1
|
Ashvin, Dhapola R, Kumari S, Sharma P, Vellingiri B, Medhi B, HariKrishnaReddy D. Unraveling the Immune Puzzle: Role of Immunomodulation in Alzheimer's Disease. J Neuroimmune Pharmacol 2025; 20:47. [PMID: 40299221 DOI: 10.1007/s11481-025-10210-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder with growing evidence highlighting the dual role of immunomodulation in its pathogenesis and potential therapeutic strategies. Disturbance in the immune system increases the inflammatory cytokines that cause tau hyperphosphorylation and neuroinflammation. Also, immune checkpoint inhibition further increases the amyloid-beta deposition. Therefore, this review examines the intricate interplay between the immune system and AD, focusing on how immunomodulatory mechanisms influence key pathological hallmarks, including amyloid-beta aggregation, tau hyperphosphorylation, neuroinflammation, and cholinergic dysfunction. We analyse critical signaling pathways involved in immune regulation, such as Toll-like receptor (TLR), Janus kinase/signal transducer and activator of transcription (JAK/STAT), phosphoinositide 3-kinase/Akt (PI3K/Akt), Wnt/β-catenin, tumor necrosis factor (TNF), and triggering receptor expressed on myeloid cells (TREM), along with immune checkpoints like programmed cell death protein 1 (PD-1). Preclinical studies of immunomodulatory agents, including salidroside, festidinol, astragalin, sulforaphane, BM-MSC, simvastatin, Ab-T1, hTREM2, and XENP345, demonstrate promising effects. Additionally, clinical investigations of drugs such as simufilam, AL002, TB006, VGL101, DNL919, XPro1595, astragalus, and IBC-Ab002 underscore the therapeutic potential of targeting immune pathways in AD. This review emphasizes how neuroinflammation, microglial activation, and peripheral immune responses contribute to disease progression. By exploring immunomodulatory mechanisms, the article sheds light on potential therapeutic targets that could help mitigate AD pathology which may pave the way for novel interventions preventing neurodegeneration in AD.
Collapse
Affiliation(s)
- Ashvin
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
2
|
Iglesias-Martínez-Almeida M, Campos-Ríos A, Freiría-Martínez L, Rivera-Baltanás T, Rodrígues-Amorím D, Diz-Chaves Y, Comis-Tuche M, Fernández-Palleiro P, Rodríguez-Jamardo C, Ramos-García S, Rodríguez-Tébar A, Del Carmen Vallejo-Curto M, Campos-Pérez JA, López-García M, de Las Heras E, García-Caballero A, Olivares JM, Lamas JA, Spuch C. Characterization and modulation of voltage-gated potassium channels in human lymphocytes in schizophrenia. Schizophr Res 2024; 270:260-272. [PMID: 38944972 DOI: 10.1016/j.schres.2024.06.034] [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: 11/09/2023] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND It is known that the immune system is dysregulated in schizophrenia, having a state similar to chronic neuroinflammation. The origin of this process is unknown, but it is known that T and B lymphocytes, which are components of the adaptive immune system, play an important role in the pathogenic mechanisms of schizophrenia. METHODS We analysed the membrane of PBMCs from patients diagnosed with schizophrenia through proteomic analysis (n = 5 schizophrenia and n = 5 control). We found the presence of the Kv1.3 voltage-gated potassium channel and its auxiliary subunit β1 (KCNAB1) and β2 (KCNAB2). From a sample of 90 participants, we carried out a study on lymphocytes with whole-cell patch-clamp experiments (n = 7 schizophrenia and n = 5 control), western blot (n = 40 schizophrenia and n = 40 control) and confocal microscopy to evaluate the presence and function of different channels. Kv in both cells. RESULTS We demonstrated the overexpression of Kv1.1, Kv1.2, Kv1.3, Kv1.6, Kv4.2, Kv4.3 and Kv7.2 channels in PBMCs from patients with schizophrenia. This study represents a groundbreaking exploration, as it involves an electrophysiological analysis performed on T and B lymphocytes from patients diagnosed of schizophrenia compared to healthy participants. We observed that B lymphocytes exhibited an increase in output current along with greater peak current amplitude and voltage conductance curves among patients with schizophrenia compared with healthy controls. CONCLUSIONS This study showed the importance of the B lymphocyte in schizophrenia. We know that the immune system is altered in schizophrenia, but the physiological mechanisms of this system are not very well known. We suggest that the B lymphocyte may be relevant in the pathophysiology of schizophrenia and that it should be investigated in more depth, opening a new field of knowledge and possibilities for new treatments combining antipsychotics and immunomodulators. The limitation is that all participants received antipsychotic medication, which may have influenced the differences observed between patients and controls. This implies that more studies need to be done where the groups can be separated according to the antipsychotic drug.
Collapse
Affiliation(s)
- Marta Iglesias-Martínez-Almeida
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; University of Vigo, Vigo, Spain
| | - Ana Campos-Ríos
- Laboratory of neuroscience, University of Vigo, Department of Functional Biology and Health Sciences, Vigo, Spain; Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), SERGAS-UVIGO, Vigo, Spain
| | - Luis Freiría-Martínez
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; University of Vigo, Vigo, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; University of Vigo, Vigo, Spain; CIBERSAM, Madrid, Spain
| | - Daniela Rodrígues-Amorím
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States
| | | | - María Comis-Tuche
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Cynthia Rodríguez-Jamardo
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; University of Vigo, Vigo, Spain
| | - Silvia Ramos-García
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | | | | | - Jose Antonio Campos-Pérez
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Marta López-García
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Elena de Las Heras
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Alejandro García-Caballero
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Jose M Olivares
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; CIBERSAM, Madrid, Spain
| | - Jose A Lamas
- Laboratory of neuroscience, University of Vigo, Department of Functional Biology and Health Sciences, Vigo, Spain; Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), SERGAS-UVIGO, Vigo, Spain
| | - Carlos Spuch
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; CIBERSAM, Madrid, Spain.
| |
Collapse
|
3
|
Cheng S, Jiang D, Lan X, Liu K, Fan C. Voltage-gated potassium channel 1.3: A promising molecular target in multiple disease therapy. Biomed Pharmacother 2024; 175:116651. [PMID: 38692062 DOI: 10.1016/j.biopha.2024.116651] [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: 02/20/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
Voltage-gated potassium channel 1.3 (Kv1.3) has emerged as a pivotal player in numerous biological processes and pathological conditions, sparking considerable interest as a potential therapeutic target across various diseases. In this review, we present a comprehensive examination of Kv1.3 channels, highlighting their fundamental characteristics and recent advancements in utilizing Kv1.3 inhibitors for treating autoimmune disorders, neuroinflammation, and cancers. Notably, Kv1.3 is prominently expressed in immune cells and implicated in immune responses and inflammation associated with autoimmune diseases and chronic inflammatory conditions. Moreover, its aberrant expression in certain tumors underscores its role in cancer progression. While preclinical studies have demonstrated the efficacy of Kv1.3 inhibitors, their clinical translation remains pending. Molecular imaging techniques offer promising avenues for tracking Kv1.3 inhibitors and assessing their therapeutic efficacy, thereby facilitating their development and clinical application. Challenges and future directions in Kv1.3 inhibitor research are also discussed, emphasizing the significant potential of targeting Kv1.3 as a promising therapeutic strategy across a spectrum of diseases.
Collapse
Affiliation(s)
- Sixuan Cheng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Kun Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Cheng Fan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
4
|
Rayala H, Kapur J, El-Haija HA, Parikh P, Zawar I. Memantine's double duty? Investigating its impact on epilepsy control in Alzheimer's disease. Epileptic Disord 2024; 26:261-263. [PMID: 38334219 PMCID: PMC11656195 DOI: 10.1002/epd2.20202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Affiliation(s)
- Harika Rayala
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jaideep Kapur
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Brain Institute, University of Virginia, Charlottesville, Virginia, USA
| | | | - Prachi Parikh
- Duke University Medical Center, Durham, North Carolina, USA
| | - Ifrah Zawar
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
5
|
Schiller K, Berrahmoune S, Dassi C, Corriveau I, Ayash TA, Osterman B, Poulin C, Shevell MI, Simard-Tremblay E, Sébire G, Myers KA. Randomized placebo-controlled crossover trial of memantine in children with epileptic encephalopathy. Brain 2023; 146:873-879. [PMID: 36256600 DOI: 10.1093/brain/awac380] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/07/2022] [Accepted: 09/25/2022] [Indexed: 02/04/2023] Open
Abstract
Memantine is an N-methyl-D-aspartate receptor antagonist, approved for dementia treatment. There is limited evidence of memantine showing benefit for paediatric neurodevelopmental phenotypes, but no randomized placebo-controlled trials in children with developmental and epileptic encephalopathy. In this randomized double-blind placebo-controlled crossover trial (Trial registration: https://clinicaltrials.gov/ct2/show/NCT03779672), patients with developmental and epileptic encephalopathy received memantine and placebo, each for a 6-week period separated by a 2-week washout phase. Electroencephalography, seizure diary, patient caregivers' global impression, serum inflammatory markers and neuropsychological evaluation were performed at baseline and after each treatment phase. The primary outcome measure was classification as a 'responder', defined as ≥2 of: >50% seizure frequency reduction, electroencephalography improvement, caregiver clinical impression improvement or clear neuropsychological testing improvement. Thirty-one patients (13 females) enrolled. Two patients withdrew prior to initiating medication and two (twins) had to be removed from analysis. Of the remaining 27 patients, nine (33%) were classified as responders to memantine versus two (7%) in the placebo group (P < 0.02). Electroencephalography improvement was seen in eight patients on memantine compared to two on placebo (P < 0.04). Seizure improvement was observed in eight patients on memantine and two on placebo (P < 0.04). Caregivers reported overall clinical improvement in 10 patients on memantine compared to seven on placebo (not significant). Statistical analysis of neuropsychological evaluation suggested improvements in symptoms of attention-deficit hyperactivity disorder and autism. Memantine is a safe and effective treatment for children with developmental and epileptic encephalopathy, having the potential to improve both seizure control and cognitive function.
Collapse
Affiliation(s)
- Katharina Schiller
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Saoussen Berrahmoune
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Christelle Dassi
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Isabelle Corriveau
- Department of Psychology, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Taghreed A Ayash
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Department of Pediatrics, McGill University, Montreal, QC H4A 3J1, Canada
| | - Bradley Osterman
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Chantal Poulin
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Michael I Shevell
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Elisabeth Simard-Tremblay
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Guillaume Sébire
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Kenneth A Myers
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
6
|
Targeted drug delivery systems to control neuroinflammation in central nervous system disorders. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
7
|
Maqoud F, Scala R, Hoxha M, Zappacosta B, Tricarico D. ATP-sensitive potassium channel subunits in the neuroinflammation: novel drug targets in neurodegenerative disorders. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:130-149. [PMID: 33463481 DOI: 10.2174/1871527320666210119095626] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/07/2020] [Accepted: 08/28/2020] [Indexed: 11/22/2022]
Abstract
Arachidonic acids and its metabolites modulate plenty of ligand-gated, voltage-dependent ion channels, and metabolically regulated potassium channels including ATP-sensitive potassium channels (KATP). KATP channels are hetero-multimeric complexes of sulfonylureas receptors (SUR1, SUR2A or SUR2B) and the pore-forming subunits (Kir6.1 and Kir6.2) likewise expressed in the pre-post synapsis of neurons and inflammatory cells, thereby affecting their proliferation and activity. KATP channels are involved in amyloid-β (Aβ)-induced pathology, therefore emerging as therapeutic targets against Alzheimer's and related diseases. The modulation of these channels can represent an innovative strategy for the treatment of neurodegenerative disorders; nevertheless, the currently available drugs are not selective for brain KATP channels and show contrasting effects. This phenomenon can be a consequence of the multiple physiological roles of the different varieties of KATP channels. Openings of cardiac and muscular KATP channel subunits, is protective against caspase-dependent atrophy in these tissues and some neurodegenerative disorders, whereas in some neuroinflammatory diseases benefits can be obtained through the inhibition of neuronal KATP channel subunits. For example, glibenclamide exerts an anti-inflammatory effect in respiratory, digestive, urological, and central nervous system (CNS) diseases, as well as in ischemia-reperfusion injury associated with abnormal SUR1-Trpm4/TNF-α or SUR1-Trpm4/ Nos2/ROS signaling. Despite this strategy is promising, glibenclamide may have limited clinical efficacy due to its unselective blocking action of SUR2A/B subunits also expressed in cardiovascular apparatus with pro-arrhythmic effects and SUR1 expressed in pancreatic beta cells with hypoglycemic risk. Alternatively, neuronal selective dual modulators showing agonist/antagonist actions on KATP channels can be an option.
Collapse
Affiliation(s)
- Fatima Maqoud
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, via Orabona 4, 70125-I. Italy
| | - Rosa Scala
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, via Orabona 4, 70125-I. Italy
| | - Malvina Hoxha
- Department of Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, "Catholic University Our Lady of Good Counsel", Tirana. Albania
| | - Bruno Zappacosta
- Department of Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, "Catholic University Our Lady of Good Counsel", Tirana. Albania
| | - Domenico Tricarico
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, via Orabona 4, 70125-I. Italy
| |
Collapse
|
8
|
Drugs Modulating CD4+ T Cells Blood-Brain Barrier Interaction in Alzheimer's Disease. Pharmaceutics 2020; 12:pharmaceutics12090880. [PMID: 32948022 PMCID: PMC7558445 DOI: 10.3390/pharmaceutics12090880] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022] Open
Abstract
The effect of Alzheimer’s disease (AD) medications on CD4+ T cells homing has not been thoroughly investigated. CD4+ T cells could both exacerbate and reduce AD symptoms based on their infiltrating subpopulations. Proinflammatory subpopulations such as Th1 and Th17 constitute a major source of proinflammatory cytokines that reduce endothelial integrity and stimulate astrocytes, resulting in the production of amyloid β. Anti-inflammatory subpopulations such as Th2 and Tregs reduce inflammation and regulate the function of Th1 and Th17. Recently, pathogenic Th17 has been shown to have a superior infiltrating capacity compared to other major CD4+ T cell subpopulations. Alzheimer’s drugs such as donepezil (Aricept), rivastigmine (Exelon), galantamine (Razadyne), and memantine (Namenda) are known to play an important part in regulating the mechanisms of the neurotransmitters. However, little is known about the effect of these drugs on CD4+ T cell subpopulations’ infiltration of the brain during AD. In this review, we focus on understanding the influence of AD drugs on CD4+ T cell subpopulation interactions with the BBB in AD. While current AD therapies improve endothelial integrity and reduce astrocytes activations, they vary according to their influence on various CD4+ T cell subpopulations. Donepezil reduces the numbers of Th1 but not Th2, Rivastigmine inhibits Th1 and Th17 but not Th2, and memantine reduces Th1 but not Treg. However, none of the current AD drugs is specifically designed to target the dysregulated balance in the Th17/Treg axis. Future drug design approaches should specifically consider inhibiting CD4+ Th17 to improve AD prognosis.
Collapse
|
9
|
Villa C, Suphesiz H, Combi R, Akyuz E. Potassium channels in the neuronal homeostasis and neurodegenerative pathways underlying Alzheimer's disease: An update. Mech Ageing Dev 2019; 185:111197. [PMID: 31862274 DOI: 10.1016/j.mad.2019.111197] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/27/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023]
Abstract
With more than 80 subunits, potassium (K+) channels represent a group of ion channels showing high degree of diversity and ubiquity. They play important role in the control of membrane depolarization and cell excitability in several tissues, including the brain. Controlling the intracellular and extracellular K+ flow in cells, they also modulate the hormone and neurotransmitter release, apoptosis and cell proliferation. It is therefore not surprising that an improper functioning of K+ channels in neurons has been associated with pathophysiology of a wide range of neurological disorders, especially Alzheimer's disease (AD). This review aims to give a comprehensive overview of the basic properties and pathophysiological functions of the main classes of K+ channels in the context of disease processes, also discussing the progress, challenges and opportunities to develop drugs targeting these channels as potential pharmacological approach for AD treatment.
Collapse
Affiliation(s)
- Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | | | - Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Enes Akyuz
- Yozgat Bozok University, Medical Faculty, Department of Biophysics, Yozgat, Turkey.
| |
Collapse
|
10
|
Hsiao HT, Lee YC, Liu YC, Kuo PC, Wu SN. Differential suppression of delayed-rectifier and inwardly rectifier K + currents by a group of ent-kaurane-type diterpenoids from Croton tonkinensis, in microglial cells. Eur J Pharmacol 2019; 856:172414. [PMID: 31129155 DOI: 10.1016/j.ejphar.2019.172414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022]
Abstract
Croton is an extensive flowering plant genus in the spurge family, Euphorbiaceae. Three croton compounds with the common ent-kaurane skeleton were purified from Croton tonkinensis. By using patch-clamp recording technique, we thoroughly examined the effect of a group of croton compounds, croton-01 (ent-18-acetoxy-7α-hydroxykaur-16-en-15-one), croton-02 (ent-7α,14β-dihydroxykaur-16-en-15-one), and croton-03 (ent-1β-acetoxy-7α,14β-dihydroxykaur-16-en-15-one), on the membrane current in SM826 and BV2 microglial cells. Although neither voltage-gated Na+ nor Ca2+ currents were present in these cells, both delayed-rectifier K+ outward (IK(DR)) and inwardly rectifying K+ currents (IK(IR)) were readily detected. Croton-03 differentially caused inhibition of IK(DR) or IK(IR) in a concentration-dependent manner. According to a minimal scheme, the shortening of the time constant in either the IK(DR)-related block or IK(IR) caused by different concentrations of croton-03 was quantitatively estimated with a dissociation constant of 6.45 and 29.5 μM, respectively. In SM826 cells differentiated with β-amyloid, inhibitory action on these K+ currents remained unaltered. In ultraviolet C-irradiated cells, the magnitude of IK(IR) was still decreased by addition of croton-03. Therefore, our study suggests that these ent-kaurane diterpenoids ought to somehow act on the cellular mechanisms by which they influence the functional activities of microglial cells.
Collapse
Affiliation(s)
- Hung-Tsung Hsiao
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Yu-Chi Lee
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan
| | - Yen-Chin Liu
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Ping-Chung Kuo
- School of Pharmacy, National Cheng Kung University, Tainan City, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City, Taiwan.
| |
Collapse
|
11
|
Abstract
Purpose of review Humoral autoimmunity has gained highest interest in neurology and psychiatry. Despite numerous recent articles on this hot topic, however, the biological significance of natural autoantibodies (AB) and the normal autoimmune repertoire of mammals remained quite obscure. AB may contribute to disorder-relevant phenotypes and are even believed to induce diseases themselves, but the circumstances under which AB become pathogenic are not fully understood. This review will focus on the highly frequent AB against the N-methyl-d-aspartate receptor 1 (NMDAR1-AB) as an illustrating example and provide a critical overview of current work (please note that the new nomenclature, GluN1, is disregarded here for consistency with the AB literature). In particular, it will demonstrate how little is known at this point and how many conclusions are drawn based on small numbers of individuals, fragmentary experimental approaches or missing controls. Recent findings NMDAR1-AB were investigated by clinicians world-wide with numerous small studies and case reports appearing yearly. Many publications were on ‘anti-NMDAR encephalitis’ cases or tried to separate those from other NMDAR1-AB associated conditions. Original exclusivity claims (e.g. electroencephalogram, EEG or functional magnetic resonance imaging, fMRI findings) turned out not to be exclusive for ‘anti-NMDAR encephalitis’. Systematic analyses of representative NMDAR1-AB positive sera of all immunoglobulin (Ig) classes showed comparable distribution of different epitopes, often polyspecific/polyclonal, across health and disease. Sophisticated imaging tools provided findings on synapse trafficking changes induced by NMDAR1-AB from psychotic subjects but still lack epitope data to support any claimed disorder link. Persistently high titers of NMDAR1-AB (IgG) in immunized mice with open blood–brain barrier (BBB)-induced psychosis-like symptoms but failed to induce inflammation in the brain. Knowledge on peripheral NMDAR, for example in the immune system, and on potential inducers of NMDAR1-AB is only slowly increasing. Summary The present knowledge on the (patho) physiological role of NMDAR1-AB is very limited and still characterized by adamant rumors. Much more experimental work and more solid and informative clinical reports, including large numbers of subjects and adequate control groups, follow-up investigations and interdisciplinary approaches will be necessary to obtain a better understanding of the significance of humoral autoimmunity in general (in focus here: NMDAR1-AB) and its disease-relevance in particular.
Collapse
|
12
|
Lowinus T, Heidel FH, Bose T, Nimmagadda SC, Schnöder T, Cammann C, Schmitz I, Seifert U, Fischer T, Schraven B, Bommhardt U. Memantine potentiates cytarabine-induced cell death of acute leukemia correlating with inhibition of K v1.3 potassium channels, AKT and ERK1/2 signaling. Cell Commun Signal 2019; 17:5. [PMID: 30651113 PMCID: PMC6335768 DOI: 10.1186/s12964-018-0317-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/28/2018] [Indexed: 12/23/2022] Open
Abstract
Background Treatment of acute leukemia is challenging and long-lasting remissions are difficult to induce. Innovative therapy approaches aim to complement standard chemotherapy to improve drug efficacy and decrease toxicity. Promising new therapeutic targets in cancer therapy include voltage-gated Kv1.3 potassium channels, but their role in acute leukemia is unclear. We reported that Kv1.3 channels of lymphocytes are blocked by memantine, which is known as an antagonist of neuronal N-methyl-D-aspartate type glutamate receptors and clinically applied in therapy of advanced Alzheimer disease. Here we evaluated whether pharmacological targeting of Kv1.3 channels by memantine promotes cell death of acute leukemia cells induced by chemotherapeutic cytarabine. Methods We analyzed acute lymphoid (Jurkat, CEM) and myeloid (HL-60, Molm-13, OCI-AML-3) leukemia cell lines and patients’ acute leukemic blasts after treatment with either drug alone or the combination of cytarabine and memantine. Patch-clamp analysis was performed to evaluate inhibition of Kv1.3 channels and membrane depolarization by memantine. Cell death was determined with propidium iodide, Annexin V and SYTOX staining and cytochrome C release assay. Molecular effects of memantine co-treatment on activation of Caspases, AKT, ERK1/2, and JNK signaling were analysed by Western blot. Kv1.3 channel expression in Jurkat cells was downregulated by shRNA. Results Our study demonstrates that memantine inhibits Kv1.3 channels of acute leukemia cells and in combination with cytarabine potentiates cell death of acute lymphoid and myeloid leukemia cell lines as well as primary leukemic blasts from acute leukemia patients. At molecular level, memantine co-application fosters concurrent inhibition of AKT, S6 and ERK1/2 and reinforces nuclear down-regulation of MYC, a common target of AKT and ERK1/2 signaling. In addition, it augments mitochondrial dysfunction resulting in enhanced cytochrome C release and activation of Caspase-9 and Caspase-3 leading to amplified apoptosis. Conclusions Our study underlines inhibition of Kv1.3 channels as a therapeutic strategy in acute leukemia and proposes co-treatment with memantine, a licensed and safe drug, as a potential approach to promote cytarabine-based cell death of various subtypes of acute leukemia. Electronic supplementary material The online version of this article (10.1186/s12964-018-0317-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Theresa Lowinus
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Present address: Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany
| | - Florian H Heidel
- Department of Hematology and Oncology, GC-I3, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Leibniz Institute on Aging, Fritz-Lipmann Institute, Jena, Germany.,Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Tanima Bose
- Leibniz Institute of Neurobiology, Magdeburg, Germany.,Present address: Institute for Clinical Neuroimmunology, Ludwigs-Maximilians-University, Munich, Germany
| | - Subbaiah Chary Nimmagadda
- Department of Hematology and Oncology, GC-I3, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Tina Schnöder
- Department of Hematology and Oncology, GC-I3, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Leibniz Institute on Aging, Fritz-Lipmann Institute, Jena, Germany.,Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Clemens Cammann
- Friedrich Loeffler Institute for Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Ingo Schmitz
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Systems-Oriented Immunology and Inflammation Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ulrike Seifert
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Friedrich Loeffler Institute for Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Fischer
- Department of Hematology and Oncology, GC-I3, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Department of Immune Control, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ursula Bommhardt
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| |
Collapse
|
13
|
Martinez B, Peplow PV. Amelioration of Alzheimer's disease pathology and cognitive deficits by immunomodulatory agents in animal models of Alzheimer's disease. Neural Regen Res 2019; 14:1158-1176. [PMID: 30804241 PMCID: PMC6425849 DOI: 10.4103/1673-5374.251192] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The most common age-related neurodegenerative disease is Alzheimer’s disease (AD) characterized by aggregated amyloid-β (Aβ) peptides in extracellular plaques and aggregated hyperphosphorylated tau protein in intraneuronal neurofibrillary tangles, together with loss of cholinergic neurons, synaptic alterations, and chronic inflammation within the brain. These lead to progressive impairment of cognitive function. There is evidence of innate immune activation in AD with microgliosis. Classically-activated microglia (M1 state) secrete inflammatory and neurotoxic mediators, and peripheral immune cells are recruited to inflammation sites in the brain. The few drugs approved by the US FDA for the treatment of AD improve symptoms but do not change the course of disease progression and may cause some undesirable effects. Translation of active and passive immunotherapy targeting Aβ in AD animal model trials had limited success in clinical trials. Treatment with immunomodulatory/anti-inflammatory agents early in the disease process, while not preventive, is able to inhibit the inflammatory consequences of both Aβ and tau aggregation. The studies described in this review have identified several agents with immunomodulatory properties that alleviated AD pathology and cognitive impairment in animal models of AD. The majority of the animal studies reviewed had used transgenic models of early-onset AD. More effort needs to be given to creat models of late-onset AD. The effects of a combinational therapy involving two or more of the tested pharmaceutical agents, or one of these agents given in conjunction with one of the cell-based therapies, in an aged animal model of AD would warrant investigation.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Molecular & Cellular Biology, University of California, Merced, Merced, CA, USA; Department of Medicine, St. Georges University School of Medicine, Grenada; Department of Physics and Engineering, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
14
|
Mulcahy MJ, Paulo JA, Hawrot E. Proteomic Investigation of Murine Neuronal α7-Nicotinic Acetylcholine Receptor Interacting Proteins. J Proteome Res 2018; 17:3959-3975. [PMID: 30285449 DOI: 10.1021/acs.jproteome.8b00618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The α7-nicotinic acetylcholine receptor (α7-nAChR) is a ligand-gated ion channel that is expressed widely in vertebrates and is the principal high-affinity α-bungarotoxin (α-bgtx) binding protein in the mammalian CNS. α7-nAChRs associate with proteins that can modulate its properties. The α7-nAChR interactome is the summation of proteins interacting or associating with α7-nAChRs in a protein complex. To identify an α7-nAChR interactome in neural tissue, we isolated α-bgtx-affinity protein complexes from wild-type and α7-nAChR knockout (α7 KO) mouse whole brain tissue homogenates using α-bgtx-affinity beads. Affinity precipitated proteins were trypsinized and analyzed with an Orbitrap Fusion mass spectrometer. Proteins isolated with the α7-nAChR specific ligand, α-bgtx, were determined to be α7-nAChR associated proteins. The α7-nAChR subunit and 120 additional proteins were identified. Additionally, 369 proteins were identified as binding to α-bgtx in the absence of α7-nAChR expression, thereby identifying nonspecific proteins for α7-nAChR investigations using α-bgtx enrichment. These results expand on our previous investigations of α7-nAChR interacting proteins using α-bgtx-affinity bead isolation by controlling for differences between α7-nAChR and α-bgtx-specific proteins, developing an improved protein isolation methodology, and incorporating the latest technology in mass spectrometry. The α7-nAChR interactome identified in this study includes proteins associated with the expression, localization, function, or modulation of α7-nAChRs, and it provides a foundation for future studies to elucidate how these interactions contribute to human disease.
Collapse
Affiliation(s)
- Matthew J Mulcahy
- Division of Biology and Biological Engineering , California Institute of Technology , 1200 East California Boulevard , Pasadena , California 91125-2900 , United States.,Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| | - Joao A Paulo
- Department of Cell Biology , Harvard Medical School , 240 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - Edward Hawrot
- Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| |
Collapse
|
15
|
Baig MH, Ahmad K, Rabbani G, Choi I. Use of Peptides for the Management of Alzheimer's Disease: Diagnosis and Inhibition. Front Aging Neurosci 2018; 10:21. [PMID: 29467644 PMCID: PMC5808296 DOI: 10.3389/fnagi.2018.00021] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/18/2018] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a form of dementia and the most common progressive neurodegenerative disease (ND). The targeting of amyloid-beta (Aβ) aggregation is one of the most widely used strategies to manage AD, and efforts are being made globally to develop peptide-based compounds for the early diagnosis and treatment of AD. Here, we briefly discuss the use of peptide-based compounds for the early diagnosis and treatment of AD and the use of peptide-based inhibitors targeting various Aβ aggregation checkpoints. In addition, we briefly discuss recent applications of peptide-based inhibitors against various AD targets including amyloid beta, β-site amyloid precursor protein cleaving enzyme 1 (BACE1), Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), tyrosine phosphatase (TP) and potassium channel KV1.3.
Collapse
Affiliation(s)
- Mohammad H Baig
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Gulam Rabbani
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
16
|
Alonso E, Vieira AC, Rodriguez I, Alvariño R, Gegunde S, Fuwa H, Suga Y, Sasaki M, Alfonso A, Cifuentes JM, Botana LM. Tetracyclic Truncated Analogue of the Marine Toxin Gambierol Modifies NMDA, Tau, and Amyloid β Expression in Mice Brains: Implications in AD Pathology. ACS Chem Neurosci 2017; 8:1358-1367. [PMID: 28125211 DOI: 10.1021/acschemneuro.7b00012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Gambierol and its two, tetra- and heptacyclic, analogues have been previously proved as promising molecules for the modulation of Alzheimer's disease (AD) hallmarks in primary cortical neurons of 3xTg-AD fetuses. In this work, the effect of the tetracyclic analogue of gambierol was tested in vivo in 3xTg-AD mice (10 months old) after 1 month of weekly treatment with 50 μg/kg. Adverse effects were not reported throughout the whole treatment period and no pathological signs were observed for the analyzed organs. The compound was found in brain samples after intraperitoneal injection. The tetracyclic analogue of gambierol elicited a decrease of amyloid β1-42 levels and a dose-dependent inhibition of β-secretase enzyme-1 activity. Moreover, this compound also reduced the phosphorylation of tau at the 181 and 159/163 residues with an increase of the inactive isoform of the glycogen synthase kinase-3β. In accordance with our in vitro neuronal model, this compound produced a reduction in the N2A subunit of the N-methyl-d-aspartate (NMDA) receptor. The combined effect of this compound on amyloid β1-42 and tau phosphorylation represents a multitarget therapeutic approach for AD which might be more effective for this multifactorial and complex neurodegenerative disease than the current treatments.
Collapse
Affiliation(s)
- Eva Alonso
- Departamento
de Farmacología, §Departamento de Anatomía, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Andrés C. Vieira
- Departamento
de Farmacología, §Departamento de Anatomía, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Inés Rodriguez
- Departamento
de Farmacología, §Departamento de Anatomía, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Rebeca Alvariño
- Departamento
de Farmacología, §Departamento de Anatomía, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Sandra Gegunde
- Departamento
de Farmacología, §Departamento de Anatomía, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Haruhiko Fuwa
- Graduate
School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Yuto Suga
- Graduate
School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Makoto Sasaki
- Graduate
School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Amparo Alfonso
- Departamento
de Farmacología, §Departamento de Anatomía, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | | | - Luis M. Botana
- Departamento
de Farmacología, §Departamento de Anatomía, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| |
Collapse
|