HDAC2 or HDAC8 expression but reduced HDAC3 protein levels. In vitro, VPA improved the survival of mouse OPCs and promoted their differentiation into oligodendrocytes following lipopolysaccharide (LPS) stimulation. LPS caused OPCs to overexpress HDAC3, which translocated from the cytoplasm into the nucleus, where it directly interacted with the nuclear transcription factor PPAR-γ and negatively regulated PPAR-γ expression. VPA decreased the expression of HDAC3 and promoted remyelination and functional neurological recovery after FDL. These findings may support the use of strategies modulating HDAC3-mediated regulation of protein acetylation for the treatment of demyelination-related cognitive dysfunction.
Collapse
|
9
|
Zhang Y, Zhang X, Wei Q, Leng S, Li C, Han B, Bai Y, Zhang H, Yao H. Activation of Sigma-1 Receptor Enhanced Pericyte Survival via the Interplay Between Apoptosis and Autophagy: Implications for Blood-Brain Barrier Integrity in Stroke. Transl Stroke Res 2020; 11:267-287. [PMID: 31290080 DOI: 10.1007/s12975-019-00711-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/21/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023]
Abstract
Stroke is a cerebrovascular disorder that affects many people worldwide. Pericytes play an important role in stroke progression and recovery. The sigma-1 receptor (σ-1R) signaling pathway has been suggested as having promising neuroprotective potential in treating stroke; however, whether σ-1R activation regulates pericyte function remains unknown. The aim of this study was to elucidate the role of σ-1R and a novel σ-1R agonist in pericytes following ischemic stroke. An ischemic stroke animal model was induced by photothrombotic middle cerebral artery occlusion (pMCAO) in σ-1R knockout (KO) and wild-type (WT) mice. After pMCAO, there was significant pericyte loss and coverage in σ-1R KO mice compared with WT mice as determined using transmission electron microscopy, immunofluorescence staining, and western blot. Interestingly, a novel σ-1R agonist decreased infarct volume and blood-brain barrier damage with a concomitant amelioration of pericyte loss, as determined by western blot. Further studies indicated that cell apoptosis and autophagy were induced in an in vivo pMCAO ischemic stroke animal model and an in vitro oxygen glucose deprivation-treatment group. Inhibition of autophagy using a pharmacological approach significantly mitigated pericyte apoptosis, suggesting that autophagy was upstream of apoptosis in pericytes. Both in vivo and in vitro studies indicated that the σ-1R agonist significantly decreased cell apoptosis via inhibition of autophagy with a subsequent enhancement of pericyte survival. This study identified the unique roles for σ-1R in mediating pericyte survival via the regulation of the interplay between apoptosis and autophagy, suggesting that a novel σ-1R agonist may be a promising therapeutic agent for the treatment of stroke patients.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
| | | | - Qiangqiang Wei
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Shuo Leng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Cai Li
- Department of Neurology, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, 276800, Shandong, China
| | - Bing Han
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Huibin Zhang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, Jiangsu, China.
| |
Collapse
|
10
|
The Role of Circular RNAs in Brain Injury. Neuroscience 2020; 428:50-59. [PMID: 31917349 DOI: 10.1016/j.neuroscience.2019.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022]
Abstract
Circular RNAs are an increasingly important topic in non-coding RNA biology, drawing considerable attention in recent years. Accumulating evidence suggests a critical role for circular RNAs in both early and latent stages of disease pathogenesis. Circular RNAs are abundantly expressed in brain tissue, with significant implications for neural development and disease progression. Disruption of these processes, including those seen in response to brain injury, can have serious consequences such as hemiplegia, aphasia, coma, and death. In this review, we describe the role of circular RNAs in the context of brain injury and explore the potential connection between circular RNAs, brain hypoxic ischemic injury, ischemia-reperfusion injury, and traumatic injury.
Collapse
|
11
|
Yang H, Gao X, Su J, Jiang H, Lei Y, Ni W, Gu Y. Pharmacokinetics and Acute Toxicity of a Histone Deacetylase Inhibitor, Scriptaid, and its Neuroprotective Effects in Mice After Intracranial Hemorrhage. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 19:55-65. [PMID: 31858907 DOI: 10.2174/1871527319666191220111126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/26/2019] [Accepted: 11/21/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND & OBJECTIVE The pharmacokinetics and acute toxicity of a histone deacetylase inhibitor, Scriptaid, was unknown in the mouse. The aim of this study was to determine the pharmacokinetics, acute toxicity, and tissue distribution of Scriptaid, a new histone deacetylase inhibitor, in mice, and its neuroprotective efficacy in a mouse intracranial hemorrhage (ICH) model. METHODS The pharmacokinetics, acute toxicity, and tissue distribution were determined in C57BL/6 male and female mice after the intraperitoneal administration of a single dose. Behavioral tests, as well as investigations of brain atrophy and white matter injury, were used to evaluate the neuroprotective effect of Scriptaid after ICH. Western blotting was used to investigate if Scriptaid could offer antiinflammatory benefits after ICH. RESULTS No significant differences were observed in body weight or brain histopathology between the group that received Scriptaid at 50 mg/kg and the group that received dimethyl sulfoxide (control). The pharmacokinetics of Scriptaid in mice was nonlinear, and it was cleared rapidly at low doses and slowly at higher doses. Consistent with the pharmacokinetic data, Scriptaid was found to distribute in several tissues, including the spleen and kidneys. In the ICH model, we found that Scriptaid could reduce neurological deficits, brain atrophy, and white matter injury in a dose-dependent manner. Western blotting results demonstrated that Scriptaid could decrease the expression of pro-inflammatory cytokines IL1β and TNFα, as well as iNOS, after ICH. CONCLUSION These findings indicate that Scriptaid is safe and can alleviate brain injury after ICH, thereby providing a foundation for the pharmacological action of Scriptaid in the treatment of brain injury after ICH.
Collapse
Affiliation(s)
- Heng Yang
- Division of Cerebrovascular Surgery and Interventional Neuroradiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xinjie Gao
- Division of Cerebrovascular Surgery and Interventional Neuroradiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jiabin Su
- Division of Cerebrovascular Surgery and Interventional Neuroradiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hanqiang Jiang
- Division of Cerebrovascular Surgery and Interventional Neuroradiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yu Lei
- Division of Cerebrovascular Surgery and Interventional Neuroradiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wei Ni
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuxiang Gu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
12
|
Bastian C, Day J, Politano S, Quinn J, Brunet S, Baltan S. Preserving Mitochondrial Structure and Motility Promotes Recovery of White Matter After Ischemia. Neuromolecular Med 2019; 21:484-492. [PMID: 31152363 PMCID: PMC6884671 DOI: 10.1007/s12017-019-08550-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/20/2019] [Indexed: 12/23/2022]
Abstract
Stroke significantly affects white matter in the brain by impairing axon function, which results in clinical deficits. Axonal mitochondria are highly dynamic and are transported via microtubules in the anterograde or retrograde direction, depending upon axonal energy demands. Recently, we reported that mitochondrial division inhibitor 1 (Mdivi-1) promotes axon function recovery by preventing mitochondrial fission only when applied during ischemia. Application of Mdivi-1 after injury failed to protect axon function. Interestingly, L-NIO, which is a NOS3 inhibitor, confers post-ischemic protection to axon function by attenuating mitochondrial fission and preserving mitochondrial motility via conserving levels of the microtubular adaptor protein Miro-2. We propose that preventing mitochondrial fission protects axon function during injury, but that restoration of mitochondrial motility is more important to promote axon function recovery after injury. Thus, Miro-2 may be a therapeutic molecular target for recovery following a stroke.
Collapse
Affiliation(s)
- Chinthasagar Bastian
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue/NC30, Cleveland, OH, 44195, USA
| | - Jerica Day
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue/NC30, Cleveland, OH, 44195, USA
| | - Stephen Politano
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue/NC30, Cleveland, OH, 44195, USA
| | - John Quinn
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue/NC30, Cleveland, OH, 44195, USA
| | - Sylvain Brunet
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue/NC30, Cleveland, OH, 44195, USA
| | - Selva Baltan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue/NC30, Cleveland, OH, 44195, USA.
| |
Collapse
|
13
|
Lu G, Zhang M, Wang J, Zhang K, Wu S, Zhao X. Epigenetic regulation of myelination in health and disease. Eur J Neurosci 2019; 49:1371-1387. [DOI: 10.1111/ejn.14337] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/22/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Guozhen Lu
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Ming Zhang
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Jian Wang
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Kaixiang Zhang
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Shengxi Wu
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Xianghui Zhao
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| |
Collapse
|
14
|
Liberale L, Carbone F, Montecucco F, Gebhard C, Lüscher TF, Wegener S, Camici GG. Ischemic stroke across sexes: What is the status quo? Front Neuroendocrinol 2018; 50:3-17. [PMID: 29753797 DOI: 10.1016/j.yfrne.2018.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/11/2018] [Accepted: 05/06/2018] [Indexed: 12/15/2022]
Abstract
Stroke prevalence is expected to increase in the next decades due to the aging of the Western population. Ischemic stroke (IS) shows an age- and sex-dependent distribution in which men represent the most affected population within 65 years of age, being passed by post-menopausal women in older age groups. Furthermore, a sexual dimorphism concerning risk factors, presentation and treatment of IS has been widely recognized. In order to address these phenomena, a number of issue have been raised involving both socio-economical and biological factors. The latter can be either dependent on sex hormones or due to intrinsic factors. Although women have poorer outcomes and are more likely to die after a cerebrovascular event, they are still underrepresented in clinical trials and this is mirrored by the lack of sex-tailored therapies. A greater effort is needed in the future to ensure improved treatment and quality of life to both sexes.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Cathérine Gebhard
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland; Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland; Cardiology, Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| | - Susanne Wegener
- Department of Neurology, University Hospital Zurich and University of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland.
| |
Collapse
|
15
|
Bastian C, Quinn J, Tripathi A, Aquila D, McCray A, Dutta R, Baltan S, Brunet S. CK2 inhibition confers functional protection to young and aging axons against ischemia by differentially regulating the CDK5 and AKT signaling pathways. Neurobiol Dis 2018; 126:47-61. [PMID: 29944965 DOI: 10.1016/j.nbd.2018.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/08/2018] [Accepted: 05/21/2018] [Indexed: 12/25/2022] Open
Abstract
White matter (WM) is injured in most strokes, which contributes to functional deficits during recovery. Casein kinase 2 (CK2) is a protein kinase that is expressed in brain, including WM. To assess the impact of CK2 inhibition on axon recovery following oxygen glucose deprivation (OGD), mouse optic nerves (MONs), which are pure WM tracts, were subjected to OGD with or without the selective CK2 inhibitor CX-4945. CX-4945 application preserved axon function during OGD and promoted axon function recovery when applied before or after OGD. This protective effect of CK2 inhibition correlated with preservation of oligodendrocytes and conservation of axon structure and axonal mitochondria. To investigate the pertinent downstream signaling pathways, siRNA targeting the CK2α subunit identified CDK5 and AKT as downstream molecules. Consequently, MK-2206 and roscovitine, which are selective AKT and CDK5 inhibitors, respectively, protected young and aging WM function only when applied before OGD. However, a novel pan-AKT allosteric inhibitor, ARQ-092, which targets both the inactive and active conformations of AKT, conferred protection to young and aging axons when applied before or after OGD. These results suggest that AKT and CDK5 signaling contribute to the WM functional protection conferred by CK2 inhibition during ischemia, while inhibition of activated AKT signaling plays the primary role in post-ischemic protection conferred by CK2 inhibition in WM independent of age. CK2 inhibitors are currently being used in clinical trials for cancer patients; therefore, our results will provide rationale for repurposing these drugs as therapeutic options for stroke patients by adding novel targets.
Collapse
Affiliation(s)
- Chinthasagar Bastian
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - John Quinn
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Ajai Tripathi
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Danielle Aquila
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Andrew McCray
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Ranjan Dutta
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Selva Baltan
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| | - Sylvain Brunet
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| |
Collapse
|
16
|
NOS3 Inhibition Confers Post-Ischemic Protection to Young and Aging White Matter Integrity by Conserving Mitochondrial Dynamics and Miro-2 Levels. J Neurosci 2018; 38:6247-6266. [PMID: 29891729 DOI: 10.1523/jneurosci.3017-17.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 01/01/2023] Open
Abstract
White matter (WM) damage following a stroke underlies a majority of the neurological disability that is subsequently observed. Although ischemic injury mechanisms are age-dependent, conserving axonal mitochondria provides consistent post-ischemic protection to young and aging WM. Nitric oxide synthase (NOS) activation is a major cause of oxidative and mitochondrial injury in gray matter during ischemia; therefore, we used a pure WM tract, isolated male mouse optic nerve, to investigate whether NOS inhibition provides post-ischemic functional recovery by preserving mitochondria. We show that pan-NOS inhibition applied before oxygen-glucose deprivation (OGD) promotes functional recovery of young and aging axons and preserves WM cellular architecture. This protection correlates with reduced nitric oxide (NO) generation, restored glutathione production, preserved axonal mitochondria and oligodendrocytes, and preserved ATP levels. Pan-NOS inhibition provided post-ischemic protection to only young axons, whereas selective inhibition of NOS3 conferred post-ischemic protection to both young and aging axons. Concurrently, genetic deletion of NOS3 conferred long-lasting protection to young axons against ischemia. OGD upregulated NOS3 levels in astrocytes, and we show for the first time that inhibition of NOS3 generation in glial cells prevents axonal mitochondrial fission and restores mitochondrial motility to confer protection to axons by preserving Miro-2 levels. Interestingly, NOS1 inhibition exerted post-ischemic protection selectively to aging axons, which feature age-dependent mechanisms of oxidative injury in WM. Our study provides the first evidence that inhibition of glial NOS activity confers long-lasting benefits to WM function and structure and suggests caution in defining the role of NO in cerebral ischemia at vascular and cellular levels.SIGNIFICANCE STATEMENT White matter (WM) injury during stroke is manifested as the subsequent neurological disability in surviving patients. Aging primarily impacts CNS WM and mechanisms of ischemic WM injury change with age. Nitric oxide is involved in various mitochondrial functions and we propose that inhibition of glia-specific nitric oxide synthase (NOS) isoforms promotes axon function recovery by preserving mitochondrial structure, function, integrity, and motility. Using electrophysiology and three-dimensional electron microscopy, we show that NOS3 inhibition provides a common target to improve young and aging axon function, whereas NOS1 inhibition selectively protects aging axons when applied after injury. This study provides the first evidence that inhibition of glial cell NOS activity confers long-lasting benefits to WM structure and function.
Collapse
|
17
|
Raffa S, Scrofani C, Valente S, Micaloni A, Forte M, Bianchi F, Coluccia R, Geurts AM, Sciarretta S, Volpe M, Torrisi MR, Rubattu S. In vitro characterization of mitochondrial function and structure in rat and human cells with a deficiency of the NADH: ubiquinone oxidoreductase Ndufc2 subunit. Hum Mol Genet 2018; 26:4541-4555. [PMID: 28973657 PMCID: PMC5886163 DOI: 10.1093/hmg/ddx333] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022] Open
Abstract
Ndufc2, a subunit of the NADH: ubiquinone oxidoreductase, plays a key role in the assembly and activity of complex I within the mitochondrial OXPHOS chain. Its deficiency has been shown to be involved in diabetes, cancer and stroke. To improve our knowledge on the mechanisms underlying the increased disease risk due to Ndufc2 reduction, we performed the present in vitro study aimed at the fine characterization of the derangements in mitochondrial structure and function consequent to Ndufc2 deficiency. We found that both fibroblasts obtained from skin of heterozygous Ndufc2 knock-out rat model showed marked mitochondrial dysfunction and PBMC obtained from subjects homozygous for the TT genotype of the rs11237379/NDUFC2 variant, previously shown to associate with reduced gene expression, demonstrated increased generation of reactive oxygen species and mitochondrial damage. The latter was associated with increased oxidative stress and significant ultrastructural impairment of mitochondrial morphology with a loss of internal cristae. In both models the exposure to stress stimuli, such as high-NaCl concentration or LPS, exacerbated the mitochondrial damage and dysfunction. Resveratrol significantly counteracted the ROS generation. These findings provide additional insights on the role of an altered pattern of mitochondrial structure–function as a cause of human diseases. In particular, they contribute to underscore a potential genetic risk factor for cardiovascular diseases, including stroke.
Collapse
Affiliation(s)
- Salvatore Raffa
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Cellular Diagnostics Unit, Azienda Sant'Andrea University Hospital, Rome, Italy
| | - Cristina Scrofani
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Sabatino Valente
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Andrea Micaloni
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Aron M Geurts
- Department of Physiology and Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sebastiano Sciarretta
- I.R.C.C.S. Neuromed, Pozzilli (Isernia), Italy.,Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,I.R.C.C.S. Neuromed, Pozzilli (Isernia), Italy
| | - Maria Rosaria Torrisi
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Cellular Diagnostics Unit, Azienda Sant'Andrea University Hospital, Rome, Italy
| | - Speranza Rubattu
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,I.R.C.C.S. Neuromed, Pozzilli (Isernia), Italy
| |
Collapse
|
18
|
Liberale L, Carbone F, Montecucco F, Gebhard C, Lüscher TF, Wegener S, Camici GG. Ischemic stroke across sexes: what is the status quo? Front Neuroendocrinol 2018:S0091-3022(18)30040-2. [PMID: 29763641 DOI: 10.1016/j.yfrne.2018.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/14/2022]
Abstract
Stroke prevalence is expected to increase in the next decades due to the aging of the Western population. Ischemic stroke (IS) shows an age- and sex-dependent distribution in which men represent the most affected population within 65 years of age, being passed by post-menopausal women in older age groups. Furthermore, a sexual dimorphism concerning risk factors, presentation and treatment of IS has been widely recognized. In order to address these phenomena, a number of issue have been raised involving both socio-economical and biological factors. The latter can be either dependent on sex hormones or due to intrinsic factors. Although women have poorer outcomes and are more likely to die after a cerebrovascular event, they are still underrepresented in clinical trials and this is mirrored by the lack of sex-tailored therapies. A greater effort is needed in the future to ensure improved treatment and quality of life to both sexes.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Cathérine Gebhard
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland; Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland; Cardiology, Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| | - Susanne Wegener
- Department of Neurology, University Hospital Zurich and University of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland.
| |
Collapse
|
19
|
Evaluation of the Neuroprotective Effect of Sirt3 in Experimental Stroke. Transl Stroke Res 2018; 10:57-66. [DOI: 10.1007/s12975-017-0603-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 12/23/2017] [Accepted: 12/27/2017] [Indexed: 11/25/2022]
|
20
|
Bai Y, Zhang Y, Han B, Yang L, Chen X, Huang R, Wu F, Chao J, Liu P, Hu G, Zhang JH, Yao H. Circular RNA DLGAP4 Ameliorates Ischemic Stroke Outcomes by Targeting miR-143 to Regulate Endothelial-Mesenchymal Transition Associated with Blood-Brain Barrier Integrity. J Neurosci 2018; 38:32-50. [PMID: 29114076 PMCID: PMC6705810 DOI: 10.1523/jneurosci.1348-17.2017] [Citation(s) in RCA: 311] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 01/01/2023] Open
Abstract
Circular RNAs (circRNAs) are highly expressed in the CNS and regulate physiological and pathophysiological processes. However, the potential role of circRNAs in stroke remains largely unknown. Here, we show that the circRNA DLGAP4 (circDLGAP4) functions as an endogenous microRNA-143 (miR-143) sponge to inhibit miR-143 activity, resulting in the inhibition of homologous to the E6-AP C-terminal domain E3 ubiquitin protein ligase 1 expression. circDLGAP4 levels were significantly decreased in the plasma of acute ischemic stroke patients (13 females and 13 males) and in a mouse stroke model. Upregulation of circDLGAP4 expression significantly attenuated neurological deficits and decreased infarct areas and blood-brain barrier damage in the transient middle cerebral artery occlusion mouse stroke model. Endothelial-mesenchymal transition contributes to blood-brain barrier disruption and circDLGAP4 overexpression significantly inhibited endothelial-mesenchymal transition by regulating tight junction protein and mesenchymal cell marker expression. Together, the results of our study are illustrative of the involvement of circDLGAP4 and its coupling mechanism in cerebral ischemia, providing translational evidence that circDLGAP4 serves as a novel therapeutic target for acute cerebrovascular protection.SIGNIFICANCE STATEMENT Circular RNAs (circRNAs) are involved in the regulation of physiological and pathophysiological processes. However, whether circRNAs are involved in ischemic injury, particularly cerebrovascular disorders, remains largely unknown. Here, we demonstrate a critical role for circular RNA DLGAP4 (circDLGAP4), a novel circular RNA originally identified as a sponge for microRNA-143 (miR-143), in ischemic stroke outcomes. Overexpression of circDLGAP4 significantly attenuated neurological deficits and decreased infarct areas and blood-brain barrier damage in the transient middle cerebral artery occlusion mouse stroke model. To our knowledge, this is the first report describing the efficacy of circRNA injection in an ischemic stroke model. Our investigation suggests that circDLGAP4 may serve as a novel therapeutic target for acute ischemic injury.
Collapse
Affiliation(s)
- Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuan Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Bing Han
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Li Yang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xufeng Chen
- Emergency Department, Jiangsu Province Hospital, Nanjing 210029, China
| | - Rongrong Huang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fangfang Wu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jie Chao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Pei Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing 210009, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - John H Zhang
- Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, California 92354, and
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China,
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| |
Collapse
|
21
|
Bastian C, Politano S, Day J, McCray A, Brunet S, Baltan S. Mitochondrial dynamics and preconditioning in white matter. CONDITIONING MEDICINE 2018; 1:64-72. [PMID: 30135960 PMCID: PMC6101249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Mechanisms of ischemic preconditioning have been extensively studied in gray matter. However, an ischemic episode affects both the gray matter (GM) and white matter (WM) portions of the brain. Inhibition of mitochondrial fission is one of the mechanisms of preconditioning neuronal cell bodies against ischemia. Although axons are anatomical extensions of neuronal cell bodies, injury mechanisms differ between GM and WM. Indeed, axonal dysfunction is responsible for much of the disability associated with clinical deficits observed after stroke; however, the signaling process underlying preconditioning remains unexplored in axons. Using mouse optic nerve, which is a pure isolated WM tract, we show that mitochondria in myelinated axons undergo rapid and profuse fission during oxygen glucose deprivation (OGD) that is mediated by translocation of cytoplasmic Dynamin Related Protein-1 (Drp-1) to mitochondria. OGD-induced mitochondrial fission correlates with reduced mitochondrial motility and loss of axon function. Mitochondrial fragmentation and loss of motility become permanent during the recovery period. Inhibiting mitochondrial fission by administering mitochondrial division inhibitor-1 (Mdivi-1) during OGD preserves mitochondrial shape and motility and promotes axon function recovery. In contrast, preconditioning WM by applying Mdivi-1 only before OGD fails to conserve mitochondrial shape or motility and fails to benefit axon function. Our findings suggest that inhibition of mitochondrial fission during ischemia promotes axon function recovery, but is not sufficient to precondition WM against ischemia. These results raise caution in that approaches to preconditioning neuronal cell bodies may not successfully translate into functional improvement following ischemia.
Collapse
Affiliation(s)
- Chinthasagar Bastian
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Stephen Politano
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Jerica Day
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Andrew McCray
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Sylvain Brunet
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| | - Selva Baltan
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, 44195
| |
Collapse
|
22
|
Potential Therapeutic Mechanisms and Tracking of Transplanted Stem Cells: Implications for Stroke Treatment. Stem Cells Int 2017; 2017:2707082. [PMID: 28904531 PMCID: PMC5585684 DOI: 10.1155/2017/2707082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/08/2017] [Accepted: 07/30/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy is a promising potential therapeutic strategy to treat cerebral ischemia in preclinical and clinical trials. Currently proposed treatments for stroke employing stem cells include the replacement of lost neurons and integration into the existing host circuitry, the release of growth factors to support and promote endogenous repair processes, and the secretion of extracellular vesicles containing proteins, noncoding RNA, or DNA to regulate gene expression in recipient cells and achieve immunomodulation. Progress has been made to elucidate the precise mechanisms underlying stem cell therapy and the homing, migration, distribution, and differentiation of transplanted stem cells in vivo using various imaging modalities. Noninvasive and safe tracer agents with high sensitivity and image resolution must be combined with long-term monitoring using imaging technology to determine the optimal therapy for stroke in terms of administration route, dosage, and timing. This review discusses potential therapeutic mechanisms of stem cell transplantation for the treatment of stroke and the limitations of current therapies. Methods to label transplanted cells and existing imaging systems for stem cell labeling and in vivo tracking will also be discussed.
Collapse
|
23
|
Yin X, Kidd GJ, Ohno N, Perkins GA, Ellisman MH, Bastian C, Brunet S, Baltan S, Trapp BD. Proteolipid protein-deficient myelin promotes axonal mitochondrial dysfunction via altered metabolic coupling. J Cell Biol 2017; 215:531-542. [PMID: 27872255 PMCID: PMC5119941 DOI: 10.1083/jcb.201607099] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/01/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022] Open
Abstract
The authors show that central nervous system myelin lacking proteolipid protein (PLP) induces mitochondrial dysfunction, including altered motility, degeneration, and ectopic smooth endoplasmic reticulum interactions, leading to axonal structural defects and degeneration. Mutated PLP occurs in hereditary spastic paraplegia, and these cellular effects provide potential insight into the pathology of the disease. Hereditary spastic paraplegia (HSP) is a neurological syndrome characterized by degeneration of central nervous system (CNS) axons. Mutated HSP proteins include myelin proteolipid protein (PLP) and axon-enriched proteins involved in mitochondrial function, smooth endoplasmic reticulum (SER) structure, and microtubule (MT) stability/function. We characterized axonal mitochondria, SER, and MTs in rodent optic nerves where PLP is replaced by the peripheral nerve myelin protein, P0 (P0-CNS mice). Mitochondrial pathology and degeneration were prominent in juxtaparanodal axoplasm at 1 mo of age. In wild-type (WT) optic nerve axons, 25% of mitochondria–SER associations occurred on extensions of the mitochondrial outer membrane. Mitochondria–SER associations were reduced by 86% in 1-mo-old P0-CNS juxtaparanodal axoplasm. 1-mo-old P0-CNS optic nerves were more sensitive to oxygen-glucose deprivation and contained less adenosine triphosphate (ATP) than WT nerves. MT pathology and paranodal axonal ovoids were prominent at 6 mo. These data support juxtaparanodal mitochondrial degeneration, reduced mitochondria–SER associations, and reduced ATP production as causes of axonal ovoid formation and axonal degeneration.
Collapse
Affiliation(s)
- Xinghua Yin
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Grahame J Kidd
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Nobuhiko Ohno
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093
| | - Chinthasagar Bastian
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Sylvain Brunet
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Selva Baltan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
24
|
Histone deacetylase inhibition is cytotoxic to oligodendrocyte precursor cells in vitro and in vivo. Int J Dev Neurosci 2016; 54:53-61. [PMID: 27587342 DOI: 10.1016/j.ijdevneu.2016.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/26/2016] [Accepted: 08/27/2016] [Indexed: 11/24/2022] Open
Abstract
Histone deacetylase (HDAC) inhibition mediated by small molecule HDAC inhibitors (HDACi) has demonstrated divergent effects including toxicity towards transformed cell lines, neuroprotection in neurological disease models, and inhibition of oligodendrocyte precursor cell (OPC) differentiation to mature oligodendrocytes (OL). However, it remains unknown if transient HDAC inhibition may promote OPC survival. Using mouse cortical OPC primary cultures, we investigated the effects of the FDA approved pan-HDACi suberoylanilide hydroxamic acid (SAHA) on OPC survival. Initial studies showed differences in the HDAC expression pattern of multiple HDAC isoforms in OPCs relative to their terminally differentiated progeny cells, OLs and astrocytes. Treatment of OPCs with SAHA for up to 72h using a maximum concentration either at or lower than those necessary for cytotoxicity in most transformed cell lines resulted in over 67% reduction in viability relative to vehicle-treated OPCs. This was at least partly due to increased apoptosis as SAHA-treated cells displayed activated caspase 3 and were protected by the general caspase inhibitor Q-VD-OPH. Additionally, SAHA treatment of whole mice at postnatal day 5 induced apoptosis of cortical OPCs. These results suggest that SAHA negatively impacts OPC survival and may be detrimental to the myelinating brain and spinal cord. Such toxicity may be relevant in a clinical context as SAHA is currently involved in numerous clinical trials and is in consideration for use in the treatment of psychiatric and neurodegenerative conditions.
Collapse
|
25
|
Saini V, Lutz D, Kataria H, Kaur G, Schachner M, Loers G. The polysialic acid mimetics 5-nonyloxytryptamine and vinorelbine facilitate nervous system repair. Sci Rep 2016; 6:26927. [PMID: 27324620 PMCID: PMC4914991 DOI: 10.1038/srep26927] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/07/2016] [Indexed: 02/05/2023] Open
Abstract
Polysialic acid (PSA) is a large negatively charged glycan mainly attached to the neural cell adhesion molecule (NCAM). Several studies have shown that it is important for correct formation of brain circuitries during development and for synaptic plasticity, learning and memory in the adult. PSA also plays a major role in nervous system regeneration following injury. As a next step for clinical translation of PSA based therapeutics, we have previously identified the small organic compounds 5-nonyloxytryptamine and vinorelbine as PSA mimetics. Activity of 5-nonyloxytryptamine and vinorelbine had been confirmed in assays with neural cells from the central and peripheral nervous system in vitro and shown to be independent of their function as serotonin receptor 5-HT1B/1D agonist or cytostatic drug, respectively. As we show here in an in vivo paradigm for spinal cord injury in mice, 5-nonyloxytryptamine and vinorelbine enhance regain of motor functions, axonal regrowth, motor neuron survival and remyelination. These data indicate that 5-nonyloxytryptamine and vinorelbine may be re-tasked from their current usage as a 5-HT1B/1D agonist or cytostatic drug to act as mimetics for PSA to stimulate regeneration after injury in the mammalian nervous system.
Collapse
Affiliation(s)
- Vedangana Saini
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
- Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - David Lutz
- Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - Hardeep Kataria
- Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Melitta Schachner
- Keck Center for Collaborative Neurosciences, Rutgers University, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, People’s Republic of China
| | - Gabriele Loers
- Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| |
Collapse
|
26
|
Wang Y, Liu G, Hong D, Chen F, Ji X, Cao G. White matter injury in ischemic stroke. Prog Neurobiol 2016; 141:45-60. [PMID: 27090751 PMCID: PMC5677601 DOI: 10.1016/j.pneurobio.2016.04.005] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/01/2016] [Accepted: 04/10/2016] [Indexed: 02/06/2023]
Abstract
Stroke is one of the major causes of disability and mortality worldwide. It is well known that ischemic stroke can cause gray matter injury. However, stroke also elicits profound white matter injury, a risk factor for higher stroke incidence and poor neurological outcomes. The majority of damage caused by stroke is located in subcortical regions and, remarkably, white matter occupies nearly half of the average infarct volume. Indeed, white matter is exquisitely vulnerable to ischemia and is often injured more severely than gray matter. Clinical symptoms related to white matter injury include cognitive dysfunction, emotional disorders, sensorimotor impairments, as well as urinary incontinence and pain, all of which are closely associated with destruction and remodeling of white matter connectivity. White matter injury can be noninvasively detected by MRI, which provides a three-dimensional assessment of its morphology, metabolism, and function. There is an urgent need for novel white matter therapies, as currently available strategies are limited to preclinical animal studies. Optimal protection against ischemic stroke will need to encompass the fortification of both gray and white matter. In this review, we discuss white matter injury after ischemic stroke, focusing on clinical features and tools, such as imaging, manifestation, and potential treatments. We also briefly discuss the pathophysiology of WMI and future research directions.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Gang Liu
- Department of Neurology, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Dandan Hong
- Department of Bioengineering, University of Pittsburgh School of Engineering, United States
| | - Fenghua Chen
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China.
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States; Geriatric Research Education and Clinical Centers, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, United States.
| |
Collapse
|
27
|
Zhao H, Han Z, Ji X, Luo Y. Epigenetic Regulation of Oxidative Stress in Ischemic Stroke. Aging Dis 2016; 7:295-306. [PMID: 27330844 PMCID: PMC4898926 DOI: 10.14336/ad.2015.1009] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/09/2015] [Indexed: 12/11/2022] Open
Abstract
The prevalence and incidence of stroke rises with life expectancy. However, except for the use of recombinant tissue-type plasminogen activator, the translation of new therapies for acute stroke from animal models into humans has been relatively unsuccessful. Oxidative DNA and protein damage following stroke is typically associated with cell death. Cause-effect relationships between reactive oxygen species and epigenetic modifications have been established in aging, cancer, acute pancreatitis, and fatty liver disease. In addition, epigenetic regulatory mechanisms during stroke recovery have been reviewed, with focuses mainly on neural apoptosis, necrosis, and neuroplasticity. However, oxidative stress-induced epigenetic regulation in vascular neural networks following stroke has not been sufficiently explored. Improved understanding of the epigenetic regulatory network upon oxidative stress may provide effective antioxidant approaches for treating stroke. In this review, we summarize the epigenetic events, including DNA methylation, histone modification, and microRNAs, that result from oxidative stress following experimental stroke in animal and cell models, and the ways in which epigenetic changes and their crosstalk influence the redox state in neurons, glia, and vascular endothelial cells, helping us to understand the foregone and vicious epigenetic regulation of oxidative stress in the vascular neural network following stroke.
Collapse
Affiliation(s)
- Haiping Zhao
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Ziping Han
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- 22Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yumin Luo
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; 3Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
| |
Collapse
|
28
|
Khoury N, Koronowski KB, Perez-Pinzon MA. Long-term window of ischemic tolerance: An evolutionarily conserved form of metabolic plasticity regulated by epigenetic modifications? ACTA ACUST UNITED AC 2016; 1:6-12. [PMID: 27796011 DOI: 10.29245/2572.942x/2016/2.1021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the absence of effective neuroprotective agents in the clinic, ischemic and pharmacological preconditioning are gaining increased interest in the field of cerebral ischemia. Our lab recently reported that resveratrol preconditioning affords tolerance against a focal cerebral ischemic insult in mice that can last for at least 14 days in vivo making it the longest window of ischemic tolerance discovered to date by a single administration of a pharmacological agent. The mechanism behind this novel extended window of ischemic tolerance remains elusive. In the below commentary we discuss potential mechanisms that could explain this novel extended window of ischemic tolerance in the context of previously identified windows and the known mechanisms behind them. We also draw parallels from the fields of hibernation and hypoxia-tolerance, which are chronic adaptations to severe conditions of hypoxia and ischemia known to be mediated by a form of metabolic depression. We also briefly discuss the importance of epigenetic modifications in maintaining this depressed state of metabolism.
Collapse
Affiliation(s)
- Nathalie Khoury
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Kevin B Koronowski
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Miguel A Perez-Pinzon
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
29
|
Glutamate and ATP at the Interface Between Signaling and Metabolism in Astroglia: Examples from Pathology. Neurochem Res 2016; 42:19-34. [PMID: 26915104 DOI: 10.1007/s11064-016-1848-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 01/21/2016] [Accepted: 01/22/2016] [Indexed: 12/17/2022]
Abstract
Glutamate is the main excitatory transmitter in the brain, while ATP represents the most important energy currency in any living cell. Yet, these chemicals play an important role in both processes, enabling them with dual-acting functions in metabolic and intercellular signaling pathways. Glutamate can fuel ATP production, while ATP can act as a transmitter in intercellular signaling. We discuss the interface between glutamate and ATP in signaling and metabolism of astrocytes. Not only do glutamate and ATP cross each other's paths in physiology of the brain, but they also do so in its pathology. We present the fabric of this process in (patho)physiology through the discussion of synthesis and metabolism of ATP and glutamate in astrocytes as well as by providing a general description of astroglial receptors for these molecules along with the downstream signaling pathways that may be activated. It is astroglial receptors for these dual-acting molecules that could hold a key for medical intervention in pathological conditions. We focus on two examples disclosing the role of activation of astroglial ATP and glutamate receptors in pathology of two kinds of brain tissue, gray matter and white matter, respectively. Interventions at the interface of metabolism and signaling show promise for translational medicine.
Collapse
|
30
|
Ransom BR, Goldberg MP, Arai K, Baltan S. White Matter Pathophysiology. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
31
|
Schmitt HM, Schlamp CL, Nickells RW. Role of HDACs in optic nerve damage-induced nuclear atrophy of retinal ganglion cells. Neurosci Lett 2015; 625:11-5. [PMID: 26733303 DOI: 10.1016/j.neulet.2015.12.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/16/2015] [Accepted: 12/08/2015] [Indexed: 12/13/2022]
Abstract
Optic neuropathies are characterized by retinal ganglion cell (RGC) death, resulting in the loss of vision. In glaucoma, the most common optic neuropathy, RGC death is initiated by axonal damage, and can be modeled by inducing acute axonal trauma through procedures such as optic nerve crush (ONC) or optic nerve axotomy. One of the early events of RGC death is nuclear atrophy, and is comprised of RGC-specific gene silencing, histone deacetylation, heterochromatin formation, and nuclear shrinkage. These early events appear to be principally regulated by epigenetic mechanisms involving histone deacetylation. Class I histone deacetylases HDACs 1, 2, and 3 are known to play important roles in the process of early nuclear atrophy in RGCs, and studies using both inhibitors and genetic ablation of Hdacs also reveal a critical role in the cell death process. Select inhibitors, such as those being developed for cancer therapy, may also provide a viable secondary treatment option for optic neuropathies.
Collapse
Affiliation(s)
- Heather M Schmitt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Cassandra L Schlamp
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert W Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
32
|
Kalani A, Kamat PK, Kalani K, Tyagi N. Epigenetic impact of curcumin on stroke prevention. Metab Brain Dis 2015; 30:427-35. [PMID: 24788895 PMCID: PMC4216637 DOI: 10.1007/s11011-014-9537-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/26/2014] [Indexed: 12/31/2022]
Abstract
The epigenetic impact of curcumin in stroke and neurodegenerative disorders is curiosity-arousing. It is derived from Curcuma longa (spice), possesses anti-oxidative, anti-inflammatory, anti-lipidemic, neuro-protective and recently shown to exhibit epigenetic modulatory properties. Epigenetic studies include DNA methylation, histone modifications and RNA-based mechanisms which regulate gene expression without altering nucleotide sequences. Curcumin has been shown to affect cancer by altering epigenetic changes but its role as an epigenetic agent in cerebral stroke has not been much explored. Although curcumin possesses remarkable medicinal properties, the bioavailability of curcumin has limited its success in epigenetic studies and clinical trials. The present review is therefore designed to look into epigenetic mechanisms that could be induced with curcumin during stroke, along with its molecular designing with different moieties that may increase its bioavailability. Curcumin has been shown to be encapsulated in exosomes, nano-vesicles (<200 nm), thereby showing its therapeutic effects in brain diseases. Curcumin delivered through nanoparticles has been shown to be neuroregenerative but the use of nanoparticles in brain has limitations. Hence, curcumin-encapsulated exosomes along with curcumin-primed exosomes (exosomes released by curcumin-treated cells) are much needed to be explored to broadly look into their use as a novel therapy for stroke.
Collapse
Affiliation(s)
- Anuradha Kalani
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Pradip K Kamat
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Komal Kalani
- Central Institute of Medicinal and Aromatic Plants-Council of Scientific and Industrial Research (CIMAP-CSIR), Lucknow-226001, India
| | - Neetu Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
33
|
Baltan S. Excitotoxicity and mitochondrial dysfunction underlie age-dependent ischemic white matter injury. ADVANCES IN NEUROBIOLOGY 2014; 11:151-70. [PMID: 25236728 PMCID: PMC8937575 DOI: 10.1007/978-3-319-08894-5_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The central nervous system white matter is damaged during an ischemic stroke and therapeutic strategies derived from experimental studies focused exclusively on young adults and gray matter have been unsuccessful in the more clinically relevant aging population. The risk for stroke increases with age and the white matter inherently becomes more susceptible to injury as a function of age. Age-related changes in the molecular architecture of white matter determine the principal injury mechanisms and the functional outcome. A prominent increase in the main plasma membrane Na(+)-dependent glutamate transporter, GLT-1/EAAT2, together with increased extracellular glutamate levels may reflect an increased need for glutamate signaling in the aging white matter to maintain its function. Mitochondria exhibit intricate dynamics to efficiently buffer Ca(2+), to produce sufficient ATP, and to effectively scavenge reactive oxygen species (ROS) in response to excitotoxicity to sustain axon function. Aging exacerbates mitochondrial fusion, leading to progressive alterations in mitochondrial dynamics and function, presumably to effectively buffer increased Ca(2+) load and ROS production. Interestingly, these adaptive adjustments become detrimental under ischemic conditions, leading to increased and early glutamate release and a rapid exhaustion of mitochondrial capacity to sustain energy status of axons. Consequently, protective interventions in young white matter become injurious or ineffective to promote recovery in aging white matter after an ischemic episode. An age-specific understanding of the mechanisms of injury processes in white matter is vital in order to design dynamic therapeutic approaches for stroke victims.
Collapse
Affiliation(s)
- Selva Baltan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NC30, Cleveland, OH, 44195, USA,
| |
Collapse
|
34
|
Du L, Risinger AL, King JB, Powell DR, Cichewicz RH. A potent HDAC inhibitor, 1-alaninechlamydocin, from a Tolypocladium sp. induces G2/M cell cycle arrest and apoptosis in MIA PaCa-2 cells. JOURNAL OF NATURAL PRODUCTS 2014; 77:1753-7. [PMID: 24999749 PMCID: PMC4113265 DOI: 10.1021/np500387h] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Indexed: 05/23/2023]
Abstract
The cyclic tetrapeptide 1-alaninechlamydocin was purified from a Great Lakes-derived fungal isolate identified as a Tolypocladium sp. Although the planar structure was previously described, a detailed analysis of its spectroscopic data and biological activity are reported here for the first time. Its absolute configuration was determined using a combination of spectroscopic ((1)H-(1)H ROESY, ECD, and X-ray diffraction) and chemical (Marfey's analysis) methods. 1-Alaninechlamydocin showed potent antiproliferative/cytotoxic activities in a human pancreatic cancer cell line (MIA PaCa-2) at low-nanomolar concentrations (GI50 5.3 nM, TGI 8.8 nM, LC50 22 nM). Further analysis revealed that 1-alaninechlamydocin induced G2/M cell cycle arrest and apoptosis. Similar to other cyclic epoxytetrapeptides, the inhibitory effects of 1-alaninechlamydocin are proposed to be produced primarily via inhibition of histone deacetylase (HDAC) activity.
Collapse
Affiliation(s)
- Lin Du
- Department of Chemistry and Biochemistry, Stephenson
Life Sciences Research Center, Natural Products Discovery Group, and Institute
for Natural Products Applications and Research Technologies, University of Oklahoma, Norman, Oklahoma 73019-5251, United States
| | - April L. Risinger
- Department of Pharmacology and Cancer Therapy & Research Center, University of Texas Health Science Center, San Antonio, Texas 78229, United States
| | - Jarrod B. King
- Department of Chemistry and Biochemistry, Stephenson
Life Sciences Research Center, Natural Products Discovery Group, and Institute
for Natural Products Applications and Research Technologies, University of Oklahoma, Norman, Oklahoma 73019-5251, United States
| | - Douglas R. Powell
- Department of Chemistry and Biochemistry, Stephenson
Life Sciences Research Center, Natural Products Discovery Group, and Institute
for Natural Products Applications and Research Technologies, University of Oklahoma, Norman, Oklahoma 73019-5251, United States
| | - Robert H. Cichewicz
- Department of Chemistry and Biochemistry, Stephenson
Life Sciences Research Center, Natural Products Discovery Group, and Institute
for Natural Products Applications and Research Technologies, University of Oklahoma, Norman, Oklahoma 73019-5251, United States
| |
Collapse
|
35
|
Kim HJ, Chuang DM. HDAC inhibitors mitigate ischemia-induced oligodendrocyte damage: potential roles of oligodendrogenesis, VEGF, and anti-inflammation. Am J Transl Res 2014; 6:206-223. [PMID: 24936215 PMCID: PMC4058304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 04/18/2014] [Indexed: 06/03/2023]
Abstract
White matter injury is an important component of stroke pathology, but its pathophysiology and potential treatment remain relatively elusive and underexplored. We previously reported that after permanent middle cerebral artery occlusion (pMCAO), sodium butyrate (SB) and trichostatin A (TSA) induced neurogenesis via histone deacetylase (HDAC) inhibition in multiple ischemic brain regions in rats; these effects-which depended on activation of brain-derived neurotrophic factor (BDNF)-TrkB signaling-contributed to behavioral improvement. The present study found that SB or TSA robustly protected against ischemia-induced loss of oligodendrocytes detected by confocal microscopy of myelin basic protein (MBP) immunostaining in the ipsilateral subventricular zone (SVZ), striatum, corpus callosum, and frontal cortex seven days post-pMCAO. Co-localization of 5-bromo-2'-deoxyuridine (BrdU)(+) and MBP(+) cells after SB treatment suggested the occurrence of oligodendrogenesis. SB also strongly upregulated vascular endothelial growth factor (VEGF), which plays a major role in neurogenesis, angiogenesis, and functional recovery after stroke. These SB-induced effects were markedly suppressed by blocking the TrkB signaling pathway with K252a. pMCAO-induced activation of microglia (OX42(+)) and macrophages/monocytes (ED1(+))-which has been linked to white matter injury-was robustly suppressed by SB in a K252a-sensitive manner. In addition, SB treatment largely blocked caspase-3(+) and OX42(+) cells in ipsilateral brain regions. Our results suggest that HDAC inhibitor-mediated protection against ischemia-induced oligodendrocyte loss may involve multiple mechanisms including oligodendrogenesis, VEGF upregulation, anti-inflammation, and caspase-3 downregulation. Taken together, the results suggest that post-insult treatment with HDAC inhibitors is a rational strategy to mitigate white matter injury following ischemic stroke.
Collapse
Affiliation(s)
- Hyeon Ju Kim
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health Bethesda, MD 20892-1363, USA
| | - De-Maw Chuang
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health Bethesda, MD 20892-1363, USA
| |
Collapse
|
36
|
Murphy SP, Lee RJ, McClean ME, Pemberton HE, Uo T, Morrison RS, Bastian C, Baltan S. MS-275, a class I histone deacetylase inhibitor, protects the p53-deficient mouse against ischemic injury. J Neurochem 2014; 129:509-15. [PMID: 24147654 PMCID: PMC8937574 DOI: 10.1111/jnc.12498] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 01/24/2023]
Abstract
The administration of pan histone deacetylase (HDAC) inhibitors reduces ischemic damage to the CNS, both in vitro and in animal models of stroke, via mechanisms which we are beginning to understand. The acetylation of p53 is regulated by Class I HDACs and, because p53 appears to play a role in ischemic pathology, the purpose of this study was to discover, using an in vitro white matter ischemia model and an in vivo cerebral ischemia model, if neuroprotection mediated by HDAC inhibition depended on p53 expression. Optic nerves were excised from wild-type and p53-deficient mice, and then subjected to oxygen-glucose deprivation in the presence and absence of a specific inhibitor of Class I HDACs (MS-275, entinostat) while compound action potentials were recorded. Furthermore, transient focal ischemia was imposed on wild-type and p53-deficient mice, which were subsequently treated with MS-275. Interestingly, and in both scenarios, the beneficial effects of MS-275 were most pronounced when p53 was absent. These results suggest that modulation of p53 activity is not responsible for MS-275-mediated neuroprotection, and further illustrate how HDAC inhibitors variably influence p53 and associated apoptotic pathways. Optic nerves from wild-type and p53-deficient mice, engineered to express cyan fluorescent protein (CFP) in neuronal mitochondria, were subjected to oxygen-glucose deprivation (OGD) in the presence and absence of a specific inhibitor of Class I histone deacetylases. The protective effect of MS-275 was evidenced by mitochondrial preservation, and this was most pronounced in the absence of p53.
Collapse
Affiliation(s)
- Sean P. Murphy
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Rona J. Lee
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Megan E. McClean
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Heather E. Pemberton
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Takuma Uo
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Richard S. Morrison
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Chinthasagar Bastian
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Selva Baltan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
37
|
Basso M, Sleiman S, Ratan RR. Looking above but not beyond the genome for therapeutics in neurology and psychiatry: epigenetic proteins and RNAs find a new focus. Neurotherapeutics 2013; 10:551-5. [PMID: 24092616 PMCID: PMC3805863 DOI: 10.1007/s13311-013-0225-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Manuela Basso
- />Burke Medical Research Institute, Weill Medical College of Cornell University, 785 Mamaroneck Avenue, White Plains, New York, 10605 NY USA
- />Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY USA
- />Centre for Integrative Biology, CIBIO, University of Trento, Trento, Italy
| | - Sama Sleiman
- />Burke Medical Research Institute, Weill Medical College of Cornell University, 785 Mamaroneck Avenue, White Plains, New York, 10605 NY USA
- />Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY USA
- />Molecular Neurobiology Program, Skirball Institute of Bimolecular Medicine, New York University School of Medicine, New York, NY USA
| | - Rajiv R. Ratan
- />Burke Medical Research Institute, Weill Medical College of Cornell University, 785 Mamaroneck Avenue, White Plains, New York, 10605 NY USA
- />Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY USA
- />Department of Neurology, Weill Medical College of Cornell University, New York, NY USA
| |
Collapse
|