1
|
Liu N, Liang X, Chen Y, Xie L. Recent trends in treatment strategies for Alzheimer 's disease and the challenges: A topical advancement. Ageing Res Rev 2024; 94:102199. [PMID: 38232903 DOI: 10.1016/j.arr.2024.102199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/12/2024] [Indexed: 01/19/2024]
Abstract
Alzheimer's Disease (AD) is an irreversible and progressive neurological disease that has affected at least 50 million people around the globe. Considering the severity of the disease and the continuous increase in the number of patients, the development of new effective drugs or intervention strategies for AD has become urgent. AD is caused by a combination of genetic, environmental, and lifestyle factors, but its exact cause has not yet been clarified. Given the current challenges being faced in the clinical treatment of AD, such as complex AD pathological network and insufficient early diagnosis, herein, we have focused on the three core pathological features of AD, including amyloid-β (Aβ) aggregation, tau phosphorylation and tangles, and activation of inflammatory factors. In this review, we have briefly underscored the primary evidence supporting each pathology and discuss AD pathological network among Aβ, tau, and inflammation. We have also comprehensively summarized the most instructive drugs and their treatment strategies against Aβ, tau, or neuroinflammation used in basic research and clinical trials. Finally, we have discussed and outlined the pros and cons of each pathological approach and looked forward to potential personalized diagnosis and treatment strategies that are beneficial to AD patients.
Collapse
Affiliation(s)
- Ni Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Xiaohan Liang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yu Chen
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Lihang Xie
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Sanchez JE, Noor S, Sun MS, Zimmerly J, Pasmay A, Sanchez JJ, Vanderwall AG, Haynes MK, Sklar LA, Escalona PR, Milligan ED. The FDA-approved compound, pramipexole and the clinical-stage investigational drug, dexpramipexole, reverse chronic allodynia from sciatic nerve damage in mice, and alter IL-1β and IL-10 expression from immune cell culture. Neurosci Lett 2023; 814:137419. [PMID: 37558176 PMCID: PMC10552878 DOI: 10.1016/j.neulet.2023.137419] [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: 04/23/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
During the onset of neuropathic pain from a variety of etiologies, nociceptors become hypersensitized, releasing neurotransmitters and other factors from centrally-projecting nerve terminals within the dorsal spinal cord. Consequently, glial cells (astrocytes and microglia) in the spinal cord are activated and mediate the release of proinflammatory cytokines that act to enhance pain transmission and sensitize mechanical non-nociceptive fibers which ultimately results in light touch hypersensitivity, clinically observed as allodynia. Pramipexole, a D2/D3 preferring agonist, is FDA-approved for the treatment of Parkinson's disease and demonstrates efficacy in animal models of inflammatory pain. The clinical-stage investigational drug, R(+) enantiomer of pramipexole, dexpramipexole, is virtually devoid of D2/D3 agonist actions and is efficacious in animal models of inflammatory and neuropathic pain. The current experiments focus on the application of a mouse model of sciatic nerve neuropathy, chronic constriction injury (CCI), that leads to allodynia and is previously characterized to generate spinal glial activation with consequent release IL-1β. We hypothesized that both pramipexole and dexpramipexole reverse CCI-induced chronic neuropathy in mice, and in human monocyte cell culture studies (THP-1 cells), pramipexole prevents IL-1β production. Additionally, we hypothesized that in rat primary splenocyte culture, dexpramixole increases mRNA for the anti-inflammatory and pleiotropic cytokine, interleukin-10 (IL-10). Results show that following intravenous pramipexole or dexpramipexole, a profound decrease in allodynia was observed by 1 hr, with allodynia returning 24 hr post-injection. Pramipexole significantly blunted IL-1β protein production from stimulated human monocytes and dexpramipexole induced elevated IL-10 mRNA expression from rat splenocytes. The data support that clinically-approved compounds like pramipexole and dexpramipexole support their application as anti-inflammatory agents to mitigate chronic neuropathy, and provide a blueprint for future, multifaceted approaches for opioid-independent neuropathic pain treatment.
Collapse
Affiliation(s)
- J E Sanchez
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - S Noor
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - M S Sun
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - J Zimmerly
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - A Pasmay
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - J J Sanchez
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - A G Vanderwall
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - M K Haynes
- Center for Molecular Discovery (CMD) Innovation, Discovery and Training Complex (IDTC), University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - L A Sklar
- Center for Molecular Discovery (CMD) Innovation, Discovery and Training Complex (IDTC), University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - P R Escalona
- Department of Psychiatry, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; New Mexico VA Health Care System, Albuquerque NM 87108, USA
| | - E D Milligan
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
3
|
Zhang Y, Fu Q, Ruan J, Shi C, Lu W, Wu J, Zhou Z. Dexpramipexole ameliorates cognitive deficits in sepsis-associated encephalopathy through suppressing mitochondria-mediated pyroptosis and apoptosis. Neuroreport 2023; 34:220-231. [PMID: 36719835 PMCID: PMC10516177 DOI: 10.1097/wnr.0000000000001882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/09/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVES This study was aimed at evaluating the effects of dexpramipexole (DPX) - a mitochondrial protectant that sustains mitochondrial function and energy production - on cognitive function in a mouse model of sepsis-associated encephalopathy (SAE) induced by peripheral administration of lipopolysaccharide (LPS) and examining the potential mechanisms. METHODS C57BL/6 male mice were randomized into one of four treatment protocols: Con+Sal, Con+DPX, LPS+Sal or LPS+DPX. The mice were intraperitoneally (i.p.) injected with LPS or equivalent volumes of normal saline once daily for 3 consecutive days. To evaluate the protective effects of DPX, we administered DPX or normal saline i.p. to the mice once daily for 6 consecutive days. Six mice in each group were decapitated on day 7, and each brain was rapidly removed and separated into two halves for biochemical and histochemical analysis. The remaining surviving mice in each group were subjected to behavioral tests from days 7 to 10. RESULTS Peripheral administration of LPS to mice led to learning and memory deficits in behavioral tests, which were associated with mitochondrial impairment and ATP depletion in the hippocampus. Repeated DPX treatment protected the mitochondria against LPS-induced morphological and functional impairment; inhibited the activation of the Nod-like receptor pyrin domain-containing 3 (NLRP3) inflammasome-caspase-1-dependent pyroptosis pathway and cytochrome c (Cyt-c)-caspase-3-dependent apoptosis pathway; and attenuated LPS-induced neuroinflammation and cell death in the hippocampus in SAE mice. CONCLUSIONS Mitochondria-mediated pyroptosis and apoptosis are involved in the pathogenesis of cognitive deficits in a mouse model of SAE and DPX protects mitochondria and suppresses the mitochondria-medicated pyroptosis and apoptosis pathways, and ameliorates LPS-induced neuroinflammation and cognitive deficits. This study provides theoretical evidence supporting DPX for the treatment of SAE.
Collapse
Affiliation(s)
- Yibao Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University
| | - Qun Fu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School
- Department of Anesthesiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiaping Ruan
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School
| | - Changxi Shi
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School
| | - Wuguang Lu
- Department of Anesthesiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Wu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Zhiqiang Zhou
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University
| |
Collapse
|
4
|
Soares P, Silva C, Chavarria D, Silva FSG, Oliveira PJ, Borges F. Drug discovery and amyotrophic lateral sclerosis: Emerging challenges and therapeutic opportunities. Ageing Res Rev 2023; 83:101790. [PMID: 36402404 DOI: 10.1016/j.arr.2022.101790] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the degeneration of upper and lower motor neurons (MNs) leading to paralysis and, ultimately, death by respiratory failure 3-5 years after diagnosis. Edaravone and Riluzole, the only drugs currently approved for ALS treatment, only provide mild symptomatic relief to patients. Extraordinary progress in understanding the biology of ALS provided new grounds for drug discovery. Over the last two decades, mitochondria and oxidative stress (OS), iron metabolism and ferroptosis, and the major regulators of hypoxia and inflammation - HIF and NF-κB - emerged as promising targets for ALS therapeutic intervention. In this review, we focused our attention on these targets to outline and discuss current advances in ALS drug development. Based on the challenges and the roadblocks, we believe that the rational design of multi-target ligands able to modulate the complex network of events behind the disease can provide effective therapies in a foreseeable future.
Collapse
Affiliation(s)
- Pedro Soares
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| | - Catia Silva
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Filomena S G Silva
- CNC - CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paulo J Oliveira
- CNC - CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; IIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
5
|
Role of Oxidative Stress in Liver Disorders. LIVERS 2022. [DOI: 10.3390/livers2040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Oxygen is vital for life as it is required for many different enzymatic reactions involved in intermediate metabolism and xenobiotic biotransformation. Moreover, oxygen consumption in the electron transport chain of mitochondria is used to drive the synthesis of ATP to meet the energetic demands of cells. However, toxic free radicals are generated as byproducts of molecular oxygen consumption. Oxidative stress ensues not only when the production of reactive oxygen species (ROS) exceeds the endogenous antioxidant defense mechanism of cells, but it can also occur as a consequence of an unbalance between antioxidant strategies. Given the important role of hepatocytes in the biotransformation and metabolism of xenobiotics, ROS production represents a critical event in liver physiology, and increasing evidence suggests that oxidative stress contributes to the development of many liver diseases. The present review, which is part of the special issue “Oxidant stress in Liver Diseases”, aims to provide an overview of the sources and targets of ROS in different liver diseases and highlights the pivotal role of oxidative stress in cell death. In addition, current antioxidant therapies as treatment options for such disorders and their limitations for future trial design are discussed.
Collapse
|
6
|
Dexpramipexole Attenuates White Matter Injury to Facilitate Locomotion and Motor Coordination Recovery via Reducing Ferroptosis after Intracerebral Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6160701. [PMID: 35965685 PMCID: PMC9371846 DOI: 10.1155/2022/6160701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/24/2022] [Accepted: 07/14/2022] [Indexed: 11/20/2022]
Abstract
Deciphering the factors causing damage to white matter fiber bundles and exploring new strategies to alleviate white matter injury (WMI) is a promising treatment to improve neurological impairments after intracerebral hemorrhage (ICH). Ferroptosis usually occurs at perihematomal region and contributes to neuronal death due to reactive oxygen species (ROS) production. Dexpramipexole (DPX) easily crosses the blood brain barrier (BBB) and exerts antioxidative properties by reducing ROS production, while the role of DPX in ferroptosis after ICH remains elusive. Here, our results indicated that ferroptosis played a significant role in WMI resulting from iron and ROS accumulation around hematoma. Further evidence demonstrated that the administration of DPX decreased iron and ROS deposition to inhibit ferroptosis at perihematomal site. With the inhibition of ferroptosis, WMI was alleviated at perihematomal site, thereafter promoting locomotion and motor coordination recovery in mice after ICH. Subsequently, the results showcased that the expression of glutathione peroxidase 4 (GPX4) and ferroptosis suppressing protein 1 (FSP1) was upregulated with the administration of DPX. Collectively, the present study uncovers the underlying mechanism and elucidates the therapeutic effect of DPX on ICH, and even in other central nervous system (CNS) diseases with the presence of ferroptosis.
Collapse
|
7
|
Vodičková A, Koren SA, Wojtovich AP. Site-specific mitochondrial dysfunction in neurodegeneration. Mitochondrion 2022; 64:1-18. [PMID: 35182728 PMCID: PMC9035127 DOI: 10.1016/j.mito.2022.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/18/2022] [Accepted: 02/14/2022] [Indexed: 02/07/2023]
Abstract
Mitochondria are essential for neuronal survival and mitochondrial dysfunction is a hallmark of neurodegeneration. The loss in mitochondrial energy production, oxidative stress, and changes in calcium handling are associated with neurodegenerative diseases; however, different sites and types of mitochondrial dysfunction are linked to distinct neuropathologies. Understanding the causal or correlative relationship between changes in mitochondria and neuropathology will lead to new therapeutic strategies. Here, we summarize the evidence of site-specific mitochondrial dysfunction and mitochondrial-related clinical trials for neurodegenerative diseases. We further discuss potential therapeutic approaches, such as mitochondrial transplantation, restoration of mitochondrial function, and pharmacological alleviation of mitochondrial dysfunction.
Collapse
Affiliation(s)
- Anežka Vodičková
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Shon A Koren
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Andrew P Wojtovich
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA; Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
8
|
Coppi E, Buonvicino D, Ranieri G, Cherchi F, Venturini M, Pugliese AM, Chiarugi A. Dexpramipexole Enhances K + Currents and Inhibits Cell Excitability in the Rat Hippocampus In Vitro. Mol Neurobiol 2021; 58:2955-2962. [PMID: 33566318 DOI: 10.1007/s12035-021-02300-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/15/2021] [Indexed: 11/28/2022]
Abstract
Dexpramipexole (DEX) has been described as the first-in-class F1Fo ATP synthase activator able to boost mitochondrial bioenergetics and provide neuroprotection in experimental models of ischemic brain injury. Although DEX failed in a phase III trial in patients with amyotrophic lateral sclerosis, it showed favorable safety and tolerability profiles. Recently, DEX emerged as a Nav1.8 Na+ channel and transient outward K+ (IA) conductance blocker, revealing therefore an unexpected, pleiotypic pharmacodynamic profile. In this study, we performed electrophysiological experiments in vitro aimed to better characterize the impact of DEX on voltage-dependent currents and synaptic transmission in the hippocampus. By means of patch-clamp recordings on isolated hippocampal neurons, we found that DEX increases outward K+ currents evoked by a voltage ramp protocol. This effect is prevented by the non-selective voltage-dependent K+ channel (Kv) blocker TEA and by the selective small-conductance Ca2+-activated K+ (SK) channel blocker apamin. In keeping with this, extracellular field recordings from rat hippocampal slices also demonstrated that the compound inhibits synaptic transmission and CA1 neuron excitability. Overall, these data further our understanding on the pharmacodynamics of DEX and disclose an additional mechanism that could underlie its neuroprotective properties. Also, they identify DEX as a lead to develop new modulators of K+ conductances.
Collapse
Affiliation(s)
- Elisabetta Coppi
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy.
| | - Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Giuseppe Ranieri
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Federica Cherchi
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Martina Venturini
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Anna Maria Pugliese
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
9
|
Sidibé H, Dubinski A, Vande Velde C. The multi-functional RNA-binding protein G3BP1 and its potential implication in neurodegenerative disease. J Neurochem 2021; 157:944-962. [PMID: 33349931 PMCID: PMC8248322 DOI: 10.1111/jnc.15280] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022]
Abstract
Ras-GTPase-activating protein (GAP)-binding protein 1 (G3BP1) is a multi-functional protein that is best known for its role in the assembly and dynamics of stress granules. Recent studies have highlighted that G3BP1 also has other functions related to RNA metabolism. In the context of disease, G3BP1 has been therapeutically targeted in cancers because its over-expression is correlated with proliferation of cancerous cells and metastasis. However, evidence suggests that G3BP1 is essential for neuronal development and possibly neuronal maintenance. In this review, we will examine the many functions that are carried out by G3BP1 in the context of neurons and speculate how these functions are critical to the progression of neurodegenerative diseases. Additionally, we will highlight the similarities and differences between G3BP1 and the closely related protein G3BP2, which is frequently overlooked. Although G3BP1 and G3BP2 have both been deemed important for stress granule assembly, their roles may differ in other cellular pathways, some of which are specific to the CNS, and presents an opportunity for further exploration.
Collapse
Affiliation(s)
- Hadjara Sidibé
- Department of NeurosciencesUniversité de Montréal, and CHUM Research CenterMontréalQCCanada
| | - Alicia Dubinski
- Department of NeurosciencesUniversité de Montréal, and CHUM Research CenterMontréalQCCanada
| | - Christine Vande Velde
- Department of NeurosciencesUniversité de Montréal, and CHUM Research CenterMontréalQCCanada
| |
Collapse
|
10
|
Nakagawa Y, Yamada S. A novel hypothesis on metal dyshomeostasis and mitochondrial dysfunction in amyotrophic lateral sclerosis: Potential pathogenetic mechanism and therapeutic implications. Eur J Pharmacol 2020; 892:173737. [PMID: 33220280 DOI: 10.1016/j.ejphar.2020.173737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/27/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by motor dysfunctions resulting from the loss of upper (UMNs) and lower (LMNs) motor neurons. While ALS symptoms are coincidental with pathological changes in LMNs and UMNs, the causal relationship between the two is unclear. For example, research on the extra-motor symptoms associated with this condition suggests that an imbalance of metals, including copper, zinc, iron, and manganese, is initially induced in the sensory ganglia due to a malfunction of metal binding proteins and transporters. It is proposed that the resultant metal dyshomeostasis may promote mitochondrial dysfunction in the satellite glial cells of these sensory ganglia, causing sensory neuron disturbances and sensory symptoms. Sensory neuron hyperactivation can result in LMN impairments, while metal dyshomeostasis in spinal cord and brain stem parenchyma induces mitochondrial dysfunction in LMNs and UMNs. These events could prompt intracellular calcium dyshomeostasis, pathological TDP-43 formation, and reactive microglia with neuroinflammation, which in turn activate the apoptosis signaling pathways within the LMNs and UMNs. Our model suggests that the degeneration of LMNs and UMNs is incidental to the metal-induced changes in the spinal cord and brain stem. Over time psychiatric symptoms may appear as the metal dyshomeostasis and mitochondrial dysfunction affect other brain regions, including the reticular formation, hippocampus, and prefrontal cortex. It is proposed that metal dyshomeostasis in combination with mitochondrial dysfunction could be the underlying mechanism responsible for the initiation and progression of the pathological changes associated with both the motor and extra-motor symptoms of ALS.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| |
Collapse
|
11
|
Blasco H, Lanznaster D, Veyrat-Durebex C, Hergesheimer R, Vourch P, Maillot F, Andres CR, Pradat PF, Corcia P. Understanding and managing metabolic dysfunction in Amyotrophic Lateral Sclerosis. Expert Rev Neurother 2020; 20:907-919. [PMID: 32583696 DOI: 10.1080/14737175.2020.1788389] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Amyotrophic Lateral Sclerosis (ALS) is a fatal motor neuron disease that leads to death after a median survival of 36 months. The development of an effective treatment has proven to be extremely difficult due to the inadequate understanding of the pathogenesis of ALS. Energy metabolism is thoroughly involved in the disease based on the discoveries of hypermetabolism, lipid/glucose metabolism, the tricarboxylic acid (TCA) cycle, and mitochondrial impairment. AREA COVERED Many perturbed metabolites within these processes have been identified as promising therapeutic targets. However, the therapeutic strategies targeting these pathways have failed to produce clinically significant results. The authors present in this review the metabolic disturbances observed in ALS and the derived-therapeutics. EXPERT OPINION The authors suggest that this is due to the insufficient knowledge of the relationship between the metabolic targets and the type of ALS of the patient, depending on genetic and environmental factors. We must improve our understanding of the pathological mechanisms and pay attention to the subtle hidden effects of changing diet, for example, and to use this strategy in addition to other drugs or to use metabolism status to determine subgroups of patients.
Collapse
Affiliation(s)
- Helene Blasco
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France.,Laboratoire de Biochimie et Biologie Moléculaire, CHRU de Tours , Tours, France
| | - Debora Lanznaster
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France
| | - Charlotte Veyrat-Durebex
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France.,Laboratoire de Biochimie et Biologie Moléculaire, CHRU de Tours , Tours, France
| | - Rudolf Hergesheimer
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France
| | - Patrick Vourch
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France.,Laboratoire de Biochimie et Biologie Moléculaire, CHRU de Tours , Tours, France
| | - Francois Maillot
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France.,Service de Médecine Interne, CHRU de Tours , Tours, France
| | - Christian R Andres
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France.,Laboratoire de Biochimie et Biologie Moléculaire, CHRU de Tours , Tours, France
| | - Pierre-François Pradat
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France.,Biomedical Imaging Laboratory, CNRS, INSERM, Sorbonne University , Paris, France.,APHP, Department of Neurology, Paris ALS Center, Pitié Salpêtrière Hospital , Paris, France
| | - Phillipe Corcia
- Unité INSERM U1253, équipe, neurogénomique et physiopathologie neuronale, Université de Tours , Tours, France.,Service de Neurologie, CHRU de Tours , Tours, France
| |
Collapse
|
12
|
Winquist RJ, Gribkoff VK. Targeting putative components of the mitochondrial permeability transition pore for novel therapeutics. Biochem Pharmacol 2020; 177:113995. [PMID: 32339494 DOI: 10.1016/j.bcp.2020.113995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Few discoveries have influenced drug discovery programs more than the finding that mitochondrial membranes undergo swings in permeability in response to cellular perturbations. The conductor of these permeability changes is the aptly named mitochondrial permeability transition pore which, although not yet precisely defined, is comprised of several integral proteins that differentially act to regulate the flux of ions, proteins and metabolic byproducts during the course of cellular physiological functions but also pathophysiological insults. Pursuit of the pore's exact identity remains a topic of keen interest, but decades of research have unearthed provocative functions for the integral proteins leading to their evaluation to develop novel therapeutics for a wide range of clinical indications. Chief amongst these targeted, integral proteins have been the Voltage Dependent Anion Channel (VDAC) and the F1FO ATP synthase. Research associated with the roles and ligands of VDAC has been extensive and we will expand upon 3 examples of ligand:VDAC interactions for consideration of drug discovery projects: Tubulin:VDAC1, Hexokinase I/II:VDAC1 and olesoxime:VDAC1. The discoveries that cyclosporine blocks mitochondrial permeability transition via binding to cyclophilin D, and that cyclophilin D is an important component of F1FO ATP synthase, has heightened interest in the F1FO ATP synthase as a focal point for drug discovery, and we will discuss 2 plausible campaigns associated with disease indications. To date no drug has emerged from prospective targeting these integral proteins; however, continued exploration such as the approaches suggested in this Commentary will increase the likelihood of providing important therapeutics for severely unmet medical needs.
Collapse
Affiliation(s)
- Raymond J Winquist
- Alkermes Pharmaceuticals Inc, 852 Winter Street, Waltham MA 02451, United States.
| | - Valentin K Gribkoff
- Yale University School of Medicine, Department of Internal Medicine, 333 Cedar St., New Haven, CT 06510, United States; TheraStat LLC, 44 Kings Grant Rd., Weston, MA 02493, United States
| |
Collapse
|
13
|
Buonvicino D, Ranieri G, Pratesi S, Gerace E, Muzzi M, Guasti D, Tofani L, Chiarugi A. Neuroprotection induced by dexpramipexole delays disease progression in a mouse model of progressive multiple sclerosis. Br J Pharmacol 2020; 177:3342-3356. [PMID: 32199028 DOI: 10.1111/bph.15058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Drugs able to counteract progressive multiple sclerosis (MS) represent a largely unmet therapeutic need. Even though the pathogenesis of disease evolution is still obscure, accumulating evidence indicates that mitochondrial dysfunction plays a causative role in neurodegeneration and axonopathy in progressive MS patients. Here, we investigated the effects of dexpramipexole, a compound with a good safety profile in humans and able to sustain mitochondria functioning and energy production, in a mouse model of progressive MS. EXPERIMENTAL APPROACH Female non-obese diabetic mice were immunized with MOG35-55 . Functional, immune and neuropathological parameters were analysed during disease evolution in animals treated or not with dexpramipexole. The compound's effects on bioenergetics and neuroprotection were also evaluated in vitro. KEY RESULTS We found that oral treatment with dexpramipexole at a dose consistent with that well tolerated in humans delayed disability progression, extended survival, counteracted reduction of spinal cord mitochondrial DNA content and reduced spinal cord axonal loss of mice. Accordingly, the drug sustained in vitro bioenergetics of mouse optic nerve and dorsal root ganglia and counteracted neurodegeneration of organotypic mouse cortical cultures exposed to the adenosine triphosphate-depleting agents oligomycin or veratridine. Dexpramipexole, however, was unable to affect the adaptive and innate immune responses both in vivo and in vitro. CONCLUSION AND IMPLICATION The present findings corroborate the hypothesis that neuroprotective agents may be of relevance to counteract MS progression and disclose the translational potential of dexpramipexole to treatment of progressive MS patients as a stand-alone or adjunctive therapy.
Collapse
Affiliation(s)
- Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Giuseppe Ranieri
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Sara Pratesi
- Centre of Immunological Research DENOTHE, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elisabetta Gerace
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Mirko Muzzi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Daniele Guasti
- Department of Clinical and Experimental Medicine, Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| | - Lorenzo Tofani
- Clinical Trials Coordinating Center of Istituto Toscano Tumori, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
14
|
Hergesheimer R, Lanznaster D, Vourc’h P, Andres C, Bakkouche S, Beltran S, Blasco H, Corcia P, Couratier P. Advances in disease-modifying pharmacotherapies for the treatment of amyotrophic lateral sclerosis. Expert Opin Pharmacother 2020; 21:1103-1110. [DOI: 10.1080/14656566.2020.1746270] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- R Hergesheimer
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France
| | - D Lanznaster
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France
| | - P Vourc’h
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France
- CHU De Tours, Service de Biochimie et Biologie Moléculaire, Tours, France
| | - Cr Andres
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France
- CHU De Tours, Service de Biochimie et Biologie Moléculaire, Tours, France
| | - Se Bakkouche
- CHU de Tours, Service de Neurologie, Tours, France
| | - S Beltran
- CHU de Tours, Service de Neurologie, Tours, France
| | - H Blasco
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France
- CHU De Tours, Service de Biochimie et Biologie Moléculaire, Tours, France
| | - P Corcia
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France
- CHU de Tours, Service de Neurologie, Tours, France
| | - P Couratier
- CHU Limoges, Service de Neurologie, Centre Expert ALS, Limoges, France
| |
Collapse
|
15
|
Lee SI, Hoeijmakers JGJ, Faber CG, Merkies ISJ, Lauria G, Waxman SG. The small fiber neuropathy NaV1.7 I228M mutation: impaired neurite integrity via bioenergetic and mitotoxic mechanisms, and protection by dexpramipexole. J Neurophysiol 2020; 123:645-657. [DOI: 10.1152/jn.00360.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Gain-of-function variants in voltage-gated sodium channel NaV1.7 that increase firing frequency and spontaneous firing of dorsal root ganglion (DRG) neurons have recently been identified in 5–10% of patients with idiopathic small fiber neuropathy (I-SFN). Our previous in vitro observations suggest that enhanced sodium channel activity can contribute to a decrease in length of peripheral sensory axons. We have hypothesized that sustained sodium influx due to the expression of SFN-associated sodium channel variants may trigger an energetic deficit in neurons that contributes to degeneration and loss of nerve fibers in SFN. Using an ATP FRET biosensor, we now demonstrate reduced steady-state levels of ATP and markedly faster ATP decay in response to membrane depolarization in cultured DRG neurons expressing an SFN-associated variant NaV1.7, I228M, compared with wild-type neurons. We also observed that I228M neurons show a significant reduction in mitochondrial density and size, indicating dysfunctional mitochondria and a reduced bioenergetic capacity. Finally, we report that exposure to dexpramipexole, a drug that improves mitochondrial energy metabolism, increases the neurite length of I228M-expressing neurons. Our data suggest that expression of gain-of-function variants of NaV1.7 can damage mitochondria and compromise cellular capacity for ATP production. The resulting bioenergetic crisis can consequently contribute to loss of axons in SFN. We suggest that, in addition to interventions that reduce ionic disturbance caused by mutant NaV1.7 channels, an alternative therapeutic strategy might target the bioenergetic burden and mitochondrial damage that occur in SFN associated with NaV1.7 gain-of-function mutations. NEW & NOTEWORTHY Sodium channel NaV1.7 mutations that increase dorsal root ganglion (DRG) neuron excitability have been identified in small fiber neuropathy (SFN). We demonstrate reduced steady-state ATP levels, faster depolarization-evoked ATP decay, and reduced mitochondrial density and size in cultured DRG neurons expressing SFN-associated variant NaV1.7 I228M. Dexpramipexole, which improves mitochondrial energy metabolism, has a protective effect. Because gain-of-function NaV1.7 variants can compromise bioenergetics, therapeutic strategies that target bioenergetic burden and mitochondrial damage merit study in SFN.
Collapse
Affiliation(s)
- Seong-il Lee
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut
- Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Janneke G. J. Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Catharina G. Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ingemar S. J. Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Neurology, Curaçao Medical Center, Willemstad, Curaçao
| | - Giuseppe Lauria
- Neuroalgology Unit, Foundazione IRCCS Istituto Neurologico “Carlo Besta,” Milan, Italy
- Department of Biomedical and Clinical Sciences “Luigi Sacco,” University of Milan, Milan, Italy
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut
- Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
16
|
Dexpramipexole blocks Nav1.8 sodium channels and provides analgesia in multiple nociceptive and neuropathic pain models. Pain 2019; 161:831-841. [DOI: 10.1097/j.pain.0000000000001774] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
17
|
Chen R, Park HA, Mnatsakanyan N, Niu Y, Licznerski P, Wu J, Miranda P, Graham M, Tang J, Boon AJW, Cossu G, Mandemakers W, Bonifati V, Smith PJS, Alavian KN, Jonas EA. Parkinson's disease protein DJ-1 regulates ATP synthase protein components to increase neuronal process outgrowth. Cell Death Dis 2019; 10:469. [PMID: 31197129 PMCID: PMC6565618 DOI: 10.1038/s41419-019-1679-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/10/2019] [Accepted: 05/19/2019] [Indexed: 12/17/2022]
Abstract
Familial Parkinson’s disease (PD) protein DJ-1 mutations are linked to early onset PD. We have found that DJ-1 binds directly to the F1FO ATP synthase β subunit. DJ-1’s interaction with the β subunit decreased mitochondrial uncoupling and enhanced ATP production efficiency while in contrast mutations in DJ-1 or DJ-1 knockout increased mitochondrial uncoupling, and depolarized neuronal mitochondria. In mesencephalic DJ-1 KO cultures, there was a progressive loss of neuronal process extension. This was ameliorated by a pharmacological reagent, dexpramipexole, that binds to ATP synthase, closing a mitochondrial inner membrane leak and enhancing ATP synthase efficiency. ATP synthase c-subunit can form an uncoupling channel; we measured, therefore, ATP synthase F1 (β subunit) and c-subunit protein levels. We found that ATP synthase β subunit protein level in the DJ-1 KO neurons was approximately half that found in their wild-type counterparts, comprising a severe defect in ATP synthase stoichiometry and unmasking c-subunit. We suggest that DJ-1 enhances dopaminergic cell metabolism and growth by its regulation of ATP synthase protein components.
Collapse
Affiliation(s)
- Rongmin Chen
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA
| | - Han-A Park
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA.,Department of Human Nutrition and Hospitality Management, University of Alabama, Tuscaloosa, AL, USA
| | - Nelli Mnatsakanyan
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA
| | - Yulong Niu
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA
| | - Pawel Licznerski
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA
| | - Jing Wu
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA
| | - Paige Miranda
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA
| | - Morven Graham
- Department of Cell Biology, Yale University, New Haven, CT, USA
| | - Jack Tang
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA
| | - Agnita J W Boon
- Department of Neurology, Erasmus MC, Rotterdam, The Netherlands
| | - Giovanni Cossu
- Neurology Service and Stroke Unit, Brotzu General Hospital, Cagliari, Italy
| | - Wim Mandemakers
- Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Vincenzo Bonifati
- Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Peter J S Smith
- Institute of Life Sciences, University of Southampton, Southampton, England.,Marine Biological Laboratory, Woods Hole, MA, USA
| | - Kambiz N Alavian
- Marine Biological Laboratory, Woods Hole, MA, USA.,Division of Brain Sciences, Department of Medicine, Imperial College, London, UK
| | - Elizabeth A Jonas
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, CT, USA. .,Marine Biological Laboratory, Woods Hole, MA, USA. .,Department of Neuroscience, Yale University, New Haven, CT, USA.
| |
Collapse
|
18
|
Mallach A, Weinert M, Arthur J, Gveric D, Tierney TS, Alavian KN. Post mortem examination of Parkinson's disease brains suggests decline in mitochondrial biomass, reversed by deep brain stimulation of subthalamic nucleus. FASEB J 2019; 33:6957-6961. [PMID: 30862197 DOI: 10.1096/fj.201802628r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is the most commonly used surgical treatment for Parkinson's disease (PD). The disease-modifying aspects of DBS at a cellular level are not fully understood, and the key question of the effect of DBS on the degeneration of the dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) remains to be answered. A major technical hurdle in determining any neuroprotective effect by DBS is its use in mid- to late-stage patients with PD when a majority of the DA neurons have been lost. In this work, we hypothesized that the long-term clinical benefits of DBS are, at least in part, due to a neuromodulatory effect on the SNpc neurons. These changes would affect cellular energetics and mitochondrial metabolism. We examined the number and volume of mitochondria as well as their vicinity to the DA presynaptic terminals postmortem caudate and putamen of 3 healthy individuals, 4 PD cases, and 3 DBS-treated patients. PD seems to have caused an increase in the mean distance between mitochondria and presynaptic terminals as well as a decrease in mean mitochondrial volume and numbers in DA projections. Although there was no difference in distance between mitochondria and presynaptic terminals of SNpc neurons in PD brains vs. DBS-treated brains, DBS treatment seemed to have inhibited or reversed the reduction in mitochondrial volume and numbers caused by PD. These results suggest enhanced metabolic plasticity leading to neuroprotection in the SNpc as a result of STN-DBS.-Mallach, A., Weinert, M., Arthur, J., Gveric, D., Tierney, T. S., Alavian, K. N. Post mortem examination of Parkinson's disease brains suggests decline in mitochondrial biomass, reversed by deep brain stimulation of subthalamic nucleus.
Collapse
Affiliation(s)
- Anna Mallach
- Division of Brain Sciences, Department of Medicine, Imperial College, London, United Kingdom
| | - Maria Weinert
- Division of Brain Sciences, Department of Medicine, Imperial College, London, United Kingdom
| | - Joy Arthur
- Division of Brain Sciences, Department of Medicine, Imperial College, London, United Kingdom
| | - Djordje Gveric
- Division of Brain Sciences, Department of Medicine, Imperial College, London, United Kingdom
| | - Travis S Tierney
- Division of Brain Sciences, Department of Medicine, Imperial College, London, United Kingdom.,Department of Neurosurgery, Nicklaus Children's Hospital, Miami, Florida, USA; and
| | - Kambiz N Alavian
- Division of Brain Sciences, Department of Medicine, Imperial College, London, United Kingdom.,Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
19
|
Coppi E, Lana D, Cherchi F, Fusco I, Buonvicino D, Urru M, Ranieri G, Muzzi M, Iovino L, Giovannini MG, Pugliese AM, Chiarugi A. Dexpramipexole enhances hippocampal synaptic plasticity and memory in the rat. Neuropharmacology 2018; 143:306-316. [PMID: 30291939 DOI: 10.1016/j.neuropharm.2018.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 01/22/2023]
Abstract
Even though pharmacological approaches able to counteract age-dependent cognitive impairment have been highly investigated, drugs improving cognition and memory are still an unmet need. It has been hypothesized that sustaining energy dynamics within the aged hippocampus can boost memory storage by sustaining synaptic functioning and long term potentiation (LTP). Dexpramipexole (DEX) is the first-in-class compound able to sustain neuronal bioenergetics by interacting with mitochondrial F1Fo-ATP synthase. In the present study, for the first time we evaluated the effects of DEX on synaptic fatigue, LTP induction, learning and memory retention. We report that DEX improved LTP maintenance in CA1 neurons of acute hippocampal slices from aged but not young rats. However, we found no evidence that DEX counteracted two classic parameters of synaptic fatigue such as fEPSP reduction or the train area during the high frequency stimulation adopted to induce LTP. Interestingly, patch-clamp recordings in rat hippocampal neurons revealed that DEX dose-dependently inhibited (IC50 814 nM) the IA current, a rapidly-inactivating K+ current that negatively regulates neuronal excitability as well as cognition and memory processes. In keeping with this, DEX counteracted both scopolamine-induced spatial memory loss in rats challenged in Morris Water Maze test and memory retention in rats undergoing Novel Object Recognition. Overall, the present study discloses the ability of DEX to boost hippocampal synaptic plasticity, learning and memory. In light of the good safety profile of DEX in humans, our findings may have a realistic translational potential to treatment of cognitive disorders.
Collapse
Affiliation(s)
- Elisabetta Coppi
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Italy.
| | - Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Federica Cherchi
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Italy
| | - Irene Fusco
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Italy
| | - Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Matteo Urru
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Giuseppe Ranieri
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Mirko Muzzi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Ludovica Iovino
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Italy
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| | - Anna Maria Pugliese
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Italy
| |
Collapse
|
20
|
Muzzi M, Buonvicino D, Urru M, Tofani L, Chiarugi A. Repurposing of dexpramipexole to treatment of neonatal hypoxic/ischemic encephalopathy. Neurosci Lett 2018; 687:234-240. [PMID: 30287306 DOI: 10.1016/j.neulet.2018.09.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/27/2018] [Accepted: 09/29/2018] [Indexed: 01/28/2023]
Abstract
Dexpramipexole (DEX) is a drug with a good safety profile in humans, known for its ability to increase mitochondrial ATP production and prompt neuroprotection in adult rodents subjected to cerebral ischemia. In the present study we evaluated the effect of DEX in rat pups subjected to common carotid artery occlusion plus hypoxia (CCAoH, the classic Rice-Vannucci model). Because of the wide range of infarct size distribution in the CCAoH model, a priori subanalysis based on the effect of DEX on mild/moderate or severe brain injuries was conducted. The subanalysis showed that the drug (3 mg/kg bid i.p, after the hypoxic insult) decreased the infarction size in pups with mild/moderate injuries. Next, we developed a distal middle cerebral artery occlusion plus hypoxia (dMCAoH) model, characterized by an intra-experimental infarct size variability lower than that of the CCAoH model. Post-ischemic treatment with DEX (3 mg/kg bid i.p, after the hypoxic insult) reduced brain infarcts in pups exposed to dMCAoH. For the first time, we show that DEX reduces brain injury in different models of neonatal HIE. In light of the favorable safety profile of DEX in humans, the drug might have a realistic translational potential to treatment of perinatal cerebrovascular disorders.
Collapse
Affiliation(s)
- Mirko Muzzi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| | - Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Matteo Urru
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Lorenzo Tofani
- Clinical Trials Coordinating Center of Istituto Toscano Tumori, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
21
|
|
22
|
Vandoorne T, De Bock K, Van Den Bosch L. Energy metabolism in ALS: an underappreciated opportunity? Acta Neuropathol 2018; 135:489-509. [PMID: 29549424 PMCID: PMC5978930 DOI: 10.1007/s00401-018-1835-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a relentlessly progressive and fatal neurodegenerative disorder that primarily affects motor neurons. Despite our increased understanding of the genetic factors contributing to ALS, no effective treatment is available. A growing body of evidence shows disturbances in energy metabolism in ALS. Moreover, the remarkable vulnerability of motor neurons to ATP depletion has become increasingly clear. Here, we review metabolic alterations present in ALS patients and models, discuss the selective vulnerability of motor neurons to energetic stress, and provide an overview of tested and emerging metabolic approaches to treat ALS. We believe that a further understanding of the metabolic biology of ALS can lead to the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Tijs Vandoorne
- Department of Neurosciences, Experimental Neurology, KU Leuven-University of Leuven, Campus Gasthuisberg O&N 4, Herestraat 49, PB 602, 3000, Leuven, Belgium
- Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000, Leuven, Belgium
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, KU Leuven-University of Leuven, Campus Gasthuisberg O&N 4, Herestraat 49, PB 602, 3000, Leuven, Belgium.
- Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000, Leuven, Belgium.
| |
Collapse
|
23
|
Muzzi M, Gerace E, Buonvicino D, Coppi E, Resta F, Formentini L, Zecchi R, Tigli L, Guasti D, Ferri M, Camaioni E, Masi A, Pellegrini‐Giampietro DE, Mannaioni G, Bani D, Pugliese AM, Chiarugi A. Dexpramipexole improves bioenergetics and outcome in experimental stroke. Br J Pharmacol 2018; 175:272-283. [PMID: 28320070 PMCID: PMC5758384 DOI: 10.1111/bph.13790] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/24/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Dexpramipexole, a drug recently tested in patients with amyotrophic lateral sclerosis (ALS,) is able to bind F1Fo ATP synthase and increase mitochondrial ATP production. Here, we have investigated its effects on experimental ischaemic brain injury. EXPERIMENTAL APPROACH The effects of dexpramipexole on bioenergetics, Ca2+ fluxes, electrophysiological functions and death were evaluated in primary neural cultures and hippocampal slices exposed to oxygen-glucose deprivation (OGD). Effects on infarct volumes and neurological functions were also evaluated in mice following proximal or distal middle cerebral artery occlusion (MCAo). Distribution of dexpramipexole within the ischaemic brain was evaluated by means of mass spectrometry imaging. KEY RESULTS Dexpramipexole increased mitochondrial ATP production in cultured neurons or glia and reduces energy failure, prevents intracellular Ca2+ overload and affords cytoprotection when cultures are exposed to OGD. This compound also counteracted ATP depletion, mitochondrial swelling, anoxic depolarization, loss of synaptic activity and neuronal death in hippocampal slices subjected to OGD. Post-ischaemic treatment with dexpramipexole, at doses consistent with those already used in ALS patients, reduced brain infarct size and ameliorated neuroscore in mice subjected to transient or permanent MCAo. Notably, the concentrations of dexpramipexole reached within the ischaemic penumbra equalled those found neuroprotective in vitro. CONCLUSION AND IMPLICATIONS Dexpramipexole, a compound able to increase mitochondrial F1Fo ATP-synthase activity reduced ischaemic brain injury. These findings, together with the excellent brain penetration and favourable safety profile in humans, make dexpramipexole a drug with realistic translational potential for the treatment of stroke. LINKED ARTICLES This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc.
Collapse
Affiliation(s)
- Mirko Muzzi
- Department of Health Sciences, Section of Clinical Pharmacology and OncologyUniversity of FlorenceFlorenceItaly
| | - Elisabetta Gerace
- Department NEUROFARBA, Division of Pharmacology and ToxicologyUniversity of FlorenceFlorenceItaly
| | - Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and OncologyUniversity of FlorenceFlorenceItaly
| | - Elisabetta Coppi
- Department NEUROFARBA, Division of Pharmacology and ToxicologyUniversity of FlorenceFlorenceItaly
| | - Francesco Resta
- Department NEUROFARBA, Division of Pharmacology and ToxicologyUniversity of FlorenceFlorenceItaly
| | - Laura Formentini
- Departamento de Biología MolecularCentro de Biología Molecular Severo Ochoa Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| | - Riccardo Zecchi
- Mass Spectrometry Service Centre (CISM)University of FlorenceFlorenceItaly
| | - Laura Tigli
- Mass Spectrometry Service Centre (CISM)University of FlorenceFlorenceItaly
| | - Daniele Guasti
- Department of Clinical and Experimental Medicine, Research Unit of Histology & EmbryologyUniversity of FlorenceFlorenceItaly
| | - Martina Ferri
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| | - Emidio Camaioni
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| | - Alessio Masi
- Department NEUROFARBA, Division of Pharmacology and ToxicologyUniversity of FlorenceFlorenceItaly
| | | | - Guido Mannaioni
- Department NEUROFARBA, Division of Pharmacology and ToxicologyUniversity of FlorenceFlorenceItaly
| | - Daniele Bani
- Department of Clinical and Experimental Medicine, Research Unit of Histology & EmbryologyUniversity of FlorenceFlorenceItaly
| | - Anna M Pugliese
- Department NEUROFARBA, Division of Pharmacology and ToxicologyUniversity of FlorenceFlorenceItaly
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and OncologyUniversity of FlorenceFlorenceItaly
| |
Collapse
|
24
|
Zhou T, Ahmad TK, Gozda K, Truong J, Kong J, Namaka M. Implications of white matter damage in amyotrophic lateral sclerosis (Review). Mol Med Rep 2017; 16:4379-4392. [PMID: 28791401 PMCID: PMC5646997 DOI: 10.3892/mmr.2017.7186] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 06/09/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease, which involves the progressive degeneration of motor neurons. ALS has long been considered a disease of the grey matter; however, pathological alterations of the white matter (WM), including axonal loss, axonal demyelination and oligodendrocyte death, have been reported in patients with ALS. The present review examined motor neuron death as the primary cause of ALS and evaluated the associated WM damage that is guided by neuronal‑glial interactions. Previous studies have suggested that WM damage may occur prior to the death of motor neurons, and thus may be considered an early indicator for the diagnosis and prognosis of ALS. However, the exact molecular mechanisms underlying early‑onset WM damage in ALS have yet to be elucidated. The present review explored the detailed anatomy of WM and identified several pathological mechanisms that may be implicated in WM damage in ALS. In addition, it associated the pathophysiological alterations of WM, which may contribute to motor neuron death in ALS, with similar mechanisms of WM damage that are involved in multiple sclerosis (MS). Furthermore, the early detection of WM damage in ALS, using neuroimaging techniques, may lead to earlier therapeutic intervention, using immunomodulatory treatment strategies similar to those used in relapsing‑remitting MS, aimed at delaying WM damage in ALS. Early therapeutic approaches may have the potential to delay motor neuron damage and thus prolong the survival of patients with ALS. The therapeutic interventions that are currently available for ALS are only marginally effective. However, early intervention with immunomodulatory drugs may slow the progression of WM damage in the early stages of ALS, thus delaying motor neuron death and increasing the life expectancy of patients with ALS.
Collapse
Affiliation(s)
- Ting Zhou
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Tina Khorshid Ahmad
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Kiana Gozda
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jessica Truong
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Michael Namaka
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
- Department of Medical Rehabilitation, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Department of Internal Medicine, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 1R9, Canada
| |
Collapse
|
25
|
Tefera TW, Borges K. Metabolic Dysfunctions in Amyotrophic Lateral Sclerosis Pathogenesis and Potential Metabolic Treatments. Front Neurosci 2017; 10:611. [PMID: 28119559 PMCID: PMC5222822 DOI: 10.3389/fnins.2016.00611] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/26/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease primarily characterized by loss of motor neurons in brain and spinal cord. The death of motor neurons leads to denervation of muscle which in turn causes muscle weakness and paralysis, decreased respiratory function and eventually death. Growing evidence indicates disturbances in energy metabolism in patients with ALS and animal models of ALS, which are likely to contribute to disease progression. Particularly, defects in glucose metabolism and mitochondrial dysfunction limit the availability of ATP to CNS tissues and muscle. Several metabolic approaches improving mitochondrial function have been investigated in vitro and in vivo and showed varying effects in ALS. The effects of metabolic approaches in ALS models encompass delays in onset of motor symptoms, protection of motor neurons and extension of survival, which signifies an important role of metabolism in the pathogenesis of the disease. There is now an urgent need to test metabolic approaches in controlled clinical trials. In addition, more detailed studies to better characterize the abnormalities in energy metabolism in patients with ALS and ALS models are necessary to develop metabolically targeted effective therapies that can slow the progression of the disease and prolong life for patients with ALS.
Collapse
Affiliation(s)
| | - Karin Borges
- Laboratory for Neurological Disorders and Metabolism, School of Biomedical Sciences, Department of Pharmacology, The University of QueenslandBrisbane, QLD, Australia
| |
Collapse
|
26
|
Biasutto L, Azzolini M, Szabò I, Zoratti M. The mitochondrial permeability transition pore in AD 2016: An update. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:2515-30. [PMID: 26902508 DOI: 10.1016/j.bbamcr.2016.02.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/13/2022]
Abstract
Over the past 30years the mitochondrial permeability transition - the permeabilization of the inner mitochondrial membrane due to the opening of a wide pore - has progressed from being considered a curious artifact induced in isolated mitochondria by Ca(2+) and phosphate to a key cell-death-inducing process in several major pathologies. Its relevance is by now universally acknowledged and a pharmacology targeting the phenomenon is being developed. The molecular nature of the pore remains to this day uncertain, but progress has recently been made with the identification of the FOF1 ATP synthase as the probable proteic substrate. Researchers sharing this conviction are however divided into two camps: these believing that only the ATP synthase dimers or oligomers can form the pore, presumably in the contact region between monomers, and those who consider that the ring-forming c subunits in the FO sector actually constitute the walls of the pore. The latest development is the emergence of a new candidate: Spastic Paraplegia 7 (SPG7), a mitochondrial AAA-type membrane protease which forms a 6-stave barrel. This review summarizes recent developments of research on the pathophysiological relevance and on the molecular nature of the mitochondrial permeability transition pore. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.
Collapse
Affiliation(s)
- Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Michele Azzolini
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy
| | - Ildikò Szabò
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biology, Viale G. Colombo 3, 35121 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35121 Padova, Italy; University of Padova, Department of Biomedical Sciences, Viale G. Colombo 3, 35121 Padova, Italy.
| |
Collapse
|
27
|
Cotroneo E, Ashek A, Wang L, Wharton J, Dubois O, Bozorgi S, Busbridge M, Alavian KN, Wilkins MR, Zhao L. Iron homeostasis and pulmonary hypertension: iron deficiency leads to pulmonary vascular remodeling in the rat. Circ Res 2015; 116:1680-90. [PMID: 25767292 DOI: 10.1161/circresaha.116.305265] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
RATIONALE Iron deficiency without anemia is prevalent in patients with idiopathic pulmonary arterial hypertension and associated with reduced exercise capacity and survival. OBJECTIVES We hypothesized that iron deficiency is involved in the pathogenesis of pulmonary hypertension and iron replacement is a possible therapeutic strategy. METHODS AND RESULTS Rats were fed an iron-deficient diet (IDD, 7 mg/kg) and investigated for 4 weeks. Iron deficiency was evident from depleted iron stores (decreased liver, serum iron, and ferritin), reduced erythropoiesis, and significantly decreased transferrin saturation and lung iron stores after 2 weeks IDD. IDD rats exhibited profound pulmonary vascular remodeling with prominent muscularization, medial hypertrophy, and perivascular inflammatory cell infiltration, associated with raised pulmonary artery pressure and right ventricular hypertrophy. IDD rat lungs demonstrated increased expression of hypoxia-induced factor-1α and hypoxia-induced factor-2α, nuclear factor of activated T cells and survivin, and signal transducers and activators of transcription-3 activation, which promote vascular cell proliferation and resistance to apoptosis. Biochemical examination showed reduced mitochondrial complex I activity and mitochondrial membrane hyperpolarization in mitochondria from IDD rat pulmonary arteries. Along with upregulation of the glucose transporter, glucose transporter 1, and glycolytic genes, hk1 and pdk1, lung fluorine-18-labeled 2-fluoro-2-deoxyglucose ligand uptake was significantly increased in IDD rats. The hemodynamic and pulmonary vascular remodeling were reversed by iron replacement (ferric carboxymaltose, 75 mg/kg) and attenuated in the presence of iron deficiency by dichloroacetate and imatinib, 2 putative treatments explored for pulmonary arterial hypertension that target aerobic glycolysis and proliferation, respectively. CONCLUSIONS These data suggest a major role for iron in pulmonary vascular homeostasis and support the clinical evaluation of iron replacement in patients with pulmonary hypertension.
Collapse
Affiliation(s)
- Emanuele Cotroneo
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - Ali Ashek
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - Lei Wang
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - John Wharton
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - Olivier Dubois
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - Sophie Bozorgi
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - Mark Busbridge
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - Kambiz N Alavian
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - Martin R Wilkins
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.)
| | - Lan Zhao
- From the Centre for Pharmacology and Therapeutics, Department of Medicine (E.C., A.A., L.W., J.W., O.D., S.B., M.R.W., L.Z.) and Division of Brain Sciences, Department of Medicine (K.N.A.), Imperial College London, Hammersmith Hospital, London, United Kingdom; and Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, United Kingdom (M.B.).
| |
Collapse
|
28
|
Bucchia M, Ramirez A, Parente V, Simone C, Nizzardo M, Magri F, Dametti S, Corti S. Therapeutic Development in Amyotrophic Lateral Sclerosis. Clin Ther 2015; 37:668-80. [DOI: 10.1016/j.clinthera.2014.12.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/18/2014] [Accepted: 12/29/2014] [Indexed: 12/12/2022]
|
29
|
Vieira FG, LaDow E, Moreno A, Kidd JD, Levine B, Thompson K, Gill A, Finkbeiner S, Perrin S. Dexpramipexole is ineffective in two models of ALS related neurodegeneration. PLoS One 2014; 9:e91608. [PMID: 25526593 PMCID: PMC4272269 DOI: 10.1371/journal.pone.0091608] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 02/11/2014] [Indexed: 12/12/2022] Open
Abstract
Treatment options for people living with amyotrophic lateral sclerosis (ALS) are limited and ineffective. Recently, dexpramipexole (RPPX) was advanced into human ALS clinical trials. In the current studies, we investigated RPPX in two parallel screening systems: 1) appropriately powered, sibling-matched, gender-balanced survival efficacy screening in high-copy B6-SJL-SOD1G93A/Gur1 mice, and 2) high-content neuronal survival screening in primary rat cortical neurons transfected with wild-type human TDP43 or mutant human TDP43. In both cases, we exposed the test systems to RPPX levels approximating those achieved in human Phase II clinical investigations. In SOD1G93A mice, no effect was observed on neuromotor disease progression or survival. In primary cortical neurons transfected with either mutant or wild-type human TDP43, a marginally significant improvement in a single indicator of neuronal survival was observed, and only at the 10 µM RPPX treatment. These systems reflect both mutant SOD1- and TDP43-mediated forms of neurodegeneration. The systems also reflect both complex non-cell autonomous and neuronal cell autonomous disease mechanisms. The results of these experiments, taken in context with results produced by other molecules tested in both screening systems, do not argue positively for further study of RPPX in ALS.
Collapse
Affiliation(s)
- Fernando G. Vieira
- ALS Therapy Development Institute, Cambridge, Massachusetts, United States of America
- * E-mail: (FGV); (SF)
| | - Eva LaDow
- Gladstone Institute of Neurological Disease, San Francisco, California, United States of America
| | - Andy Moreno
- ALS Therapy Development Institute, Cambridge, Massachusetts, United States of America
| | - Joshua D. Kidd
- ALS Therapy Development Institute, Cambridge, Massachusetts, United States of America
| | - Beth Levine
- ALS Therapy Development Institute, Cambridge, Massachusetts, United States of America
| | - Kenneth Thompson
- ALS Therapy Development Institute, Cambridge, Massachusetts, United States of America
| | - Alan Gill
- ALS Therapy Development Institute, Cambridge, Massachusetts, United States of America
| | - Steven Finkbeiner
- Gladstone Institute of Neurological Disease, San Francisco, California, United States of America
- Taube-Koret Center for Neurodegenerative Disease Research and the Hellman Family Foundation Alzheimer's Disease Research Program, San Francisco, California, United States of America
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (FGV); (SF)
| | - Steven Perrin
- ALS Therapy Development Institute, Cambridge, Massachusetts, United States of America
| |
Collapse
|
30
|
Martin LJ, Fancelli D, Wong M, Niedzwiecki M, Ballarini M, Plyte S, Chang Q. GNX-4728, a novel small molecule drug inhibitor of mitochondrial permeability transition, is therapeutic in a mouse model of amyotrophic lateral sclerosis. Front Cell Neurosci 2014; 8:433. [PMID: 25565966 PMCID: PMC4271619 DOI: 10.3389/fncel.2014.00433] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 12/01/2014] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder in humans characterized by progressive degeneration of skeletal muscle and motor neurons in spinal cord, brainstem, and cerebral cortex causing skeletal muscle paralysis, respiratory insufficiency, and death. There are no cures or effective treatments for ALS. ALS can be inherited, but most cases are not associated with a family history of the disease. Mitochondria have been implicated in the pathogenesis but definitive proof of causal mechanisms is lacking. Identification of new clinically translatable disease mechanism-based molecular targets and small molecule drug candidates are needed for ALS patients. We tested the hypothesis in an animal model that drug modulation of the mitochondrial permeability transition pore (mPTP) is therapeutic in ALS. A prospective randomized placebo-controlled drug trial was done in a transgenic (tg) mouse model of ALS. We explored GNX-4728 as a therapeutic drug. GNX-4728 inhibits mPTP opening as evidenced by increased mitochondrial calcium retention capacity (CRC) both in vitro and in vivo. Chronic systemic treatment of G37R-human mutant superoxide dismutase-1 (hSOD1) tg mice with GNX-4728 resulted in major therapeutic benefits. GNX-4728 slowed disease progression and significantly improved motor function. The survival of ALS mice was increased significantly by GNX-4728 treatment as evidence by a nearly 2-fold extension of lifespan (360 days-750 days). GNX-4728 protected against motor neuron degeneration and mitochondrial degeneration, attenuated spinal cord inflammation, and preserved neuromuscular junction (NMJ) innervation in the diaphragm in ALS mice. This work demonstrates that a mPTP-acting drug has major disease-modifying efficacy in a preclinical mouse model of ALS and establishes mitochondrial calcium retention, and indirectly the mPTP, as targets for ALS drug development.
Collapse
Affiliation(s)
- Lee J. Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Pathobiology Graduate Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | | | - Margaret Wong
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Mark Niedzwiecki
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | | | | | - Qing Chang
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|
31
|
Scarpelli M, Todeschini A, Rinaldi F, Rota S, Padovani A, Filosto M. Strategies for treating mitochondrial disorders: an update. Mol Genet Metab 2014; 113:253-60. [PMID: 25458518 DOI: 10.1016/j.ymgme.2014.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 09/30/2014] [Accepted: 09/30/2014] [Indexed: 12/12/2022]
Abstract
Mitochondrial diseases are a heterogeneous group of disorders resulting from primary dysfunction of the respiratory chain due to both nuclear and mitochondrial DNA mutations. The wide heterogeneity of biochemical dysfunctions and pathogenic mechanisms typical of this group of diseases has hindered therapy trials; therefore, available treatment options remain limited. Therapeutic strategies aimed at increasing mitochondrial functions (by enhancing biogenesis and electron transport chain function), improving the removal of reactive oxygen species and noxious metabolites, modulating aberrant calcium homeostasis and repopulating mitochondrial DNA could potentially restore the respiratory chain dysfunction. The challenge that lies ahead is the translation of some promising laboratory results into safe and effective therapies for patients. In this review we briefly update and discuss the most feasible therapeutic approaches for mitochondrial diseases.
Collapse
Affiliation(s)
- Mauro Scarpelli
- Section of Neurology, Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Alice Todeschini
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - Fabrizio Rinaldi
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - Silvia Rota
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - Alessandro Padovani
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - Massimiliano Filosto
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy.
| |
Collapse
|
32
|
Wei D, Wu C, He P, Kerr D, Stecher S, Yang L. Chiral liquid chromatography–tandem mass spectrometry assay to determine that dexpramipexole is not converted to pramipexole in vivo after administered in humans. J Chromatogr B Analyt Technol Biomed Life Sci 2014; 971:133-40. [DOI: 10.1016/j.jchromb.2014.09.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/04/2014] [Accepted: 09/21/2014] [Indexed: 10/24/2022]
|
33
|
Berry JD, Cudkowicz ME, Shefner JM. Predicting success: optimizing phase II ALS trials for the transition to phase III. Amyotroph Lateral Scler Frontotemporal Degener 2014; 15:1-8. [PMID: 24588460 DOI: 10.3109/21678421.2013.838969] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) research is advancing quickly, but the transition from phase II to phase III trials remains particularly challenging. In part, this is because of the paradox of phase II ALS trials - they are expected to inform researchers about safety, tolerability, dosage selection, and efficacy using a small number of patients, and relying on essentially the same outcome measures used in phase III trials. We examined pharmacokinetics in the cerebrospinal fluid and pharmacodynamic markers to demonstrate target engagement. In addition, primary outcome measures are often not specified or do not reach pre-specified goals for significance. We conclude that future trials should include pharmacokinetic (preferably in CSF) and pharmacodynamic markers of target engagement when possible. Primary endpoints should be pre-specified. Inclusion criteria should be used to reduce heterogeneity and target a relevant subpopulation of people with ALS when possible. Multiple phase II trials might be required before moving to a large phase III trial.
Collapse
Affiliation(s)
- James D Berry
- Massachusetts General Hospital, Neurological Clinical Research Center , Boston, Massachusetts
| | | | | |
Collapse
|
34
|
Alavian KN, Dworetzky SI, Bonanni L, Zhang P, Sacchetti S, Li H, Signore AP, Smith PJS, Gribkoff VK, Jonas EA. The mitochondrial complex V-associated large-conductance inner membrane current is regulated by cyclosporine and dexpramipexole. Mol Pharmacol 2014; 87:1-8. [PMID: 25332381 DOI: 10.1124/mol.114.095661] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Inefficiency of oxidative phosphorylation can result from futile leak conductance through the inner mitochondrial membrane. Stress or injury may exacerbate this leak conductance, putting cells, and particularly neurons, at risk of dysfunction and even death when energy demand exceeds cellular energy production. Using a novel method, we have recently described an ion conductance consistent with mitochondrial permeability transition pore (mPTP) within the c-subunit of the ATP synthase. Excitotoxicity, reactive oxygen species-producing stimuli, or elevated mitochondrial matrix calcium opens the channel, which is inhibited by cyclosporine A and ATP/ADP. Here we show that ATP and the neuroprotective drug dexpramipexole (DEX) inhibited an ion conductance consistent with this c-subunit channel (mPTP) in brain-derived submitochondrial vesicles (SMVs) enriched for F1FO ATP synthase (complex V). Treatment of SMVs with urea denatured extramembrane components of complex V, eliminated DEX- but not ATP-mediated current inhibition, and reduced binding of [(14)C]DEX. Direct effects of DEX on the synthesis and hydrolysis of ATP by complex V suggest that interaction of the compound with its target results in functional conformational changes in the enzyme complex. [(14)C]DEX bound specifically to purified recombinant b and oligomycin sensitivity-conferring protein subunits of the mitochondrial F1FO ATP synthase. Previous data indicate that DEX increased the efficiency of energy production in cells, including neurons. Taken together, these studies suggest that modulation of a complex V-associated inner mitochondrial membrane current is metabolically important and may represent an avenue for the development of new therapeutics for neurodegenerative disorders.
Collapse
Affiliation(s)
- Kambiz N Alavian
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Steven I Dworetzky
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Laura Bonanni
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Ping Zhang
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Silvio Sacchetti
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Hongmei Li
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Armando P Signore
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Peter J S Smith
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Valentin K Gribkoff
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| | - Elizabeth A Jonas
- Department of Internal Medicine (K.N.A., P.Z., S.S., H.L., E.A.J.) and Department of Neurobiology (E.A.J.), Yale University School of Medicine, New Haven, Connecticut; Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom (K.N.A.); Department of Neuroscience, Imaging and Clinical Sciences, University G.d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy (L.B.); Knopp Biosciences LLC, Pittsburgh, Pennsylvania (S.I.D., A.P.S., V.K.G.); and Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, Massachusetts (P.J.S.S.)
| |
Collapse
|
35
|
Goyal NA, Mozaffar T. Experimental trials in amyotrophic lateral sclerosis: a review of recently completed, ongoing and planned trials using existing and novel drugs. Expert Opin Investig Drugs 2014; 23:1541-51. [DOI: 10.1517/13543784.2014.933807] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
36
|
Muyderman H, Chen T. Mitochondrial dysfunction in amyotrophic lateral sclerosis - a valid pharmacological target? Br J Pharmacol 2014; 171:2191-205. [PMID: 24148000 PMCID: PMC3976630 DOI: 10.1111/bph.12476] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/10/2013] [Accepted: 09/23/2013] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease characterized by the selective death of upper and lower motor neurons which ultimately leads to paralysis and ultimately death. Pathological changes in ALS are closely associated with pronounced and progressive changes in mitochondrial morphology, bioenergetics and calcium homeostasis. Converging evidence suggests that impaired mitochondrial function could be pivotal in the rapid neurodegeneration of this condition. In this review, we provide an update of recent advances in understanding mitochondrial biology in the pathogenesis of ALS and highlight the therapeutic value of pharmacologically targeting mitochondrial biology to slow disease progression.
Collapse
Affiliation(s)
- H Muyderman
- Centre for Neuroscience, Discipline of Medical Biochemistry, Flinders Medical Science and Technology, School of Medicine, Flinders UniversityAdelaide, SA, Australia
| | - T Chen
- Centre for Neuroscience, Discipline of Medical Biochemistry, Flinders Medical Science and Technology, School of Medicine, Flinders UniversityAdelaide, SA, Australia
| |
Collapse
|
37
|
Turner MR, Bowser R, Bruijn L, Dupuis L, Ludolph A, McGrath M, Manfredi G, Maragakis N, Miller RG, Pullman SL, Rutkove SB, Shaw PJ, Shefner J, Fischbeck KH. Mechanisms, models and biomarkers in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2013; 14 Suppl 1:19-32. [PMID: 23678877 DOI: 10.3109/21678421.2013.778554] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The last 30 years have seen a major advance in the understanding of the clinical and pathological heterogeneity of amyotrophic lateral sclerosis (ALS), and its overlap with frontotemporal dementia. Multiple, seemingly disparate biochemical pathways converge on a common clinical syndrome characterized by progressive loss of upper and lower motor neurons. Pathogenic themes in ALS include excitotoxicity, oxidative stress, mitochondrial dysfunction, neuroinflammation, altered energy metabolism, and most recently RNA mis-processing. The transgenic rodent, overexpressing mutant superoxide dismutase-1, is now only one of several models of ALS pathogenesis. The nematode, fruit fly and zebrafish all offer fresh insight, and the development of induced pluripotent stem cell-derived motor neurons holds promise for the screening of candidate therapeutics. The lack of useful biomarkers in ALS contributes to diagnostic delay, and the inability to stratify patients by prognosis may be an important factor in the failure of therapeutic trials. Biomarkers sensitive to disease activity might lessen reliance on clinical measures and survival as trial endpoints and reduce study length. Emerging proteomic markers of neuronal loss and glial activity in cerebrospinal fluid, a cortical signature derived from advanced structural and functional MRI, and the development of more sensitive measurements of lower motor neuron physiology are leading a new phase of biomarker-driven therapeutic discovery.
Collapse
Affiliation(s)
- Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Dexpramipexole versus placebo for patients with amyotrophic lateral sclerosis (EMPOWER): a randomised, double-blind, phase 3 trial. Lancet Neurol 2013; 12:1059-67. [DOI: 10.1016/s1474-4422(13)70221-7] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
39
|
Boscolo A, Ori C, Bennett J, Wiltgen B, Jevtovic-Todorovic V. Mitochondrial protectant pramipexole prevents sex-specific long-term cognitive impairment from early anaesthesia exposure in rats. Br J Anaesth 2013; 110 Suppl 1:i47-52. [PMID: 23616588 DOI: 10.1093/bja/aet073] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Exposure to general anaesthesia during critical stages of brain development results in long-lasting cognitive impairment. Co-administration of protective agents could minimize the detrimental effects of anaesthesia. Co-administration of R(+)pramipexole (PPX), a synthetic aminobenzothiazol derivative that restores mitochondrial integrity, prevents anaesthesia-induced mitochondrial and neuronal damage and prevents early development of cognitive impairment. Here, we determine the protective effects of PPX into late adulthood in male and female rats. METHODS Postnatal day 7 rats of both sexes were exposed to mock anaesthesia or combined midazolam, nitrous oxide, and isoflurane anaesthesia for 6 h with or without PPX. Cognitive abilities were assessed between 5 and 7 months of age using Morris water maze spatial navigation tasks. RESULTS Examination of spatial reference memory revealed that female, but not male, neonatal rats exposed to anaesthesia showed slowing of acquisition rates, which was significantly improved with PPX treatment. Examination of memory retention revealed that both male and female anaesthesia-treated rats have impaired memory retention performance compared with sham controls. Co-treatment with PPX resulted in improvement in memory retention in both sexes. CONCLUSION PPX provides long-lasting protection against cognitive impairment known to occur when very young animals are exposed to anaesthesia during the peak of brain development. Anaesthesia-induced cognitive impairment appears to be sex-specific with females being more vulnerable than males, suggesting that they could benefit more from early prevention.
Collapse
Affiliation(s)
- A Boscolo
- Department of Anaesthesiology, University of Virginia, Charlottesville, VA, USA
| | | | | | | | | |
Collapse
|
40
|
Goldstein A, Wolfe LA. The elusive magic pill: finding effective therapies for mitochondrial disorders. Neurotherapeutics 2013; 10:320-8. [PMID: 23355364 PMCID: PMC3625379 DOI: 10.1007/s13311-012-0175-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The incidence of mitochondrial diseases has been estimated at 11.5/100,000 (1:8500) worldwide. In the USA up to 4000 newborns annually are expected to develop a mitochondrial disease. More than 50 million adults in the USA also suffer from diseases in which primary or secondary mitochondrial dysfunction is involved. Mitochondrial dysfunction has been identified in cancer, infertility, diabetes, heart diseases, blindness, deafness, kidney disease, liver disease, stroke, migraine, dwarfism, and resulting from numerous medication toxicities. Mitochondrial dysfunction is also involved in normal aging and age-related neurodegenerative diseases, such as Parkinson and Alzheimer diseases. Yet most treatments available are based on empiric data and clinician experience because of the lack of randomized controlled clinical trials to provide evidence-based treatments for these disorders. Here we explore the current state of research for the treatment of mitochondrial disorders.
Collapse
Affiliation(s)
- Amy Goldstein
- />Division of Child Neurology, Childrens Hospital of Pittsburgh of UPMC, Pittsburgh, PA USA
| | - Lynne A. Wolfe
- />Undiagnosed Diseases Program, National Institutes of Health, 10 Center DR, MSC 1205, RM# 3-2551, Bethesda, MD 20892 USA
| |
Collapse
|
41
|
Zhao W, Varghese M, Vempati P, Dzhun A, Cheng A, Wang J, Lange D, Bilski A, Faravelli I, Pasinetti GM. Caprylic triglyceride as a novel therapeutic approach to effectively improve the performance and attenuate the symptoms due to the motor neuron loss in ALS disease. PLoS One 2012; 7:e49191. [PMID: 23145119 PMCID: PMC3492315 DOI: 10.1371/journal.pone.0049191] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/05/2012] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder of motor neurons causing progressive muscle weakness, paralysis, and finally death. ALS patients suffer from asthenia and their progressive weakness negatively impacts quality of life, limiting their daily activities. They have impaired energy balance linked to lower activity of mitochondrial electron transport chain enzymes in ALS spinal cord, suggesting that improving mitochondrial function may present a therapeutic approach for ALS. When fed a ketogenic diet, the G93A ALS mouse shows a significant increase in serum ketones as well as a significantly slower progression of weakness and lower mortality rate. In this study, we treated SOD1-G93A mice with caprylic triglyceride, a medium chain triglyceride that is metabolized into ketone bodies and can serve as an alternate energy substrate for neuronal metabolism. Treatment with caprylic triglyceride attenuated progression of weakness and protected spinal cord motor neuron loss in SOD1-G93A transgenic animals, significantly improving their performance even though there was no significant benefit regarding the survival of the ALS transgenic animals. We found that caprylic triglyceride significantly promoted the mitochondrial oxygen consumption rate in vivo. Our results demonstrated that caprylic triglyceride alleviates ALS-type motor impairment through restoration of energy metabolism in SOD1-G93A ALS mice, especially during the overt stage of the disease. These data indicate the feasibility of using caprylic acid as an easily administered treatment with a high impact on the quality of life of ALS patients.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
- GRECC, James J Peters Veterans Affairs Medical Center, New York, New York, United States of America
| | - Merina Varghese
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Prashant Vempati
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Anastasiya Dzhun
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Alice Cheng
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Jun Wang
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
- GRECC, James J Peters Veterans Affairs Medical Center, New York, New York, United States of America
| | - Dale Lange
- Hospital for Special Surgery, New York, New York, United States of America
| | - Amanda Bilski
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Irene Faravelli
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Giulio Maria Pasinetti
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
- GRECC, James J Peters Veterans Affairs Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
42
|
Corcia P, Gordon PH. Amyotrophic lateral sclerosis and the clinical potential of dexpramipexole. Ther Clin Risk Manag 2012; 8:359-66. [PMID: 22956874 PMCID: PMC3431958 DOI: 10.2147/tcrm.s21981] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder that leads to progressive weakness from loss of motor neurons and death on average in less than 3 years after symptom onset. No clear causes have been found and just one medication, riluzole, extends survival. Researchers have identified some of the cellular processes that occur after disease onset, including mitochondrial dysfunction, protein aggregation, oxidative stress, excitotoxicity, inflammation, and apoptosis. Mitochondrial disease may be a primary event in neurodegeneration or occur secondary to other cellular processes, and may itself contribute to oxidative stress, excitotoxicity, and apoptosis. Clinical trials currently aim to slow disease progression by testing drugs that impact one or more of these pathways. While every agent tested in the 18 years after the approval of riluzole has been ineffective, basic and clinical research methods in ALS have become dramatically more sophisticated. Dexpramipexole (RPPX), the R(+) enantiomer of pramiprexole, which is approved for symptomatic treatment of Parkinson disease, carries perhaps the currently largest body of pre-and early clinical data that support testing in ALS. The neuroprotective properties of RPPX in various models of neurodegeneration, including the ALS murine model, may be produced through protective actions on mitochondria. Early phase trials in human ALS suggest that the drug can be taken safely by patients in doses that provide neuroprotection in preclinical models. A Phase III trial to test the efficacy of RPPX in ALS is underway.
Collapse
Affiliation(s)
- Philippe Corcia
- Centre SLA, CHRU de Tours, Tours, France; UMR INSERM U930, Université François Rabelais de Tours (PC), Tours, France
| | | |
Collapse
|
43
|
Dunkel P, Chai CL, Sperlágh B, Huleatt PB, Mátyus P. Clinical utility of neuroprotective agents in neurodegenerative diseases: current status of drug development for Alzheimer's, Parkinson's and Huntington's diseases, and amyotrophic lateral sclerosis. Expert Opin Investig Drugs 2012; 21:1267-308. [PMID: 22741814 DOI: 10.1517/13543784.2012.703178] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION According to the definition of the Committee to Identify Neuroprotective Agents in Parkinson's Disease (CINAPS), "neuroprotection would be any intervention that favourably influences the disease process or underlying pathogenesis to produce enduring benefits for patients" [Meissner W, et al. Trends Pharmacol Sci 2004;25:249-253]. Preferably, neuroprotective agents should be used before or eventually during the prodromal phase of the diseases that could start decades before the appearance of symptoms. Although several symptomatic drugs are available, a disease-modifying agent is still elusive. AREAS COVERED The aim of the present review is to give an overview of neuroprotective agents being currently investigated for the treatment of AD, PD, HD and ALS in clinical phases. EXPERT OPINION Development of effective neuroprotective therapies resulting in clinically meaningful results is hampered by several factors in all research stages, both conceptual and methodological. Novel solutions might be offered by evaluation of new targets throughout clinical studies, therapies emerging from drug repositioning approaches, multi-target approaches and network pharmacology.
Collapse
Affiliation(s)
- Petra Dunkel
- Semmelweis University, Department of Organic Chemistry, Budapest, Hungary
| | | | | | | | | |
Collapse
|