1
|
Joshi DC, Chavan MB, Gurow K, Gupta M, Dhaliwal JS, Ming LC. The role of mitochondrial dysfunction in Huntington's disease: Implications for therapeutic targeting. Biomed Pharmacother 2025; 183:117827. [PMID: 39854819 DOI: 10.1016/j.biopha.2025.117827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Huntington's disease (HD) is a progressive, autosomal dominant neurodegenerative disorder characterized by cognitive decline, motor dysfunction, and psychiatric disturbances. A common feature of neurodegenerative disorders is mitochondrial dysfunction, which affects the brain's sensitivity to oxidative damage and its high oxygen demand. This dysfunction may plays a significant role in the pathogenesis of Huntington's disease. HD is caused by a CAG repeat expansion in the huntingtin gene, which leads to the production of a toxic mutant huntingtin (mHTT) protein. This disruption in mitochondrial function compromises energy metabolism and increases oxidative stress, resulting in mitochondrial DNA abnormalities, impaired calcium homeostasis, and altered mitochondrial dynamics. These effects ultimately may contribute to neuronal dysfunction and cell death, underscoring the importance of targeting mitochondrial function in developing therapeutic strategies for HD. This review discusses the mechanistic role of mitochondrial dysfunction in Huntington's disease. Mitochondrial dysfunction is a crucial factor in HD, making mitochondrial-targeted therapies a promising approach for treatment. We explore therapies that address bioenergy deficits, antioxidants that reduce reactive oxygen species, calcium modulators that restore calcium homeostasis, and treatments that enhance mitochondrial dynamics to rejuvenate mitochondrial function. We also highlight innovative treatment approaches such as gene editing and stem cell therapy, which offer hope for more personalized strategies. In conclusion, understanding mitochondrial dysfunction in Huntington's disease may guide potential treatment strategies. Targeting this dysfunction may help to slow disease progression and enhance the quality of life for individuals affected by Huntington's disease.
Collapse
Affiliation(s)
- Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Dist., Ajmer, Rajasthan, India.
| | - Mayuri Bapu Chavan
- TMV's Lokmanya Tilak Institute of Pharmaceutical Sciences, Pune, Maharashtra, India.
| | - Kajal Gurow
- Department of Pharmacology, Gurukul Pharmacy college, Ranpur, Kota, Rajasthan, India
| | - Madhu Gupta
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India.
| | | | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia; Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (deemed to be University), Sawangi (M), Wardha, India.
| |
Collapse
|
2
|
Marupudi N, Xiong MP. Genetic Targets and Applications of Iron Chelators for Neurodegeneration with Brain Iron Accumulation. ACS BIO & MED CHEM AU 2024; 4:119-130. [PMID: 38911909 PMCID: PMC11191567 DOI: 10.1021/acsbiomedchemau.3c00066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 06/25/2024]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of neurodegenerative diseases that are typically caused by a monogenetic mutation, leading to development of disordered movement symptoms such as dystonia, hyperreflexia, etc. Brain iron accumulation can be diagnosed through MRI imaging and is hypothesized to be the cause of oxidative stress, leading to the degeneration of brain tissue. There are four main types of NBIA: pantothenate kinase-associated neurodegeneration (PKAN), PLA2G6-associated neurodegeneration (PLAN), mitochondrial membrane protein-associated neurodegeneration (MKAN), and beta-propeller protein-associated neurodegeneration (BPAN). There are no causative therapies for these diseases, but iron chelators have been shown to have potential toward treating NBIA. Three chelators are investigated in this Review: deferoxamine (DFO), desferasirox (DFS), and deferiprone (DFP). DFO has been investigated to treat neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD); however, dose-related toxicity in these studies, as well as in PKAN studies, have shown that the drug still requires more development before it can be applied toward NBIA cases. Iron chelation therapies other than the ones currently in clinical use have not yet reached clinical studies, but they may possess characteristics that would allow them to access the brain in ways that current chelators cannot. Intranasal formulations are an attractive dosage form to study for chelation therapy, as this method of delivery can bypass the blood-brain barrier and access the CNS. Gene therapy differs from iron chelation therapy as it is a causal treatment of the disease, whereas iron chelators only target the disease progression of NBIA. Because the pathophysiology of NBIA diseases is still unclear, future courses of action should be focused on causative treatment; however, iron chelation therapy is the current best course of action.
Collapse
Affiliation(s)
- Neharika Marupudi
- Department of Pharmaceutical
& Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602-2352, United States
| | - May P. Xiong
- Department of Pharmaceutical
& Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602-2352, United States
| |
Collapse
|
3
|
Xie Q, Hu X, Zhao X, Xiang Z, Chen Q, Xie Z, Wang H, Zhao Y, Cheng X, Wang C. Effects and mechanism of extracts rich in phenylpropanoids-polyacetylenes and polysaccharides from Codonopsis Radix on improving scopolamine-induced memory impairment of mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117106. [PMID: 37652198 DOI: 10.1016/j.jep.2023.117106] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is a progressive developmental neurodegenerative disease that primarily develops in old age. Memory impairment is an important manifestation of AD. It has been demonstrated that inflammation and oxidative stress are important mediators in the development and progression of AD. Codonopsis Radix (CR) has a long history of consumption, exhibiting lots of beneficial health effects, including anti-ageing, antioxidant, and anti-inflammatory properties. However, studies on the effects of CR on scopolamine-induced amnesia have rarely been reported. AIM OF THE STUDY The aim of this study was to investigate the ameliorative effect of macromolecular portion (polysaccharides, POL) and small molecule portion (fine extract rich in phenylpropanoids-polyacetylenes, EPP) from CR on improving scopolamine-induced memory impairment and to elucidate the potential mechanism of action. MATERIALS AND METHODS C57BL/6 mice were pretreated with EPP (0.2, 0.4, and 0.6 g/kg), POL (0.3, 0.6, and 0.9 g/kg), and donepezil (5 mg/kg) by gavage for 7 days, followed by intraperitoneal injection of scopolamine (1 mg/kg) to induce memory impairment. The 16S rRNA gene sequencing, histopathological, western blotting, and biochemical analysis (various biochemical markers and protein expressions related to cholinergic system, oxidative stress, and neuroinflammation) were performed to further elucidate the mechanism of action. Moreover, the acetylcholinesterase (AChE) inhibitory activities of POL, EPP, and its main compounds tangshenoside I, lobetyol, lobetyolin, and lobetyolinin were evaluated. RESULTS Experiments have confirmed that both POL and EPP from CR could improve scopolamine-induced spatial learning memory deficits. Both of them could regulate cholinergic function by inhibiting AChE and activating choline acetyltransferase (ChAT) activities. They also could enhance antioxidant defense via increasing the activities of superoxide dismutase and glutathione peroxidase, and anti-inflammatory function through suppressing inflammatory factors (nitric oxide, TNF-α, and IL-6) and regulating gut flora. Besides, in vitro experiments demonstrated that four monomeric compounds and EPP, except POL, exhibited inhibition of AChE activity. CONCLUSION EPP and POL from CR exert a beneficial effect on learning and memory processes in mice with scopolamine-induced memory impairment. CR may be a promising medicine for preventing and improving learning memory.
Collapse
Affiliation(s)
- Qi Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Xianrun Hu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Xiang Zhao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Zedong Xiang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Qianping Chen
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Zhejun Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Hanxue Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Yonglin Zhao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Xuemei Cheng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
4
|
Cha J, Kim HS, Kwon G, Cho SY, Kim JM. Acute effects of (-)-gallocatechin gallate-rich green tea extract on the cerebral hemodynamic response of the prefrontal cortex in healthy humans. FRONTIERS IN NEUROERGONOMICS 2023; 4:1136362. [PMID: 38234497 PMCID: PMC10790935 DOI: 10.3389/fnrgo.2023.1136362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 11/01/2023] [Indexed: 01/19/2024]
Abstract
Objective The benefits of long-term consumption of green tea on the brain are well known. However, among many ingredients of green tea, the acute effects of (-)-gallocatechin gallate-rich green tea extract (GCG-GTE), have received comparatively less attention. Herein, we investigated the acute effects of oral ingestion of green tea with GCG-GTE, which contains close replicas of the ingredients of hot green tea, on task-dependent hemodynamics in the prefrontal cortex of healthy adult human brains. Methods In this randomized, double-blind, placebo-controlled, parallel group trial, 35 healthy adults completed computerized cognitive tasks that demand activation of the prefrontal cortex at baseline and 1 h after consumption of placebo and 900 mg of GCG-GTE extract supplement. During cognitive testing, hemodynamic responses (change in HbO2 concentration) in the prefrontal cortex were assessed using functional near-infrared spectroscopy (fNIRS). Results In fNIRS data, significant group x session interactions were found in the left (p = 0.035) and right (p = 0.036) dorsolateral prefrontal cortex (DLPFC). In behavioral data, despite the numerical increase in the GCG-GTE group and the numerical decrease in the Placebo group, no significant differences were observed in the cognitive performance measure between the groups. Conclusion The result suggests a single dose of orally administered GCG-GTE can reduce DLPFC activation in healthy humans even with increased task demand. GCG-GTE is a promising functional material that can affect neural efficiency to lower mental workload during cognitively demanding tasks. However, further studies are needed to verify this.
Collapse
Affiliation(s)
- Jihyun Cha
- Department of Research and Development, OBELAB Inc., Seoul, Republic of Korea
| | - Hyung-Su Kim
- Amorepacific R&I Center, Yongin-si, Republic of Korea
| | - Gusang Kwon
- Amorepacific R&I Center, Yongin-si, Republic of Korea
| | - Si-Young Cho
- Amorepacific R&I Center, Yongin-si, Republic of Korea
| | - Jae-Myoung Kim
- Department of Research and Development, OBELAB Inc., Seoul, Republic of Korea
| |
Collapse
|
5
|
Martínez-Iglesias O, Naidoo V, Carrera I, Corzo L, Cacabelos R. Natural Bioactive Products as Epigenetic Modulators for Treating Neurodegenerative Disorders. Pharmaceuticals (Basel) 2023; 16:216. [PMID: 37259364 PMCID: PMC9967112 DOI: 10.3390/ph16020216] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 08/27/2023] Open
Abstract
Neurodegenerative disorders (NDDs) are major health issues in Western countries. Despite significant efforts, no effective therapeutics for NDDs exist. Several drugs that target epigenetic mechanisms (epidrugs) have been recently developed for the treatment of NDDs, and several of these are currently being tested in clinical trials. Furthermore, various bioproducts have shown important biological effects for the potential prevention and treatment of these disorders. Here, we review the use of natural products as epidrugs to treat NDDs in order to explore the epigenetic effects and benefits of functional foods and natural bioproducts on neurodegeneration.
Collapse
Affiliation(s)
- Olaia Martínez-Iglesias
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, 15165 Bergondo, Corunna, Spain
| | | | | | | | | |
Collapse
|
6
|
Lange KW, Lange KM, Nakamura Y. Green tea, epigallocatechin gallate and the prevention of Alzheimer’s disease: Clinical evidence. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
7
|
Dietary and nutraceutical-based therapeutic approaches to combat the pathogenesis of Huntington’s disease. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
|
8
|
Prifti E, Tsakiri EN, Vourkou E, Stamatakis G, Samiotaki M, Skoulakis EMC, Papanikolopoulou K. Mical modulates Tau toxicity via cysteine oxidation in vivo. Acta Neuropathol Commun 2022; 10:44. [PMID: 35379354 PMCID: PMC8981811 DOI: 10.1186/s40478-022-01348-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/18/2022] [Indexed: 12/24/2022] Open
Abstract
Tau accumulation is clearly linked to pathogenesis in Alzheimer’s disease and other Tauopathies. However, processes leading to Tau fibrillization and reasons for its pathogenicity remain largely elusive. Mical emerged as a novel interacting protein of human Tau expressed in Drosophila brains. Mical is characterized by the presence of a flavoprotein monooxygenase domain that generates redox potential with which it can oxidize target proteins. In the well-established Drosophila Tauopathy model, we use genetic interactions to show that Mical alters Tau interactions with microtubules and the Actin cytoskeleton and greatly affects Tau aggregation propensity and Tau-associated toxicity and dysfunction. Exploration of the mechanism was pursued using a Mical inhibitor, a mutation in Mical that selectively disrupts its monooxygenase domain, Tau transgenes mutated at cysteine residues targeted by Mical and mass spectrometry analysis to quantify cysteine oxidation. The collective evidence strongly indicates that Mical’s redox activity mediates the effects on Tau via oxidation of Cys322. Importantly, we also validate results from the fly model in human Tauopathy samples by showing that MICAL1 is up-regulated in patient brains and co-localizes with Tau in Pick bodies. Our work provides mechanistic insights into the role of the Tau cysteine residues as redox-switches regulating the process of Tau self-assembly into inclusions in vivo, its function as a cytoskeletal protein and its effect on neuronal toxicity and dysfunction.
Collapse
|
9
|
Roham PH, Save SN, Sharma S. Human islet amyloid polypeptide: A therapeutic target for the management of type 2 diabetes mellitus. J Pharm Anal 2022; 12:556-569. [PMID: 36105173 PMCID: PMC9463490 DOI: 10.1016/j.jpha.2022.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 12/22/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) and other metabolic disorders are often silent and go unnoticed in patients because of the lack of suitable prognostic and diagnostic markers. The current therapeutic regimens available for managing T2DM do not reverse diabetes; instead, they delay the progression of diabetes. Their efficacy (in principle) may be significantly improved if implemented at earlier stages. The misfolding and aggregation of human islet amyloid polypeptide (hIAPP) or amylin has been associated with a gradual decrease in pancreatic β-cell function and mass in patients with T2DM. Hence, hIAPP has been recognized as a therapeutic target for managing T2DM. This review summarizes hIAPP's role in mediating dysfunction and apoptosis in pancreatic β-cells via induction of endoplasmic reticulum stress, oxidative stress, mitochondrial dysfunction, inflammatory cytokine secretion, autophagy blockade, etc. Furthermore, it explores the possibility of using intermediates of the hIAPP aggregation pathway as potential drug targets for T2DM management. Finally, the effects of common antidiabetic molecules and repurposed drugs; other hIAPP mimetics and peptides; small organic molecules and natural compounds; nanoparticles, nanobodies, and quantum dots; metals and metal complexes; and chaperones that have demonstrated potential to inhibit and/or reverse hIAPP aggregation and can, therefore, be further developed for managing T2DM have been discussed. Misfolded species of hIAPP form toxic oligomers in pancreatic β-cells. hIAPP amyloids has been detected in the pancreas of about 90% subjects with T2DM. Inhibitors of hIAPP aggregation can help manage T2DM.
Collapse
|
10
|
Nurkenov T, Tsoy A, Olzhayev F, Abzhanova E, Turgambayeva A, Zhussupova A, Avula B, Ross S, Aituarova A, Kassymova D, Zhusupova G, Shalakhmetova T, Tokay T, Lee JC, Askarova S. Plant Extract of Limonium gmelinii Attenuates Oxidative Responses in Neurons, Astrocytes, and Cerebral Endothelial Cells In Vitro and Improves Motor Functions of Rats after Middle Cerebral Artery Occlusion. Antioxidants (Basel) 2021; 10:antiox10111814. [PMID: 34829685 PMCID: PMC8614848 DOI: 10.3390/antiox10111814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
There are numerous publications demonstrating that plant polyphenols can reduce oxidative stress and inflammatory processes in the brain. In the present study we have investigated the neuroprotective effect of plant extract isolated from the roots of L. gmelinii since it contains a rich source of polyphenols and other biologically active compounds. We have applied an oxidative and inflammatory model induced by NMDA, H2O2, and TNF-α in human primary neurons and astrocytes, and mouse cerebral endothelial cell (CECs) line in vitro. The levels of ROS generation, NADPH oxidase activation, P-selectin expression, and activity of ERK1/2 were evaluated by quantitative immunofluorescence analysis, confocal microscopy, and MAPK assay. In vivo, sensorimotor functions in rats with middle cerebral artery occlusion (MCAO) were assessed. In neurons NMDA induced overproduction of ROS, in astrocytes TNF-α initiated ROS generation, NADPH oxidase activation, and phosphorylation of ERK1/2. In CECs, the exposure by TNF-α induced oxidative stress and triggered the accumulation of P-selectin on the surface of the cells. In turn, pre-treatment of the cells with the extract of L. gmelinii suppressed oxidative stress in all cell types and pro-inflammatory responses in astrocytes and CECs. In vivo, the treatment with L. gmelinii extract improved motor activity in rats with MCAO.
Collapse
Affiliation(s)
- Tulendy Nurkenov
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (T.N.); (A.Z.); (A.A.); (D.K.); (G.Z.); (T.S.)
- Department of Biology, Institute of Natural Science and Geography, Abai Kazakh National Pedagogical University, Almaty 050010, Kazakhstan
| | - Andrey Tsoy
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.T.); (F.O.); (E.A.); (A.T.)
| | - Farkhad Olzhayev
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.T.); (F.O.); (E.A.); (A.T.)
| | - Elvira Abzhanova
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.T.); (F.O.); (E.A.); (A.T.)
| | - Anel Turgambayeva
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.T.); (F.O.); (E.A.); (A.T.)
| | - Aizhan Zhussupova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (T.N.); (A.Z.); (A.A.); (D.K.); (G.Z.); (T.S.)
| | - Bharathi Avula
- School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (S.R.)
| | - Samir Ross
- School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (S.R.)
| | - Aigerim Aituarova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (T.N.); (A.Z.); (A.A.); (D.K.); (G.Z.); (T.S.)
| | - Dariya Kassymova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (T.N.); (A.Z.); (A.A.); (D.K.); (G.Z.); (T.S.)
| | - Galiya Zhusupova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (T.N.); (A.Z.); (A.A.); (D.K.); (G.Z.); (T.S.)
| | - Tamara Shalakhmetova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (T.N.); (A.Z.); (A.A.); (D.K.); (G.Z.); (T.S.)
| | - Tursonjan Tokay
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - James C. Lee
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Sholpan Askarova
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.T.); (F.O.); (E.A.); (A.T.)
- Correspondence:
| |
Collapse
|
11
|
Goyal S, Seth B, Chaturvedi RK. Polyphenols and Stem Cells for Neuroregeneration in Parkinson's Disease and Amyotrophic Lateral Sclerosis. Curr Pharm Des 2021; 28:806-828. [PMID: 34781865 DOI: 10.2174/1381612827666211115154450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/02/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS) are neurological disorders, pathologically characterized by chronic degeneration of dopaminergic neurons and motor neurons respectively. There is still no cure or effective treatment against the disease progression and most of the treatments are symptomatic. The present review offers an overview of the different factors involved in the pathogenesis of these diseases. Subsequently, we focused on the recent advanced studies of dietary polyphenols and stem cell therapies, which have made it possible to slow down the progression of neurodegeneration. To date, stem cells and different polyphenols have been used for the directional induction of neural stem cells into dopaminergic neurons and motor neurons. We have also discussed their involvement in the modulation of different signal transduction pathways and growth factor levels in various in vivo and in vitro studies. Likewise stem cells, polyphenols also exhibit the potential of neuroprotection by their anti-apoptotic, anti-inflammatory, anti-oxidant properties regulating the growth factors levels and molecular signaling events. Overall this review provides a detailed insight into recent strategies that promise the use of polyphenol with stem cell therapy for the possible treatment of PD and ALS.
Collapse
Affiliation(s)
- Shweta Goyal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001. India
| | - Brashket Seth
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001. India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001. India
| |
Collapse
|
12
|
Samec M, Liskova A, Koklesova L, Zhai K, Varghese E, Samuel SM, Šudomová M, Lucansky V, Kassayova M, Pec M, Biringer K, Brockmueller A, Kajo K, Hassan STS, Shakibaei M, Golubnitschaja O, Büsselberg D, Kubatka P. Metabolic Anti-Cancer Effects of Melatonin: Clinically Relevant Prospects. Cancers (Basel) 2021; 13:3018. [PMID: 34208645 PMCID: PMC8234897 DOI: 10.3390/cancers13123018] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic reprogramming characterized by alterations in nutrient uptake and critical molecular pathways associated with cancer cell metabolism represents a fundamental process of malignant transformation. Melatonin (N-acetyl-5-methoxytryptamine) is a hormone secreted by the pineal gland. Melatonin primarily regulates circadian rhythms but also exerts anti-inflammatory, anti-depressant, antioxidant and anti-tumor activities. Concerning cancer metabolism, melatonin displays significant anticancer effects via the regulation of key components of aerobic glycolysis, gluconeogenesis, the pentose phosphate pathway (PPP) and lipid metabolism. Melatonin treatment affects glucose transporter (GLUT) expression, glucose-6-phosphate dehydrogenase (G6PDH) activity, lactate production and other metabolic contributors. Moreover, melatonin modulates critical players in cancer development, such as HIF-1 and p53. Taken together, melatonin has notable anti-cancer effects at malignancy initiation, progression and metastasing. Further investigations of melatonin impacts relevant for cancer metabolism are expected to create innovative approaches supportive for the effective prevention and targeted therapy of cancers.
Collapse
Affiliation(s)
- Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.); (K.B.)
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.); (K.B.)
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.); (K.B.)
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (K.Z.); (E.V.); (S.M.S.)
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (K.Z.); (E.V.); (S.M.S.)
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (K.Z.); (E.V.); (S.M.S.)
| | - Miroslava Šudomová
- Museum of Literature in Moravia, Klašter 1, 66461 Rajhrad, Czech Republic;
| | - Vincent Lucansky
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia;
| | - Monika Kassayova
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P. J. Šafarik University, 04001 Košice, Slovakia;
| | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.); (K.B.)
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Karol Kajo
- Department of Pathology, St. Elizabeth Cancer Institute Hospital, 81250 Bratislava, Slovakia;
- Biomedical Research Centre, Slovak Academy of Sciences, 81439 Bratislava, Slovakia
| | - Sherif T. S. Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Czech Republic;
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Olga Golubnitschaja
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1160 Brussels, Belgium;
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (K.Z.); (E.V.); (S.M.S.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1160 Brussels, Belgium;
| |
Collapse
|
13
|
Bellmann-Strobl J, Paul F, Wuerfel J, Dörr J, Infante-Duarte C, Heidrich E, Körtgen B, Brandt A, Pfüller C, Radbruch H, Rust R, Siffrin V, Aktas O, Heesen C, Faiss J, Hoffmann F, Lorenz M, Zimmermann B, Groppa S, Wernecke KD, Zipp F. Epigallocatechin Gallate in Relapsing-Remitting Multiple Sclerosis: A Randomized, Placebo-Controlled Trial. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:e981. [PMID: 33762428 PMCID: PMC8054966 DOI: 10.1212/nxi.0000000000000981] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/07/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To assess the safety and efficacy of epigallocatechin-3-gallate (EGCG) add-on to glatiramer acetate (GA) in patients with relapsing-remitting multiple sclerosis (RRMS). METHODS We enrolled patients with RRMS (aged 18-60 years, Expanded Disability Status Scale [EDSS] score 0-6.5), receiving stable GA treatment in a multicenter, prospective, double-blind, phase II, randomized controlled trial. Participants received up to 800 mg oral EGCG daily over a period of 18 months. The primary outcome was the proportion of patients without new hyperintense lesions on T2-weighted (T2w) brain MRI within 18 months. Secondary end points included additional MRI and clinical parameters. Immunologic effects of EGCG were investigated in exploratory experiments. RESULTS A total of 122 patients on GA were randomly assigned to EGCG treatment (n = 62) or placebo (n = 60). We could not demonstrate a difference between groups after 18 months for the primary outcome or other radiologic (T2w lesion volume, T1w hypointense lesion number or volume, number of cumulative contrast-enhancing lesions, percent brain volume change), or clinical (EDSS, MS functional composite, and annualized relapse rate) parameter. EGCG treatment did not affect immune response to GA. Pharmacologic analysis revealed wide ranging EGCG plasma levels. The treatment was well tolerated with a similar incidence of mostly mild adverse events similar in both groups. CONCLUSION In RRMS, oral EGCG add-on to GA was not superior to placebo in influencing MRI and clinical disease activity over 18 months. The treatment was safe at a daily dosage up to 800 mg EGCG. It did not influence immune parameters, despite indication of EGCG being bioavailable in patients. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that for patients with RRMS, EGCG added to GA did not significantly affect the development of new hyperintense lesions on T2-weighted brain MRI. TRIAL REGISTRATION INFORMATION Clinical trial registration number: NCT00525668.
Collapse
Affiliation(s)
- Judith Bellmann-Strobl
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin.
| | - Friedemann Paul
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Jens Wuerfel
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Jan Dörr
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Carmen Infante-Duarte
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Elmira Heidrich
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Benedict Körtgen
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Alexander Brandt
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Caspar Pfüller
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Helena Radbruch
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Rebekka Rust
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Volker Siffrin
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Orhan Aktas
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Christoph Heesen
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Jürgen Faiss
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Frank Hoffmann
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Mario Lorenz
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Benno Zimmermann
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Sergiu Groppa
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Klaus-Dieter Wernecke
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| | - Frauke Zipp
- From the NeuroCure Clinical Research Center (J.B.-S., F.P., J.D., A.B., V.S.), Charité-Universitätsmedizin Berlin; Medical Image Analysis Center (J.W.), University Basel; Institut for Medical Immunology (C.I.-D., E.H.), Charité-Universitätsmedizin Berlin; Department of Neurology and Neuroimaging Center (B.K.), Johannes Gutenberg University, Mainz; Charité-Universitätsmedizin Berlin (C.P.); NeuroCure Clinical Research Center (H.R., R.R.), Charité-Universitätsmedizin Berlin, Germany; Department of Neurology (O.A.), Medical Faculty, Heinrich Heine University Düsseldorf; Institut für Neuroimmunologie und Multiple Sklerose (C.H.), Universitätsklinikum Hamburg-Eppendorf, Hamburg; Klinik für Neurologie (J.F.), Asklepios Klinik Lübben/Teupitz; Department of Neurology (F.H.), Krankenhaus Martha-Maria Halle-Dölau, Halle/Saale; Medizinische Klinik für Kardiologie und Angiologie (M.L.), Campus Mitte, Charité-Universitätsmedizin Berlin; Institute of Nutritional and Food Sciences (B.Z.), University of Bonn; Department of Neurology and Neuroimaging Center (NIC) (S.G., F.Z.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz; and Charité-Universitätsmedizin Berlin and SOSTANA GmbH (K.-D.W.), Berlin
| |
Collapse
|
14
|
Rust R, Chien C, Scheel M, Brandt AU, Dörr J, Wuerfel J, Klumbies K, Zimmermann H, Lorenz M, Wernecke KD, Bellmann-Strobl J, Paul F. Epigallocatechin Gallate in Progressive MS: A Randomized, Placebo-Controlled Trial. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:e964. [PMID: 33622766 PMCID: PMC7954462 DOI: 10.1212/nxi.0000000000000964] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/17/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To examine whether treatment with epigallocatechin gallate (EGCG) influences progression of brain atrophy, reduces clinical and further radiologic disease activity markers, and is safe in patients with progressive multiple sclerosis (PMS). METHODS We enrolled 61 patients with primary or secondary PMS in a randomized double-blind, parallel-group, phase II trial on oral EGCG (up to 1,200 mg daily) or placebo for 36 months with an optional open-label EGCG treatment extension (OE) of 12-month duration. The primary end point was the rate of brain atrophy, quantified as brain parenchymal fraction (BPF). The secondary end points were radiologic and clinical disease parameters and safety assessments. RESULTS In our cohort, 30 patients were randomized to EGCG treatment and 31 to placebo. Thirty-eight patients (19 from each group) completed the study. The primary endpoint was not met, as in 36 months the rate of decrease in BPF was 0.0092 ± 0.0152 in the treatment group and -0.0078 ± 0.0159 in placebo-treated patients. None of the secondary MRI and clinical end points revealed group differences. Adverse events of EGCG were mostly mild and occurred with a similar incidence in the placebo group. One patient in the EGCG group had to stop treatment due to elevated aminotransferases (>3.5 times above normal limit). CONCLUSIONS In a phase II trial including patients with multiple sclerosis (MS) with progressive disease course, we were unable to demonstrate a treatment effect of EGCG on the primary and secondary radiologic and clinical disease parameters while confirming on overall beneficial safety profile. CLINICALTRIALGOV IDENTIFIER NCT00799890. CLASSIFICATION OF EVIDENCE This phase II trial provides Class II evidence that for patients with PMS, EGCG was safe, well tolerated, and did not significantly reduce the rate of brain atrophy.
Collapse
Affiliation(s)
- Rebekka Rust
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Claudia Chien
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Michael Scheel
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Alexander U Brandt
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Jan Dörr
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Jens Wuerfel
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Katharina Klumbies
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Hanna Zimmermann
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Mario Lorenz
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Klaus-Dieter Wernecke
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Judith Bellmann-Strobl
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland
| | - Friedemann Paul
- From the Charité - Universitätsmedizin Berlin (R.R., C.C., M.S., A.U.B., J.D., K.K., H.Z., M.L., K.-D.W., J.B.-S., F.P.), Berlin, Germany; and Jens Würfel, University Basel, Basel, Switzerland.
| |
Collapse
|
15
|
Klumbies K, Rust R, Dörr J, Konietschke F, Paul F, Bellmann-Strobl J, Brandt AU, Zimmermann HG. Retinal Thickness Analysis in Progressive Multiple Sclerosis Patients Treated With Epigallocatechin Gallate: Optical Coherence Tomography Results From the SUPREMES Study. Front Neurol 2021; 12:615790. [PMID: 33995239 PMCID: PMC8113620 DOI: 10.3389/fneur.2021.615790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/25/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Epigallocatechin gallate (EGCG) is an anti-inflammatory agent and has proven neuroprotective properties in animal models of multiple sclerosis (MS). Optical coherence tomography (OCT) assessed retinal thickness analysis can reflect treatment responses in MS. Objective: To analyze the influence of EGCG treatment on retinal thickness analysis as secondary and exploratory outcomes of the randomized controlled Sunphenon in Progressive Forms of MS trial (SUPREMES, NCT00799890). Methods: SUPREMES patients underwent OCT with the Heidelberg Spectralis device at a subset of visits. We determined peripapillary retinal nerve fiber layer (pRNFL) thickness from a 12° ring scan around the optic nerve head and thickness of the ganglion cell/inner plexiform layer (GCIP) and inner nuclear layer (INL) within a 6 mm diameter grid centered on the fovea from a macular volume scan. Longitudinal OCT data were available for exploratory analysis from 31 SUPREMES participants (12/19 primary/secondary progressive MS (PPMS/SPMS); mean age 51 ± 7 years; 12 female; mean time since disease onset 16 ± 11 years). We tested the null hypothesis of no treatment*time interaction using nonparametric analysis of longitudinal data in factorial experiments. Results: After 2 years, there were no significant differences in longitudinal retinal thickness changes between EGCG treated and placebo arms in any OCT parameter (Mean change [confidence interval] ECGC vs. Placebo: pRNFL: -0.83 [1.29] μm vs. -0.64 [1.56] μm, p = 0.156; GCIP: -0.67 [0.67] μm vs. -0.14 [0.47] μm, p = 0.476; INL: -0.06 [0.58] μm vs. 0.22 [0.41] μm, p = 0.455). Conclusion: Retinal thickness analysis did not reveal a neuroprotective effect of EGCG. While this is in line with the results of the main SUPREMES trial, our study was probably underpowered to detect an effect. Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT00799890.
Collapse
Affiliation(s)
- Katharina Klumbies
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rebekka Rust
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jan Dörr
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Neurology Department, Oberhavel Clinic, Hennigsdorf, Germany
| | - Frank Konietschke
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Judith Bellmann-Strobl
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander U Brandt
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Neurology, University of California, Irvine, Irvine, CA, United States
| | - Hanna G Zimmermann
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
16
|
Singh P, Sivanandam TM, Konar A, Thakur MK. Role of nutraceuticals in cognition during aging and related disorders. Neurochem Int 2020; 143:104928. [PMID: 33285273 DOI: 10.1016/j.neuint.2020.104928] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/05/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
Abstract
Cognitive abilities are compromised with advancing age posing a great risk for the development of dementia and other related brain disorders. Genetic susceptibility as well as environmental exposures determine the fate of cognitive aging and its transition to pathological states. Emerging epidemiological and observational studies have revealed the importance of lifestyle factors including dietary patterns and nutritional intake in the maintenance of cognitive health and reducing the risk of neurodegenerative disorders. In this context, nutraceutical interventions have gained considerable attention in preventing age-related cognitive deficits and counteracting pathological processes. Nutraceuticals include dietary plants and derivatives, food supplements and processed foods with nutritional and pharmaceutical values. The present review highlights the importance of nutraceuticals in attenuating cognitive aging and its progression to dementia, with specific emphasis on chemical constituents, neurocognitive properties and mechanism of action.
Collapse
Affiliation(s)
- Padmanabh Singh
- Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Thamil Mani Sivanandam
- Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Arpita Konar
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India.
| | - M K Thakur
- Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
17
|
Olajide OA, Sarker SD. Alzheimer's disease: natural products as inhibitors of neuroinflammation. Inflammopharmacology 2020; 28:1439-1455. [PMID: 32930914 PMCID: PMC7572326 DOI: 10.1007/s10787-020-00751-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/28/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer’s disease (AD) is the most common form of dementia and affects 44 million people worldwide. New emerging evidence from pre-clinical and clinical investigations shows that neuroinflammation is a major pathological component of AD suggesting that anti-inflammatory strategies are important in delaying the onset or slowing the progression of the disease. However, efforts to employ current anti-inflammatory agents in AD clinical trials have produced limited success. Consequently, there is a need to explore anti-inflammatory natural products, which target neuroinflammatory pathways relevant to AD pathogenesis. This review summarises important druggable molecular targets of neuroinflammation and presents classes of anti-neuroinflammatory natural products with potentials for preventing and reducing symptoms of AD.
Collapse
Affiliation(s)
- Olumayokun A Olajide
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK.
| | - Satyajit D Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool, L3 3AF, UK
| |
Collapse
|
18
|
Khan H, Ullah H, Tundis R, Belwal T, Devkota HP, Daglia M, Cetin Z, Saygili EI, Campos MDG, Capanoglu E, Du M, Dar P, Xiao J. Dietary Flavonoids in the Management of Huntington’s Disease: Mechanism and Clinical Perspective. EFOOD 2020. [DOI: 10.2991/efood.k.200203.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
19
|
|
20
|
Dorababu A. Critical evaluation of current Alzheimer's drug discovery (2018-19) & futuristic Alzheimer drug model approach. Bioorg Chem 2019; 93:103299. [PMID: 31586701 DOI: 10.1016/j.bioorg.2019.103299] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/14/2019] [Accepted: 09/16/2019] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD), a neurodegenerative disease responsible for death of millions of people worldwide is a progressive clinical disorder which causes neurons to degenerate and ultimately die. It is one of the common causes of dementia wherein a person's incapability to independently think, behave and decline in social skills can be quoted as major symptoms. However the early signs include the simple non-clinical symptoms such as forgetting recent events and conversations. Onset of these symptoms leads to worsened conditions wherein the AD patient suffers severe memory impairment and eventually becomes unable to work out everyday tasks. Even though there is no complete cure for AD, rigorous research has been going on to reduce the progress of AD. Currently, a very few clinical drugs are prevailing for AD treatment. So this is the need of hour to design, develop and discovery of novel anti-AD drugs. The main factors for the cause of AD according to scientific research reveals structural changes in brain proteins such as beta amyloid, tau proteins into plaques and tangles respectively. The abnormal proteins distort the neurons. Despite the high potencies of the synthesized molecules; they could not get on the clinical tests up to human usage. In this review article, the recent research carried out with respect to inhibition of AChE, BuChE, NO, BACE1, MAOs, Aβ, H3R, DAPK, CSF1R, 5-HT4R, PDE, σ1R and GSK-3β is compiled and organized. The summary is focused mainly on cholinesterases, Aβ, BACE1 and MAOs classes of potential inhibitors. The review also covers structure activity relationship of most potent compounds of each class of inhibitors alongside redesign and remodeling of the most significant inhibitors in order to expect cutting edge inhibitory properties towards AD. Alongside the molecular docking studies of the some final compounds are discussed.
Collapse
Affiliation(s)
- Atukuri Dorababu
- Department of Studies in Chemistry, SRMPP Govt. First Grade College, Huvinahadagali 583219, Karnataka, India.
| |
Collapse
|
21
|
Complex of EGCG with Cu(II) Suppresses Amyloid Aggregation and Cu(II)-Induced Cytotoxicity of α-Synuclein. Molecules 2019; 24:molecules24162940. [PMID: 31416122 PMCID: PMC6719089 DOI: 10.3390/molecules24162940] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 12/14/2022] Open
Abstract
Accumulation of α-synuclein (α-Syn) is a remarkable pathology for Parkinson’s disease (PD), therefore clearing it is possibly a promising strategy for treating PD. Aberrant copper (Cu(II)) homeostasis and oxidative stress play critical roles in the abnormal aggregation of α-Syn in the progress of PD. It is reported that the polyphenol (−)-epi-gallocatechin gallate (EGCG) can inhibit α-Syn fibrillation and aggregation, disaggregate α-Syn mature fibrils, as well as protect α-Syn overexpressed-PC12 cells against damage. Also, previous studies have reported that EGCG can chelate many divalent metal ions. What we investigate here is whether EGCG can interfere with the Cu(II) induced fibrillation of α-Syn and protect the cell viability. In this work, on a molecular and cellulaire basis, we demonstrated that EGCG can form a Cu(II)/EGCG complex, leading to the inhibition of Cu(II)-induced conformation transition of α-Syn from random coil to β-sheet, which is a dominant structure in α-Syn fibrils and aggregates. Moreover, we found that the mixture of Cu(II) and EGCG in a molar ratio from 0.5 to 2 can efficiently inhibit this process. Furthermore, we demonstrated that in the α-Syn transduced-PC12 cells, EGCG can inhibit the overexpression and fibrillation of α-Syn in the cells, and reduce Cu(II)-induced reactive oxygen species (ROS), protecting the cells against Cu(II)-mediated toxicity.
Collapse
|
22
|
Kevadiya BD, Ottemann BM, Thomas MB, Mukadam I, Nigam S, McMillan J, Gorantla S, Bronich TK, Edagwa B, Gendelman HE. Neurotheranostics as personalized medicines. Adv Drug Deliv Rev 2019; 148:252-289. [PMID: 30421721 PMCID: PMC6486471 DOI: 10.1016/j.addr.2018.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022]
Abstract
The discipline of neurotheranostics was forged to improve diagnostic and therapeutic clinical outcomes for neurological disorders. Research was facilitated, in largest measure, by the creation of pharmacologically effective multimodal pharmaceutical formulations. Deployment of neurotheranostic agents could revolutionize staging and improve nervous system disease therapeutic outcomes. However, obstacles in formulation design, drug loading and payload delivery still remain. These will certainly be aided by multidisciplinary basic research and clinical teams with pharmacology, nanotechnology, neuroscience and pharmaceutic expertise. When successful the end results will provide "optimal" therapeutic delivery platforms. The current report reviews an extensive body of knowledge of the natural history, epidemiology, pathogenesis and therapeutics of neurologic disease with an eye on how, when and under what circumstances neurotheranostics will soon be used as personalized medicines for a broad range of neurodegenerative, neuroinflammatory and neuroinfectious diseases.
Collapse
Affiliation(s)
- Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brendan M Ottemann
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Midhun Ben Thomas
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saumya Nigam
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn McMillan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
23
|
Avenanthramide-C Restores Impaired Plasticity and Cognition in Alzheimer's Disease Model Mice. Mol Neurobiol 2019; 57:315-330. [PMID: 31332763 DOI: 10.1007/s12035-019-01707-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/10/2019] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline and dementia with no effective treatment. Here, we investigated a novel compound from oats named avenanthramide-C (Avn-C), on AD-related memory impairment and behavioral deficits in transgenic mouse models. Acute hippocampal slices of wild-type or AD transgenic mice were treated with Avn-C in the presence or absence of oligomeric Aβ42. LTP analyses and immunoblotting were performed to assess the effect of Avn-C on Aβ-induced memory impairment. To further investigate the effect of Avn-C on impaired memory and Aβ pathology, two different AD transgenic mice (Tg2576 and 5XFAD) models were orally treated with either Avn-C or vehicle for 2 weeks. They were then assessed for the effect of the treatment on neuropathologies and behavioral impairments. Avn-C reversed impaired LTP in both ex vivo- and in vivo-treated AD mice hippocampus. Oral administration (6 mg/kg per day) for 2 weeks in AD mice leads to improved recognition and spatial memory, reduced caspase-3 cleavage, reversed neuroinflammation, and to accelerated glycogen synthase kinase-3β (pS9GSK-3β) and interleukin (IL-10) levels. Avn-C exerts its beneficial effects by binding to α1A adrenergic receptors to stimulate adenosine monophosphate-activated kinase (AMPK). All of the beneficial effects of Avn-C on LTP retrieval could be blocked by prazosin hydrochloride, a specific inhibitor of α1A adrenergic receptors. Our findings provide evidence, for the first time, that oats' Avn-C reverses the AD-related memory and behavioral impairments, and establish it as a potential candidate for Alzheimer's disease drug development.
Collapse
|
24
|
Levin J, Maaß S, Schuberth M, Giese A, Oertel WH, Poewe W, Trenkwalder C, Wenning GK, Mansmann U, Südmeyer M, Eggert K, Mollenhauer B, Lipp A, Löhle M, Classen J, Münchau A, Kassubek J, Gandor F, Berg D, Egert-Schwender S, Eberhardt C, Paul F, Bötzel K, Ertl-Wagner B, Huppertz HJ, Ricard I, Höglinger GU. Safety and efficacy of epigallocatechin gallate in multiple system atrophy (PROMESA): a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2019; 18:724-735. [PMID: 31278067 DOI: 10.1016/s1474-4422(19)30141-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 03/24/2019] [Accepted: 03/27/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Multiple system atrophy is a rare neurodegenerative disease characterised by aggregation of α-synuclein in oligodendrocytes and neurons. The polyphenol epigallocatechin gallate inhibits α-synuclein aggregation and reduces associated toxicity. We aimed to establish if epigallocatechin gallate could safely slow disease progression in patients with multiple system atrophy. METHODS We did a randomised, double-blind, parallel group, placebo-controlled clinical trial at 12 specialist centres in Germany. Eligible participants were older than 30 years; met consensus criteria for possible or probable multiple system atrophy and could ambulate independently (ie, were at Hoehn and Yahr stages 1-3); and were on stable anti-Parkinson's, anti-dysautonomia, anti-dementia, and anti-depressant regimens (if necessary) for at least 1 month. Participants were randomly assigned (1:1) to epigallocatechin gallate or placebo (mannitol) via a web-generated permuted blockwise randomisation list (block size=2) that was stratified by disease subtype (parkinsonism-predominant disease vs cerebellar-ataxia-predominant disease). All participants and study personnel were masked to treatment assignment. Participants were given one hard gelatin capsule (containing either 400 mg epigallocatechin gallate or mannitol) orally once daily for 4 weeks, then one capsule twice daily for 4 weeks, and then one capsule three times daily for 40 weeks. After 48 weeks, all patients underwent a 4-week wash-out period. The primary endpoint was change in motor examination score of the Unified Multiple System Atrophy Rating Scale (UMSARS) from baseline to 52 weeks. Efficacy analyses were done in all people who received at least one dose of study medication. Safety was analysed in all people who received at least one dose of the study medication to which they had been randomly assigned. This trial is registered with ClinicalTrials.gov (NCT02008721) and EudraCT (2012-000928-18), and is completed. FINDINGS Between April 23, 2014, and Sept 3, 2015, 127 participants were screened and 92 were randomly assigned-47 to epigallocatechin gallate and 45 to placebo. Of these, 67 completed treatment and 64 completed the study (altough one of these patients had a major protocol violation). There was no evidence of a difference in the mean change from baseline to week 52 in motor examination scores on UMSARS between the epigallocatechin gallate (5·66 [SE 1·01]) and placebo (6·60 [0·99]) groups (mean difference -0·94 [SE 1·41; 95% CI -3·71 to 1·83]; p=0·51). Four patients in the epigallocatechin gallate group and two in the placebo group died. Two patients in the epigallocatechin gallate group had to stop treatment because of hepatotoxicity. INTERPRETATION 48 weeks of epigallocatechin gallate treatment did not modify disease progression in patients with multiple system atrophy. Epigallocatechin gallate was overall well tolerated but was associated with hepatotoxic effects in some patients, and thus doses of more than 1200 mg should not be used. FUNDING ParkinsonFonds Deutschland, German Parkinson Society, German Neurology Foundation, Lüneburg Foundation, Bischof Dr Karl Golser Foundation, and Dr Arthur Arnstein Foundation.
Collapse
Affiliation(s)
- Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany
| | - Sylvia Maaß
- German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany; Department of Neurology, Technical University Munich, Munich, Germany
| | - Madeleine Schuberth
- Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Armin Giese
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wolfgang H Oertel
- Department of Neurology, Philipps-Universität Marburg, Marburg, Germany
| | - Werner Poewe
- Department of Neurobiology, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany; Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| | - Gregor K Wenning
- Department of Neurobiology, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Ulrich Mansmann
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martin Südmeyer
- Institute of Clinical Neuroscience and Medical Psychology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Karla Eggert
- Department of Neurology, Philipps-Universität Marburg, Marburg, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany; Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Axel Lipp
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthias Löhle
- Department of Neurology, University of Rostock, Rostock, Germany; German Center for Neurodegenerative Diseases, Rostock, Germany; Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Joseph Classen
- Department of Neurology, University of Leipzig, Leipzig Germany
| | | | - Jan Kassubek
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Florin Gandor
- Movement Disorders Hospital, Beelitz-Heilstätten, Germany
| | - Daniela Berg
- Department of Neurodegeneration, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| | | | - Cornelia Eberhardt
- Pharmacy Department, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friedemann Paul
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Max Delbrueck Center for Molecular Medicine, NeuroCure Experimental and Clinical Research Center, Berlin, Germany
| | - Kai Bötzel
- Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Birgit Ertl-Wagner
- Department of Radiology, Ludwig-Maximilians-University Munich, Munich, Germany; Department of Radiology, The Hopsital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | - Ingrid Ricard
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases, Munich, Germany; Department of Neurology, Technical University Munich, Munich, Germany; Department of Neurology, Hanover Medical School, Hanover, Germany.
| |
Collapse
|
25
|
Bhat J, Dubin S, Dananberg A, Quabius ES, Fritsch J, Dowds CM, Saxena A, Chitadze G, Lettau M, Kabelitz D. Histone Deacetylase Inhibitor Modulates NKG2D Receptor Expression and Memory Phenotype of Human Gamma/Delta T Cells Upon Interaction With Tumor Cells. Front Immunol 2019; 10:569. [PMID: 30972064 PMCID: PMC6445873 DOI: 10.3389/fimmu.2019.00569] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 03/04/2019] [Indexed: 12/31/2022] Open
Abstract
The functional plasticity and anti-tumor potential of human γδ T cells have been widely studied. However, the epigenetic regulation of γδ T-cell/tumor cell interactions has been poorly investigated. In the present study, we show that treatment with the histone deacetylase inhibitor Valproic acid (VPA) significantly enhanced the expression and/or release of the NKG2D ligands MICA, MICB and ULBP-2, but not ULBP-1 in the pancreatic carcinoma cell line Panc89 and the prostate carcinoma cell line PC-3. Under in vitro tumor co-culture conditions, the expression of full length and the truncated form of the NKG2D receptor in γδ T cells was significantly downregulated. Furthermore, using a newly established flow cytometry-based method to analyze histone acetylation (H3K9ac) in γδ T cells, we showed constitutive H3K9aclow and inducible H3K9achigh expression in Vδ2 T cells. The detailed analysis of H3K9aclow Vδ2 T cells revealed a significant reversion of TEMRA to TEM phenotype during in vitro co-culture with pancreatic ductal adenocarcinoma cells. Our study uncovers novel mechanisms of how epigenetic modifiers modulate γδ T-cell differentiation during interaction with tumor cells. This information is important when considering combination therapy of VPA with the γδ T-cell-based immunotherapy for the treatment of certain types of cancer.
Collapse
Affiliation(s)
- Jaydeep Bhat
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Samuel Dubin
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Alexandra Dananberg
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Elgar Susanne Quabius
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Oto-Rhino-Laryngology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Juergen Fritsch
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - C. Marie Dowds
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Ankit Saxena
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Guranda Chitadze
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Marcus Lettau
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
26
|
A review on iron chelators as potential therapeutic agents for the treatment of Alzheimer’s and Parkinson’s diseases. Mol Divers 2018; 23:509-526. [DOI: 10.1007/s11030-018-9878-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/25/2018] [Indexed: 12/19/2022]
|
27
|
Hofer S, Kainz K, Zimmermann A, Bauer MA, Pendl T, Poglitsch M, Madeo F, Carmona-Gutierrez D. Studying Huntington's Disease in Yeast: From Mechanisms to Pharmacological Approaches. Front Mol Neurosci 2018; 11:318. [PMID: 30233317 PMCID: PMC6131589 DOI: 10.3389/fnmol.2018.00318] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder that leads to progressive neuronal loss, provoking impaired motor control, cognitive decline, and dementia. So far, HD remains incurable, and available drugs are effective only for symptomatic management. HD is caused by a mutant form of the huntingtin protein, which harbors an elongated polyglutamine domain and is highly prone to aggregation. However, many aspects underlying the cytotoxicity of mutant huntingtin (mHTT) remain elusive, hindering the efficient development of applicable interventions to counteract HD. An important strategy to obtain molecular insights into human disorders in general is the use of eukaryotic model organisms, which are easy to genetically manipulate and display a high degree of conservation regarding disease-relevant cellular processes. The budding yeast Saccharomyces cerevisiae has a long-standing and successful history in modeling a plethora of human maladies and has recently emerged as an effective tool to study neurodegenerative disorders, including HD. Here, we summarize some of the most important contributions of yeast to HD research, specifically concerning the elucidation of mechanistic features of mHTT cytotoxicity and the potential of yeast as a platform to screen for pharmacological agents against HD.
Collapse
Affiliation(s)
- Sebastian Hofer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Katharina Kainz
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Andreas Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Maria A. Bauer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Tobias Pendl
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Michael Poglitsch
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | | |
Collapse
|
28
|
Bansal S, Maurya IK, Yadav N, Thota CK, Kumar V, Tikoo K, Chauhan VS, Jain R. C-Terminal Fragment, Aβ 39-42-Based Tetrapeptides Mitigates Amyloid-β Aggregation-Induced Toxicity. ACS OMEGA 2018; 3:10019-10032. [PMID: 31459130 PMCID: PMC6645473 DOI: 10.1021/acsomega.8b01522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/20/2018] [Indexed: 06/07/2023]
Abstract
Since the introduction of acetyl cholinesterase inhibitors as the first approved drugs by the US Food and Drug Administration for Alzheimer's disease (AD) in clinics, less than satisfactory success in the design of anti-AD agents has impelled the scientists to also focus toward inhibition of Aβ aggregation. Considering the specific binding of fragments for their parent peptide, herein, we synthesized more than 40 new peptides based on a C-terminus tetrapeptide fragment of Aβ1-42. Initial screening by MTT cell viability assay and supportive results by ThT fluorescence assay led us to identify a tetrapeptide showing complete inhibition for Aβ1-42 aggregation. Peptide 20 displayed 100% cell viability at 20 μM concentration, while at lower concentrations of 10 and 2 μM 76.6 and 70% of cells were viable. Peptide 20 was found to restrict the conformational transition of Aβ1-42 peptide toward β-sheet structure. Inhibitory activity of tetrapeptide 20 was further evidenced by the absence of Aβ1-42 aggregates in electron microscopy. Peptide 20 and other significantly active tetrapeptide analogues could prove imperative in the future design of anti-AD agents.
Collapse
Affiliation(s)
- Sunil Bansal
- Department
of Medicinal Chemistry and Department of Pharmacology and
Toxicology, National Institute of Pharmaceutical
Education and Research, Sector 67, S.A.S Nagar, 160 062 Punjab, India
| | - Indresh Kumar Maurya
- Department
of Microbial Biotechnology, Punjab University, Sector 14, Chandigarh 160 014, India
| | - Nitin Yadav
- International
Center for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110 067, India
| | - Chaitanya Kumar Thota
- International
Center for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110 067, India
| | - Vinod Kumar
- Department
of Medicinal Chemistry and Department of Pharmacology and
Toxicology, National Institute of Pharmaceutical
Education and Research, Sector 67, S.A.S Nagar, 160 062 Punjab, India
| | - Kulbhushan Tikoo
- Department
of Medicinal Chemistry and Department of Pharmacology and
Toxicology, National Institute of Pharmaceutical
Education and Research, Sector 67, S.A.S Nagar, 160 062 Punjab, India
| | - Virander Singh Chauhan
- International
Center for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110 067, India
| | - Rahul Jain
- Department
of Medicinal Chemistry and Department of Pharmacology and
Toxicology, National Institute of Pharmaceutical
Education and Research, Sector 67, S.A.S Nagar, 160 062 Punjab, India
| |
Collapse
|
29
|
Wang W, Wang W, Yao G, Ren Q, Wang D, Wang Z, Liu P, Gao P, Zhang Y, Wang S, Song S. Novel sarsasapogenin-triazolyl hybrids as potential anti-Alzheimer's agents: Design, synthesis and biological evaluation. Eur J Med Chem 2018; 151:351-362. [DOI: 10.1016/j.ejmech.2018.03.082] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 03/24/2018] [Accepted: 03/30/2018] [Indexed: 12/22/2022]
|
30
|
Propyl Gallate Exerts an Antimigration Effect on Temozolomide-Treated Malignant Glioma Cells through Inhibition of ROS and the NF- κB Pathway. J Immunol Res 2017; 2017:9489383. [PMID: 29062841 PMCID: PMC5618759 DOI: 10.1155/2017/9489383] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 01/12/2023] Open
Abstract
In this study, we demonstrated that temozolomide (TMZ) and propyl gallate (PG) combination enhanced the inhibition of migration in human U87MG glioma cells. PG inhibited the TMZ-induced reactive oxygen species (ROS) generation. The mitochondrial complex III and NADPH oxidase are two critical sites that can be considered to regulate antimigration in TMZ-treated U87MG cells. PG can enhance the antimigration effect of TMZ through suppression of metalloproteinase-2 and metalloproteinase-9 activities, ROS generation, and the NF-κB pathway and possibly provide a novel prospective strategy for treating malignant glioma.
Collapse
|
31
|
Hasani P, Tehrani HS, Yaghmaei P, Roudbari NH. Effects of Camellia Sinensis Extract on Passive Avoidance Learning and Hippocampal Neurogenesis in Rats. NEUROPHYSIOLOGY+ 2017. [DOI: 10.1007/s11062-017-9642-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Cascella M, Bimonte S, Muzio MR, Schiavone V, Cuomo A. The efficacy of Epigallocatechin-3-gallate (green tea) in the treatment of Alzheimer's disease: an overview of pre-clinical studies and translational perspectives in clinical practice. Infect Agent Cancer 2017; 12:36. [PMID: 28642806 PMCID: PMC5477123 DOI: 10.1186/s13027-017-0145-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/06/2017] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most common form of dementia characterized by cognitive and memory impairment. One of the mechanism involved in the pathogenesis of AD, is the oxidative stress being involved in AD's development and progression. In addition, several studies proved that chronic viral infections, mainly induced by Human herpesvirus 1 (HHV-1), Cytomegalovirus (CMV), Human herpesvirus 2 (HHV-2), and Hepatitis C virus (HCV) could be responsible for AD's neuropathology. Despite the large amount of data regarding the pathogenesis of Alzheimer's disease (AD), a very limited number of therapeutic drugs and/or pharmacological approaches, have been developed so far. It is important to underline that, in recent years, natural compounds, due their antioxidants and anti-inflammatory properties have been largely studied and identified as promising agents for the prevention and treatment of neurodegenerative diseases, including AD. The ester of epigallocatechin and gallic acid, (-)-Epigallocatechin-3-Gallate (EGCG), is the main and most significantly bioactive polyphenol found in solid green tea extract. Several studies showed that this compound has important anti-inflammatory and antiatherogenic properties as well as protective effects against neuronal damage and brain edema. To date, many studies regarding the potential effects of EGCG in AD's treatment have been reported in literature. The purpose of this review is to summarize the in vitro and in vivo pre-clinical studies on the use of EGCG in the prevention and the treatment of AD as well as to offer new insights for translational perspectives into clinical practice.
Collapse
Affiliation(s)
- Marco Cascella
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori – IRCCS - “Fondazione G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Sabrina Bimonte
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori – IRCCS - “Fondazione G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Maria Rosaria Muzio
- Division of Infantile Neuropsychiatry, UOMI - Maternal and Infant Health, ASL NA 3 SUD, Torre del Greco, Via Marconi, 80059 Naples, Italy
| | - Vincenzo Schiavone
- Division of Anesthesia and Intensive Care, Presidio Ospedaliero “Pineta Grande”, Castel Voltuno, 81100 Caserta, Italy
| | - Arturo Cuomo
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori – IRCCS - “Fondazione G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| |
Collapse
|
33
|
Shin JW, Wu Y, Kang YG, Kim JK, Choi HJ, Shin JW. The Effects of Epigallocatechin-3-Gallate and Mechanical Stimulation on Osteogenic Differentiation of Human Mesenchymal Stem Cells: Individual or Synergistic Effects. Tissue Eng Regen Med 2017; 14:307-315. [PMID: 30603487 DOI: 10.1007/s13770-017-0040-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 08/28/2016] [Accepted: 09/09/2016] [Indexed: 12/21/2022] Open
Abstract
This study aims to investigate the roles and effects of EGCG (epigallocatechin-3-gallate) during the osteogenic differentiation of human mesenchymal stem cells (hMSCs) in vitro. Recent studies have shown that proper mechanical stimuli can induce osteogenic differentiation of hMSCs apart from biochemical factors. In this study, the hMSC cultures were subjected to: (1) 25 uM EGCG alone or (2) 3% mechanical stretching (0.2 Hz for 4 h/day for 4 days) or (3) in combination with 3% mechanical stretching (0.2 Hz for 4 h/day for 4 days). The two factors were applied to the cell cultures separately and in combination to investigate the individual and synergistic effect of both mechanical stimulation and ECGC in the osteogenic differentiation of hMSCs. Utilizing real time PCR, we measured various osteogenic markers and even those related to intracellular signalings. Further investigation of mitochondria was performed that mitochondria biogenesis, antioxidant capacity, and morphological related markers were measured. hMSCs were to be osteogenic or myogenic differentiated when they were under 3% stretching only. However, when EGCG was applied along with stretching they were to be osteogenic differentiated rather than to be myogenic differentiated. This was supported by evaluating intracellular signalings: BMP-2 and VEGF. Therefore, the synergistical effects of simultaneous employment of stretching and EGCG on osteogenic differentiation were confirmed. Moreover, simultaneous employment was found positive in mitochondria biogenesis, antioxidant capacity, and morphological changes. Through this study, we came into the conclusion that the combination of proper mechanical stretching, 3% in this study, and EGCG promote osteogenic differentiation. Reflecting that EGCG can be obtained from plants not from the chemical syntheses, it is worth to be studied further either by animal tests or long-term experiments for clinical applications.
Collapse
Affiliation(s)
- Ji Won Shin
- 1Department of Biomedical Engineering, Inje University, 197 Inje-ro, Gimhae, 50834 Korea
| | - Yanru Wu
- 2Department of Health Science and Technology, Inje University, 197 Inje-ro, Gimhae, 50834 Korea
| | - Yun Gyeong Kang
- 1Department of Biomedical Engineering, Inje University, 197 Inje-ro, Gimhae, 50834 Korea
| | - Jeong Koo Kim
- 1Department of Biomedical Engineering, Inje University, 197 Inje-ro, Gimhae, 50834 Korea
| | - Hyun Ju Choi
- Research and Development Team, Gimhae Biomedical Center, Gimhae, Gyeongnam 50834 Korea
| | - Jung-Woog Shin
- 1Department of Biomedical Engineering, Inje University, 197 Inje-ro, Gimhae, 50834 Korea.,2Department of Health Science and Technology, Inje University, 197 Inje-ro, Gimhae, 50834 Korea.,4Cardiovascular and Metabolic Disease Center/Institute of Aged Life Redesign/UHARC, Inje University, 197 Inje-ro, Gimhae, 50834 Korea
| |
Collapse
|
34
|
Ren B, Zhang M, Hu R, Chen H, Wang M, Lin Y, Sun Y, Jia L, Liang G, Zheng J. Identification of a New Function of Cardiovascular Disease Drug 3-Morpholinosydnonimine Hydrochloride as an Amyloid-β Aggregation Inhibitor. ACS OMEGA 2017; 2:243-250. [PMID: 30023514 PMCID: PMC6044715 DOI: 10.1021/acsomega.6b00397] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/13/2017] [Indexed: 05/29/2023]
Abstract
Cardiovascular disease (CVD) and Alzheimer's disease (AD) have a mutual cause-and-effect relationship, and they share some common risk factors. Although numerous Food and Drug Administration (FDA)-approved drugs have been developed for CVD treatment, no drugs are clinically available for AD treatment. Given the common disease-causing factors and links between the two diseases and the well-demonstrated drugs for CVD, we propose to re-examine the new potential of the existing CVD drugs as amyloid-β (Aβ) inhibitors. 3-Morpholinosydnonimine hydrochloride (SIN-1) is an FDA-approved drug for inhibiting platelet aggregation in CVD. Herein, we examine the inhibition activity of SIN-1 on the aggregation and toxicity of Aβ1-42 using combined experimental and computational approaches. Collective experimental data from ThT, circular dichroism, and atomic force microscopy demonstrate that SIN-1 can effectively inhibit amyloid formation at every stage of Aβ aggregation by prolonging lag phase, slowing down aggregation rate, and reducing final fibril formation. The cell viability assay also shows that SIN-1 enables the protection of SH-SY5Y cells from Aβ-induced cell toxicity. Such an inhibition effect is attributed to interference with the structural transition of Aβ toward a β-sheet structure by SIN-1. Furthermore, molecular dynamic simulations confirm that SIN-1 preferentially binds to the C-terminal β-sheet grooves of an Aβ oligomer and consequently disrupts the β-sheet structure of Aβ and Aβ-Aβ association, explaining experimental observations. This work discovers a new function of SIN-1, making it a promising compound with dual protective roles in inhibiting both platelet and Aβ aggregations against CVD and AD.
Collapse
Affiliation(s)
- Baiping Ren
- Department
of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
- School
of Life Science and Biotechnology, Dalian
University of Technology, Dalian 116024, China
| | - Mingzhen Zhang
- Department
of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Rundong Hu
- Department
of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Hong Chen
- Department
of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Manli Wang
- Mingyuan
Staff-Worker Hospital of Xinjiang Oil Field Company, 789 Youhao Beilu, Urumqi, Xinjiang 830000, China
| | - Yufeng Lin
- Mingyuan
Staff-Worker Hospital of Xinjiang Oil Field Company, 789 Youhao Beilu, Urumqi, Xinjiang 830000, China
| | - Yan Sun
- Department
of Biochemical Engineering and Key Laboratory of Systems Bioengineering
of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Lingyun Jia
- School
of Life Science and Biotechnology, Dalian
University of Technology, Dalian 116024, China
| | - Guizhao Liang
- Department
of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
- Key
Laboratory of Biorheological Science and Technology, Ministry of Education,
School of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jie Zheng
- Department
of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
35
|
Bansal S, Maurya IK, Shenmar K, Yadav N, Thota CK, Kumar V, Tikoo K, Chauhan VS, Jain R. Aβ1–42 C-terminus fragment derived peptides prevent the self-assembly of the parent peptide. RSC Adv 2017. [DOI: 10.1039/c6ra26295c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A series of peptides derived from the C-terminus fragment (Aβ38–42) of Aβ showed significant to complete reduction in Aβ-induced toxicity.
Collapse
Affiliation(s)
- Sunil Bansal
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- S. A. S. Nagar
- India
| | | | - Kitika Shenmar
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- S. A. S. Nagar
- India
| | - Nitin Yadav
- International Center for Genetic Engineering and Biotechnology
- New Delhi
- India
| | | | - Vinod Kumar
- Department of Pharmacology and Toxicology
- National Institute of Pharmaceutical Education and Research
- S. A. S Nagar
- India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology
- National Institute of Pharmaceutical Education and Research
- S. A. S Nagar
- India
| | | | - Rahul Jain
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- S. A. S. Nagar
- India
| |
Collapse
|
36
|
Biophysical Approach to Mechanisms of Cancer Prevention and Treatment with Green Tea Catechins. Molecules 2016; 21:molecules21111566. [PMID: 27869750 PMCID: PMC6273158 DOI: 10.3390/molecules21111566] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/26/2016] [Accepted: 11/09/2016] [Indexed: 01/25/2023] Open
Abstract
Green tea catechin and green tea extract are now recognized as non-toxic cancer preventives for humans. We first review our brief historical development of green tea cancer prevention. Based on exciting evidence that green tea catechin, (−)-epigallocatechin gallate (EGCG) in drinking water inhibited lung metastasis of B16 melanoma cells, we and other researchers have studied the inhibitory mechanisms of metastasis with green tea catechins using biomechanical tools, atomic force microscopy (AFM) and microfluidic optical stretcher. Specifically, determination of biophysical properties of cancer cells, low cell stiffness, and high deformability in relation to migration, along with biophysical effects, were studied by treatment with green tea catechins. The study with AFM revealed that low average values of Young’s moduli, indicating low cell stiffness, are closely associated with strong potential of cell migration and metastasis for various cancer cells. It is important to note that treatments with EGCG and green tea extract elevated the average values of Young’s moduli resulting in increased stiffness (large elasticity) of melanomas and various cancer cells. We discuss here the biophysical basis of multifunctions of green tea catechins and green tea extract leading to beneficial effects for cancer prevention and treatment.
Collapse
|
37
|
Melki R. Role of Different Alpha-Synuclein Strains in Synucleinopathies, Similarities with other Neurodegenerative Diseases. JOURNAL OF PARKINSONS DISEASE 2016; 5:217-27. [PMID: 25757830 PMCID: PMC4923763 DOI: 10.3233/jpd-150543] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Misfolded protein aggregates are the hallmark of several neurodegenerative diseases in humans. The main protein constituent of these aggregates and the regions within the brain that are affected differ from one neurodegenerative disorder to another. A plethora of reports suggest that distinct diseases have in common the ability of protein aggregates to spread and amplify within the central nervous system. This review summarizes briefly what is known about the nature of the protein aggregates that are infectious and the reason they are toxic to cells. The chameleon property of polypeptides which aggregation into distinct high-molecular weight assemblies is associated to different diseases, in particular, that of alpha-synuclein which aggregation is the hallmark of distinct synucleinopathies, is discussed. Finally, strategies targeting the formation and propagation of structurally distinct alpha-synuclein assemblies associated to different synucleinopathies are presented and their therapeutic and diagnostic potential is discussed.
Collapse
Affiliation(s)
- Ronald Melki
- Correspondence to: Ronald Melki, Neuro Psi, CNRS, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France. Tel.: +33 169823503; Fax: +33 169823129;
| |
Collapse
|
38
|
Bansal S, Maurya IK, Yadav N, Thota CK, Kumar V, Tikoo K, Chauhan VS, Jain R. C-Terminal Fragment, Aβ32-37, Analogues Protect Against Aβ Aggregation-Induced Toxicity. ACS Chem Neurosci 2016; 7:615-23. [PMID: 26835536 DOI: 10.1021/acschemneuro.6b00006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Amyloid-β aggregation is a major etiological phenomenon in Alzheimer's disease. Herein, we report peptide-based inhibitors that diminish the amyloid load by obviating Aβ aggregation. Taking the hexapeptide fragment, Aβ32-37, as lead, more than 40 new peptides were synthesized. Upon evaluation of the newly synthesized hexapeptides as inhibitors of Aβ toxicity by the MTT-based cell viability assay, a number of peptides exhibited significant Aβ aggregation inhibitory activity at sub-micromolar concentration range. A hexapeptide (1) showed complete mitigation of Aβ toxicity in the cell culture assay at 2 μM. In the ThT fluorescence assay, upon incubation of Aβ with this peptide, we observed no increase in the ThT fluorescence relative to control. The secondary structure estimation by circular dichroism spectroscopy and morphological examination by transmission electron microscopy further confirmed the results.
Collapse
Affiliation(s)
- Sunil Bansal
- Department
of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S Nagar 160 062, Punjab India
| | - Indresh Kumar Maurya
- Department
of Microbial Biotechnology, Panjab University, Sector 14, Chandigarh 160 014, India
| | - Nitin Yadav
- International Center for Genetic Engineering and Biotechnology, Aruna Asif Ali Marg, New
Delhi 110 067, India
| | - Chaitanya Kumar Thota
- International Center for Genetic Engineering and Biotechnology, Aruna Asif Ali Marg, New
Delhi 110 067, India
| | - Vinod Kumar
- Department
of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S Nagar 160 062, Punjab India
| | - Kulbhushan Tikoo
- Department
of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S Nagar 160 062, Punjab India
| | - Virander Singh Chauhan
- International Center for Genetic Engineering and Biotechnology, Aruna Asif Ali Marg, New
Delhi 110 067, India
| | - Rahul Jain
- Department
of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S Nagar 160 062, Punjab India
| |
Collapse
|
39
|
Stotani S, Giordanetto F, Medda F. DYRK1A inhibition as potential treatment for Alzheimer's disease. Future Med Chem 2016; 8:681-96. [PMID: 27073990 DOI: 10.4155/fmc-2016-0013] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
In total, 47,500,000 people worldwide are affected by dementia and this number is estimated to double by 2030 and triple within 2050 resulting in a huge burden on public health. Alzheimer's disease (AD), a progressive neurodegenerative disorder, is the most common cause of dementia, accounting for 60-70% of all the cases. The cause of AD is still poorly understood but several brain abnormalities (e.g., loss of neuronal connections and neuronal death) have been identified in affected patients. In addition to the accumulation of β-amyloid plaques in the brain tissue, aberrant phosphorylation of tau proteins has proved to increase neuronal death. DYRK1A phosphorylates tau on 11 different Ser/Thr residues, resulting in the formation of aggregates called 'neurofibrillary tangles' which, together with amyloid plaques, could be responsible for dementia, neuronal degeneration and cell death. Small molecule inhibition of DYRK1A could thus represent an interesting approach toward the treatment of Alzheimer's and other neurodegenerative diseases. Herein we review the current progress in the identification and development of DYRK1A inhibitors.
Collapse
Affiliation(s)
- Silvia Stotani
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Str. 76a, 44227 Dortmund, Germany
| | - Fabrizio Giordanetto
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Str. 76a, 44227 Dortmund, Germany
- DE Shaw Research, 120W 45th Street, New York, NY 10036, USA
| | - Federico Medda
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Str. 76a, 44227 Dortmund, Germany
| |
Collapse
|
40
|
Ramachandran B, Jayavelu S, Murhekar K, Rajkumar T. Repeated dose studies with pure Epigallocatechin-3-gallate demonstrated dose and route dependant hepatotoxicity with associated dyslipidemia. Toxicol Rep 2016; 3:336-345. [PMID: 28959554 PMCID: PMC5615837 DOI: 10.1016/j.toxrep.2016.03.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/15/2016] [Accepted: 03/02/2016] [Indexed: 12/22/2022] Open
Abstract
EGCG (Epigallocatechin-3-gallate) is the major active principle catechin found in green tea. Skepticism regarding the safety of consuming EGCG is gaining attention, despite the fact that it is widely being touted for its potential health benefits, including anti-cancer properties. The lack of scientific data on safe dose levels of pure EGCG is of concern, while EGCG has been commonly studied as a component of GTE (Green tea extract) and not as a single active constituent. This study has been carried out to estimate the maximum tolerated non-toxic dose of pure EGCG and to identify the treatment related risk factors. In a fourteen day consecutive treatment, two different administration modalities were compared, offering an improved [i.p (intraperitoneal)] and limited [p.o (oral)] bioavailability. A trend of dose and route dependant hepatotoxicity was observed particularly with i.p treatment and EGCG increased serum lipid profile in parallel to hepatotoxicity. Fourteen day tolerable dose of EGCG was established as 21.1 mg/kg for i.p and 67.8 mg/kg for p.o. We also observed that, EGCG induced effects by both treatment routes are reversible, subsequent to an observation period for further fourteen days after cessation of treatment. It was demonstrated that the severity of EGCG induced toxicity appears to be a function of dose, route of administration and period of treatment.
Collapse
Affiliation(s)
- Balaji Ramachandran
- Department of Molecular Oncology, Cancer Institute (W.I.A), No. 38, Sardar Patel Road, Adyar, 600 036 Chennai, India
| | - Subramani Jayavelu
- Department of Molecular Oncology, Cancer Institute (W.I.A), No. 38, Sardar Patel Road, Adyar, 600 036 Chennai, India
| | - Kanchan Murhekar
- Department of Oncopathology, Cancer Institute (W.I.A), No. 38, Sardar Patel Road, Adyar, 600 036 Chennai, India
| | - Thangarajan Rajkumar
- Department of Molecular Oncology, Cancer Institute (W.I.A), No. 38, Sardar Patel Road, Adyar, 600 036 Chennai, India
| |
Collapse
|
41
|
The PROMESA-protocol: progression rate of multiple system atrophy under EGCG supplementation as anti-aggregation-approach. J Neural Transm (Vienna) 2016; 123:439-45. [PMID: 26809243 DOI: 10.1007/s00702-016-1507-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/11/2016] [Indexed: 12/11/2022]
Abstract
Formation of toxic α-synuclein oligomers appears to be a key underlying pathological mechanism of synucleinopathies such as Parkinson's disease or multiple system atrophy (MSA). Given that Epigallocatechin-gallate has been shown to inhibit α-synuclein aggregation, it might represent a causal treatment option. Therefore, we set out to evaluate the safety, tolerability and a potential disease-modifying effect of Epigallocatechin-gallate in patients with MSA after 48 weeks of treatment. Power calculation was performed on existing natural history data on the progression of the Unified MSA Rating Scale as primary readout parameter. To assess the efficacy of Epigallocatechin-gallate versus placebo regarding the reduction of disease progression measured during the study period (80 % power, 5 % p level, 50 % effect size) 36 patients per group are needed. Considering a drop-out rate of 20 % a total of 86 patients will be recruited in this multicentre study. These data provide a solid rationale to investigate whether supplementation of Epigallocatechin-gallate can delay the progression of the MSA-related disability.
Collapse
|
42
|
Pithadia A, Brender JR, Fierke CA, Ramamoorthy A. Inhibition of IAPP Aggregation and Toxicity by Natural Products and Derivatives. J Diabetes Res 2016; 2016:2046327. [PMID: 26649317 PMCID: PMC4662995 DOI: 10.1155/2016/2046327] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/15/2015] [Indexed: 01/10/2023] Open
Abstract
Fibrillar aggregates of human islet amyloid polypeptide, hIAPP, a pathological feature seen in some diabetes patients, are a likely causative agent for pancreatic beta-cell toxicity, leading to a transition from a state of insulin resistance to type II diabetes through the loss of insulin producing beta-cells by hIAPP induced toxicity. Because of the probable link between hIAPP and the development of type II diabetes, there has been strong interest in developing reagents to study the aggregation of hIAPP and possible therapeutics to block its toxic effects. Natural products are a class of compounds with interesting pharmacological properties against amyloids which have made them interesting targets to study hIAPP. Specifically, the ability of polyphenolic natural products, EGCG, curcumin, and resveratrol, to modulate the aggregation of hIAPP is discussed. Furthermore, we have outlined possible mechanistic discoveries of the interaction of these small molecules with the peptide and how they may mitigate toxicity associated with peptide aggregation. These abundantly found agents have been long used to combat diseases for many years and may serve as useful templates toward developing therapeutics against hIAPP aggregation and toxicity.
Collapse
Affiliation(s)
- Amit Pithadia
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Jeffrey R. Brender
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Carol A. Fierke
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | - Ayyalusamy Ramamoorthy
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, MI 48109-1055, USA
- *Ayyalusamy Ramamoorthy:
| |
Collapse
|
43
|
Tofolean IT, Ganea C, Ionescu D, Filippi A, Garaiman A, Goicea A, Gaman MA, Dimancea A, Baran I. Cellular determinants involving mitochondrial dysfunction, oxidative stress and apoptosis correlate with the synergic cytotoxicity of epigallocatechin-3-gallate and menadione in human leukemia Jurkat T cells. Pharmacol Res 2015; 103:300-17. [PMID: 26687095 DOI: 10.1016/j.phrs.2015.12.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 12/09/2015] [Accepted: 12/09/2015] [Indexed: 12/17/2022]
Abstract
We have investigated the growth-suppressive action of epigallocatechin-3-gallate (EGCG) on human leukemia Jurkat T cells. Results show a strong correlation between the dose-dependent reduction of clonogenic survival following acute EGCG treatments and the EGCG-induced decline of the mitochondrial level of Ca(2+). The cell killing ability of EGCG was synergistically enhanced by menadione. In addition, the cytotoxic effect of EGCG applied alone or in combination with menadione was accompanied by apoptosis induction. We also observed that in acute treatments EGCG displays strong antioxidant properties in the intracellular milieu, but concurrently triggers some oxidative stress generating mechanisms that can fully develop on a longer timescale. In parallel, EGCG dose-dependently induced mitochondrial depolarization during exposure, but this condition was subsequently reversed to a persistent hyperpolarized mitochondrial state that was dependent on the activity of respiratory Complex I. Fluorimetric measurements suggest that EGCG is a mitochondrial Complex III inhibitor and indicate that EGCG evokes a specific cellular fluorescence with emission at 400nm and two main excitation bands (at 330nm and 350nm) that may originate from a mitochondrial supercomplex containing dimeric Complex III and dimeric ATP-synthase, and therefore could provide a valuable means to characterize the functional properties of the respiratory chain.
Collapse
Affiliation(s)
- Ioana Teodora Tofolean
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania
| | - Constanta Ganea
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania
| | - Diana Ionescu
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania
| | - Alexandru Filippi
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania
| | - Alexandru Garaiman
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania
| | - Alexandru Goicea
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania
| | - Mihnea-Alexandru Gaman
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania
| | - Alexandru Dimancea
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania
| | - Irina Baran
- "Carol Davila" University of Medicine and Pharmacy, Dept. of Biophysics, 8 Eroii Sanitari, 050474 Bucharest, Romania.
| |
Collapse
|
44
|
Mossakowski AA, Pohlan J, Bremer D, Lindquist R, Millward JM, Bock M, Pollok K, Mothes R, Viohl L, Radbruch M, Gerhard J, Bellmann-Strobl J, Behrens J, Infante-Duarte C, Mähler A, Boschmann M, Rinnenthal JL, Füchtemeier M, Herz J, Pache FC, Bardua M, Priller J, Hauser AE, Paul F, Niesner R, Radbruch H. Tracking CNS and systemic sources of oxidative stress during the course of chronic neuroinflammation. Acta Neuropathol 2015; 130:799-814. [PMID: 26521072 PMCID: PMC4654749 DOI: 10.1007/s00401-015-1497-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 11/30/2022]
Abstract
The functional dynamics and cellular sources of oxidative stress are central to understanding MS pathogenesis but remain elusive, due to the lack of appropriate detection methods. Here we employ NAD(P)H fluorescence lifetime imaging to detect functional NADPH oxidases (NOX enzymes) in vivo to identify inflammatory monocytes, activated microglia, and astrocytes expressing NOX1 as major cellular sources of oxidative stress in the central nervous system of mice affected by experimental autoimmune encephalomyelitis (EAE). This directly affects neuronal function in vivo, indicated by sustained elevated neuronal calcium. The systemic involvement of oxidative stress is mirrored by overactivation of NOX enzymes in peripheral CD11b+ cells in later phases of both MS and EAE. This effect is antagonized by systemic intake of the NOX inhibitor and anti-oxidant epigallocatechin-3-gallate. Together, this persistent hyper-activation of oxidative enzymes suggests an “oxidative stress memory” both in the periphery and CNS compartments, in chronic neuroinflammation.
Collapse
Affiliation(s)
- Agata A Mossakowski
- German Rheumatism Research Center, Berlin, Germany
- Department of Neurology, NeuroCure Clinical Research Center, Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Pohlan
- German Rheumatism Research Center, Berlin, Germany
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Intravital Imaging and Immune Dynamics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Jason M Millward
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Bock
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karolin Pollok
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Intravital Imaging and Immune Dynamics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ronja Mothes
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Intravital Imaging and Immune Dynamics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Leonard Viohl
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Intravital Imaging and Immune Dynamics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Radbruch
- German Rheumatism Research Center, Berlin, Germany
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Judith Bellmann-Strobl
- Department of Neurology, NeuroCure Clinical Research Center, Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Janina Behrens
- Department of Neurology, NeuroCure Clinical Research Center, Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Infante-Duarte
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Mähler
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Boschmann
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jan Leo Rinnenthal
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Josephine Herz
- Department of Paediatrics I, Neonatology, University Hospital Essen, Essen, 45122, Germany
| | - Florence C Pache
- German Rheumatism Research Center, Berlin, Germany
- Department of Neurology, NeuroCure Clinical Research Center, Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Intravital Imaging and Immune Dynamics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Cluster of Excellence NeuroCure and BIH, Berlin, Germany
| | - Anja E Hauser
- German Rheumatism Research Center, Berlin, Germany
- Intravital Imaging and Immune Dynamics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Department of Neurology, NeuroCure Clinical Research Center, Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Helena Radbruch
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
45
|
Cardoso A, Magano S, Marrana F, Andrade JP. D-Galactose High-Dose Administration Failed to Induce Accelerated Aging Changes in Neurogenesis, Anxiety, and Spatial Memory on Young Male Wistar Rats. Rejuvenation Res 2015; 18:497-507. [PMID: 25936362 DOI: 10.1089/rej.2015.1684] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The model of accelerated senescence with the prolonged administration of d-galactose is used in anti-aging studies because it mimics several aging-associated alterations such as increase of oxidative stress and decline of cognition. However, there is no standardized protocol for this aging model, and recently some reports have questioned its effectiveness. To clarify this issue, we used a model of high-dose d-galactose on 1-month-old male Wistar rats and studied the hippocampus, one of the most affected brain regions. In one group (n = 10), d-galactose was daily administered intraperitoneally (300 mg/kg) during 8 weeks whereas age-matched controls (n = 10) were injected intraperitoneally with saline. A third group (n = 10) was treated with the same dose of d-galactose and with oral epigallocatechin-3-gallate (EGCG) (2 grams/L), a green tea catechin with anti-oxidant and neuroprotective properties. After treatments, animals were submitted to open-field, elevated plus-maze and Morris water maze tests, and neurogenesis in the dentate gyrus subgranular layer was quantified. There were no significant alterations when the three groups were compared in the number of doublecortin- and Ki-67-immunoreactive cells, and also on anxiety levels, spatial learning, and memory. Therefore, d-galactose was not effective in the induction of accelerated aging, and EGCG administered to d-galactose-treated animals did not improve behavior and had no effects on neurogenesis. We conclude that daily 300 mg/kg of d-galactose administered intraperitoneally may not be a suitable model for inducing age-related neurobehavioral alterations in young male Wistar rats. More studies are necessary to obtain a reliable and reproducible model of accelerated senescence in rodents using d-galactose.
Collapse
Affiliation(s)
- Armando Cardoso
- 1 Department of Anatomy, Faculty of Medicine, University of Porto , Porto, Portugal .,2 Center of Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto , Porto, Portugal
| | - Sara Magano
- 1 Department of Anatomy, Faculty of Medicine, University of Porto , Porto, Portugal
| | - Francisco Marrana
- 1 Department of Anatomy, Faculty of Medicine, University of Porto , Porto, Portugal
| | - José P Andrade
- 1 Department of Anatomy, Faculty of Medicine, University of Porto , Porto, Portugal .,2 Center of Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto , Porto, Portugal
| |
Collapse
|
46
|
Janssen A, Fiebiger S, Bros H, Hertwig L, Romero-Suarez S, Hamann I, Chanvillard C, Bellmann-Strobl J, Paul F, Millward JM, Infante-Duarte C. Treatment of Chronic Experimental Autoimmune Encephalomyelitis with Epigallocatechin-3-Gallate and Glatiramer Acetate Alters Expression of Heme-Oxygenase-1. PLoS One 2015; 10:e0130251. [PMID: 26114502 PMCID: PMC4482710 DOI: 10.1371/journal.pone.0130251] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 05/18/2015] [Indexed: 01/17/2023] Open
Abstract
We previously demonstrated that epigallocatechin-3-gallate (EGCG) synergizes with the immunomodulatory agent glatiramer acetate (GA) in eliciting anti-inflammatory and neuroprotective effects in the relapsing-remitting EAE model. Thus, we hypothesized that mice with chronic EAE may also benefit from this combination therapy. We first assessed how a treatment with a single dose of GA together with daily application of EGCG may modulate EAE. Although single therapies with a suboptimal dose of GA or EGCG led to disease amelioration and reduced CNS inflammation, the combination therapy had no effects. While EGCG appeared to preserve axons and myelin, the single GA dose did not improve axonal damage or demyelination. Interestingly, the neuroprotective effect of EGCG was abolished when GA was applied in combination. To elucidate how a single dose of GA may interfere with EGCG, we focused on the anti-inflammatory, iron chelating and anti-oxidant properties of EGCG. Surprisingly, we observed that while EGCG induced a downregulation of the gene expression of heme oxygenase-1 (HO-1) in affected CNS areas, the combined therapy of GA+EGCG seems to promote an increased HO-1 expression. These data suggest that upregulation of HO-1 may contribute to diminish the neuroprotective benefits of EGCG alone in this EAE model. Altogether, our data indicate that neuroprotection by EGCG in chronic EAE may involve regulation of oxidative processes, including downmodulation of HO-1. Further investigation of the re-dox balance in chronic neuroinflammation and in particular functional studies on HO-1 are warranted to understand its role in disease progression.
Collapse
Affiliation(s)
- Antonia Janssen
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sebastian Fiebiger
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Helena Bros
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
- NeuroCure Clinical Research Center, Charité—Universitätmedizin Berlin, Berlin, Germany
| | - Laura Hertwig
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Silvina Romero-Suarez
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Isabell Hamann
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Coralie Chanvillard
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Judith Bellmann-Strobl
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
- NeuroCure Clinical Research Center, Charité—Universitätmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
- NeuroCure Clinical Research Center, Charité—Universitätmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jason M. Millward
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Carmen Infante-Duarte
- Institute for Medical Immunology, Charité—Universitätmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
- * E-mail:
| |
Collapse
|
47
|
Tomaszewska E, Winiarska-Mieczan A, Dobrowolski P. Hematological and serum biochemical parameters of blood in adolescent rats and histomorphological changes in the jejunal epithelium and liver after chronic exposure to cadmium and lead in the case of supplementation with green tea vs black, red or white tea. ACTA ACUST UNITED AC 2015; 67:331-9. [DOI: 10.1016/j.etp.2015.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 02/16/2015] [Accepted: 02/26/2015] [Indexed: 12/31/2022]
|
48
|
Mähler A, Steiniger J, Bock M, Klug L, Parreidt N, Lorenz M, Zimmermann BF, Krannich A, Paul F, Boschmann M. Metabolic response to epigallocatechin-3-gallate in relapsing-remitting multiple sclerosis: a randomized clinical trial. Am J Clin Nutr 2015; 101:487-95. [PMID: 25733633 DOI: 10.3945/ajcn.113.075309] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Muscle weakness and fatigue are common symptoms in multiple sclerosis (MS). Green tea catechins such as (-)epigallocatechin-3-gallate (EGCG) are known to improve energy metabolism at rest and during exercise. OBJECTIVE We tested the hypothesis that EGCG improves energy metabolism and substrate utilization in patients with MS. DESIGN Eighteen patients (8 men) with relapsing-remitting MS (expanded disability status scale score <4.5, all receiving glatiramer acetate) participated in this randomized, double-blind, placebo-controlled, crossover trial at a clinical research center. All patients received EGCG (600 mg/d) and placebo over 12 wk (4-wk washout in between). After each intervention, fasting and postprandial energy expenditure (EE), as well as fat oxidation (FAOx) and carbohydrate oxidation (CHOx) rates, were measured either at rest or during 40 min of exercise (0.5 W/kg). At rest, blood samples and microdialysates from adipose tissue and skeletal muscle were also taken. RESULTS At rest, postprandial EE and CHOx, as well as adipose tissue perfusion and glucose supply, were significantly lower in men but higher in women receiving EGCG compared with placebo. During exercise, postprandial EE was lower after EGCG than after placebo, indicating an increased working efficiency (men > women). After placebo, exercise EE was mainly fueled by FAOx in both men and women. After EGCG, there was a shift to a higher and more stable CHOx during exercise in men but not in women. CONCLUSIONS Our data indicate that EGCG given to patients with MS over 12 wk improves muscle metabolism during moderate exercise to a greater extent in men than in women, possibly because of sex-specific effects on autonomic and endocrine control.
Collapse
Affiliation(s)
- Anja Mähler
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Jochen Steiniger
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Markus Bock
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Lars Klug
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Nadine Parreidt
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Mario Lorenz
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Benno F Zimmermann
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Alexander Krannich
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Friedemann Paul
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| | - Michael Boschmann
- From the Experimental & Clinical Research Center-a joint cooperation between Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany (AM, JS, M Bock, LK, NP, and M Boschmann); NeuroCure Clinical Research Center (AM, M Bock, and FP) and Medical Clinic for Cardiology and Angiology Campus Mitte (ML), Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (ML); University of Bonn, Institute of Nutritional and Food Sciences, Bonn, Germany (BFZ); Institute Prof. Dr. Georg Kurz GmbH, Köln, Germany (BFZ); Department of Biostatistics, Clinical Research Unit of Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany (AK); and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany (FP)
| |
Collapse
|
49
|
Golubnitschaja O, Costigliola V. EPMA summit 2014 under the auspices of the presidency of Italy in the EU: professional statements. EPMA J 2015; 6:4. [PMID: 25878761 PMCID: PMC4397737 DOI: 10.1186/s13167-015-0026-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/04/2015] [Indexed: 01/02/2023]
Abstract
Over the next 10-20 years, a pessimistic prognosis considers pandemic scenario for type 2 diabetes mellitus, neurodegenerative disorders and some types of cancer followed by the economic disaster of healthcare systems in a global scale. Well-recognised deficits of currently provided medical services result from the delayed 'disease care'. Herewith EPMA releases the long-term strategies for the effective promotion of predictive, preventive and personalised medicine (PPPM) considered as the medicine of the future. Under the EPMA-umbrella, an international forum of currently 45 countries is actively contributing to the development and implementation of the innovative PPPM concepts. EPMA is open for collaboration with other leading European and global professional networks relevant for the effective promotion of PPPM in sciences and practical implementation.
Collapse
Affiliation(s)
- Olga Golubnitschaja
- Department of Radiology, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany ; The European Association for Predictive, Preventive and Personalised Medicine, 1150 Brussels, Belgium
| | - Vincenzo Costigliola
- The European Association for Predictive, Preventive and Personalised Medicine, 1150 Brussels, Belgium
| | | |
Collapse
|
50
|
Fangueiro JF, Andreani T, Fernandes L, Garcia ML, Egea MA, Silva AM, Souto EB. Physicochemical characterization of epigallocatechin gallate lipid nanoparticles (EGCG-LNs) for ocular instillation. Colloids Surf B Biointerfaces 2014; 123:452-60. [PMID: 25303852 DOI: 10.1016/j.colsurfb.2014.09.042] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/04/2014] [Accepted: 09/19/2014] [Indexed: 11/15/2022]
Abstract
The encapsulation of epigallocatechin gallate (EGCG) in lipid nanoparticles (LNs) could be a suitable approach to avoid drug oxidation and epimerization, which are common processes that lead to low bioavailability of the drug limiting its therapeutic efficacy. The human health benefits of EGCG gained much interest in the pharmaceutical field, and so far there are no studies reporting its encapsulation in LNs. The purpose of this study has been the development of an innovative system for the ocular delivery of EGCG using LNs as carrier for the future treatment of several diseases, such as dry eye, age-related macular degeneration (AMD), glaucoma, diabetic retinopathy and macular oedema. LNs dispersions have been produced by multiple emulsion technique and previously optimized by a factorial design. In order to increase ocular retention time and mucoadhesion by electrostatic attraction, two distinct cationic lipids were used, namely, cetyltrimethylammonium bromide (CTAB) and dimethyldioctadecylammonium bromide (DDAB). EGCG has been successfully loaded in the LNs dispersions and the nanoparticles analysis over 30 days of storage time predicted a good physicochemical stability. The particles were found to be in the nanometer range (<300 nm) and all the evaluated parameters, namely pH, osmolarity and viscosity, were compatible to the ocular administration. The evaluation of the cationic lipid used was compared regarding physical and chemical parameters, lipid crystallization and polymorphism, and stability of dispersion during storage. The results show that different lipids lead to different characteristics mainly associated with the acyl chain composition, i.e. double lipid shows to have influence in the crystallization and stability. Despite the recorded differences between DTAB and DDAB, both cationic LNs seem to fit the parameters for ocular drug delivery.
Collapse
Affiliation(s)
- Joana F Fangueiro
- CEBIMED, Research Centre for Biomedicine, Fernando Pessoa University, UFP-FCS, Praça 9 de Abril, 349, P-4249-004 Porto, Portugal; Faculty of Health Sciences, Fernando Pessoa University, UFP-FCS, Rua Carlos da Maia, 296, 4200-150 Porto, Portugal
| | - Tatiana Andreani
- Faculty of Health Sciences, Fernando Pessoa University, UFP-FCS, Rua Carlos da Maia, 296, 4200-150 Porto, Portugal; Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, University of Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal; Department of Biology and Environment, University of Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Lisete Fernandes
- Electron Microscopy Unit, University of Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Maria L Garcia
- Department of Physical Chemistry, Faculty of Pharmacy, University of Barcelona, Av. Joan XXIII s/n, 08028 Barcelona, Spain; Institute of Nanoscience and Nanotechnology, University of Barcelona, Av. Joan XXIII s/n, 08028 Barcelona, Spain
| | - Maria A Egea
- Department of Physical Chemistry, Faculty of Pharmacy, University of Barcelona, Av. Joan XXIII s/n, 08028 Barcelona, Spain; Institute of Nanoscience and Nanotechnology, University of Barcelona, Av. Joan XXIII s/n, 08028 Barcelona, Spain
| | - Amélia M Silva
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, University of Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal; Department of Biology and Environment, University of Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Eliana B Souto
- CEBIMED, Research Centre for Biomedicine, Fernando Pessoa University, UFP-FCS, Praça 9 de Abril, 349, P-4249-004 Porto, Portugal; Faculty of Pharmacy of University of Coimbra (FFUC), Polo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|