1
|
Mitrofanis J, Stone J, Hamblin MR, Magistretti P, Benabid AL, Jeffery G. A spotlight on dosage and subject selection for effective neuroprotection: exploring the central role of mitochondria. Neural Regen Res 2025; 20:1081-1082. [PMID: 38989941 PMCID: PMC11438325 DOI: 10.4103/nrr.nrr-d-24-00222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/30/2024] [Indexed: 07/12/2024] Open
Affiliation(s)
- John Mitrofanis
- Fonds Clinatec, Université Grenoble Alpes, Grenoble, France (Mitrofanis J, Benabid AL)
- Institute of Ophthalmology, University College London, London, UK (Mitrofanis J, Jeffery G)
| | - Jonathan Stone
- School of Medical Science, University of Sydney, Sydney, Australia (Stone J)
| | - Michael R Hamblin
- Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa (Hamblin MR)
| | - Pierre Magistretti
- King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (Magistretti P)
| | - Alim-Louis Benabid
- Fonds Clinatec, Université Grenoble Alpes, Grenoble, France (Mitrofanis J, Benabid AL)
| | - Glen Jeffery
- Institute of Ophthalmology, University College London, London, UK (Mitrofanis J, Jeffery G)
| |
Collapse
|
2
|
Sisalli MJ, D'Apolito E, Cuomo O, Lombardi G, Tufano M, Annunziato L, Scorziello A. The E3-ligase Siah2 activates mitochondrial quality control in neurons to maintain energy metabolism during ischemic brain tolerance. Cell Death Dis 2025; 16:52. [PMID: 39875361 PMCID: PMC11775118 DOI: 10.1038/s41419-025-07339-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025]
Abstract
Mitochondrial quality control is crucial for the homeostasis of the mitochondrial network. The balance between mitophagy and biogenesis is needed to reduce cerebral ischemia-induced cell death. Ischemic preconditioning (IPC) represents an adaptation mechanism of CNS that increases tolerance to lethal cerebral ischemia. It has been demonstrated that hypoxia-induced Seven in absentia Homolog 2 (Siah2) E3-ligase activation influences mitochondrial dynamics promoting the degradation of mitochondrial proteins. Therefore, in the present study, we investigated the role of Siah2 in the IPC-induced neuroprotection in in vitro and in vivo models of IPC. To this aim, cortical neurons were exposed to 30-min oxygen and glucose deprivation (OGD, sublethal insult) followed by 3 h OGD plus reoxygenation (lethal insult). Our results revealed that the mitochondrial depolarization induced by hypoxia activates Siah2 at the mitochondrial level and increases LC3-II protein expression, a marker of mitophagy, an effect counteracted by the reoxygenation phase. By contrast, hypoxia reduced the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a marker of mitochondrial biogenesis, whereas its expression was increased after reoxygenation thus improving mitochondrial membrane potential, mitochondrial calcium content, and mitochondrial morphology, hence leading to neuroprotection in IPC. Furthermore, Siah2 silencing confirmed these results. Collectively, these findings indicate that the balance between mitophagy and mitochondrial biogenesis, due to the activation of the Siah2-E3-ligase, might play a role in IPC-induced neuroprotection.
Collapse
Affiliation(s)
- Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Elena D'Apolito
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Giovanna Lombardi
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Michele Tufano
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | | | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
3
|
Okonkwo E, Saha B, Sahu G, Bera A, Sharma P. Blood-Based Lateral-Flow Immunoassays Dipstick Test for Damaged Mitochondrial Electron Transport Chain in Pyruvate Treated Rats with Combined Blast Exposure and Hemorrhagic Shock. J Clin Med 2025; 14:754. [PMID: 39941423 PMCID: PMC11818850 DOI: 10.3390/jcm14030754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Blast trauma presents a unique challenge due to its complex mechanism of injury, which impacts the brain and other vital organs through overpressure waves and internal bleeding. Severe blood loss leads to an inadequate oxygen supply and insufficient fuel delivery to cells, impairing ATP production by mitochondria-essential for cell survival. While clinical symptoms of metabolic disruption are evident soon after injury, the molecular, cellular, and systemic damage persists for days to years post-injury. Current challenges in treating traumatic brain injury (TBI) stem from (1) the lack of early blood-based biomarkers for detecting metabolic failure and mitochondrial damage and (2) the limited success of mitochondrial-targeted therapeutic strategies. Objectives: To identify blood-based mitochondrial biomarkers for evaluating the severity of brain injuries and to investigate therapeutic strategies targeting mitochondria. Methods: A preclinical rat model subjected to blast exposure, with or without hemorrhagic shock (HS), followed by resuscitation was utilized. Blood samples were obtained at baseline (T0), post-injury (T60), and at the conclusion of the experiment (T180), and analyzed using a validated dipstick assay to measure mitochondrial enzyme activity. Results: Blast and HS injuries led to a significant decrease in the activity of mitochondrial enzymes, including complex I, complex IV, and the pyruvate dehydrogenase complex (PDH), compared to baseline (p < 0.05). Concurrently, blood lactate concentrations were significantly elevated (p < 0.001). An inverse correlation was observed between mitochondrial enzyme dysfunction and blood lactate levels (p < 0.05). Treatment with sodium pyruvate post-injury restored complex I, complex IV, and PDH activity to near-baseline levels, corrected hyperlactatemia, and reduced reactive oxygen species (ROS) production by mitochondria. Conclusions: Serial monitoring of blood mitochondrial enzyme activity, such as complex I, complex IV, and PDH, may serve as a valuable tool for prognostication and guiding the use of mitochondrial-targeted therapies. Additionally, mitochondrial enzyme assays in blood samples can provide insights into the global redox status, potentially paving the way for novel therapeutic interventions in TBI.
Collapse
Affiliation(s)
| | | | | | | | - Pushpa Sharma
- Department of Anesthesiology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
4
|
Serikbaeva A, Li Y, Ma S, Yi D, Kazlauskas A. Resilience to diabetic retinopathy. Prog Retin Eye Res 2024; 101:101271. [PMID: 38740254 PMCID: PMC11262066 DOI: 10.1016/j.preteyeres.2024.101271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Chronic elevation of blood glucose at first causes relatively minor changes to the neural and vascular components of the retina. As the duration of hyperglycemia persists, the nature and extent of damage increases and becomes readily detectable. While this second, overt manifestation of diabetic retinopathy (DR) has been studied extensively, what prevents maximal damage from the very start of hyperglycemia remains largely unexplored. Recent studies indicate that diabetes (DM) engages mitochondria-based defense during the retinopathy-resistant phase, and thereby enables the retina to remain healthy in the face of hyperglycemia. Such resilience is transient, and its deterioration results in progressive accumulation of retinal damage. The concepts that co-emerge with these discoveries set the stage for novel intellectual and therapeutic opportunities within the DR field. Identification of biomarkers and mediators of protection from DM-mediated damage will enable development of resilience-based therapies that will indefinitely delay the onset of DR.
Collapse
Affiliation(s)
- Anara Serikbaeva
- Department of Physiology and Biophysics, University of Illinois at Chicago, 1905 W Taylor St, Chicago, IL 60612, USA
| | - Yanliang Li
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W Taylor St, Chicago, IL 60612, USA
| | - Simon Ma
- Department of Bioengineering, University of Illinois at Chicago, 1905 W Taylor St, Chicago, IL 60612, USA
| | - Darvin Yi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W Taylor St, Chicago, IL 60612, USA; Department of Bioengineering, University of Illinois at Chicago, 1905 W Taylor St, Chicago, IL 60612, USA
| | - Andrius Kazlauskas
- Department of Physiology and Biophysics, University of Illinois at Chicago, 1905 W Taylor St, Chicago, IL 60612, USA; Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W Taylor St, Chicago, IL 60612, USA.
| |
Collapse
|
5
|
Baskaran K, Johnson JT, Prem PN, Ravindran S, Kurian GA. Evaluation of prophylactic efficacy of sodium thiosulfate in combating I/R injury in rat brain: exploring its efficiency further in vascular calcified brain slice model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2587-2598. [PMID: 37058187 DOI: 10.1007/s00210-023-02481-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
Cerebral ischemia reperfusion injury (CIR) is one of the clinical manifestations encountered during the management of stroke. High prevalence of intracranial arterial calcification is reported in stroke patients. However, the impact of vascular calcification (VC) in the outcome of CIR and the efficacy of mechanical preconditioning (IPC) and pharmacological conditioning with sodium thiosulphate (STS) in ameliorating IR remains unclear. Two experimental models namely carotid artery occlusion (n = 36) and brain slice models (n = 18) were used to evaluate the efficacy of STS in male Wistar rats. IR was inflicted in rat by occluding carotid artery for 30 min followed by 24-h reperfusion after STS (100 mg/kg) administration. Brain slice model was used to reconfirm the results to account blood brain barrier permeability. Further, brain slice tissue was utilised to evaluate the efficacy of STS in VC rat brain by measuring the histological alterations and biochemical parameters. Pre-treatment of STS prior to CIR in intact animal significantly reduced the IR-associated histopathological alterations in brain, declined oxidative stress and improved the mitochondrial function found to be similar to IPC. Brain slice model data also confirmed the neuroprotective effect of STS similar to IPC in IR challenged tissue slice. Higher tissue injury was noted in VC brain IR tissue than normal IR tissue. Therapeutic efficacy of STS was evident in VC rat brain tissues and normal tissues subjected to IR. On the other hand, IPC-mediated protection was noted only in IR normal and adenine-induced VC brain tissues not in high-fat diet (HFD) induced VC brain tissues. Based on the results, we concluded that similar to IPC, STS was effective in attenuating IR injury in CIR rat brain. Vascular calcification adversely affected the recovery protocol of brain tissues from ischemic insult. STS was found to be an effective agent in ameliorating the IR injury in both adenine and HFD induced vascular calcified rat brain, but IPC-mediated neuroprotection was absent in HFD-induced VC brain tissues.
Collapse
Affiliation(s)
- Keerthana Baskaran
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
| | - Jefri Thimoathi Johnson
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
| | - Priyanka N Prem
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
- School of Chemical and Biotechnology, SASTRA Deemed University, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
| | - Sriram Ravindran
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
| | - Gino A Kurian
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India.
- School of Chemical and Biotechnology, SASTRA Deemed University, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India.
| |
Collapse
|
6
|
Rahman SO, Khan T, Iqubal A, Agarwal S, Akhtar M, Parvez S, Shah ZA, Najmi AK. Association between insulin and Nrf2 signalling pathway in Alzheimer's disease: A molecular landscape. Life Sci 2023:121899. [PMID: 37394097 DOI: 10.1016/j.lfs.2023.121899] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/17/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
Insulin, a well-known hormone, has been implicated as a regulator of blood glucose levels for almost a century now. Over the past few decades, the non-glycemic actions of insulin i.e. neuronal growth and proliferation have been extensively studied. In 2005, Dr. Suzanne de La Monte and her team reported that insulin might be involved in the pathogenesis of Alzheimer's Disease (AD) and thus coined a term "Type-3 diabetes" This hypothesis was supported by several subsequent studies. The nuclear factor erythroid 2- related factor 2 (Nrf2) triggers a cascade of events under the regulation of distinct mechanisms including protein stability, phosphorylation and nuclear cytoplasmic shuttling, finally leading to the protection against oxidative damage. The Nrf2 pathway has been investigated extensively in relevance to neurodegenerative disorders, particularly AD. Many studies have indicated a strong correlation between insulin and Nrf2 signalling pathways both in the periphery and the brainbut merely few of them have focused on elucidating their inter-connective role in AD. The present review emphasizes key molecular pathways that correlate the role of insulin with Nrf2 during AD. The review has also identified key unexplored areas that could be investigated in future to further establish the insulin and Nrf2 influence in AD.
Collapse
Affiliation(s)
- Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Tahira Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Shivani Agarwal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Neurobehavioral Pharmacology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Zahoor Ahmad Shah
- Department of Medicinal and Biological Chemistry, University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
7
|
Ikeno Y, Ghincea CV, Roda GF, Cheng L, Aftab M, Meng X, Weyant MJ, Cleveland JC, Fullerton DA, Reece TB. Direct and indirect activation of the adenosine triphosphate-sensitive potassium channel to induce spinal cord ischemic metabolic tolerance. J Thorac Cardiovasc Surg 2023; 165:e90-e99. [PMID: 34763893 DOI: 10.1016/j.jtcvs.2021.08.085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/07/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The mitochondrial adenosine triphosphate-sensitive potassium channel is central to pharmacologically induced tolerance to spinal cord injury. We hypothesized that both direct and nitric oxide-dependent indirect activation of the adenosine triphosphate-sensitive potassium channel contribute to the induction of ischemic metabolic tolerance. METHODS Spinal cord injury was induced in adult male C57BL/6 mice through 7 minutes of thoracic aortic crossclamping. Pretreatment consisted of intraperitoneal injection 3 consecutive days before injury. Experimental groups were sham (no pretreatment or ischemia, n = 10), spinal cord injury control (pretreatment with normal saline, n = 27), Nicorandil 1.0 mg/kg (direct and indirect adenosine triphosphate-sensitive potassium channel opener, n = 20), Nicorandil 1 mg/kg + carboxy-PTIO 1 mg/kg (nitric oxide scavenger, n = 21), carboxy-PTIO (n = 12), diazoxide 5 mg/kg (selective direct adenosine triphosphate-sensitive potassium channel opener, n = 25), and DZ 5 mg/kg+ carboxy-PTIO 1 mg/kg, carboxy-PTIO (n = 23). Limb motor function was assessed using the Basso Mouse Score (0-9) at 12-hour intervals for 48 hours after ischemia. RESULTS Motor function was significantly preserved at all time points after ischemia in the Nicorandil pretreatment group compared with ischemic control. The addition of carboxy-PTIO partially attenuated Nicorandil's motor-preserving effect. Motor function in the Nicorandil + carboxy-PTIO group was significantly preserved compared with the spinal cord injury control group (P < .001), but worse than in the Nicorandil group (P = .078). Motor preservation in the diazoxide group was similar to the Nicorandil + carboxy-PTIO group. There was no significant difference between the diazoxide and diazoxide + carboxy-PTIO groups. CONCLUSIONS Both direct and nitric oxide-dependent indirect activation of the mitochondrial adenosine triphosphate-sensitive potassium channel play an important role in pharmacologically induced motor function preservation.
Collapse
Affiliation(s)
- Yuki Ikeno
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - Christian V Ghincea
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - Gavriel F Roda
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - Linling Cheng
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - Muhammad Aftab
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - Xianzhong Meng
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - Michael J Weyant
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - Joseph C Cleveland
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - David A Fullerton
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo
| | - T Brett Reece
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado, Aurora, Colo.
| |
Collapse
|
8
|
Papiri G, D’Andreamatteo G, Cacchiò G, Alia S, Silvestrini M, Paci C, Luzzi S, Vignini A. Multiple Sclerosis: Inflammatory and Neuroglial Aspects. Curr Issues Mol Biol 2023; 45:1443-1470. [PMID: 36826039 PMCID: PMC9954863 DOI: 10.3390/cimb45020094] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Multiple sclerosis (MS) represents the most common acquired demyelinating disorder of the central nervous system (CNS). Its pathogenesis, in parallel with the well-established role of mechanisms pertaining to autoimmunity, involves several key functions of immune, glial and nerve cells. The disease's natural history is complex, heterogeneous and may evolve over a relapsing-remitting (RRMS) or progressive (PPMS/SPMS) course. Acute inflammation, driven by infiltration of peripheral cells in the CNS, is thought to be the most relevant process during the earliest phases and in RRMS, while disruption in glial and neural cells of pathways pertaining to energy metabolism, survival cascades, synaptic and ionic homeostasis are thought to be mostly relevant in long-standing disease, such as in progressive forms. In this complex scenario, many mechanisms originally thought to be distinctive of neurodegenerative disorders are being increasingly recognized as crucial from the beginning of the disease. The present review aims at highlighting mechanisms in common between MS, autoimmune diseases and biology of neurodegenerative disorders. In fact, there is an unmet need to explore new targets that might be involved as master regulators of autoimmunity, inflammation and survival of nerve cells.
Collapse
Affiliation(s)
- Giulio Papiri
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Giordano D’Andreamatteo
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Gabriella Cacchiò
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Sonila Alia
- Section of Biochemistry, Biology and Physics, Department of Clinical Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Mauro Silvestrini
- Neurology Unit, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Cristina Paci
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Simona Luzzi
- Neurology Unit, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Arianna Vignini
- Section of Biochemistry, Biology and Physics, Department of Clinical Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| |
Collapse
|
9
|
Wu X, You J, Chen X, Zhou M, Ma H, Zhang T, Huang C. An overview of hyperbaric oxygen preconditioning against ischemic stroke. Metab Brain Dis 2023; 38:855-872. [PMID: 36729260 PMCID: PMC10106353 DOI: 10.1007/s11011-023-01165-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/23/2022] [Accepted: 01/12/2023] [Indexed: 02/03/2023]
Abstract
Ischemic stroke (IS) has become the second leading cause of morbidity and mortality worldwide, and the prevention of IS should be given high priority. Recent studies have indicated that hyperbaric oxygen preconditioning (HBO-PC) may be a protective nonpharmacological method, but its underlying mechanisms remain poorly defined. This study comprehensively reviewed the pathophysiology of IS and revealed the underlying mechanism of HBO-PC in protection against IS. The preventive effects of HBO-PC against IS may include inducing antioxidant, anti-inflammation, and anti-apoptosis capacity; activating autophagy and immune responses; upregulating heat shock proteins, hypoxia-inducible factor-1, and erythropoietin; and exerting protective effects upon the blood-brain barrier. In addition, HBO-PC may be considered a safe and effective method to prevent IS in combination with stem cell therapy. Although the benefits of HBO-PC on IS have been widely observed in recent research, the implementation of this technique is still controversial due to regimen differences. Transferring the results to clinical application needs to be taken carefully, and screening for the optimal regimen would be a daunting task. In addition, whether we should prescribe an individualized preconditioning regimen to each stroke patient needs further exploration.
Collapse
Affiliation(s)
- Xuyi Wu
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Jiuhong You
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xinxin Chen
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Mei Zhou
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Hui Ma
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Tianle Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Huang
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Serikbaeva A, Li Y, Ganesh B, Zelkha R, Kazlauskas A. Hyperglycemia Promotes Mitophagy and Thereby Mitigates Hyperglycemia-Induced Damage. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1779-1794. [PMID: 36063899 PMCID: PMC9765315 DOI: 10.1016/j.ajpath.2022.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/02/2022] [Accepted: 08/17/2022] [Indexed: 12/31/2022]
Abstract
The observation that diabetic retinopathy (DR) typically takes decades to develop suggests the existence of an endogenous system that protects from diabetes-induced damage. To investigate the existance of such a system, primary human retinal endothelial cells were cultured in either normal glucose (5 mmol/L) or high glucose (30 mmol/L; HG). Prolonged exposure to HG was beneficial instead of detrimental. Although tumor necrosis factor-α-induced expression of vascular cell adhesion molecule 1 and intercellular adhesion molecule 1 was unaffected after 1 day of HG, it waned as the exposure to HG was extended. Similarly, oxidative stress-induced death decreased with prolonged exposure to HG. Furthermore, mitochondrial functionality, which was compromised by 1 day of HG, was improved by 10 days of HG, and this change required increased clearance of damaged mitochondria (mitophagy). Finally, antagonizing mitochondrial dynamics compromised the cells' ability to endure HG: susceptibility to cell death increased, and basal barrier function and responsiveness to vascular endothelial growth factor deteriorated. These observations indicate the existence of an endogenous system that protects human retinal endothelial cells from the deleterious effects of HG. Hyperglycemia-induced mitochondrial adaptation is a plausible contributor to the mechanism responsible for the delayed onset of DR; loss of hyperglycemia-induced mitochondrial adaptation may set the stage for the development of DR.
Collapse
Affiliation(s)
- Anara Serikbaeva
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Yueru Li
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Balaji Ganesh
- Research Resources Center, University of Illinois at Chicago, Chicago, Illinois
| | - Ruth Zelkha
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Andrius Kazlauskas
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois; Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
11
|
Correia SC, Alves MG, Oliveira PF, Casadesus G, LaManna J, Perry G, Moreira PI. Hypoxic Preconditioning Averts Sporadic Alzheimer's Disease-Like Phenotype in Rats: A Focus on Mitochondria. Antioxid Redox Signal 2022; 37:739-757. [PMID: 35316086 DOI: 10.1089/ars.2019.8007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aims: Brief episodes of sublethal hypoxia reprogram brain response to face possible subsequent lethal stimuli by triggering adaptive and prosurvival events-a phenomenon denominated hypoxic preconditioning (HP). To date, the potential therapeutic implications of HP to forestall sporadic Alzheimer's disease (sAD) pathology remain unexplored. Using a well-established protocol of HP and focusing on hippocampus as a first brain region affected in AD, this study was undertaken to investigate the potential protective effects of HP in a sAD rat model induced by the intracerebroventricular (icv) administration of streptozotocin (STZ) and to uncover the mitochondrial adaptations underlying this nonpharmacological strategy. Results: HP prevented the memory and learning deficits as well as tau pathology in the icvSTZ rat model. HP also attenuated icvSTZ-related reactive astrogliosis, as noted by increased glial fibrillary acidic protein immunoreactivity and myo-inositol levels. Notably, HP abrogated the icvSTZ-related impaired energy metabolism and oxidative damage. Particularly, HP averted increased lactate, glutamate, and succinate levels, and decreased mitochondrial respiratory chain function and mitochondrial DNA content. Concerning mitochondrial adaptations underlying HP-triggered tolerance to icvSTZ, preconditioned hippocampal mitochondria displayed an enhanced complex II-energized mitochondrial respiration, which resulted from a coordinated interaction between mitochondrial biogenesis and fusion-fission. Mitochondrial biogenesis was stimulated immediately after HP, whereas in a latter phase mitochondrial fusion-fission events are modulated favoring the generation of elongated mitochondria. Innovation and Conclusion: Overall, these results demonstrate for the first time that HP prevents the sAD-like phenotype, in part, by targeting mitochondria emerging as a preventive strategy in the context of AD. Antioxid. Redox Signal. 37, 739-757.
Collapse
Affiliation(s)
- Sónia C Correia
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, and University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Marco G Alves
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- QOPNA & LAQV, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Joseph LaManna
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| | - George Perry
- Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Paula I Moreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, and University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
12
|
Correia SC, Moreira PI. Oxygen Sensing and Signaling in Alzheimer's Disease: A Breathtaking Story! Cell Mol Neurobiol 2022; 42:3-21. [PMID: 34510330 PMCID: PMC11441261 DOI: 10.1007/s10571-021-01148-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Oxygen sensing and homeostasis is indispensable for the maintenance of brain structural and functional integrity. Under low-oxygen tension, the non-diseased brain has the ability to cope with hypoxia by triggering a homeostatic response governed by the highly conserved hypoxia-inducible family (HIF) of transcription factors. With the advent of advanced neuroimaging tools, it is now recognized that cerebral hypoperfusion, and consequently hypoxia, is a consistent feature along the Alzheimer's disease (AD) continuum. Of note, the reduction in cerebral blood flow and tissue oxygenation detected during the prodromal phases of AD, drastically aggravates as disease progresses. Within this scenario a fundamental question arises: How HIF-driven homeostatic brain response to hypoxia "behaves" during the AD continuum? In this sense, the present review is aimed to critically discuss and summarize the current knowledge regarding the involvement of hypoxia and HIF signaling in the onset and progression of AD pathology. Importantly, the promises and challenges of non-pharmacological and pharmacological strategies aimed to target hypoxia will be discussed as a new "hope" to prevent and/or postpone the neurodegenerative events that occur in the AD brain.
Collapse
Affiliation(s)
- Sónia C Correia
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, Polo I, 1st Floor, 3004-504, Coimbra, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, Polo I, 1st Floor, 3004-504, Coimbra, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| |
Collapse
|
13
|
A Rationale for Hypoxic and Chemical Conditioning in Huntington's Disease. Int J Mol Sci 2021; 22:ijms22020582. [PMID: 33430140 PMCID: PMC7826574 DOI: 10.3390/ijms22020582] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
Neurodegenerative diseases are characterized by adverse cellular environments and pathological alterations causing neurodegeneration in distinct brain regions. This development is triggered or facilitated by conditions such as hypoxia, ischemia or inflammation and is associated with disruptions of fundamental cellular functions, including metabolic and ion homeostasis. Targeting intracellular downstream consequences to specifically reverse these pathological changes proved difficult to translate to clinical settings. Here, we discuss the potential of more holistic approaches with the purpose to re-establish a healthy cellular environment and to promote cellular resilience. We review the involvement of important molecular pathways (e.g., the sphingosine, δ-opioid receptor or N-Methyl-D-aspartate (NMDA) receptor pathways) in neuroprotective hypoxic conditioning effects and how these pathways can be targeted for chemical conditioning. Despite the present scarcity of knowledge on the efficacy of such approaches in neurodegeneration, the specific characteristics of Huntington’s disease may make it particularly amenable for such conditioning techniques. Not only do classical features of neurodegenerative diseases like mitochondrial dysfunction, oxidative stress and inflammation support this assumption, but also specific Huntington’s disease characteristics: a relatively young age of neurodegeneration, molecular overlap of related pathologies with hypoxic adaptations and sensitivity to brain hypoxia. The aim of this review is to discuss several molecular pathways in relation to hypoxic adaptations that have potential as drug targets in neurodegenerative diseases. We will extract the relevance for Huntington’s disease from this knowledge base.
Collapse
|
14
|
Millet GP, Debevec T, Brocherie F, Burtscher M, Burtscher J. Altitude and COVID-19: Friend or foe? A narrative review. Physiol Rep 2021; 8:e14615. [PMID: 33340275 PMCID: PMC7749581 DOI: 10.14814/phy2.14615] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
Recent reports suggest that high-altitude residence may be beneficial in the novel coronavirus disease (COVID-19) implicating that traveling to high places or using hypoxic conditioning thus could be favorable as well. Physiological high-altitude characteristics and symptoms of altitude illnesses furthermore seem similar to several pathologies associated with COVID-19. As a consequence, high altitude and hypoxia research and related clinical practices are discussed for potential applications in COVID-19 prevention and treatment. We summarize the currently available evidence on the relationship between altitude/hypoxia conditions and COVID-19 epidemiology and pathophysiology. The potential for treatment strategies used for altitude illnesses is evaluated. Symptomatic overlaps in the pathophysiology of COVID-19 induced ARDS and high altitude illnesses (i.e., hypoxemia, dyspnea…) have been reported but are also common to other pathologies (i.e., heart failure, pulmonary embolism, COPD…). Most treatments of altitude illnesses have limited value and may even be detrimental in COVID-19. Some may be efficient, potentially the corticosteroid dexamethasone. Physiological adaptations to altitude/hypoxia can exert diverse effects, depending on the constitution of the target individual and the hypoxic dose. In healthy individuals, they may optimize oxygen supply and increase mitochondrial, antioxidant, and immune system function. It is highly debated if these physiological responses to hypoxia overlap in many instances with SARS-CoV-2 infection and may exert preventive effects under very specific conditions. The temporal overlap of SARS-CoV-2 infection and exposure to altitude/hypoxia may be detrimental. No evidence-based knowledge is presently available on whether and how altitude/hypoxia may prevent, treat or aggravate COVID-19. The reported lower incidence and mortality of COVID-19 in high-altitude places remain to be confirmed. High-altitude illnesses and COVID-19 pathologies exhibit clear pathophysiological differences. While potentially effective as a prophylactic measure, altitude/hypoxia is likely associated with elevated risks for patients with COVID-19. Altogether, the different points discussed in this review are of possibly some relevance for individuals who aim to reach high-altitude areas. However, due to the ever-changing state of understanding of COVID-19, all points discussed in this review may be out of date at the time of its publication.
Collapse
Affiliation(s)
| | - Tadej Debevec
- Faculty of SportUniversity of LjubljanaLjubljanaSlovenia
| | | | | | | |
Collapse
|
15
|
Manouchehri N, Khodagholi F, Dargahi L, Ahmadiani A. Mitochondrial Complex I Is an Essential Player in LPS-Induced Preconditioning in Differentiated PC12 Cells. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 18:1445-1455. [PMID: 32641953 PMCID: PMC6934967 DOI: 10.22037/ijpr.2019.1100711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Preconditioning (PC) as a protective strategy against noxious insults can decline cell death and apoptosis. It has been approved that mitochondria play a key role in PC mechanism. The critical role of complex I (CI) in oxidative phosphorylation machinery and intracellular ROS production, particularly in the brain, accentuates its possible role in PC-induced neuroprotection. Here, differentiated PC12 cells were preconditioned with ultra-low dose LPS (ULD, 3 μg/mL) prior to exposure to high concentration of LPS (HD, 750 μg/mL). Our results showed that HD LPS treatment reduces cell viability and CI activity, and intensifies expression of cleaved caspase 3 compared to the control group. Intriguingly, PC induction resulted in enhancement of cell viability and CI activity and reduction of caspase3 cleavage compared to HD LPS group. In order to explore the role of CI in PC, we combined the ULD LPS with rotenone, a CI inhibitor. Following rotenone administration, cell viability significantly reduced while caspase3 cleavage increased compared to PC induction group. Taken together, cell survival and reduction of apoptosis followed by PC can be at least partially attributed to the preservation of mitochondrial CI function.
Collapse
Affiliation(s)
- Nasim Manouchehri
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Single-Channel Properties of the ROMK-Pore-Forming Subunit of the Mitochondrial ATP-Sensitive Potassium Channel. Int J Mol Sci 2019; 20:ijms20215323. [PMID: 31731540 PMCID: PMC6862428 DOI: 10.3390/ijms20215323] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
An increased flux of potassium ions into the mitochondrial matrix through the ATP-sensitive potassium channel (mitoKATP) has been shown to provide protection against ischemia-reperfusion injury. Recently, it was proposed that the mitochondrial-targeted isoform of the renal outer medullary potassium channel (ROMK) protein creates a pore-forming subunit of mitoKATP in heart mitochondria. Our research focuses on the properties of mitoKATP from heart-derived H9c2 cells. For the first time, we detected single-channel activity and describe the pharmacology of mitoKATP in the H9c2 heart-derived cells. The patch-clamping of mitoplasts from wild type (WT) and cells overexpressing ROMK2 revealed the existence of a potassium channel that exhibits the same basic properties previously attributed to mitoKATP. ROMK2 overexpression resulted in a significant increase of mitoKATP activity. The conductance of both channels in symmetric 150/150 mM KCl was around 97 ± 2 pS in WT cells and 94 ± 3 pS in cells overexpressing ROMK2. The channels were inhibited by 5-hydroxydecanoic acid (a mitoKATP inhibitor) and by Tertiapin Q (an inhibitor of both the ROMK-type channels and mitoKATP). Additionally, mitoKATP from cells overexpressing ROMK2 were inhibited by ATP/Mg2+ and activated by diazoxide. We used an assay based on proteinase K to examine the topology of the channel in the inner mitochondrial membrane and found that both termini of the protein localized to the mitochondrial matrix. We conclude that the observed activity of the channel formed by the ROMK protein corresponds to the electrophysiological and pharmacological properties of mitoKATP.
Collapse
|
17
|
Gleave JA, Arathoon LR, Trinh D, Lizal KE, Giguère N, Barber JH, Najarali Z, Khan MH, Thiele SL, Semmen MS, Koprich JB, Brotchie JM, Eubanks JH, Trudeau LE, Nash JE. Sirtuin 3 rescues neurons through the stabilisation of mitochondrial biogenetics in the virally-expressing mutant α-synuclein rat model of parkinsonism. Neurobiol Dis 2017; 106:133-146. [DOI: 10.1016/j.nbd.2017.06.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/14/2017] [Indexed: 01/01/2023] Open
|
18
|
Chen L, Nye DM, Stone MC, Weiner AT, Gheres KW, Xiong X, Collins CA, Rolls MM. Mitochondria and Caspases Tune Nmnat-Mediated Stabilization to Promote Axon Regeneration. PLoS Genet 2016; 12:e1006503. [PMID: 27923046 PMCID: PMC5173288 DOI: 10.1371/journal.pgen.1006503] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 12/20/2016] [Accepted: 11/22/2016] [Indexed: 11/24/2022] Open
Abstract
Axon injury can lead to several cell survival responses including increased stability and axon regeneration. Using an accessible Drosophila model system, we investigated the regulation of injury responses and their relationship. Axon injury stabilizes the rest of the cell, including the entire dendrite arbor. After axon injury we found mitochondrial fission in dendrites was upregulated, and that reducing fission increased stabilization or neuroprotection (NP). Thus axon injury seems to both turn on NP, but also dampen it by activating mitochondrial fission. We also identified caspases as negative regulators of axon injury-mediated NP, so mitochondrial fission could control NP through caspase activation. In addition to negative regulators of NP, we found that nicotinamide mononucleotide adenylyltransferase (Nmnat) is absolutely required for this type of NP. Increased microtubule dynamics, which has previously been associated with NP, required Nmnat. Indeed Nmnat overexpression was sufficient to induce NP and increase microtubule dynamics in the absence of axon injury. DLK, JNK and fos were also required for NP. Because NP occurs before axon regeneration, and NP seems to be actively downregulated, we tested whether excessive NP might inhibit regeneration. Indeed both Nmnat overexpression and caspase reduction reduced regeneration. In addition, overexpression of fos or JNK extended the timecourse of NP and dampened regeneration in a Nmnat-dependent manner. These data suggest that NP and regeneration are conflicting responses to axon injury, and that therapeutic strategies that boost NP may reduce regeneration. Unlike many other cell types, most neurons last a lifetime. When injured, these cells often activate survival and repair strategies rather than dying. One such response is regeneration of the axon after it is injured. Axon regeneration is a conserved process activated by the same signaling cascade in worms, flies and mammals. Surprisingly we find that this signaling cascade first initiates a different response. This first response stabilizes the cell, and its downregulation by mitochondrial fission and caspases allows for maximum regeneration at later times. We propose that neurons respond to axon injury in a multi-step process with an early lock-down phase in which the cell is stabilized, followed by a more plastic state in which regeneration is maximized.
Collapse
Affiliation(s)
- Li Chen
- Huck Institutes of the Life Sciences, and Biochemistry and Molecular Biology,The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Derek M. Nye
- Huck Institutes of the Life Sciences, and Biochemistry and Molecular Biology,The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Michelle C. Stone
- Huck Institutes of the Life Sciences, and Biochemistry and Molecular Biology,The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Alexis T. Weiner
- Huck Institutes of the Life Sciences, and Biochemistry and Molecular Biology,The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Kyle W. Gheres
- Huck Institutes of the Life Sciences, and Biochemistry and Molecular Biology,The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Xin Xiong
- Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Catherine A. Collins
- Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Melissa M. Rolls
- Huck Institutes of the Life Sciences, and Biochemistry and Molecular Biology,The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
19
|
Ramsay RR, Majekova M, Medina M, Valoti M. Key Targets for Multi-Target Ligands Designed to Combat Neurodegeneration. Front Neurosci 2016; 10:375. [PMID: 27597816 PMCID: PMC4992697 DOI: 10.3389/fnins.2016.00375] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022] Open
Abstract
HIGHLIGHTS Compounds that interact with multiple targets but minimally with the cytochrome P450 system (CYP) address the many factors leading to neurodegeneration.Acetyl- and Butyryl-cholineEsterases (AChE, BChE) and Monoamine Oxidases A/B (MAO A, MAO B) are targets for Multi-Target Designed Ligands (MTDL).ASS234 is an irreversible inhibitor of MAO A >MAO B and has micromolar potency against the cholinesterases.ASS234 is a poor CYP substrate in human liver, yielding the depropargylated metabolite.SMe1EC2, a stobadine derivative, showed high radical scavenging property, in vitro and in vivo giving protection in head trauma and diabetic damage of endothelium.Control of mitochondrial function and morphology by manipulating fission and fusion is emerging as a target area for therapeutic strategies to decrease the pathological outcome of neurodegenerative diseases. Growing evidence supports the view that neurodegenerative diseases have multiple and common mechanisms in their aetiologies. These multifactorial aspects have changed the broadly common assumption that selective drugs are superior to "dirty drugs" for use in therapy. This drives the research in studies of novel compounds that might have multiple action mechanisms. In neurodegeneration, loss of neuronal signaling is a major cause of the symptoms, so preservation of neurotransmitters by inhibiting the breakdown enzymes is a first approach. Acetylcholinesterase (AChE) inhibitors are the drugs preferentially used in AD and that one of these, rivastigmine, is licensed also for PD. Several studies have shown that monoamine oxidase (MAO) B, located mainly in glial cells, increases with age and is elevated in Alzheimer (AD) and Parkinson's Disease's (PD). Deprenyl, a MAO B inhibitor, significantly delays the initiation of levodopa treatment in PD patients. These indications underline that AChE and MAO are considered a necessary part of multi-target designed ligands (MTDL). However, both of these targets are simply symptomatic treatment so if new drugs are to prevent degeneration rather than compensate for loss of neurotransmitters, then oxidative stress and mitochondrial events must also be targeted. MAO inhibitors can protect neurons from apoptosis by mechanisms unrelated to enzyme inhibition. Understanding the involvement of MAO and other proteins in the induction and regulation of the apoptosis in mitochondria will aid progress toward strategies to prevent the loss of neurons. In general, the oxidative stress observed both in PD and AD indicate that antioxidant properties are a desirable part of MTDL molecules. After two or more properties are incorporated into one molecule, the passage from a lead compound to a therapeutic tool is strictly linked to its pharmacokinetic and toxicity. In this context the interaction of any new molecules with cytochrome P450 and other xenobiotic metabolic processes is a crucial point. The present review covers the biochemistry of enzymes targeted in the design of drugs against neurodegeneration and the cytochrome P450-dependent metabolism of MTDLs.
Collapse
Affiliation(s)
- Rona R. Ramsay
- Biomedical Sciences Research Complex, University of St. AndrewsSt. Andrews, UK
| | - Magdalena Majekova
- Department of Biochemical Pharmacology, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of SciencesBratislava, Slovakia
| | - Milagros Medina
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias and BIFI, Universidad de ZaragozaZaragoza, Spain
| | - Massimo Valoti
- Dipartimento di Scienze della Vita, Università degli Studi di SienaSiena, Italy
| |
Collapse
|
20
|
Villoslada P, Rovira A, Montalban X, Arroyo R, Paul F, Meca-Lallana V, Ramo C, Fernandez O, Saiz A, Garcia-Merino A, Ramió-Torrentà L, Casanova B, Oreja-Guevara C, Muñoz D, Martinez-Rodriguez JE, Lensch E, Prieto JM, Meuth SG, Nuñez X, Campás C, Pugliese M. Effects of diazoxide in multiple sclerosis: A randomized, double-blind phase 2 clinical trial. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e147. [PMID: 26405686 PMCID: PMC4567455 DOI: 10.1212/nxi.0000000000000147] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/15/2015] [Indexed: 11/15/2022]
Abstract
Objective: The aim of this study was to test the safety of diazoxide and to search for signs of efficacy in patients with relapsing-remitting multiple sclerosis (RRMS). Methods: In this multicenter, randomized, placebo-controlled, double-blind trial (treatment allocation was concealed), 102 patients with RRMS were randomized to receive a daily oral dose of diazoxide (0.3 and 4 mg/d) or placebo for 24 weeks (NCT01428726). The primary endpoint was the cumulative number of new T1 gadolinium-enhancing lesions per patient, recorded every 4 weeks from week 4 to week 24. Secondary endpoints included brain MRI variables such as the number of new/enlarging T2 lesions and the percentage brain volume change (PBVC); clinical variables such as the percentage of relapse-free patients, relapse rate, and change in the Expanded Disability Status Scale score; and safety and tolerability. Results: Diazoxide was well-tolerated and it produced no serious adverse events other than 1 case of Hashimoto disease. At the 2 doses tested, diazoxide did not improve the primary endpoint or the MRI and clinical variables related to the presence of new lesions or relapses. Patients treated with diazoxide showed reduced PBVC compared with the placebo group, although such changes could be confounded by the higher disease activity of the treated group and the vascular effects of diazoxide. Conclusion: At the doses tested, oral diazoxide did not decrease the appearance of new lesions evident by MRI. The effects in slowing the progression of brain atrophy require further validation. Classification of evidence: This study provides Class I evidence that for patients with RRMS, diazoxide (0.3 and 4 mg/d) does not significantly change the number of new MRI T1 gadolinium-enhancing lesions.
Collapse
Affiliation(s)
- Pablo Villoslada
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Alex Rovira
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Xavier Montalban
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Rafael Arroyo
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Friedemann Paul
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Virginia Meca-Lallana
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Cristina Ramo
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Oscar Fernandez
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Albert Saiz
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Antonio Garcia-Merino
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Lluís Ramió-Torrentà
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Bonaventura Casanova
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Celia Oreja-Guevara
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Delicias Muñoz
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Jose Enrique Martinez-Rodriguez
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Eckart Lensch
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Jose Maria Prieto
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Sven G Meuth
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Xavier Nuñez
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Clara Campás
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | - Marco Pugliese
- Institut d' Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clinic (P.V., A.S.), Barcelona, Spain; Unitat de RM (Servei de Radiologia) (A.R., X.M.), Departamento de Neurología-Neuroinmunología, Centro de Esclerosis Múltiple de Cataluña (Cemcat), Hospital Vall d'Hebron, Barcelona, Spain; Hospital Clinico San Carlos (R.A., C.O.-G.), Madrid, Spain; NeuroCure Clinical Research Center and Department of Neurology (F.P.), Charité University Medicine Berlin, Berlin, Germany; Hospital de La Princesa (V.M.-L.), Madrid, Spain; Hospital Germans Trias i Pujol (C.R.), Badalona, Spain; Hospital Regional Universitario (IBIMA) (O.F.), Malaga, Spain; Hospital Puerta de Hierro (A.G.-M.), Madrid, Spain; Hospital Universitari Dr Josep Trueta (L.R.-T.), IDIBGI, Girona, Spain; Hospital La Fe (B.C.), Valencia, Spain; Hospital Xeral-Cies (D.M.), Vigo, Spain; Hospital del Mar (J.E.M.-R.), Barcelona, Spain; Deutsche Klinik für Diagnostik (E.L.), Wiesbaden, Germany; Hospital Universitario Santiago de Compostela (J.M.P.), Spain; Department of Neurology (S.G.M.), University of Munster, Germany; TrialFormSupport (X.N.), Barcelona, Spain; Advancell, Advanced In Vitro Cell Technologies, S.A (C.C.), Barcelona, Spain; and Neurotec Pharma S.L (M.P.), Barcelona, Spain
| | | |
Collapse
|
21
|
Sisalli MJ, Annunziato L, Scorziello A. Novel Cellular Mechanisms for Neuroprotection in Ischemic Preconditioning: A View from Inside Organelles. Front Neurol 2015; 6:115. [PMID: 26074868 PMCID: PMC4443717 DOI: 10.3389/fneur.2015.00115] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/05/2015] [Indexed: 11/16/2022] Open
Abstract
Ischemic preconditioning represents an important adaptation mechanism of CNS, which results in its increased tolerance to the lethal cerebral ischemia. The molecular mechanisms responsible for the induction and maintenance of ischemic tolerance in the brain are complex and not yet completely clarified. In the last 10 years, great attention has been devoted to unravel the intracellular pathways activated by preconditioning and responsible for the establishing of the tolerant phenotype. Indeed, recent papers have been published supporting the hypothesis that mitochondria might act as master regulators of preconditioning-triggered endogenous neuroprotection due to their ability to control cytosolic calcium homeostasis. More interestingly, the demonstration that functional alterations in the ability of mitochondria and endoplasmic reticulum (ER) managing calcium homeostasis during ischemia, opened a new line of research focused to the role played by mitochondria and ER cross-talk in the pathogenesis of cerebral ischemia in order to identify new molecular mechanisms involved in the ischemic tolerance. In line with these findings and considering that the expression of the three isoforms of the sodium calcium exchanger (NCX), NCX1, NCX2, and NCX3, mainly responsible for the regulation of Ca2+ homeostasis, was reduced during cerebral ischemia, it was investigated whether these proteins might play a role in neuroprotection induced by ischemic tolerance. In this review, evidence supporting the involvement of ER and mitochondria interaction within the preconditioning paradigm will be provided. In particular, the key role played by NCXs in the regulation of Ca2+-homeostasis at the different subcellular compartments will be discussed as new molecular mechanism proposed for the establishing of ischemic tolerant phenotype.
Collapse
Affiliation(s)
- Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Science, School of Medicine, Federico II University of Naples , Naples , Italy
| | - Lucio Annunziato
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Science, School of Medicine, Federico II University of Naples , Naples , Italy ; Fondazione IRCSS SDN , Naples , Italy
| | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Science, School of Medicine, Federico II University of Naples , Naples , Italy
| |
Collapse
|
22
|
Rocha-Santos V, Figueira ERR, Rocha-Filho JA, Coelho AMM, Pinheiro RS, Bacchella T, Machado MCC, D'Albuquerque LAC. Pentoxifylline enhances the protective effects of hypertonic saline solution on liver ischemia reperfusion injury through inhibition of oxidative stress. Hepatobiliary Pancreat Dis Int 2015; 14:194-200. [PMID: 25865693 DOI: 10.1016/s1499-3872(15)60348-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Liver ischemia reperfusion (IR) injury triggers a systemic inflammatory response and is the main cause of organ dysfunction and adverse postoperative outcomes after liver surgery. Pentoxifylline (PTX) and hypertonic saline solution (HTS) have been identified to have beneficial effects against IR injury. This study aimed to investigate if the addition of PTX to HTS is superior to HTS alone for the prevention of liver IR injury. METHODS Male Wistar rats were allocated into three groups. Control rats underwent 60 minutes of partial liver ischemia, HTS rats were treated with 0.4 mL/kg of intravenous 7.5% NaCl 15 minutes before reperfusion, and HPTX group were treated with 7.5% NaCl plus 25 mg/kg of PTX 15 minutes before reperfusion. Samples were collected after reperfusion for determination of ALT, AST, TNF-alpha, IL-6, IL-10, mitochondrial respiration, lipid peroxidation, pulmonary permeability and myeloperoxidase. RESULTS HPTX significantly decreased TNF-alpha 30 minutes after reperfusion. HPTX and HTS significantly decreased ALT, AST, IL-6, mitochondrial dysfunction and pulmonary myeloperoxidase 4 hours after reperfusion. Compared with HTS only, HPTX significantly decreased hepatic oxidative stress 4 hours after reperfusion and pulmonary permeability 4 and 12 hours after reperfusion. CONCLUSION This study showed that PTX added the beneficial effects of HTS on liver IR injury through decreases of hepatic oxidative stress and pulmonary permeability.
Collapse
Affiliation(s)
- Vinicius Rocha-Santos
- Department of Gastroenterology, Laboratory of Medical Investigations LIM37 Discipline of Liver and Gastrointestinal Transplantation and Discipline of Anesthesiology, Hospital das Clinicas, University of Sao Paulo School of Medicine, Sao Paulo, SP, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Hao YH, Zhao L, Peng RY. Effects of microwave radiation on brain energy metabolism and related mechanisms. Mil Med Res 2015; 2:4. [PMID: 26000171 PMCID: PMC4440565 DOI: 10.1186/s40779-015-0033-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/29/2015] [Indexed: 11/10/2022] Open
Abstract
With the rapid development of electronic technologies, anxiety regarding the potential health hazards induced by microwave radiation (MW) has been growing in recent years. The brain is one of the most sensitive target organs for microwave radiation, where mitochondrial injury occurs earlier and more severely than in other organs. Energy metabolism disorders do play an important role during the process of microwave radiation-induced brain damage. In this paper, we will review the biological effects of microwave radiation, the features of brain energy supply and consumption and the effects of microwave radiation on mitochondrial energy metabolism and potential related mechanisms.
Collapse
Affiliation(s)
- Yan-Hui Hao
- Beijing Institute of Radiation Medicine, Beijing, 100850 China
| | - Li Zhao
- Beijing Institute of Radiation Medicine, Beijing, 100850 China
| | - Rui-Yun Peng
- Beijing Institute of Radiation Medicine, Beijing, 100850 China
| |
Collapse
|
24
|
Abstract
Oxidative stress is characterized by imbalanced reactive oxygen species (ROS) production and antioxidant defenses. Two main antioxidant systems exist. The nonenzymatic system relies on molecules to directly quench ROS and the enzymatic system is composed of specific enzymes that detoxify ROS. Among the latter, the superoxide dismutase (SOD) family is important in oxidative stress modulation. Of these, manganese-dependent SOD (MnSOD) plays a major role due to its mitochondrial location, i.e., the main site of superoxide (O(2)(·-)) production. As such, extensive research has focused on its capacity to modulate oxidative stress. Early data demonstrated the relevance of MnSOD as an O(2)(·-) scavenger. More recent research has, however, identified a prominent role for MnSOD in carcinogenesis. In addition, SOD downregulation appears associated with health risk in heart and brain. A single nucleotide polymorphism which alters the mitochondria signaling sequence for the cytosolic MnSOD form has been identified. Transport into the mitochondria was differentially affected by allelic presence and a new chapter in MnSOD research thus begun. As a result, an ever-increasing number of diseases appear associated with this allelic variation including metabolic and cardiovascular disease. Although diet and exercise upregulate MnSOD, the relationship between environmental and genetic factors remains unclear.
Collapse
|
25
|
Fu Q, Sun Z, Zhang J, Gao N, Qi F, Che F, Ma G. Diazoxide preconditioning antagonizes cytotoxicity induced by epileptic seizures. Neural Regen Res 2014; 8:1000-6. [PMID: 25206393 PMCID: PMC4145886 DOI: 10.3969/j.issn.1673-5374.2013.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 02/05/2013] [Indexed: 01/15/2023] Open
Abstract
Diazoxide, an activator of mitochondrial ATP-sensitive potassium channels, can protect neurons and astrocytes against oxidative stress and apoptosis. In this study, we established a cellular model of epilepsy by culturing hippocampal neurons in magnesium-free medium, and used this to investigate effects of diazoxide preconditioning on the expression of inwardly rectifying potassium channel (Kir) subunits of the ATP-sensitive potassium. We found that neuronal viability was significantly reduced in the epileptic cells, whereas it was enhanced by diazoxide preconditioning. Double immunofluorescence and western blot showed a significant increase in the expression of Kir6.1 and Kir6.2 in epileptic cells, especially at 72 hours after seizures. Diazoxide pretreatment completely reversed this effect at 24 hours after seizures. In addition, Kir6.1 expression was significantly upregulated compared with Kir6.2 in hippocampal neurons after seizures. These findings indicate that diazoxide pretreatment may counteract epileptiform discharge-induced cytotoxicity by suppressing the expression of Kir subunits.
Collapse
Affiliation(s)
- Qingxi Fu
- Department of Neurology, Linyi People's Hospital, Linyi 276003, Shandong Province, China
| | - Zhiqing Sun
- Department of Neurology, Linyi People's Hospital, Linyi 276003, Shandong Province, China
| | - Jinling Zhang
- Department of Neurology, Linyi People's Hospital, Linyi 276003, Shandong Province, China
| | - Naiyong Gao
- Department of Neurology, Linyi People's Hospital, Linyi 276003, Shandong Province, China
| | - Faying Qi
- Department of Neurology, Linyi People's Hospital, Linyi 276003, Shandong Province, China
| | - Fengyuan Che
- Department of Neurology, Linyi People's Hospital, Linyi 276003, Shandong Province, China
| | - Guozhao Ma
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong Province, China
| |
Collapse
|
26
|
Fu Q, Gao N, Yu J, Ma G, Du Y, Wang F, Su Q, Che F. Diazoxide pretreatment prevents Aβ1-42 induced oxidative stress in cholinergic neurons via alleviating NOX2 expression. Neurochem Res 2014; 39:1313-21. [PMID: 24771316 DOI: 10.1007/s11064-014-1313-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 04/09/2014] [Accepted: 04/16/2014] [Indexed: 12/19/2022]
Abstract
The aggregation and accumulation of amyloid-β (Aβ) plays a significant role in the pathogenesis of Alzheimer's disease. Aβ is known to increase free radical production in neuronal cells, leading to oxidative stress and cell death. Diazoxide (DZ), a highly selective drug capable of opening mitochondrial ATP-sensitive potassium channels, has neuroprotective effects against neuronal cell death. However, the mechanism through which DZ protects cholinergic neurons against Aβ-induced oxidative injury is still unclear. The present study was designed to investigate the effects of DZ pretreatment against Aβ1-42 induced oxidative damage and cytotoxicity. Through measures of DZ effects on Aβ1-42 induced cellular damage, reactive oxygen species (ROS) and MDA generation and expressions of gp91phox and p47phox in cholinergic neurons, new insights into the neuroprotective mechanisms can be derived. Aβ1-42 significantly decreased 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide levels and increased ROS and MDA production; all effects were attenuated by pretreatment with DZ or diphenyleneiodonium chloride (a NOX2 inhibitor). Pretreatment with DZ also attenuated the upregulation of NOX2 subunits (gp91phox and p47phox) induced by Aβ1-42. Since NOX2 is one of the main sources of free radicals, these results suggest that DZ can counteract Aβ1-42 induced oxidative stress and associated cell death by reducing the level of ROS and MDA, in part, by alleviating NOX2 expression.
Collapse
Affiliation(s)
- Qingxi Fu
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Liu W, Liu K, Tao H, Chen C, Zhang JH, Sun X. Hyperoxia preconditioning: the next frontier in neurology? Neurol Res 2013; 34:415-21. [DOI: 10.1179/1743132812y.0000000034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Wenwu Liu
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Kan Liu
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Hengyi Tao
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Chunhua Chen
- Department of Anatomy and EmbryologyPeking University Health Science Center, Beijing, China
| | - John H Zhang
- Department of AnesthesiologyLoma Linda Medical Center, Loma Linda, CA, USA
| | - Xuejun Sun
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| |
Collapse
|
28
|
K(ATP) channel opener diazoxide prevents neurodegeneration: a new mechanism of action via antioxidative pathway activation. PLoS One 2013; 8:e75189. [PMID: 24040400 PMCID: PMC3770693 DOI: 10.1371/journal.pone.0075189] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/11/2013] [Indexed: 11/26/2022] Open
Abstract
Pharmacological modulation of ATP-sensitive potassium channels has become a promising new therapeutic approach for the treatment of neurodegenerative diseases due to their role in mitochondrial and cellular protection. For instance, diazoxide, a well-known ATP-sensitive potassium channel activator with high affinity for mitochondrial component of the channel has been proved to be effective in animal models for different diseases such as Alzheimer’s disease, stroke or multiple sclerosis. Here, we analyzed the ability of diazoxide for protecting neurons front different neurotoxic insults in vitro and ex vivo. Results showed that diazoxide effectively protects NSC-34 motoneurons from glutamatergic, oxidative and inflammatory damage. Moreover, diazoxide decreased neuronal death in organotypic hippocampal slice cultures after exicitotoxicity and preserved myelin sheath in organotypic cerebellar cultures exposed to pro-inflammatory demyelinating damage. In addition, we demonstrated that one of the mechanisms of actions implied in the neuroprotective role of diazoxide is mediated by the activation of Nrf2 expression and nuclear translocation. Nrf2 expression was increased in NSC-34 neurons in vitro as well as in the spinal cord of experimental autoimmune encephalomyelitis animals orally administered with diazoxide. Thus, diazoxide is a neuroprotective agent against oxidative stress-induced damage and cellular dysfunction that can be beneficial for diseases such as multiple sclerosis.
Collapse
|
29
|
Stetler RA, Leak RK, Yin W, Zhang L, Wang S, Gao Y, Chen J. Mitochondrial biogenesis contributes to ischemic neuroprotection afforded by LPS pre-conditioning. J Neurochem 2012; 123 Suppl 2:125-37. [PMID: 23050650 DOI: 10.1111/j.1471-4159.2012.07951.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although alterations in mitochondrial dynamics are associated with cellular responses to injury, the functional role of these dynamic changes in ischemic neurons is underexplored. One of these dynamic responses to injury includes mitochondrial biogenesis. Various sublethal pre-conditioning stimuli that induce an ischemic-tolerant state [e.g., lipopolysaccharide (LPS)] may also induce mitochondrial biogenesis. Using neuron-enriched cultures, we found that sublethal LPS pre-conditioning induced both ischemic tolerance and markers of mitochondrial biogenesis with overlapping dose-response temporal kinetics. Sublethal LPS transiently increased the expression of critical components of the mitochondrial transcriptional machinery, including nuclear respiratory factor 1 (NRF1) and mitochondrial transcription factor A (TFAM), as well as mtDNA copy number, mitochondrial protein levels, and markers of functional mitochondria, such as increased cellular ATP content, citrate synthase activity, and maximal respiration capacity. Importantly, knockdown of TFAM abrogated both the induction of mitochondrial biogenesis and the neuroprotective pre-conditioning effects of LPS. Several signaling pathways coordinated these events. AMPK inhibition suppressed NRF1 and TFAM expression by LPS, whereas PI3K/Akt signaling was necessary for the nuclear translocation of NRF1 and subsequent induction of TFAM. This is the first demonstration that LPS pre-conditioning initiates multiple signaling pathways leading to mitochondrial biogenesis in neurons and that these dynamic changes contribute to ischemic tolerance.
Collapse
Affiliation(s)
- R Anne Stetler
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
30
|
Yokobori S, Mazzeo AT, Hosein K, Gajavelli S, Dietrich WD, Bullock MR. Preconditioning for traumatic brain injury. Transl Stroke Res 2012; 4:25-39. [PMID: 24323189 DOI: 10.1007/s12975-012-0226-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 10/23/2012] [Accepted: 10/29/2012] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI) treatment is now focused on the prevention of primary injury and reduction of secondary injury. However, no single effective treatment is available as yet for the mitigation of traumatic brain damage in humans. Both chemical and environmental stresses applied before injury have been shown to induce consequent protection against post-TBI neuronal death. This concept termed "preconditioning" is achieved by exposure to different pre-injury stressors to achieve the induction of "tolerance" to the effect of the TBI. However, the precise mechanisms underlying this "tolerance" phenomenon are not fully understood in TBI, and therefore even less information is available about possible indications in clinical TBI patients. In this review, we will summarize TBI pathophysiology, and discuss existing animal studies demonstrating the efficacy of preconditioning in diffuse and focal type of TBI. We will also review other non-TBI preconditioning studies, including ischemic, environmental, and chemical preconditioning, which maybe relevant to TBI. To date, no clinical studies exist in this field, and we speculate on possible future clinical situations, in which pre-TBI preconditioning could be considered.
Collapse
Affiliation(s)
- Shoji Yokobori
- Department of Neurosurgery, Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA,
| | | | | | | | | | | |
Collapse
|
31
|
Irobi J, Holmgren A, Winter VD, Asselbergh B, Gettemans J, Adriaensen D, Groote CCD, Coster RV, Jonghe PD, Timmerman V. Mutant HSPB8 causes protein aggregates and a reduced mitochondrial membrane potential in dermal fibroblasts from distal hereditary motor neuropathy patients. Neuromuscul Disord 2012; 22:699-711. [DOI: 10.1016/j.nmd.2012.04.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 03/23/2012] [Accepted: 04/16/2012] [Indexed: 02/08/2023]
|
32
|
von Bernhardi R, Eugenín J. Alzheimer's disease: redox dysregulation as a common denominator for diverse pathogenic mechanisms. Antioxid Redox Signal 2012; 16:974-1031. [PMID: 22122400 DOI: 10.1089/ars.2011.4082] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and a progressive neurodegeneration that appears to result from multiple pathogenic mechanisms (including protein misfolding/aggregation, involved in both amyloid β-dependent senile plaques and tau-dependent neurofibrillary tangles), metabolic and mitochondrial dysfunction, excitoxicity, calcium handling impairment, glial cell dysfunction, neuroinflammation, and oxidative stress. Oxidative stress, which could be secondary to several of the other pathophysiological mechanisms, appears to be a major determinant of the pathogenesis and progression of AD. The identification of oxidized proteins common for mild cognitive impairment and AD suggests that key oxidation pathways are triggered early and are involved in the initial progression of the neurodegenerative process. Abundant data support that oxidative stress, also considered as a main factor for aging, the major risk factor for AD, can be a common key element capable of articulating the divergent nature of the proposed pathogenic factors. Pathogenic mechanisms influence each other at different levels. Evidence suggests that it will be difficult to define a single-target therapy resulting in the arrest of progression or the improvement of AD deterioration. Since oxidative stress is present from early stages of disease, it appears as one of the main targets to be included in a clinical trial. Exploring the articulation of AD pathogenic mechanisms by oxidative stress will provide clues for better understanding the pathogenesis and progression of this dementing disorder and for the development of effective therapies to treat this disease.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | |
Collapse
|
33
|
Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer's disease and other disorders. Nat Rev Neurosci 2011; 12:723-38. [PMID: 22048062 DOI: 10.1038/nrn3114] [Citation(s) in RCA: 2125] [Impact Index Per Article: 151.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neurovascular unit (NVU) comprises brain endothelial cells, pericytes or vascular smooth muscle cells, glia and neurons. The NVU controls blood-brain barrier (BBB) permeability and cerebral blood flow, and maintains the chemical composition of the neuronal 'milieu', which is required for proper functioning of neuronal circuits. Recent evidence indicates that BBB dysfunction is associated with the accumulation of several vasculotoxic and neurotoxic molecules within brain parenchyma, a reduction in cerebral blood flow, and hypoxia. Together, these vascular-derived insults might initiate and/or contribute to neuronal degeneration. This article examines mechanisms of BBB dysfunction in neurodegenerative disorders, notably Alzheimer's disease, and highlights therapeutic opportunities relating to these neurovascular deficits.
Collapse
Affiliation(s)
- Berislav V Zlokovic
- Department of Physiology and Biophysics, and Center for Neurodegeneration and Regeneration at the Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, 1501 San Pablo Street, Los Angeles, California 90089, USA.
| |
Collapse
|
34
|
Ischemic preconditioning-like effect of polyunsaturated fatty acid-rich diet on hepatic ischemia/reperfusion injury. J Gastrointest Surg 2011; 15:1679-88. [PMID: 21826546 DOI: 10.1007/s11605-011-1648-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 07/26/2011] [Indexed: 01/31/2023]
Abstract
AIM The aim of this study was to investigate a possible preconditioning effect of oral diet enriched with polyunsaturated fatty acids (PUFAs) on liver ischemia/reperfusion (I/R) injuries. METHODS Wistar male rats were fed a standard diet or polyunsaturated fatty acid-rich diet (PRD) enriched with (GII) or without (GIII) ω-3 PUFA. Rats were submitted to partial liver ischemia during 1 h and evaluated in pre- and post-I/R conditions. In pre-I/R condition, livers were collected for determination of fatty acid composition, liver mitochondrial function, malondialdehyde (MDA) content, and histological analysis. Four hours after liver reperfusion serum activities of aspartate aminotransferase (AST) and alanine aminotransferase (ALT), serum levels of tumor necrosis factor-alpha, interleukin-6, interleukin-10, and prostaglandin-E2, liver mitochondrial function, MDA content, and histology were evaluated. RESULTS In the pre-I/R condition, GII and GIII groups had an increase on PUFA content and exhibited slight increased macrosteatosis and microsteatosis in the liver. After 4 h of reperfusion, PRD-fed rats showed a marked decrease on steatosis, diminished necrosis, an increase in MDA formation, and mitochondrial uncoupling. We also observed a marked decrease in plasma levels of cytokines and ALT and AST activities in post-I/R condition in PRD groups. CONCLUSION In this experimental model in the rat, PRD has a preconditioning effect protecting the liver from I/R injury and should be object of future clinical studies.
Collapse
|
35
|
Correia SC, Santos RX, Cardoso SM, Santos MS, Oliveira CR, Moreira PI. Cyanide preconditioning protects brain endothelial and NT2 neuron-like cells against glucotoxicity: role of mitochondrial reactive oxygen species and HIF-1α. Neurobiol Dis 2011; 45:206-18. [PMID: 21854848 DOI: 10.1016/j.nbd.2011.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 07/27/2011] [Accepted: 08/03/2011] [Indexed: 01/06/2023] Open
Abstract
The current study was undertaken to address the role of mitochondrial reactive oxygen species (ROS), and hypoxia inducible factor-1 alpha (HIF-1α) signaling pathway in the protection against high glucose levels in brain endothelial and NT2 neuron-like cells. Rat brain endothelial cells (RBE4) treated with non-toxic concentrations of cyanide (≤1 μM; 1h) exhibited an increase in ROS levels, particularly hydrogen peroxide (H(2)O(2)). Cyanide also induced a modest mitochondrial depolarization, an increase in oxygen consumption and a structural (smaller mitochondria) and spatial (perinuclear region) reorganization of mitochondrial network. The stabilization and nuclear activation of HIF-1α in the presence of cyanide were also observed, which resulted in an increase in vascular endothelial growth factor (VEGF), endothelial nitric oxide synthase (eNOS) and erythropoietin (EPO) protein levels reflecting an adaptive response. Importantly, preconditioning induced by cyanide protected brain endothelial cells against high glucose-mediated damage by the prevention of apoptotic cell death. In mitochondrial DNA-depleted NT2 (NT2 ρ0) cells, cyanide (0.1 μM) was unable to stimulate ROS production and, consequently, protect against glucotoxicity. Conversely, in NT2 cells, the parental cells with functional mitochondria, cyanide significantly increased ROS levels protecting against high glucose-induced neuronal cell loss and activation of caspase-3. The free radical scavenger N-acetyl-L-cysteine and the specific HIF-1α inhibitor 2-methoxyestradiol completely abolished the protective effects of cyanide preconditioning. Altogether our results demonstrate that mitochondrial preconditioning induced by cyanide triggers a protective response mediated by mitochondrial ROS and HIF-1α activation and signaling, which render brain endothelial and neuronal cells resistant against glucotoxicity.
Collapse
Affiliation(s)
- Sónia C Correia
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | | | | | | | | | | |
Collapse
|
36
|
The possible impact of noise-induced Ca 2+ -dependent activity in the central auditory pathway: A manganese-enhanced MRI study. Neuroimage 2011; 57:190-197. [DOI: 10.1016/j.neuroimage.2011.04.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 04/05/2011] [Accepted: 04/08/2011] [Indexed: 11/19/2022] Open
|
37
|
Cardoso S, Santos RX, Carvalho C, Correia S, Santos MS, Moreira PI. Mitochondrial Uncoupling Proteins and Oxidative Stress: Implications for Diabetes and Neurodegeneration. ACTA ACUST UNITED AC 2011. [DOI: 10.5530/ax.2011.2.3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Rapposelli S. Novel adenosine 5'-triphosphate-sensitive potassium channel ligands: a patent overview (2005-2010). Expert Opin Ther Pat 2011; 21:355-79. [PMID: 21269236 DOI: 10.1517/13543776.2011.553601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION ATP-sensitive potassium channels are important metabolic regulators that link cellular metabolism to excitability. Their wide distribution in various tissues and organs makes them significant and topical targets in a large number of diseases. AREAS COVERED This review summarizes the current understanding of the molecular biology and pharmacology of K(ATP) channels, and the pathological states that result from aberrant expression or function of these proteins. In particular, relevant research, patents and patent applications of the past 5 years are discussed. EXPERT OPINION The tissue-specific K(ATP) channel modulation reflects an early discovery stage in drug design. The wide distribution of K(ATP) channels lets us consider them as valid targets for several pathologies, but on other hand the ubiquitous nature is a relevant drawback in developing an effective therapy because of the onset of side effects related to the lack of selectivity. On this basis, further investigations on both the structures and the localization of each receptor subtype should be carried out either exploring the structure-activity relationship of the already existing K(ATP) ligands or developing new selective fluorescent probes. To date, this research area still strives to design new tissue-targeted ligands that could pave the way to the development of innovative and effective drugs for clinical use.
Collapse
Affiliation(s)
- Simona Rapposelli
- Dipartimento di Scienze Farmaceutiche - Università di Pisa, Via Bonanno, 6, 56126 Pisa, Italy.
| |
Collapse
|