1
|
Jammes M, Tabasi A, Bach T, Ritter T. Healing the Cornea: Exploring the Therapeutic Solutions Offered by MSCs and MSC-derived EVs. Prog Retin Eye Res 2024:101325. [PMID: 39709150 DOI: 10.1016/j.preteyeres.2024.101325] [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: 09/18/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Affecting a large proportion of the population worldwide, corneal disorders constitute a concerning health hazard associated to compromised eyesight or blindness for most severe cases. In the last decades, mesenchymal stem/stromal cells (MSCs) demonstrated promising abilities in improving symptoms associated to corneal diseases or alleviating these affections, especially through their anti-inflammatory, immunomodulatory and pro-regenerative properties. More recently, MSC therapeutic potential was shown to be mediated by the molecules they release, and particularly by their extracellular vesicles (EVs; MSC-EVs). Consequently, using MSC-EVs emerged as a pioneering strategy to mitigate the risks related to cell therapy while providing MSC therapeutic benefits. Despite the promises given by MSC- and MSC-EV-based approaches, many improvements are considered to optimize the therapeutic significance of these therapies. This review aspires to provide a comprehensive and detailed overview of current knowledge on corneal therapies involving MSCs and MSC-EVs, the strategies currently under evaluation, and the gaps remaining to be addressed for clinical implementation. From encapsulating MSCs or their EVs into biomaterials to enhance the ocular retention time to loading MSC-EVs with therapeutic drugs, a wide range of ground-breaking strategies are currently contemplated to lead to the safest and most effective treatments. Promising research initiatives also include diverse gene therapies and the targeting of specific cell types through the modification of the EV surface, paving the way for future therapeutic innovations. As one of the most important challenges, MSC-EV large-scale production strategies are extensively investigated and offer a wide array of possibilities to meet the needs of clinical applications.
Collapse
Affiliation(s)
- Manon Jammes
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Abbas Tabasi
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Trung Bach
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland; CURAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
2
|
Chib S, Dutta BJ, Chalotra R, Abubakar M, Kumar P, Singh TG, Singh R. Role of Flavonoids in Mitigating the Pathological Complexities and Treatment Hurdles in Alzheimer's Disease. Phytother Res 2024. [PMID: 39660432 DOI: 10.1002/ptr.8406] [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: 07/10/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
With the passage of time, people step toward old age and become more prone to several diseases associated with the age. One such is Alzheimer's disease (AD) which results into neuronal damage and dementia with the progression of age. The existing therapeutics has been hindered by various enkindles like less eminent between remote populations, affordability issues and toxicity profiles. Moreover, lack of suitable therapeutic option further worsens the quality of life in older population. Developing an efficient therapeutic intervention to cure AD is still a challenge for medical fraternity. Recently, alternative approaches attain the attention of researchers to focus on plant-based therapy in mitigating AD. In this context, flavonoids gained centrality as a feasible treatment in modifying various neurological deficits. This review mainly focuses on the pathological facets and economic burden of AD. Furthermore, we have explored the possible mechanism of flavonoids with the preclinical and clinical aspects for curing AD. Flavonoids being potential therapeutic, target the pathogenic factors of AD such as oxidative stress, inflammation, metal toxicity, Aβ accumulation, modulate neurotransmission and insulin signaling. In this review, we emphasized on potential neuroprotective effects of flavonoids in AD pathology, with focus on both experimental and clinical findings. While preclinical studies suggest promising therapeutic benefits, clinical data remains limited and inconclusive. Thus, further high-quality clinical trials are necessary to validate the efficacy of flavonoids in AD. The study aim is to promote the plant-based therapies and encourage people to add flavonoids to regular diet to avail the beneficial effects in preventive therapy for AD.
Collapse
Affiliation(s)
- Shivani Chib
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Bhaskar Jyoti Dutta
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Rishabh Chalotra
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Md Abubakar
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | | | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
3
|
Zhang J, Jiang Y, Dong X, Meng Z, Ji L, Kang Y, Liu M, Zhou W, Song W. Alpha-lipoic acid alleviates cognitive deficits in transgenic APP23/PS45 mice through a mitophagy-mediated increase in ADAM10 α-secretase cleavage of APP. Alzheimers Res Ther 2024; 16:160. [PMID: 39030577 PMCID: PMC11264788 DOI: 10.1186/s13195-024-01527-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/13/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Alpha-lipoic acid (ALA) has a neuroprotective effect on neurodegenerative diseases. In the clinic, ALA can improve cognitive impairments in patients with Alzheimer's disease (AD) and other dementias. Animal studies have confirmed the anti-amyloidosis effect of ALA, but its underlying mechanism remains unclear. In particular, the role of ALA in amyloid-β precursor protein (APP) metabolism has not been fully elucidated. OBJECTIVE To investigate whether ALA can reduce the amyloidogenic effect of APP in a transgenic mouse model of AD, and to study the mechanism underlying this effect. METHODS ALA was infused into 2-month-old APP23/PS45 transgenic mice for 4 consecutive months and their cognitive function and AD-like pathology were then evaluated. An ALA drug concentration gradient was applied to 20E2 cells in vitro to evaluate its effect on the expression of APP proteolytic enzymes and metabolites. The mechanism by which ALA affects APP processing was studied using GI254023X, an inhibitor of A Disintegrin and Metalloproteinase 10 (ADAM10), as well as the mitochondrial toxic drug carbonyl cyanide m-chlorophenylhydrazone (CCCP). RESULTS Administration of ALA ameliorated amyloid plaque neuropathology in the brain tissue of APP23/PS45 mice and reduced learning and memory impairment. ALA also increased the expression of ADAM10 in 20E2 cells and the non-amyloidogenic processing of APP to produce the 83 amino acid C-terminal fragment (C83). In addition to activating autophagy, ALA also significantly promoted mitophagy. BNIP3L-knockdown reduced the mat/pro ratio of ADAM10. By using CCCP, ALA was found to regulate BNIP3L-mediated mitophagy, thereby promoting the α-cleavage of APP. CONCLUSIONS The enhanced α-secretase cleavage of APP by ADAM10 is the primary mechanism through which ALA ameliorates the cognitive deficits in APP23/PS45 transgenic mice. BNIP3L-mediated mitophagy contributes to the anti-amyloid properties of ALA by facilitating the maturation of ADAM10. This study provides novel experimental evidence for the treatment of AD with ALA.
Collapse
Affiliation(s)
- Jie Zhang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanshuang Jiang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangjun Dong
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zijun Meng
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Liangye Ji
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Kang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Mingjing Liu
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihui Zhou
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Weihong Song
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325001, China.
| |
Collapse
|
4
|
Rodnyy AY, Kondaurova EM, Tsybko AS, Popova NK, Kudlay DA, Naumenko VS. The brain serotonin system in autism. Rev Neurosci 2024; 35:1-20. [PMID: 37415576 DOI: 10.1515/revneuro-2023-0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/10/2023] [Indexed: 07/08/2023]
Abstract
Autism spectrum disorders (ASDs) are among the most common neurodevelopmental diseases. These disorders are characterized by lack of social interaction, by repetitive behavior, and often anxiety and learning disabilities. The brain serotonin (5-HT) system is known to be crucially implicated in a wide range of physiological functions and in the control of different kinds of normal and pathological behavior. A growing number of studies indicate the involvement of the brain 5-HT system in the mechanisms underlying both ASD development and ASD-related behavioral disorders. There are some review papers describing the role of separate key players of the 5-HT system in an ASD and/or autistic-like behavior. In this review, we summarize existing data on the participation of all members of the brain 5-HT system, namely, 5-HT transporter, tryptophan hydroxylase 2, MAOA, and 5-HT receptors, in autism in human and various animal models. Additionally, we describe the most recent studies involving modern techniques for in vivo regulation of gene expression that are aimed at identifying exact roles of 5-HT receptors, MAOA, and 5-HT transporter in the mechanisms underlying autistic-like behavior. Altogether, results of multiple research articles show that the brain 5-HT system intimately partakes in the control of some types of ASD-related behavior, and that specific changes in a function of a certain 5-HT receptor, transporter, and/or enzyme may normalize this aberrant behavior. These data give hope that some of clinically used 5-HT-related drugs have potential for ASD treatment.
Collapse
Affiliation(s)
- Alexander Ya Rodnyy
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| | - Elena M Kondaurova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| | - Anton S Tsybko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| | - Nina K Popova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| | - Dmitry A Kudlay
- NRC Institute of Immunology FMBA of Russia, Kashirskoe Highway 24, Moscow 115522, Russia
- Sechenov's University, 8-2 Trubetskaya Str., Moscow 119991, Russia
| | - Vladimir S Naumenko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| |
Collapse
|
5
|
Santillán-Morales V, Rodriguez-Espinosa N, Muñoz-Estrada J, Alarcón-Elizalde S, Acebes Á, Benítez-King G. Biomarkers in Alzheimer's Disease: Are Olfactory Neuronal Precursors Useful for Antemortem Biomarker Research? Brain Sci 2024; 14:46. [PMID: 38248261 PMCID: PMC10813897 DOI: 10.3390/brainsci14010046] [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: 11/16/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's disease (AD), as the main cause of dementia, affects millions of people around the world, whose diagnosis is based mainly on clinical criteria. Unfortunately, the diagnosis is obtained very late, when the neurodegenerative damage is significant for most patients. Therefore, the exhaustive study of biomarkers is indispensable for diagnostic, prognostic, and even follow-up support. AD is a multifactorial disease, and knowing its underlying pathological mechanisms is crucial to propose new and valuable biomarkers. In this review, we summarize some of the main biomarkers described in AD, which have been evaluated mainly by imaging studies in cerebrospinal fluid and blood samples. Furthermore, we describe and propose neuronal precursors derived from the olfactory neuroepithelium as a potential resource to evaluate some of the widely known biomarkers of AD and to gear toward searching for new biomarkers. These neuronal lineage cells, which can be obtained directly from patients through a non-invasive and outpatient procedure, display several characteristics that validate them as a surrogate model to study the central nervous system, allowing the analysis of AD pathophysiological processes. Moreover, the ease of obtaining and harvesting endows them as an accessible and powerful resource to evaluate biomarkers in clinical practice.
Collapse
Affiliation(s)
- Valeria Santillán-Morales
- Laboratory of Neuropharmacology, Clinical Research, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (V.S.-M.); (S.A.-E.)
| | - Norberto Rodriguez-Espinosa
- Department of Neurology, University Hospital Nuestra Señora de Candelaria, 38010 Tenerife, Spain;
- Department of Internal Medicine, Dermatology and Psychiatry, Faculty of Health Sciences, University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Jesús Muñoz-Estrada
- Department of Computational Biomedicine, Cedars Sinai Medical Center, Los Angeles, CA 90069, USA;
| | - Salvador Alarcón-Elizalde
- Laboratory of Neuropharmacology, Clinical Research, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (V.S.-M.); (S.A.-E.)
| | - Ángel Acebes
- Department of Basic Medical Sciences, Institute of Biomedical Technologies (ITB), University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Gloria Benítez-King
- Laboratory of Neuropharmacology, Clinical Research, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (V.S.-M.); (S.A.-E.)
| |
Collapse
|
6
|
Singh L, Kaur H, Chandra Arya G, Bhatti R. Neuroprotective potential of formononetin, a naturally occurring isoflavone phytoestrogen. Chem Biol Drug Des 2024; 103:e14353. [PMID: 37722967 DOI: 10.1111/cbdd.14353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/17/2023] [Accepted: 09/04/2023] [Indexed: 09/20/2023]
Abstract
The increased prevalence of neurological illnesses is a burgeoning challenge to the public healthcare system and presents greater financial pressure. Formononetin, an O-methylated isoflavone, has gained a lot of attention due to its neuroprotective potential explored in several investigations. Formononetin is widely found in legumes and several types of clovers including Trifolium pratense L., Astragalus membranaceus, Sophora tomentosa, etc. Formononetin modulates various endogenous mediators to confer neuroprotection. It prevents RAGE activation that results in the inhibition of neuronal damage via downregulating the level of ROS and proinflammatory cytokines. Furthermore, formononetin also increases the expression of ADAM-10, which affects the pathology of neurodegenerative disease by lowering tau phosphorylation, maintaining synaptic plasticity, and boosting hippocampus neurogenesis. Besides these, formononetin also increases the expression of antioxidants, Nrf-2, PI3K, ApoJ, and LRP1. Whereas, reduces the expression of p65-NF-κB and proinflammatory cytokines. It also inhibits the deposition of Aβ and MAO-B activity. An inhibition of Aβ/RAGE-induced activation of MAPK and NOX governs the protection elicited by formononetin against inflammatory and oxidative stress-induced neuronal damage. Besides this, PI3K/Akt and ER-α-mediated activation of ADAM10, ApoJ/LRP1-mediated clearance of Aβ, and MAO-B inhibition-mediated preservation of dopaminergic neurons integrity are the major modulations produced by formononetin. This review covers the biosynthesis of formononetin and key molecular pathways modulated by formononetin to confer neuroprotection.
Collapse
Affiliation(s)
- Lovedeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
- University Institute of Pharma Sciences, Chandigarh University, Mohali, India
| | - Harpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Girish Chandra Arya
- University Institute of Pharma Sciences, Chandigarh University, Mohali, India
| | - Rajbir Bhatti
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
7
|
Eremin DV, Kondaurova EM, Rodnyy AY, Molobekova CA, Kudlay DA, Naumenko VS. Serotonin Receptors as a Potential Target in the Treatment of Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:2023-2042. [PMID: 38462447 DOI: 10.1134/s0006297923120064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 03/12/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide that has an increasing impact on aging societies. Besides its critical role in the control of various physiological functions and behavior, brain serotonin (5-HT) system is involved in the regulation of migration, proliferation, differentiation, maturation, and programmed death of neurons. At the same time, a growing body of evidence indicates the involvement of 5-HT neurotransmission in the formation of insoluble aggregates of β-amyloid and tau protein, the main histopathological signs of AD. The review describes the role of various 5-HT receptors and intracellular signaling cascades induced by them in the pathological processes leading to the development of AD, first of all, in protein aggregation. Changes in the functioning of certain types of 5-HT receptors or associated intracellular signaling mediators prevent accumulation of β-amyloid plaques and tau protein neurofibrillary tangles. Based on the experimental data, it can be suggested that the use of 5-HT receptors as new drug targets will not only improve cognitive performance in AD, but will be also important in treating the causes of AD-related dementia.
Collapse
Affiliation(s)
- Dmitrii V Eremin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | - Elena M Kondaurova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Aleksander Ya Rodnyy
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Camilla A Molobekova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Dmitrii A Kudlay
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Vladimir S Naumenko
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| |
Collapse
|
8
|
Hershkovits AS, Gelley S, Hanna R, Kleifeld O, Shulman A, Fishman A. Shifting the balance: soluble ADAM10 as a potential treatment for Alzheimer's disease. Front Aging Neurosci 2023; 15:1171123. [PMID: 37266401 PMCID: PMC10229884 DOI: 10.3389/fnagi.2023.1171123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/20/2023] [Indexed: 06/03/2023] Open
Abstract
Introduction Accumulation of amyloid β in the brain is regarded as a key initiator of Alzheimer's disease pathology. Processing of the amyloid precursor protein (APP) in the amyloidogenic pathway yields neurotoxic amyloid β species. In the non-amyloidogenic pathway, APP is processed by membrane-bound ADAM10, the main α-secretase in the nervous system. Here we present a new enzymatic approach for the potential treatment of Alzheimer's disease using a soluble form of ADAM10. Methods The ability of the soluble ADAM10 to shed overexpressed and endogenous APP was determined with an ADAM10 knockout cell line and a human neuroblastoma cell line, respectively. We further examined its effect on amyloid β aggregation by thioflavin T fluorescence, HPLC, and confocal microscopy. Using N-terminal and C-terminal enrichment proteomic approaches, we identified soluble ADAM10 substrates. Finally, a truncated soluble ADAM10, based on the catalytic domain, was expressed in Escherichia coli for the first time, and its activity was evaluated. Results The soluble enzyme hydrolyzes APP and releases the neuroprotective soluble APPα when exogenously added to cell cultures. The soluble ADAM10 inhibits the formation and aggregation of characteristic amyloid β extracellular neuronal aggregates. The proteomic investigation identified new and verified known substrates, such as VGF and N-cadherin, respectively. The truncated variant also exhibited α-secretase capacity as shown with a specific ADAM10 fluorescent substrate in addition to shedding overexpressed and endogenous APP. Discussion Our in vitro study demonstrates that exogenous treatment with a soluble variant of ADAM10 would shift the balance toward the non-amyloidogenic pathway, thus utilizing its natural neuroprotective effect and inhibiting the main neurotoxic amyloid β species. The potential of such a treatment for Alzheimer's disease needs to be further evaluated in vivo.
Collapse
Affiliation(s)
- Ayelet Sarah Hershkovits
- Department of Biotechnology and Food Engineering Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Sivan Gelley
- Department of Biotechnology and Food Engineering Technion-Israel Institute of Technology, Haifa, Israel
| | - Rawad Hanna
- Department of Biology Technion-Israel Institute of Technology, Haifa, Israel
| | - Oded Kleifeld
- Department of Biology Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Ayelet Fishman
- Department of Biotechnology and Food Engineering Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
9
|
Urriola-Muñoz P, Pattison LA, Smith ESJ. Dysregulation of ADAM10 shedding activity in naked mole-rat fibroblasts is due to deficient phosphatidylserine externalization. J Cell Physiol 2023; 238:761-775. [PMID: 36790936 DOI: 10.1002/jcp.30972] [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: 10/02/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/16/2023]
Abstract
The naked mole-rat (NMR, Heterocephalus glaber) is of significant interest to biogerontological research, rarely developing age-associated diseases, such as cancer. The transmembrane glycoprotein CD44 is upregulated in certain cancers and CD44 cleavage by a disintegrin and metalloproteinase 10 (ADAM10) regulates cellular migration. Here we provide evidence that mature ADAM10 is expressed in NMR primary skin fibroblasts (NPSF), and that ionomycin increases cell surface ADAM10 localization. However, we observed an absence of ADAM10 mediated CD44 cleavage, as well as shedding of exogenous and overexpressed betacellulin in NPSF, whereas in mouse primary skin fibroblasts ionomycin induced ADAM10-dependent cleavage of both CD44 and betacellulin. Overexpressing a hyperactive form of the Ca2+ -dependent phospholipid scramblase ANO6 in NPSF increased phosphatidylserine (PS) externalization, which rescued the ADAM10 sheddase activity and promoted cell migration in NPSF in an ADAM10-dependent manner. These findings suggest that dysregulation of ADAM10 shedding activity is due to a deficient PS externalization in NMR.
Collapse
Affiliation(s)
| | - Luke A Pattison
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
A Disintegrin and Metalloproteinase 10 (ADAM10) Is Essential for Oligodendrocyte Precursor Development and Myelination in the Mouse Brain. Mol Neurobiol 2023; 60:1675-1689. [PMID: 36550333 PMCID: PMC9899191 DOI: 10.1007/s12035-022-03163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
A disintegrin and metalloproteinase 10 (ADAM10) plays an essential role in the regulation of survival, proliferation, migration, and differentiation of various neural cells. Nevertheless, the role of ADAM10 in oligodendrocyte precursors (OPCs) and myelination in the central nervous system (CNS) of developing and adult mouse brains is still unknown. We generated ADAM10 conditional knockout (ADAM10 cKO) mice lacking the ADAM10 gene primarily in OPCs by crossing NG2-Cre mice with ADAM10 loxp/loxp mice. We found that OPCs expressed ADAM10 in the mouse corpus callosum and the hippocampus. ADAM10 cKO mice showed significant loss of back hair and reduction in weight and length on postnatal (30 ± 2.1) day, died at (65 ± 5) days after birth, and exhibited the "anxiety and depression-like" performances. Conditional knockout of ADAM10 in OPCs resulted in a prominent increase in myelination and a decrease in the number of OPCs in the corpus callosum at P30 owing to premyelination and lack of proliferation of OPCs. Moreover, the number of proliferating OPCs and mature oligodendrocytes (OLs) also decreased with age in the corpus callosum of ADAM10 cKO mice from P30 to P60. Western blot and RT-PCR results showed that the activation of Notch-1 and its four target genes, Hes1, Hes5, Hey1, and Hey2, was inhibited in the corpus callosum tissue of ADAM10 knockout mice. In our study, we provided experimental evidence to demonstrate that ADAM10 is essential for modulating CNS myelination and OPC development by activating Notch-1 signaling in the developing and adult mouse brain.
Collapse
|
11
|
Bok J, Ha J, Ahn BJ, Jang Y. Disease-Modifying Effects of Non-Invasive Electroceuticals on β-Amyloid Plaques and Tau Tangles for Alzheimer's Disease. Int J Mol Sci 2022; 24:ijms24010679. [PMID: 36614120 PMCID: PMC9821138 DOI: 10.3390/ijms24010679] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Electroceuticals refer to various forms of electronic neurostimulators used for therapy. Interdisciplinary advances in medical engineering and science have led to the development of the electroceutical approach, which involves therapeutic agents that specifically target neural circuits, to realize precision therapy for Alzheimer's disease (AD). To date, extensive studies have attempted to elucidate the disease-modifying effects of electroceuticals on areas in the brain of a patient with AD by the use of various physical stimuli, including electric, magnetic, and electromagnetic waves as well as ultrasound. Herein, we review non-invasive stimulatory systems and their effects on β-amyloid plaques and tau tangles, which are pathological molecular markers of AD. Therefore, this review will aid in better understanding the recent technological developments, applicable methods, and therapeutic effects of electronic stimulatory systems, including transcranial direct current stimulation, 40-Hz gamma oscillations, transcranial magnetic stimulation, electromagnetic field stimulation, infrared light stimulation and ionizing radiation therapy, and focused ultrasound for AD.
Collapse
Affiliation(s)
- Junsoo Bok
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
| | - Juchan Ha
- Department of Biomedical Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
| | - Bum Ju Ahn
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, Republic of Korea
| | - Yongwoo Jang
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, Republic of Korea
- Correspondence: ; Tel.: +82-2-2220-0655
| |
Collapse
|
12
|
Maglione AV, do Nascimento BPP, Ribeiro MO, de Souza TJL, da Silva REC, Sato MA, Penatti CAA, Britto LRG, de Souza JS, Maciel RMB, da Conceição RR, Laureano-Melo R, Giannocco G. Triiodothyronine Treatment reverses Depression-Like Behavior in a triple-transgenic animal model of Alzheimer's Disease. Metab Brain Dis 2022; 37:2735-2750. [PMID: 35951206 DOI: 10.1007/s11011-022-01055-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022]
Abstract
Alzheimer disease's (AD) is a neurodegenerative disorder characterized by cognitive and behavioral impairment. The central nervous system is an important target of thyroid hormones (TH). An inverse association between serum triiodothyronine (T3) levels and the risk of AD symptoms and progression has been reported. We investigated the effects of T3 treatment on the depression-like behavior in male transgenic 3xTg-AD mice. Animals were divided into 2 groups treated with daily intraperitoneal injections of 20 ng/g of body weight (b.w.) L-T3 (T3 group) or saline (vehicle, control group). The experimental protocol lasted 21 days, and behavioral tests were conducted on days 18-20. At the end of the experiment, the TH profile and hippocampal gene expression were evaluated. The T3-treated group significantly increased serum T3 and decreased thyroxine (T4) levels. When compared to control hippocampal samples, the T3 group exhibited attenuated glycogen synthase kinase-3 (GSK3), metalloproteinase 10 (ADAM10), amyloid-beta precursor-protein (APP), serotonin transporter (SERT), 5HT1A receptor, monocarboxylate transporter 8 (MCT8) and bone morphogenetic protein 7 (BMP-7) gene expression, whereas augmented superoxide dismutase 2 (SOD2) and Hairless gene expression. T3-treated animals also displayed reduced immobility time in both the tail suspension and forced swim tests, and in the latter presented a higher latency time compared to the control group. Therefore, our findings suggest that in an AD mouse model, T3 supplementation promotes improvements in depression-like behavior, through the modulation of the serotonergic related genes involved in the transmission mediated by 5HT1A receptors and serotonin reuptake, and attenuated disease progression.
Collapse
Affiliation(s)
- Andréa V Maglione
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Bruna P P do Nascimento
- Laboratory of Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
- Developmental Disorders Program, Center of Biological Science and Health, Mackenzie Presbyterian University, São Paulo, Brazil
| | - Miriam O Ribeiro
- Developmental Disorders Program, Center of Biological Science and Health, Mackenzie Presbyterian University, São Paulo, Brazil
| | - Talytha J L de Souza
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Renata E C da Silva
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Monica A Sato
- Dept. Morphology and Physiology, Faculdade de Medicina do ABC, Centro Universitário FMABC, Santo André- Brazil, São Paulo, Santo André, Brazil
| | - Carlos A A Penatti
- Laboratory of Human Physiology, Universidade Nove de Julho, São Paulo, Brazil
| | - Luiz R G Britto
- Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Janaina S de Souza
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Rui M B Maciel
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Rodrigo Rodrigues da Conceição
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil.
| | - Roberto Laureano-Melo
- Laboratory of Physiopharmacoly and Behavior, Universidade de Barra Mansa, Rio de Janeiro, Brazil
| | - Gisele Giannocco
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil.
| |
Collapse
|
13
|
Aydin N, Turkez H, Tozlu OO, Arslan ME, Yavuz M, Sonmez E, Ozpolat OF, Cacciatore I, Di Stefano A, Mardinoglu A. Ameliorative Effects by Hexagonal Boron Nitride Nanoparticles against Beta Amyloid Induced Neurotoxicity. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12152690. [PMID: 35957121 PMCID: PMC9370266 DOI: 10.3390/nano12152690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 05/28/2023]
Abstract
Alzheimer’s disease (AD) is considered as the most common neurodegenerative disease. Extracellular amyloid beta (Aβ) deposition is a hallmark of AD. The options based on degradation and clearance of Aβ are preferred as promising therapeutic strategies for AD. Interestingly, recent findings indicate that boron nanoparticles not only act as a carrier but also play key roles in mediating biological effects. In the present study, the aim was to investigate the effects of different concentrations (0−500 mg/L) of hexagonal boron nitride nanoparticles (hBN-NPs) against neurotoxicity by beta amyloid (Aβ1-42) in differentiated human SH-SY5Y neuroblastoma cell cultures for the first time. The synthesized hBN-NPs were characterized by X-ray diffraction (XRD) measurements, scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Aβ1-42-induced neurotoxicity and therapeutic potential by hBN-NPs were assessed on differentiated SH-SY5Y cells using MTT and LDH release assays. Levels of total antioxidant capacity (TAC) and total oxidant status (TOS), expression levels of genes associated with AD and cellular morphologies were examined. The exposure to Aβ1-42 significantly decreased the rates of viable cells which was accompanied by elevated TOS level. Aβ1-42 induced both apoptotic and necrotic cell death. Aβ exposure led to significant increases in expression levels of APOE, BACE 1, EGFR, NCTSN and TNF-α genes and significant decreases in expression levels of ADAM 10, APH1A, BDNF, PSEN1 and PSENEN genes (p < 0.05). All the Aβ1-42-induced neurotoxic insults were inhibited by the applications with hBN-NPs. hBN-NPs also suppressed the remarkable elevation in the signal for Aβ following exposure to Aβ1-42 for 48 h. Our results indicated that hBN-NPs could significantly prevent the neurotoxic damages by Aβ. Thus, hBN-NPs could be a novel and promising anti-AD agent for effective drug development, bio-nano imaging or drug delivery strategies.
Collapse
Affiliation(s)
- Nursah Aydin
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25050, Turkey
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey
- East Anatolia High Technology Application and Research Center (DAYTAM), Ataturk University, Erzurum 25240, Turkey
| | - Ozlem Ozdemir Tozlu
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25050, Turkey
| | - Mehmet Enes Arslan
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25050, Turkey
| | - Mehmet Yavuz
- REEM Neuropsychiatry Clinics, İstanbul 34245, Turkey
| | - Erdal Sonmez
- Department of Nanoscience and Nanoengineering, Graduate School of Natural and Applied Sciences, Ataturk University, Erzurum 25240, Turkey
- Department of Physics, Kazım Karabekir Education Faculty, Atatürk University, Erzurum 25240, Turkey
| | - Ozgur Fırat Ozpolat
- Computer Sciences Research and Application Center, Atatürk University, Erzurum 25240, Turkey
| | - Ivana Cacciatore
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti Scalo, CH, Italy
| | - Antonio Di Stefano
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti Scalo, CH, Italy
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, SE-17121 Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| |
Collapse
|
14
|
Sirin S, Nigdelioglu Dolanbay S, Aslim B. The relationship of early- and late-onset Alzheimer’s disease genes with COVID-19. J Neural Transm (Vienna) 2022; 129:847-859. [PMID: 35429259 PMCID: PMC9012910 DOI: 10.1007/s00702-022-02499-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/02/2022] [Indexed: 12/13/2022]
Abstract
Individuals with Alzheimer’s disease and other neurodegenerative diseases have been exposed to excess risk by the COVID-19 pandemic. COVID-19’s main manifestations include high body temperature, dry cough, and exhaustion. Nevertheless, some affected individuals may have an atypical presentation at diagnosis but suffer neurological signs and symptoms as the first disease manifestation. These findings collectively show the neurotropic nature of SARS-CoV-2 virus and its ability to involve the central nervous system. In addition, Alzheimer’s disease and COVID-19 has a number of common risk factors and comorbid conditions including age, sex, hypertension, diabetes, and the expression of APOE ε4. Until now, a plethora of studies have examined the COVID-19 disease but only a few studies has yet examined the relationship of COVID-19 and Alzheimer’s disease as risk factors of each other. This review emphasizes the recently published evidence on the role of the genes of early- or late-onset Alzheimer’s disease in the susceptibility of individuals currently suffering or recovered from COVID-19 to Alzheimer’s disease or in the susceptibility of individuals at risk of or with Alzheimer’s disease to COVID-19 or increased COVID-19 severity and mortality. Furthermore, the present review also draws attention to other uninvestigated early- and late-onset Alzheimer’s disease genes to elucidate the relationship between this multifactorial disease and COVID-19.
Collapse
|
15
|
Musardo S, Therin S, Pelucchi S, D'Andrea L, Stringhi R, Ribeiro A, Manca A, Balducci C, Pagano J, Sala C, Verpelli C, Grieco V, Edefonti V, Forloni G, Gardoni F, Meli G, Di Marino D, Di Luca M, Marcello E. The development of ADAM10 endocytosis inhibitors for the treatment of Alzheimer's disease. Mol Ther 2022; 30:2474-2490. [PMID: 35390543 DOI: 10.1016/j.ymthe.2022.03.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/15/2021] [Accepted: 03/31/2022] [Indexed: 10/18/2022] Open
Abstract
The development of new therapeutic avenues that target the early stages of Alzheimer's disease (AD) is urgently necessary. ADAM10 is a sheddase that is involved in dendritic spine shaping and limits the generation of amyloid-β. ADAM10 endocytosis increases in the hippocampus of AD patients, resulting in the decreased postsynaptic localization of the enzyme. To restore this altered pathway, we developed a cell-permeable peptide (PEP3) with a strong safety profile that is able to interfere with ADAM10 endocytosis, upregulating the postsynaptic localization and activity of ADAM10. After extensive validation, experiments in a relevant animal model clarified the optimal timing of the treatment window. PEP3 administration was effective for the rescue of cognitive defects in APP/PS1 mice only if administered at an early disease stage. Increased ADAM10 activity promoted synaptic plasticity, as revealed by changes in the molecular compositions of synapses and the spine morphology. Even though further studies are required to evaluate efficacy and safety issues of long-term administration of PEP3, these results provide preclinical evidence to support the therapeutic potential of PEP3 in AD.
Collapse
Affiliation(s)
- Stefano Musardo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Sebastien Therin
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Laura D'Andrea
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Ana Ribeiro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Annalisa Manca
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Jessica Pagano
- CNR Neuroscience Institute, Via Raoul Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Carlo Sala
- CNR Neuroscience Institute, Via Raoul Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Via Raoul Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Valeria Grieco
- Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Via Dell'Università 6, 26900 Lodi, Italy
| | - Valeria Edefonti
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry, and Epidemiology "G.A. Maccacaro", Universita` degli Studi di Milano, via Celoria 22, 20133 Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Giovanni Meli
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Daniele Di Marino
- Department of Life and Environmental Sciences, New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy.
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy.
| |
Collapse
|
16
|
Scramblases as Regulators of Proteolytic ADAM Function. MEMBRANES 2022; 12:membranes12020185. [PMID: 35207106 PMCID: PMC8880048 DOI: 10.3390/membranes12020185] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/25/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022]
Abstract
Proteolytic ectodomain release is a key mechanism for regulating the function of many cell surface proteins. The sheddases ADAM10 and ADAM17 are the best-characterized members of the family of transmembrane disintegrin-like metalloproteinase. Constitutive proteolytic activities are low but can be abruptly upregulated via inside-out signaling triggered by diverse activating events. Emerging evidence indicates that the plasma membrane itself must be assigned a dominant role in upregulation of sheddase function. Data are discussed that tentatively identify phospholipid scramblases as central players during these events. We propose that scramblase-dependent externalization of the negatively charged phospholipid phosphatidylserine (PS) plays an important role in the final activation step of ADAM10 and ADAM17. In this manuscript, we summarize the current knowledge on the interplay of cell membrane changes, PS exposure, and proteolytic activity of transmembrane proteases as well as the potential consequences in the context of immune response, infection, and cancer. The novel concept that scramblases regulate the action of ADAM-proteases may be extendable to other functional proteins that act at the cell surface.
Collapse
|
17
|
Pelucchi S, Gardoni F, Di Luca M, Marcello E. Synaptic dysfunction in early phases of Alzheimer's Disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:417-438. [PMID: 35034752 DOI: 10.1016/b978-0-12-819410-2.00022-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The synapse is the locus of plasticity where short-term alterations in synaptic strength are converted to long-lasting memories. In addition to the presynaptic terminal and the postsynaptic compartment, a more holistic view of the synapse includes the astrocytes and the extracellular matrix to form a tetrapartite synapse. All these four elements contribute to synapse health and are crucial for synaptic plasticity events and, thereby, for learning and memory processes. Synaptic dysfunction is a common pathogenic trait of several brain disorders. In Alzheimer's Disease, the degeneration of synapses can be detected at the early stages of pathology progression before neuronal degeneration, supporting the hypothesis that synaptic failure is a major determinant of the disease. The synapse is the place where amyloid-β peptides are generated and is the target of the toxic amyloid-β oligomers. All the elements constituting the tetrapartite synapse are altered in Alzheimer's Disease and can synergistically contribute to synaptic dysfunction. Moreover, the two main hallmarks of Alzheimer's Disease, i.e., amyloid-β and tau, act in concert to cause synaptic deficits. Deciphering the mechanisms underlying synaptic dysfunction is relevant for the development of the next-generation therapeutic strategies aimed at modifying the disease progression.
Collapse
Affiliation(s)
- Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
18
|
Lichtenthaler SF, Tschirner SK, Steiner H. Secretases in Alzheimer's disease: Novel insights into proteolysis of APP and TREM2. Curr Opin Neurobiol 2021; 72:101-110. [PMID: 34689040 DOI: 10.1016/j.conb.2021.09.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
Secretases are a group of proteases that are major drug targets considered for the prevention and treatment of Alzheimer's disease (AD). Secretases do not only process the AD-linked neuronal amyloid precursor protein (APP) but also the triggering receptor expressed on myeloid cells 2 (TREM2), thereby controlling microglial functions. This review highlights selected recent discoveries for the α-secretases a disintegrin and metalloprotease 10 (ADAM10) and a disintegrin and metalloprotease 17 (ADAM17), the β-secretase β-site APP cleaving enzyme 1 (BACE1) and γ-secretase and their link to AD. New genetic evidence strengthens the role of α-secretases in AD through cleavage of APP and TREM2. Novel proteins were linked to AD, which control α- and β-secretase activity through transcriptional and post-translational mechanisms. Finally, new opportunities but also challenges are discussed for pharmacologically targeting β- and γ-secretase cleavage of APP and α-secretase cleavage of TREM2 with the aim to prevent or treat AD.
Collapse
Affiliation(s)
- Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Sarah K Tschirner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Harald Steiner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, LMU Munich, Germany.
| |
Collapse
|
19
|
D’Andrea L, Stringhi R, Di Luca M, Marcello E. Looking at Alzheimer's Disease Pathogenesis from the Nuclear Side. Biomolecules 2021; 11:biom11091261. [PMID: 34572474 PMCID: PMC8467578 DOI: 10.3390/biom11091261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 01/22/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder representing the most common form of dementia. It is biologically characterized by the deposition of extracellular amyloid-β (Aβ) senile plaques and intracellular neurofibrillary tangles, constituted by hyperphosphorylated tau protein. The key protein in AD pathogenesis is the amyloid precursor protein (APP), which is cleaved by secretases to produce several metabolites, including Aβ and APP intracellular domain (AICD). The greatest genetic risk factor associated with AD is represented by the Apolipoprotein E ε4 (APOE ε4) allele. Importantly, all of the above-mentioned molecules that are strictly related to AD pathogenesis have also been described as playing roles in the cell nucleus. Accordingly, evidence suggests that nuclear functions are compromised in AD. Furthermore, modulation of transcription maintains cellular homeostasis, and alterations in transcriptomic profiles have been found in neurodegenerative diseases. This report reviews recent advancements in the AD players-mediated gene expression. Aβ, tau, AICD, and APOE ε4 localize in the nucleus and regulate the transcription of several genes, part of which is involved in AD pathogenesis, thus suggesting that targeting nuclear functions might provide new therapeutic tools for the disease.
Collapse
|
20
|
Ma D, Li Y, Zhu Y, Wei W, Zhang L, Li Y, Li L, Zhang L. Cornel Iridoid Glycoside Ameliorated Alzheimer's Disease-Like Pathologies and Necroptosis through RIPK1/MLKL Pathway in Young and Aged SAMP8 Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:9920962. [PMID: 34475966 PMCID: PMC8407981 DOI: 10.1155/2021/9920962] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/17/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Aging is an important risk factor for sporadic Alzheimer's disease (AD) and other neurodegenerative diseases. Senescence-accelerated mouse-prone 8 (SAMP8) is used as an animal model for brain aging and sporadic AD research studies. The aim of the current study was to investigate the pharmacological effects of cornel iridoid glycoside (CIG), an active ingredient of Cornus officinalis, on AD-type pathological changes in young and aged SAMP8 mice. METHODS Locomotor activity test was used to detect the aging process of SAMP8 mice. Nissl staining and immunohistochemical staining were applied to detect neurons and myelin basic protein-labelled myelin sheath. Western blotting was used to detect the expression levels of related proteins of synapse, APP processing, and necroptosis. RESULTS The results showed that SAMP8 mice at the age of 6 and 14 months exhibited lower locomotor activity, age-related neuronal loss, demyelination, synaptic damage, and APP amyloidogenic processing. In addition, the increased levels of receptor-interacting protein kinase-1 (RIPK1), mixed lineage kinase domain-like protein (MLKL), and p-MLKL indicating necroptosis were found in the brain of SAMP8 mice. Intragastric administration of CIG for 2 months improved locomotor activity; alleviated neuronal loss and demyelination; increased the expression of synaptophysin, postsynaptic density protein 95, and AMPA receptor subunit 1; elevated the levels of soluble APPα fragment and disintegrin and metalloproteinase 10 (ADAM10); and decreased the levels of RIPK1, p-MLKL, and MLKL in the brain of young and aged SAMP8 mice. CONCLUSION This study denoted that CIG might be a potential drug for aging-related neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Denglei Ma
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Yanzheng Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
- Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Yanqiu Zhu
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Weipeng Wei
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Yali Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| |
Collapse
|
21
|
Hsia HE, Tüshaus J, Feng X, Hofmann LI, Wefers B, Marciano DK, Wurst W, Lichtenthaler SF. Endoglycan (PODXL2) is proteolytically processed by ADAM10 (a disintegrin and metalloprotease 10) and controls neurite branching in primary neurons. FASEB J 2021; 35:e21813. [PMID: 34390512 DOI: 10.1096/fj.202100475r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/22/2021] [Accepted: 07/07/2021] [Indexed: 01/24/2023]
Abstract
Cell adhesion is tightly controlled in multicellular organisms, for example, through proteolytic ectodomain shedding of the adhesion-mediating cell surface transmembrane proteins. In the brain, shedding of cell adhesion proteins is required for nervous system development and function, but the shedding of only a few adhesion proteins has been studied in detail in the mammalian brain. One such adhesion protein is the transmembrane protein endoglycan (PODXL2), which belongs to the CD34-family of highly glycosylated sialomucins. Here, we demonstrate that endoglycan is broadly expressed in the developing mouse brains and is proteolytically shed in vitro in mouse neurons and in vivo in mouse brains. Endoglycan shedding in primary neurons was mediated by the transmembrane protease a disintegrin and metalloprotease 10 (ADAM10), but not by its homolog ADAM17. Functionally, endoglycan deficiency reduced the branching of neurites extending from primary neurons in vitro, whereas deletion of ADAM10 had the opposite effect and increased neurite branching. Taken together, our study discovers a function for endoglycan in neurite branching, establishes endoglycan as an ADAM10 substrate and suggests that ADAM10 cleavage of endoglycan may contribute to neurite branching.
Collapse
Affiliation(s)
- Hung-En Hsia
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Xiao Feng
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Laura I Hofmann
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg/Munich, Germany
| | - Denise K Marciano
- Departments of Cell Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg/Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Technical University of Munich-Weihenstephan, Neuherberg/Munich, Neuherberg, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
22
|
Redox-Active Metal Ions and Amyloid-Degrading Enzymes in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22147697. [PMID: 34299316 PMCID: PMC8307724 DOI: 10.3390/ijms22147697] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
Redox-active metal ions, Cu(I/II) and Fe(II/III), are essential biological molecules for the normal functioning of the brain, including oxidative metabolism, synaptic plasticity, myelination, and generation of neurotransmitters. Dyshomeostasis of these redox-active metal ions in the brain could cause Alzheimer’s disease (AD). Thus, regulating the levels of Cu(I/II) and Fe(II/III) is necessary for normal brain function. To control the amounts of metal ions in the brain and understand the involvement of Cu(I/II) and Fe(II/III) in the pathogenesis of AD, many chemical agents have been developed. In addition, since toxic aggregates of amyloid-β (Aβ) have been proposed as one of the major causes of the disease, the mechanism of clearing Aβ is also required to be investigated to reveal the etiology of AD clearly. Multiple metalloenzymes (e.g., neprilysin, insulin-degrading enzyme, and ADAM10) have been reported to have an important role in the degradation of Aβ in the brain. These amyloid degrading enzymes (ADE) could interact with redox-active metal ions and affect the pathogenesis of AD. In this review, we introduce and summarize the roles, distributions, and transportations of Cu(I/II) and Fe(II/III), along with previously invented chelators, and the structures and functions of ADE in the brain, as well as their interrelationships.
Collapse
|
23
|
Koh YQ, Ng DQ, Ng CC, Boey A, Wei M, Sze SK, Ho HK, Acharya M, Limoli CL, Chan A. Extracellular Vesicle Proteome of Breast Cancer Patients with and Without Cognitive Impairment Following Anthracycline-based Chemotherapy: An Exploratory Study. Biomark Insights 2021; 16:11772719211018204. [PMID: 34103887 PMCID: PMC8150437 DOI: 10.1177/11772719211018204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
Cognitive impairment due to cancer and its therapy is a major concern among cancer patients and survivors. Extracellular vesicle (EVs) composition altered by cancer and chemotherapy may affect neurological processes such as neuroplasticity, potentially impacting the cognitive abilities of cancer patients and survivors. We investigated the EV proteome of breast cancer patients with and without cognitive impairment following anthracycline-based chemotherapy from longitudinally collected plasma. EVs were cup-shaped and positive for Flotillin-1 and TSG-101. We identified 517 differentially expressed EV proteins between the cognitive impaired and non-impaired groups during and post-chemotherapy. The observed decreased expression of p2X purinoceptor, cofilin-1, ADAM 10, and dynamin-1 in the plasma EVs of the cognitive impaired group may suggest alterations in the mechanisms underlying synaptic plasticity. The reduced expression of tight junction proteins among cognitive-impaired patients may imply weakening of the blood-brain barrier. These EV protein signatures may serve as a fingerprint that underscores the mechanisms underlying cognitive impairment in cancer patients and survivors.
Collapse
Affiliation(s)
- Yong Qin Koh
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, University of California, Irvine, CA, USA
| | - Chiu Chin Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Adrian Boey
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Meng Wei
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Munjal Acharya
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, University of California, Irvine, CA, USA.,Department of Oncology Pharmacy, National Cancer Centre Singapore, Singapore
| |
Collapse
|
24
|
Noori T, Dehpour AR, Sureda A, Sobarzo-Sanchez E, Shirooie S. Role of natural products for the treatment of Alzheimer's disease. Eur J Pharmacol 2021; 898:173974. [PMID: 33652057 DOI: 10.1016/j.ejphar.2021.173974] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 12/19/2022]
Abstract
Negative psychological and physiological consequences of neurodegenerative disorders represent a high social and health cost. Among the neurodegenerative disorders Alzheimer's disease (AD) is recognized as a leading neurodegenerative condition and a primary cause of dementia in the elderlys. AD is considered as neurodegenerative disorder that progressively impairs cognitive function and memory. According to current epidemiological data, about 50 milLion people worldwide are suffering from AD. The primary symptoms of AD are almost inappreciable and usually comprise forgetfulness of recent events. Numerous processes are involved in the development of AD, for example oxidative stress (OS) mainly due to mitochondrial dysfunction, intracellular the accumulation of hyperphosphorylated tau (τ) proteins in the form of neurofibrillary tangles, excessive the accumulation of extracellular plaques of beta-amyloid (Aβ), genetic and environmental factors. Running treatments only attenuate symptoms and temporarily reduce the rate of cognitive progression associated with AD. This means that most treatments focus only on controlLing symptoms, particularly in the initial stages of the disease. In the past, the first choice of treatment was based on natural ingredients. In this sense, diverse natural products (NPs) are capable to decrease the symptoms and alleviate the development of several diseases including AD attracting the attention of the scientific community and the pharmaceutical industry. Specifically, numerous NPs including flavonoids, gingerols, tannins, anthocyanins, triterpenes and alkaloids have been shown anti-inflammatory, antioxidant, anti-amyloidogenic, and anti-choLinesterase properties. This review provide a summary of the pathogenesis and the therapeutic goals of AD. It also discusses the available data on various plants and isolated natural compounds used to prevent and diminish the symptoms of AD.
Collapse
Affiliation(s)
- Tayebeh Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, TUMS, Tehran, Iran
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX), University Research Institute of Health Sciences (IUNICS), and Health Research Institute of Balearic Islands (IdISBa), University of Balearic Islands, Palma de Mallorca E-07122, Balearic Islands, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Eduardo Sobarzo-Sanchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile; Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Spain
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
25
|
Wang H, Muthu Karuppan MK, Nair M, Lakshmana MK. Autophagy-Dependent Increased ADAM10 Mature Protein Induced by TFEB Overexpression Is Mediated Through PPARα. Mol Neurobiol 2021; 58:2269-2283. [PMID: 33417226 DOI: 10.1007/s12035-020-02230-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Nonamyloidogenic processing of amyloid precursor protein (APP) by augmenting ADAM10 is a promising therapeutic strategy for Alzheimer's disease (AD). Therefore identification of molecular pathways that regulate ADAM10 expression is crucial. Autophagy is strongly dysregulated in AD, and TFEB was recently shown to be a master regulator of autophagy-lysosome pathway (ALP). Here, we report that TFEB expression in HeLa cells increased ADAM10 mature form by 72% (p < 0.01, n = 4), while TFEB knockdown by CRISPR strategy reduced ADAM10 mature form by 36% (p < 0.05, n = 4). Autophagy inhibition by 3-methyladenine (3-MA), but not bafilomycin A1 (BAF1), reduced ADAM10 mature form by 49% (p < 0.05, n = 4) in the TFEB expressing HeLa cells. Autophagy activation by 3 h of starvation increased ADAM10 to 91% (p < 0.001, n = 6) relative to 51% (p < 0.01, n = 6) in the nutrient-fed cells. Further, siRNAs targeted against PPARα in HeLa cells decreased ADAM10 levels by 28% (p < 0.05, n = 6) relative to the cells treated with scrambled siRNAs. Further, incubation of EGFP-TFEB expressing HeLa cells with PPARα antagonist, but not PPARβ or PPARγ antagonists, prevented TFEB-induced increase in ADAM10 levels. Importantly, flag-TFEB expression in the brain also increased ADAM10 by 60% (p < 0.05, n = 3) in the cortical and 34% (p < 0.001, n = 3) in the hippocampal homogenates. ADAM10 activity also increased by 57% (p < 0.01, n = 3) in the HeLa cells. Finally, TFEB-induced ADAM10 potentiation led to increased secretion of sAPPα by 154% (p < 0.001, n = 3) in the cortex and 62% (p < 0.001, n = 3) in the hippocampus. Thus, TFEB expression enhances nonamyloidogenic processing of APP. In conclusion, TFEB expression induces ADAM10 in an autophagy-dependent manner through PPARα.
Collapse
Affiliation(s)
- Hongjie Wang
- Institute for Human Health & Disease Intervention (I-HEALTH), Department of Chemistry and Biochemistry, Center for Molecular Biology and Biotechnology, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Mohan Kumar Muthu Karuppan
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200, 8th Street, University Park, Miami, FL, 33199, USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200, 8th Street, University Park, Miami, FL, 33199, USA
| | - Madepalli K Lakshmana
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200, 8th Street, University Park, Miami, FL, 33199, USA.
| |
Collapse
|
26
|
Zheng Y, Verhoeff TA, Perez Pardo P, Garssen J, Kraneveld AD. The Gut-Brain Axis in Autism Spectrum Disorder: A Focus on the Metalloproteases ADAM10 and ADAM17. Int J Mol Sci 2020; 22:ijms22010118. [PMID: 33374371 PMCID: PMC7796333 DOI: 10.3390/ijms22010118] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a spectrum of disorders that are characterized by problems in social interaction and repetitive behavior. The disease is thought to develop from changes in brain development at an early age, although the exact mechanisms are not known yet. In addition, a significant number of people with ASD develop problems in the intestinal tract. A Disintegrin And Metalloproteases (ADAMs) include a group of enzymes that are able to cleave membrane-bound proteins. ADAM10 and ADAM17 are two members of this family that are able to cleave protein substrates involved in ASD pathogenesis, such as specific proteins important for synapse formation, axon signaling and neuroinflammation. All these pathological mechanisms are involved in ASD. Besides the brain, ADAM10 and ADAM17 are also highly expressed in the intestines. ADAM10 and ADAM17 have implications in pathways that regulate gut permeability, homeostasis and inflammation. These metalloproteases might be involved in microbiota-gut-brain axis interactions in ASD through the regulation of immune and inflammatory responses in the intestinal tract. In this review, the potential roles of ADAM10 and ADAM17 in the pathology of ASD and as targets for new therapies will be discussed, with a focus on the gut-brain axis.
Collapse
Affiliation(s)
- Yuanpeng Zheng
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Tessa A. Verhoeff
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Paula Perez Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
- Global Centre of Excellence Immunology, Danone Nutricia Research B.V., 3584CT Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
- Correspondence: ; Tel.: +31-(0)3-02534509
| |
Collapse
|
27
|
Wang ZG, He ZY, Liang S, Yang Q, Cheng P, Chen AM. Comprehensive proteomic analysis of exosomes derived from human bone marrow, adipose tissue, and umbilical cord mesenchymal stem cells. Stem Cell Res Ther 2020; 11:511. [PMID: 33246507 PMCID: PMC7694919 DOI: 10.1186/s13287-020-02032-8] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stem cell (MSC)-derived exosomes have shown comprehensive application prospects over the years. Despite performing similar functions, exosomes from different origins present heterogeneous characteristics and components; however, the relative study remains scarce. Lacking of a valuable reference, researchers select source cells for exosome studies mainly based on accessibility and personal preference. Methods In this study, exosomes secreted by MSCs derived from different tissues were isolated, by ultracentrifugation, and proteomics analysis was performed. A total of 1014 proteins were detected using a label-free method. Results Bioinformatics analysis revealed their shared function in the extracellular matrix receptor. Bone marrow MSC-derived exosomes showed superior regeneration ability, and adipose tissue MSC-derived exosomes played a significant role in immune regulation, whereas umbilical cord MSC-derived exosomes were more prominent in tissue damage repair. Conclusions This study systematically and comprehensively analyzes the human MSC-derived exosomes via proteomics, which reveals their potential applications in different fields, so as to provide a reference for researchers to select optimal source cells in future exosome-related studies. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-02032-8.
Collapse
Affiliation(s)
- Zheng-Gang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Zhi-Yi He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Shuang Liang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Qing Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China.
| | - An-Min Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People's Republic of China.
| |
Collapse
|
28
|
Target Enzymes Considered for the Treatment of Alzheimer's Disease and Parkinson's Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2010728. [PMID: 33224974 PMCID: PMC7669341 DOI: 10.1155/2020/2010728] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/15/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022]
Abstract
Various amyloidogenic proteins have been suggested to be involved in the onset and progression of neurodegenerative diseases (ND) such as Alzheimer's disease (AD) and Parkinson's disease (PD). Particularly, the aggregation of misfolded amyloid-β and hyperphosphorylated tau and α-synuclein are linked to the pathogenesis of AD and PD, respectively. In order to care the diseases, multiple small molecules have been developed to regulate the aggregation pathways of these amyloid proteins. In addition to controlling the aggregation of amyloidogenic proteins, maintaining the levels of the proteins in the brain by amyloid degrading enzymes (ADE; neprilysin (NEP), insulin-degrading enzyme (IDE), asparagine endopeptidase (AEP), and ADAM10) is also essential to cure AD and PD. Therefore, numerous biological molecules and chemical agents have been investigated as either inducer or inhibitor against the levels and activities of ADE. Although the side effect of enhancing the activity of ADE could occur, the removal of amyloidogenic proteins could result in a relatively good strategy to treat AD and PD. Furthermore, since the causes of ND are diverse, various multifunctional (multitarget) chemical agents have been designed to control the actions of multiple risk factors of ND, including amyloidogenic proteins, metal ions, and reactive oxygen species. Many of them, however, were invented without considerations of regulating ADE levels and actions. Incorporation of previously created molecules with the chemical agents handling ADE could be a promising way to treat AD and PD. This review introduces the ADE and molecules capable of modulating the activity and expression of ADE.
Collapse
|
29
|
Cuffaro D, Nuti E, D’Andrea F, Rossello A. Developments in Carbohydrate-Based Metzincin Inhibitors. Pharmaceuticals (Basel) 2020; 13:ph13110376. [PMID: 33182755 PMCID: PMC7696829 DOI: 10.3390/ph13110376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 01/03/2023] Open
Abstract
Matrix metalloproteinases (MMPs) and A disintegrin and Metalloproteinase (ADAMs) are zinc-dependent endopeptidases belonging to the metzincin superfamily. Upregulation of metzincin activity is a major feature in many serious pathologies such as cancer, inflammations, and infections. In the last decades, many classes of small molecules have been developed directed to inhibit these enzymes. The principal shortcomings that have hindered clinical development of metzincin inhibitors are low selectivity for the target enzyme, poor water solubility, and long-term toxicity. Over the last 15 years, a novel approach to improve solubility and bioavailability of metzincin inhibitors has been the synthesis of carbohydrate-based compounds. This strategy consists of linking a hydrophilic sugar moiety to an aromatic lipophilic scaffold. This review aims to describe the development of sugar-based and azasugar-based derivatives as metzincin inhibitors and their activity in several pathological models.
Collapse
|
30
|
Yumoto T, Kimura M, Nagatomo R, Sato T, Utsunomiya S, Aoki N, Kitaura M, Takahashi K, Takemoto H, Watanabe H, Okano H, Yoshida F, Nao Y, Tomita T. Autism-associated variants of neuroligin 4X impair synaptogenic activity by various molecular mechanisms. Mol Autism 2020; 11:68. [PMID: 32873342 PMCID: PMC7465329 DOI: 10.1186/s13229-020-00373-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Several genetic alterations, including point mutations and copy number variations in NLGN genes, have been associated with psychiatric disorders, such as autism spectrum disorder (ASD) and X-linked mental retardation (XLMR). NLGN genes encode neuroligin (NL) proteins, which are adhesion molecules that are important for proper synaptic formation and maturation. Previously, we and others found that the expression level of murine NL1 is regulated by proteolytic processing in a synaptic activity-dependent manner. METHODS In this study, we analyzed the effects of missense variants associated with ASD and XLMR on the metabolism and function of NL4X, a protein which is encoded by the NLGN4X gene and is expressed only in humans, using cultured cells, primary neurons from rodents, and human induced pluripotent stem cell-derived neurons. RESULTS NL4X was found to undergo proteolytic processing in human neuronal cells. Almost all NL4X variants caused a substantial decrease in the levels of mature NL4X and its synaptogenic activity in a heterologous culture system. Intriguingly, the L593F variant of NL4X accelerated the proteolysis of mature NL4X proteins located on the cell surface. In contrast, other variants decreased the cell-surface trafficking of NL4X. Notably, protease inhibitors as well as chemical chaperones rescued the expression of mature NL4X. LIMITATIONS Our study did not reveal whether these dysfunctional phenotypes occurred in individuals carrying NLGN4X variant. Moreover, though these pathological mechanisms could be exploited as potential drug targets for ASD, it remains unclear whether these compounds would have beneficial effects on ASD model animals and patients. CONCLUSIONS These data suggest that reduced amounts of the functional NL4X protein on the cell surface is a common mechanism by which point mutants of the NL4X protein cause psychiatric disorders, although different molecular mechanisms are thought to be involved. Furthermore, these results highlight that the precision medicine approach based on genetic and cell biological analyses is important for the development of therapeutics for psychiatric disorders.
Collapse
Affiliation(s)
- Takafumi Yumoto
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Misaki Kimura
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Ryota Nagatomo
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tsukika Sato
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Shun Utsunomiya
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi, Osaka, Japan
- Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Natsue Aoki
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi, Osaka, Japan
- Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Motoji Kitaura
- Research Administration SPRC, R&D General Administration Unit, General Administration Division, Shionogi Administration Service, Osaka, Japan
| | - Koji Takahashi
- Drug Discovery Technology 3, Laboratory for Innovative Therapy Research, Shionogi, Osaka, Japan
| | - Hiroshi Takemoto
- Neuroscience 2, Laboratory for Drug Discovery and Disease Research, Shionogi, Osaka, Japan
- Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Hirotaka Watanabe
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Fumiaki Yoshida
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yosuke Nao
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
31
|
Xie Z, Shen P, Qu Y, Xu J, Zheng C, Gao Y, Wang B. MiR-20a inhibits the progression of human arthritis fibroblast-like synoviocytes and inflammatory factor expression by targeting ADAM10. ENVIRONMENTAL TOXICOLOGY 2020; 35:867-878. [PMID: 32198911 DOI: 10.1002/tox.22923] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 06/10/2023]
Abstract
MiR-20a has been reported as a key regulator to pro-inflammatory factor release in fibroblast-like synoviocytes (FLS), which caused rheumatoid arthritis (RA). However, the molecular mechanism of miR-20a in RA remains to be further elucidated. This study aimed to investigate the roles of miR-20a in RA pathology. RA (n = 24) and osteoarthritis (OA, n = 20) and normal healthy tissues (n = 16) were collected from operation. TargetScan and dual-luciferase reporter were performed to predict and confirm the potential binding sites of miR-20a on ADAM metallopeptidase domain 10 (ADAM10). Pearson's analysis was adopted to evaluate the correlation between miR-20a and ADAM10 expression. It was found that MiR-20a was downregulated in RA tissues, and overexpressed miR-20a inhibited cell viability, migration and invasion, and the expression of inflammatory factors in RA-FLS MH7A cells. ADAM10 was identified as the target gene of miR-20a, and upregulation of ADAM10 reversed the inhibitory effects of miR-20a. In conclusion, miR-20a inhibits the progression of RA-FLS as well as the inflammatory factor expression by targeting ADAM10.
Collapse
Affiliation(s)
- Zikang Xie
- Department of Orthopedics, Changzhou Traditional Chinese Medicine Hospital, Changzhou, Jiangsu, China
| | - Pengfei Shen
- Department of Orthopedics, Changzhou Traditional Chinese Medicine Hospital, Changzhou, Jiangsu, China
| | - Yuxing Qu
- Department of Orthopedics, Changzhou Traditional Chinese Medicine Hospital, Changzhou, Jiangsu, China
| | - Jianda Xu
- Department of Orthopedics, Changzhou Traditional Chinese Medicine Hospital, Changzhou, Jiangsu, China
| | - Chong Zheng
- Department of Orthopedics, Changzhou Traditional Chinese Medicine Hospital, Changzhou, Jiangsu, China
| | - Yi Gao
- Department of Orthopedics, Changzhou Traditional Chinese Medicine Hospital, Changzhou, Jiangsu, China
| | - Bin Wang
- Department of Orthopedics, Changzhou Traditional Chinese Medicine Hospital, Changzhou, Jiangsu, China
| |
Collapse
|
32
|
Bleibaum F, Sommer A, Veit M, Rabe B, Andrä J, Kunzelmann K, Nehls C, Correa W, Gutsmann T, Grötzinger J, Bhakdi S, Reiss K. ADAM10 sheddase activation is controlled by cell membrane asymmetry. J Mol Cell Biol 2020; 11:979-993. [PMID: 30753537 PMCID: PMC6927242 DOI: 10.1093/jmcb/mjz008] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 11/20/2018] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Dysregulation of the disintegrin-metalloproteinase ADAM10 may contribute to the development of diseases including tumorigenesis and Alzheimer's disease. The mechanisms underlying ADAM10 sheddase activation are incompletely understood. Here, we show that transient exposure of the negatively charged phospholipid phosphatidylserine (PS) is necessarily required. The soluble PS headgroup was found to act as competitive inhibitor of substrate cleavage. Overexpression of the Ca2+-dependent phospholipid scramblase Anoctamin-6 (ANO6) led to increased PS externalization and substrate release. Transfection with a constitutively active form of ANO6 resulted in maximum sheddase activity in the absence of any stimulus. Calcium-dependent ADAM10 activation could not be induced in lymphocytes of patients with Scott syndrome harbouring a missense mutation in ANO6. A putative PS-binding motif was identified in the conserved stalk region. Replacement of this motif resulted in strong reduction of sheddase activity. In conjunction with the recently described 3D structure of the ADAM10 extracellular domain, a model is advanced to explain how surface-exposed PS triggers ADAM10 sheddase function.
Collapse
Affiliation(s)
| | - Anselm Sommer
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - Martin Veit
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - Björn Rabe
- Institute of Biochemistry, University of Kiel, Olshausenstraße 40, Kiel, Germany
| | - Jörg Andrä
- Hamburg University of Applied Science, Ulmenliet 20, Hamburg, Germany
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, Universitätsstraße 31, Regensburg, Germany
| | - Christian Nehls
- Forschungszentrum Borstel, Leibniz-Zentrum für Medizin und Biowissenschaften, Parkallee 10, Borstel, Germany
| | - Wilmar Correa
- Forschungszentrum Borstel, Leibniz-Zentrum für Medizin und Biowissenschaften, Parkallee 10, Borstel, Germany
| | - Thomas Gutsmann
- Forschungszentrum Borstel, Leibniz-Zentrum für Medizin und Biowissenschaften, Parkallee 10, Borstel, Germany
| | - Joachim Grötzinger
- Institute of Biochemistry, University of Kiel, Olshausenstraße 40, Kiel, Germany
| | - Sucharit Bhakdi
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - Karina Reiss
- Department of Dermatology, University of Kiel, Kiel, Germany
| |
Collapse
|
33
|
Brummer T, Müller SA, Pan-Montojo F, Yoshida F, Fellgiebel A, Tomita T, Endres K, Lichtenthaler SF. NrCAM is a marker for substrate-selective activation of ADAM10 in Alzheimer's disease. EMBO Mol Med 2020; 11:emmm.201809695. [PMID: 30833305 PMCID: PMC6460357 DOI: 10.15252/emmm.201809695] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The metalloprotease ADAM10 is a drug target in Alzheimer's disease, where it cleaves the amyloid precursor protein (APP) and lowers amyloid‐beta. Yet, ADAM10 has additional substrates, which may cause mechanism‐based side effects upon therapeutic ADAM10 activation. However, they may also serve—in addition to APP—as biomarkers to monitor ADAM10 activity in patients and to develop APP‐selective ADAM10 activators. Our study demonstrates that one such substrate is the neuronal cell adhesion protein NrCAM. ADAM10 controlled NrCAM surface levels and regulated neurite outgrowth in vitro in an NrCAM‐dependent manner. However, ADAM10 cleavage of NrCAM, in contrast to APP, was not stimulated by the ADAM10 activator acitretin, suggesting that substrate‐selective ADAM10 activation may be feasible. Indeed, a whole proteome analysis of human CSF from a phase II clinical trial showed that acitretin, which enhanced APP cleavage by ADAM10, spared most other ADAM10 substrates in brain, including NrCAM. Taken together, this study demonstrates an NrCAM‐dependent function for ADAM10 in neurite outgrowth and reveals that a substrate‐selective, therapeutic ADAM10 activation is possible and may be monitored with NrCAM.
Collapse
Affiliation(s)
- Tobias Brummer
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stephan A Müller
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany
| | - Francisco Pan-Montojo
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Fumiaki Yoshida
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Andreas Fellgiebel
- Department of Psychiatry and Psychotherapy, University Medical Center JGU, Mainz, Germany
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center JGU, Mainz, Germany
| | - Stefan F Lichtenthaler
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany .,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute for Advanced Study, Technische Universität München, Garching, Germany
| |
Collapse
|
34
|
Manohar S, Russo FY, Seigel GM, Salvi R. Dynamic Changes in Synaptic Plasticity Genes in Ipsilateral and Contralateral Inferior Colliculus Following Unilateral Noise-induced Hearing Loss. Neuroscience 2020; 436:136-153. [PMID: 32278721 DOI: 10.1016/j.neuroscience.2020.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/07/2020] [Accepted: 04/03/2020] [Indexed: 12/27/2022]
Abstract
Unilateral noise-induced hearing loss reduces the input to the central auditory pathway disrupting the excitatory and inhibitory inputs to the inferior colliculus (IC), an important binaural processing center. Little is known about the compensatory synaptic changes that occur in the IC as a consequence of unilateral noise-induced hearing loss. To address this issue, Sprague-Dawley rats underwent unilateral noise exposure resulting in severe unilateral hearing loss. IC tissues from the contralateral and ipsilateral IC were evaluated for acute (2-d) and chronic (28-d) changes in the expression of 84 synaptic plasticity genes on a PCR array. Arc and Egr1 genes were further visualized by in situ hybridization to validate the PCR results. None of the genes were upregulated, but many were downregulated post-exposure. At 2-d post-exposure, more than 75% of the genes were significantly downregulated in the contralateral IC, while only two were downregulated in the ipsilateral IC. Many of the downregulated genes were related to long-term depression, long-term potentiation, cell adhesion, immediate early genes, neural receptors and postsynaptic density. At 28-d post-exposure, the gene expression pattern was reversed with more than 85% of genes in the ipsilateral IC now downregulated. Most genes previously downregulated in the contralateral IC 2-d post-exposure had recovered; less than 15% remained downregulated. These time-dependent, asymmetric changes in synaptic plasticity gene expression could shed new light on the perceptual deficits associated with unilateral hearing loss and the dynamic structural and functional changes that occur in the IC days and months following unilateral noise-induced hearing loss.
Collapse
Affiliation(s)
| | | | - Gail M Seigel
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY 14214, USA
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
35
|
Shang P, Zhang Y, Ma D, Hao Y, Wang X, Xin M, Zhang Y, Zhu M, Feng J. Inflammation resolution and specialized pro-resolving lipid mediators in CNS diseases. Expert Opin Ther Targets 2019; 23:967-986. [PMID: 31711309 DOI: 10.1080/14728222.2019.1691525] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Inflammation resolution induced by specialized pro-resolving lipid mediators (SPMs) is a new concept. The application of SPMs is a promising therapeutic strategy that can potentially supersede anti-inflammatory drugs. Most CNS diseases are associated with hyperreactive inflammatory damage. CNS inflammation causes irreversible neuronal loss and permanent functional impairments. Given the high mortality and morbidity rates, the investigation of therapeutic strategies to ameliorate inflammatory damage is necessary.Areas covered: In this review, we explore inflammation resolution in CNS disorders. We discuss the underlying mechanisms and dynamic changes of SPMs and their precursors in neurological diseases and examine how this can potentially be incorporated into the clinic. References were selected from PubMed; most were published between 2010 and 2019.Expert opinion: Inflammation resolution is a natural process that emerges after acute or chronic inflammation. The evidence that SPMs can effectively ameliorate hyperreactive inflammation, shorten resolution time and accelerate tissue regeneration in CNS disorders. Adjuvants and nanotechnology offer opportunities for SPM drug design; however, more preclinical studies are necessary to investigate basic, critical issues such as safety.
Collapse
Affiliation(s)
- Pei Shang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Di Ma
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yulei Hao
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyu Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Meiying Xin
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yunhai Zhang
- Jiangsu Key Laboratory of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Mingqin Zhu
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiachun Feng
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
36
|
Castro MA, Hadziselimovic A, Sanders CR. The vexing complexity of the amyloidogenic pathway. Protein Sci 2019; 28:1177-1193. [PMID: 30897251 PMCID: PMC6566549 DOI: 10.1002/pro.3606] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/18/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
Abstract
The role of the amyloidogenic pathway in the etiology of Alzheimer's disease (AD), particularly the common sporadic late onset forms of the disease, is controversial. To some degree, this is a consequence of the failure of drug and therapeutic antibody trials based either on targeting the proteases in this pathway or its amyloid end products. Here, we explore the formidable complexity of the biochemistry and cell biology associated with this pathway. For example, we review evidence that the immediate precursor of amyloid-β, the C99 domain of the amyloid precursor protein (APP), may itself be toxic. We also review important new results that appear to finally establish a direct genetic link between mutations in APP and the sporadic forms of AD. Based on the complexity of amyloidogenesis, it seems possible that a major contributor to the failure of related drug trials is that we have an incomplete understanding of this pathway and how it is linked to Alzheimer's pathogenesis. If so, this highlights a need for further characterization of this pathway, not its abandonment.
Collapse
Affiliation(s)
- Manuel A. Castro
- Departments of Biochemistry and MedicineVanderbilt University School of MedicineNashvilleTennessee 37240
| | - Arina Hadziselimovic
- Departments of Biochemistry and MedicineVanderbilt University School of MedicineNashvilleTennessee 37240
| | - Charles R. Sanders
- Departments of Biochemistry and MedicineVanderbilt University School of MedicineNashvilleTennessee 37240
| |
Collapse
|
37
|
Mahasenan KV, Ding D, Gao M, Nguyen TT, Suckow MA, Schroeder VA, Wolter WR, Chang M, Mobashery S. In Search of Selectivity in Inhibition of ADAM10. ACS Med Chem Lett 2018; 9:708-713. [PMID: 30034605 DOI: 10.1021/acsmedchemlett.8b00163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
The metalloproteinase ADAM10 has been reported as an important target for drug discovery in several human diseases. In this vein, (6S,7S)-N-hydroxy-5-methyl-6-(4-(5-(trifluoromethyl)pyridin-2-yl)piperazine-1-carbonyl)-5-azaspiro[2.5]octane-7-carboxamide (compound 1) has been reported as a selective ADAM10 inhibitor. We synthesized this compound and document that it lacks both potency and selectivity in inhibition of ADAM10. This finding necessitated a structure-based computational analysis to investigate potency and selectivity of ADAM10 inhibition. The model that emerged indeed excluded compound 1 as an inhibitor for ADAM10, while suggesting another reported compound, (1R,3S,4S)-3-(hydroxycarbamoyl)-4-(4-phenylpiperidine-1-carbonyl)cyclohexyl pyrrolidine-1-carboxylate (compound 2), as an ADAM10 selective inhibitor. Compound 2 was synthesized and its potency, and selectivity in inhibition of ADAM10 were documented with a panel of several related enzymes. Pharmacokinetic studies of compound 2 in mice documented that the compound crosses the blood-brain barrier and may be useful as a pharmacological agent or mechanistic tool to delineate the role of ADAM10 in neurological diseases.
Collapse
Affiliation(s)
- Kiran V. Mahasenan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Derong Ding
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Ming Gao
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Trung T. Nguyen
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mark A. Suckow
- Freimann Life Science Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Valerie A. Schroeder
- Freimann Life Science Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - William R. Wolter
- Freimann Life Science Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
38
|
Alpha-Secretase ADAM10 Regulation: Insights into Alzheimer's Disease Treatment. Pharmaceuticals (Basel) 2018; 11:ph11010012. [PMID: 29382156 PMCID: PMC5874708 DOI: 10.3390/ph11010012] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/23/2018] [Accepted: 01/23/2018] [Indexed: 02/07/2023] Open
Abstract
ADAM (a disintegrin and metalloproteinase) is a family of widely expressed, transmembrane and secreted proteins of approximately 750 amino acids in length with functions in cell adhesion and proteolytic processing of the ectodomains of diverse cell-surface receptors and signaling molecules. ADAM10 is the main α-secretase that cleaves APP (amyloid precursor protein) in the non-amyloidogenic pathway inhibiting the formation of β-amyloid peptide, whose accumulation and aggregation leads to neuronal degeneration in Alzheimer’s disease (AD). ADAM10 is a membrane-anchored metalloprotease that sheds, besides APP, the ectodomain of a large variety of cell-surface proteins including cytokines, adhesion molecules and notch. APP cleavage by ADAM10 results in the production of an APP-derived fragment, sAPPα, which is neuroprotective. As increased ADAM10 activity protects the brain from β-amyloid deposition in AD, this strategy has been proved to be effective in treating neurodegenerative diseases, including AD. Here, we describe the physiological mechanisms regulating ADAM10 expression at different levels, aiming to propose strategies for AD treatment. We report in this review on the physiological regulation of ADAM10 at the transcriptional level, by epigenetic factors, miRNAs and/or translational and post-translational levels. In addition, we describe the conditions that can change ADAM10 expression in vitro and in vivo, and discuss how this knowledge may help in AD treatment. Regulation of ADAM10 is achieved by multiple mechanisms that include transcriptional, translational and post-translational strategies, which we will summarize in this review.
Collapse
|