51
|
Richards T, Perron JC, Patel K, Wurpel J, Reznik SE, Schanne F. Therapeutic Intervention of Neuroinflammatory Alzheimer Disease Model by Inhibition of Classical Complement Pathway with the Use of Anti-C1r Loaded Exosomes. RESEARCH SQUARE 2023:rs.3.rs-3399248. [PMID: 37886595 PMCID: PMC10602145 DOI: 10.21203/rs.3.rs-3399248/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease associated with memory decline, cognitive impairment, amyloid plaque formation and tau tangles. Neuroinflammation has been shown to be a precursor to apparent amyloid plaque accumulation and subsequent synaptic loss and cognitive decline. In this study, the ability of a novel, small molecule, T-ALZ01, to inhibit neuroinflammatory processes was analyzed. T-ALZ01, an inhibitor of complement component C1r, demonstrated a significant reduction in the levels of the inflammatory cytokines, IL-6 and TNF-α in vitro. An LPS-induced animal model, whereby animals were injected intraperitoneally with 0.5 mg/kg LPS, was used to analyze the effect of T-ALZ01 on neuroinflammation in vivo. Moreover, exosomes (nanosized, endogenous extracellular vehicles) were used as drug delivery vehicles to facilitate intranasal administration of T-ALZ01 across the blood-brain barrier. T-ALZ01 demonstrated significant reduction in degenerating neurons and the activation of resident microglia and astrocytes, as well as inflammatory markers in vivo. This study demonstrates a significant use of small molecule complement inhibitors via exosome drug delivery as a possible therapeutic in disorders characterized by neuroinflammation, such AD.
Collapse
|
52
|
Ronström JW, Williams SB, Payne A, Obray DJ, Hafen C, Burris M, Scott Weber K, Steffensen SC, Yorgason JT. Interleukin-10 enhances activity of ventral tegmental area dopamine neurons resulting in increased dopamine release. Brain Behav Immun 2023; 113:145-155. [PMID: 37453452 PMCID: PMC10530119 DOI: 10.1016/j.bbi.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/06/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023] Open
Abstract
Dopamine transmission from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) regulates important aspects of motivation and is influenced by the neuroimmune system. The neuroimmune system is a complex network of leukocytes, microglia and astrocytes that detect and remove foreign threats like bacteria or viruses and communicate with each other to regulate non-immune (e.g neuronal) cell activity through cytokine signaling. Inflammation is a key regulator of motivational states, though the effects of specific cytokines on VTA circuitry and motivation are largely unknown. Therefore, electrophysiology, neurochemical, immunohistochemical and behavioral studies were performed to determine the effects of the anti-inflammatory cytokine interleukin-10 (IL-10) on mesolimbic activity, dopamine transmission and conditioned behavior. IL-10 enhanced VTA dopamine firing and NAc dopamine levels via decreased VTA GABA currents in dopamine neurons. The IL-10 receptor was localized on VTA dopamine and non-dopamine cells. The IL-10 effects on dopamine neurons required post-synaptic phosphoinositide 3-kinase activity, and IL-10 appeared to have little-to-no efficacy on presynaptic GABA terminals. Intracranial IL-10 enhanced NAc dopamine levels in vivo and produced conditioned place aversion. Together, these studies identify the IL-10R on VTA dopamine neurons as a potential regulator of motivational states.
Collapse
Affiliation(s)
- Joakim W Ronström
- Brigham Young University, Department of Psychology/Neuroscience, Provo, UT 84602, United States
| | - Stephanie B Williams
- Brigham Young University, Department of Psychology/Neuroscience, Provo, UT 84602, United States
| | - Andrew Payne
- Brigham Young University, Department of Psychology/Neuroscience, Provo, UT 84602, United States
| | - Daniel J Obray
- Brigham Young University, Department of Psychology/Neuroscience, Provo, UT 84602, United States
| | - Caylor Hafen
- Brigham Young University, Department of Psychology/Neuroscience, Provo, UT 84602, United States
| | - Matthew Burris
- Brigham Young University, Department of Cellular Biology and Physiology, Provo, UT 84602, United States
| | - K Scott Weber
- Brigham Young University, Department of Microbiology and Molecular Biology, Provo, UT 84602, United States
| | - Scott C Steffensen
- Brigham Young University, Department of Psychology/Neuroscience, Provo, UT 84602, United States
| | - Jordan T Yorgason
- Brigham Young University, Department of Psychology/Neuroscience, Provo, UT 84602, United States; Brigham Young University, Department of Cellular Biology and Physiology, Provo, UT 84602, United States.
| |
Collapse
|
53
|
Ramos V, Reis M, Ferreira L, Silva AM, Ferraz R, Vieira M, Vasconcelos V, Martins R. Stalling the Course of Neurodegenerative Diseases: Could Cyanobacteria Constitute a New Approach toward Therapy? Biomolecules 2023; 13:1444. [PMID: 37892126 PMCID: PMC10604708 DOI: 10.3390/biom13101444] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Neurodegenerative diseases (NDs) are characterized by progressive and irreversible neuronal loss, accompanied by a range of pathological pathways, including aberrant protein aggregation, altered energy metabolism, excitotoxicity, inflammation, and oxidative stress. Some of the most common NDs include Alzheimer's Disease (AD), Parkinson's Disease (PD), Multiple Sclerosis (MS), Amyotrophic Lateral Sclerosis (ALS), and Huntington's Disease (HD). There are currently no available cures; there are only therapeutic approaches that ameliorate the progression of symptoms, which makes the search for new drugs and therapeutic targets a constant battle. Cyanobacteria are ancient prokaryotic oxygenic phototrophs whose long evolutionary history has resulted in the production of a plethora of biomedically relevant compounds with anti-inflammatory, antioxidant, immunomodulatory, and neuroprotective properties, that can be valuable in this field. This review summarizes the major NDs and their pathophysiology, with a focus on the anti-neurodegenerative properties of cyanobacterial compounds and their main effects.
Collapse
Affiliation(s)
- Vitória Ramos
- School of Health, Polytechnic Institute of Porto (ESS/P.PORTO), Rua Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal; (V.R.); (A.M.S.); (R.F.); (M.V.)
| | - Mariana Reis
- Interdisciplinary Centre of Marine and Environmental Research, University of Porto (CIIMAR/CIMAR), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; (M.R.); (L.F.); (V.V.)
| | - Leonor Ferreira
- Interdisciplinary Centre of Marine and Environmental Research, University of Porto (CIIMAR/CIMAR), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; (M.R.); (L.F.); (V.V.)
- Department of Biology, Faculty of Sciences, University of Porto (FCUP), Rua do Campo Alegre, Edifício FC4, 4169-007 Porto, Portugal
| | - Ana Margarida Silva
- School of Health, Polytechnic Institute of Porto (ESS/P.PORTO), Rua Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal; (V.R.); (A.M.S.); (R.F.); (M.V.)
| | - Ricardo Ferraz
- School of Health, Polytechnic Institute of Porto (ESS/P.PORTO), Rua Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal; (V.R.); (A.M.S.); (R.F.); (M.V.)
- Associated Laboratory for Green Chemistry—Network of Chemistry and Technology (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal
| | - Mónica Vieira
- School of Health, Polytechnic Institute of Porto (ESS/P.PORTO), Rua Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal; (V.R.); (A.M.S.); (R.F.); (M.V.)
- Center for Translational Health and Medical Biotechnology Research (TBIO/ESS/P.PORTO), Rua Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal
| | - Vitor Vasconcelos
- Interdisciplinary Centre of Marine and Environmental Research, University of Porto (CIIMAR/CIMAR), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; (M.R.); (L.F.); (V.V.)
- Department of Biology, Faculty of Sciences, University of Porto (FCUP), Rua do Campo Alegre, Edifício FC4, 4169-007 Porto, Portugal
| | - Rosário Martins
- School of Health, Polytechnic Institute of Porto (ESS/P.PORTO), Rua Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal; (V.R.); (A.M.S.); (R.F.); (M.V.)
- Interdisciplinary Centre of Marine and Environmental Research, University of Porto (CIIMAR/CIMAR), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; (M.R.); (L.F.); (V.V.)
| |
Collapse
|
54
|
Stoyanova K, Stoyanov D, Petrov S, Baldzhieva A, Bozhkova M, Murdzheva M, Kalfova T, Andreeva H, Taskov H, Vassilev P, Todev A. Conversion and Obsessive-Phobic Symptoms Predict IL-33 and IL-28A Levels in Individuals Diagnosed with COVID-19. Brain Sci 2023; 13:1271. [PMID: 37759873 PMCID: PMC10526257 DOI: 10.3390/brainsci13091271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The first epidemiological wave of the incidence of COVID-19 in Bulgaria was registered in June 2020. After the wave peak, we conducted a study in persons diagnosed with COVID-19 (N = 52). They were assessed with the anxiety-depressive scale (ADS), including basic (BS), vegetative (VS), conversion (CS), obsessive-phobic (OPS), and depressive (DS) symptoms. ADS assessment of individuals diagnosed with SARS-CoV-2 indicated a correlation between OPS and IL-33 values. IL-10 levels were higher than reference ranges in all patients. Multiple linear regression analyses demonstrated that combination of CS and OPS explained 28% of IL-33 levels, while combination of symptoms from all ADS dimensions explained 24% of IL-33 levels. It was also found that 21% of IL-28A levels was explained from the combination by all ADS dimensions, whereas OPS was the predictor for lower concentrations. The obtained results revealed meaningful correlations between psycho neuro-immunological factors in pathogenesis of illness from the coronavirus infection.
Collapse
Affiliation(s)
- Kristina Stoyanova
- Research Institute at Medical University of Plovdiv, Research Group “Translational and Computational Neuroscience”, SRIPD, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Drozdstoy Stoyanov
- Research Institute at Medical University of Plovdiv, Research Group “Translational and Computational Neuroscience”, SRIPD, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
- Department of Psychiatry and Medical Psychology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Steliyan Petrov
- Department of Medical Microbiology and Immunology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (S.P.); (A.B.); (M.B.); (M.M.); (T.K.); (H.A.)
- Laboratory of Clinical Immunology, University Hospital “St. George”, 4002 Plovdiv, Bulgaria
| | - Alexandra Baldzhieva
- Department of Medical Microbiology and Immunology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (S.P.); (A.B.); (M.B.); (M.M.); (T.K.); (H.A.)
- Laboratory of Clinical Immunology, University Hospital “St. George”, 4002 Plovdiv, Bulgaria
| | - Martina Bozhkova
- Department of Medical Microbiology and Immunology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (S.P.); (A.B.); (M.B.); (M.M.); (T.K.); (H.A.)
- Laboratory of Clinical Immunology, University Hospital “St. George”, 4002 Plovdiv, Bulgaria
| | - Mariana Murdzheva
- Department of Medical Microbiology and Immunology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (S.P.); (A.B.); (M.B.); (M.M.); (T.K.); (H.A.)
- Laboratory of Clinical Immunology, University Hospital “St. George”, 4002 Plovdiv, Bulgaria
| | - Teodora Kalfova
- Department of Medical Microbiology and Immunology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (S.P.); (A.B.); (M.B.); (M.M.); (T.K.); (H.A.)
- Laboratory of Clinical Immunology, University Hospital “St. George”, 4002 Plovdiv, Bulgaria
| | - Hristina Andreeva
- Department of Medical Microbiology and Immunology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (S.P.); (A.B.); (M.B.); (M.M.); (T.K.); (H.A.)
- Laboratory of Clinical Immunology, University Hospital “St. George”, 4002 Plovdiv, Bulgaria
| | - Hristo Taskov
- Research Institute at Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Petar Vassilev
- Department of Infectious Disease, Parasitology, and Tropical Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (P.V.); (A.T.)
| | - Angel Todev
- Department of Infectious Disease, Parasitology, and Tropical Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (P.V.); (A.T.)
| |
Collapse
|
55
|
Sentyabreva AV, Miroshnichenko EA, Melnikova EA, Tsvetkov IS, Kosyreva AM. Morphofunctional Changes in Brain and Peripheral Blood in Adult and Aged Wistar Rats with AlCl 3-Induced Neurodegeneration. Biomedicines 2023; 11:2336. [PMID: 37760778 PMCID: PMC10526012 DOI: 10.3390/biomedicines11092336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND the general lifespan has been prolonged greatly during the past century, and the incidence of age-associated diseases, including neurodegenerative ones, has increased as well. However, modelling of age-related pathologies is mostly conducted on adult rodents. We studied morphofunctional changes in the brain and peripheral blood of adult Wistar rats in comparison with old Wistar rats to determine age-related physiological changes and differences in adaptive reactions to AlCl3 exposure. METHODS the work was performed on adult and old male Wistar rats. The animals consumed a 100 mg/kg solution of AlCl3 each day for 60 days. Morphological changes of neurons and microglia, mRNA expression levels of pro-inflammatory and anti-inflammatory cytokines, microglia activation markers, amyloid-related proteins, and hallmarks of cellular senescence, monocyte, and lymphocyte subpopulations in the peripheral blood were examined. RESULTS old rats showed increasing hyperchromic neurons in the hippocampus; activation of microglia; upregulation of pro-inflammatory cytokines and cellular senescence markers; downregulation of anti-inflammatory cytokines; and Hif-1a and a decrease in B-cells and monocyte in peripheral blood. CONCLUSION compared to young animals, aged rats respond to aluminum exposure with a severe decline of most cells' function and irreversible neuronal loss. Regarding all reported data, neurodegeneration modelling and investigating of factors capable of accelerating or preventing it should be performed in experimental work on aged animals.
Collapse
Affiliation(s)
- Alexandra Vladislavovna Sentyabreva
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Alexandrovna Miroshnichenko
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Andreevna Melnikova
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| | - Ivan Sergeevich Tsvetkov
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| | - Anna Mikhailovna Kosyreva
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
56
|
Kalashnikova I, Cambell H, Kolpek D, Park J. Optimization and characterization of miRNA-129-5p-encapsulated poly (lactic- co-glycolic acid) nanoparticles to reprogram activated microglia. NANOSCALE ADVANCES 2023; 5:3439-3452. [PMID: 37383067 PMCID: PMC10295030 DOI: 10.1039/d3na00149k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/05/2023] [Indexed: 06/30/2023]
Abstract
Microglia have become a therapeutic target of many inflammation-mediated diseases in the central nervous system (CNS). Recently, microRNA (miRNA) has been proposed as an important regulator of immune responses. Specifically, miRNA-129-5p has been shown to play critical roles in the regulation of microglia activation. We have demonstrated that biodegradable poly (lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs) modulated innate immune cells and limited neuroinflammation after injury to the CNS. In this study, we optimized and characterized PLGA-based NPs for miRNA-129-5p delivery to utilize their synergistic immunomodulatory features for activated microglia modulation. A series of nanoformulations employing multiple excipients including epigallocatechin gallate (EGCG), spermidine (Sp), or polyethyleneimine (PEI) for miRNA-129-5p complexation and miRNA-129-5p conjugation to PLGA (PLGA-miR) were utilized. We characterized a total of six nanoformulations through physicochemical, biochemical, and molecular biological methods. In addition, we investigated the immunomodulatory effects of multiple nanoformulations. The data indicated that the immunomodulatory effects of nanoformulation, PLGA-miR with the excipient Sp (PLGA-miR+Sp) and PEI (PLGA-miR+PEI) were significant compared to other nanoformulations including naked PLGA-based NP. These nanoformulations promoted a sustained release of miRNA-129-5p and polarization of activated microglia into a more pro-regenerative phenotype. Moreover, they enhanced the expression of multiple regeneration-associated factors, while alleviating the expression of pro-inflammatory factors. Collectively, the proposed nanoformulations in this study highlight the promising therapeutic tools for synergistic immunomodulatory effects between PLGA-based NPs and miRNA-129-5p to modulate activated microglia which will have numerous applications for inflammation-derived diseases.
Collapse
Affiliation(s)
- Irina Kalashnikova
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky 789 S. Limestone Lexington KY 40506 USA +1-859-257-1850
| | - Heather Cambell
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky 789 S. Limestone Lexington KY 40506 USA +1-859-257-1850
| | - Daniel Kolpek
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky 789 S. Limestone Lexington KY 40506 USA +1-859-257-1850
| | - Jonghyuck Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky 789 S. Limestone Lexington KY 40506 USA +1-859-257-1850
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky Lexington KY USA
| |
Collapse
|
57
|
Merchant RA, Chan YH, Anbarasan D, Aprahamian I. Association of Motoric Cognitive Risk Syndrome with Sarcopenia and Systemic Inflammation in Pre-Frail Older Adults. Brain Sci 2023; 13:936. [PMID: 37371414 DOI: 10.3390/brainsci13060936] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/04/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Motoric cognitive risk syndrome (MCR) is defined by the presence of slow gait and subjective cognitive decline. It is well recognized as a prodrome for dementia, but the biological mechanism and trajectory for MCR are still lacking. The objective of this study was to explore the association of MCR with body composition, including sarcopenia and systemic inflammation, in pre-frail older adults in a cross-sectional study of 397 pre-frail community-dwelling older adults. Data on demographics, physical function, frailty, cognition (Montreal Cognitive Assessment (MoCA)), perceived health and depression were collected. Body composition was measured using a bioelectrical impedance analyzer. Systemic inflammatory biomarkers, such as progranulin, growth differentiation factor-15 (GDF-15), interleukin-10 (IL-10), interleukin-6 and tumor necrosis factor-α (TNF-α), were collected. Univariate and multivariate logistic regression were used to analyze the association between MCR, body composition, sarcopenia and systemic inflammatory biomarkers. The prevalence of MCR was 14.9%. They were significantly older and there were more females, depression, functional impairment, lower education, physical activity and MoCA scores. Body fat percentage (BF%), fat mass index, fat to fat free mass ratio (FM/FFM) and sarcopenia prevalence were significantly higher in MCR. Serum GDF-15 and TNF-α levels were highest with progranulin/TNF-α and IL-10/TNF-α ratio lowest in MCR. Compared to healthy patients, MCR was significantly associated with sarcopenia (aOR 2.62; 95% CI 1.46-3.17), BF% (aOR 1.06; 95% CI 1.01-1.12), FMI (aOR 1.16; 95% CI 1.02-1.30) and FM/FFM (aOR 6.38; 95% CI 1.20-33.98). The association of IL-10 to TNF-α ratio (aOR 0.98, 95% CI 0.97-0.99) and IL-10 (aOR 2.22, 95% CI 0.05-0.98) with MCR were independent of sarcopenia and BF%. Longitudinal population studies are needed to understand the role of body fat indices and IL-10 in pre-frail older adults with MCR and trajectory to dementia.
Collapse
Affiliation(s)
- Reshma Aziz Merchant
- Division of Geriatric Medicine, Department of Medicine, National University Hospital, 1E Kent Ridge Road, Singapore 119228, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Yiong Huak Chan
- Biostatistics Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Denishkrshna Anbarasan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Ivan Aprahamian
- Geriatrics Division, Department of Internal Medicine, Jundiai Medical School, Jundiai 13202-550, SP, Brazil
| |
Collapse
|
58
|
Carlini V, Noonan DM, Abdalalem E, Goletti D, Sansone C, Calabrone L, Albini A. The multifaceted nature of IL-10: regulation, role in immunological homeostasis and its relevance to cancer, COVID-19 and post-COVID conditions. Front Immunol 2023; 14:1161067. [PMID: 37359549 PMCID: PMC10287165 DOI: 10.3389/fimmu.2023.1161067] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Interleukin-10 (IL-10) is a pleiotropic cytokine that has a fundamental role in modulating inflammation and in maintaining cell homeostasis. It primarily acts as an anti-inflammatory cytokine, protecting the body from an uncontrolled immune response, mostly through the Jak1/Tyk2 and STAT3 signaling pathway. On the other hand, IL-10 can also have immunostimulating functions under certain conditions. Given the pivotal role of IL-10 in immune modulation, this cytokine could have relevant implications in pathologies characterized by hyperinflammatory state, such as cancer, or infectious diseases as in the case of COVID-19 and Post-COVID-19 syndrome. Recent evidence proposed IL-10 as a predictor of severity and mortality for patients with acute or post-acute SARS-CoV-2 infection. In this context, IL-10 can act as an endogenous danger signal, released by tissues undergoing damage in an attempt to protect the organism from harmful hyperinflammation. Pharmacological strategies aimed to potentiate or restore IL-10 immunomodulatory action may represent novel promising avenues to counteract cytokine storm arising from hyperinflammation and effectively mitigate severe complications. Natural bioactive compounds, derived from terrestrial or marine photosynthetic organisms and able to increase IL-10 expression, could represent a useful prevention strategy to curb inflammation through IL-10 elevation and will be discussed here. However, the multifaceted nature of IL-10 has to be taken into account in the attempts to modulate its levels.
Collapse
Affiliation(s)
- Valentina Carlini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Douglas M. Noonan
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Eslam Abdalalem
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Clementina Sansone
- Stazione Zoologica Anton Dohrn, Istituto Nazionale di Biologia, Ecologia e Biotecnologie Marine, Napoli, Italy
| | - Luana Calabrone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Adriana Albini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) European Institute of Oncology IEO-, Milan, Italy
| |
Collapse
|
59
|
Ispoglou T, Ferentinos P, Prokopidis K, Blake C, Aldrich L, Elia A, Lees M, Hind K. Exploring the impact of exercise and essential amino acid plus cholecalciferol supplementation on physical fitness and body composition in multiple sclerosis: A case study. Clin Case Rep 2023; 11:e7548. [PMID: 37323260 PMCID: PMC10264925 DOI: 10.1002/ccr3.7548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023] Open
Abstract
Key Clinical Message In MS patients, especially those frail or malnourished, combining home-based exercise twice weekly with essential amino acids and vitamin D may improve body composition, strength, and physical performance, enabling long-term functional improvements. Abstract Multiple sclerosis (MS) is associated with reduced bone and muscle strength and function. We aimed to investigate the effectiveness of a 24-week intervention in a 57-year-old frail female with MS. The participant completed a 2×/week exercise intervention and ingested 2×/day a supplement containing 7.5 g essential amino acids and 500 IU cholecalciferol. Body composition, 6-m gait speed (GS), handgrip strength (HGS), 30-sec arm-curl test (30ACT), 6-min walking test (6MWT), 30-sec chair-stand test (30CST), and plasma concentrations of 25-hydroxyvitamin D3 [25(OH)D3], insulin-like growth factor 1 (IGF-1), and amino acids were assessed at baseline, and at Weeks 12 and 24. Plasma 25(OH)D3 increased from 23.2 to 41.3 ng/mL and IGF-1 from 131.6 to 140.7 ng/mL from baseline to post-intervention. BMI, total lean tissue mass (LTM), fat mass, bone mineral content, and the sum of 17 amino acids increased by 3.8, 1.0, 3.5, 0.2, and 19%, respectively, at Week 24. There were clinically significant increases in regional LTM (6.9% arms and 6.3% legs) and large increases in GS (67.3%), dominant HGS (31.5%), non-dominant HGS (11.8%), dominant 30ACT (100%), non-dominant 30ACT (116.7%), 6MWT (125.6%), and 30CST (44.4%). The current intervention was effective in improving components of physical fitness and body composition in a female with MS.
Collapse
Affiliation(s)
| | | | | | - Cameron Blake
- Carnegie School of SportLeeds Beckett UniversityLeedsUK
| | - Luke Aldrich
- Carnegie School of SportLeeds Beckett UniversityLeedsUK
| | - Antonis Elia
- Division of Environmental PhysiologyRoyal Institute of TechnologyStockholmSweden
| | - Matthew Lees
- Faculty of Kinesiology and Physical EducationUniversity of TorontoTorontoOntarioCanada
| | - Karen Hind
- Wolfson Research Institute for Health and Well‐beingDurhamUK
| |
Collapse
|
60
|
Cyr B, de Rivero Vaccari JP. Sex Differences in the Inflammatory Profile in the Brain of Young and Aged Mice. Cells 2023; 12:1372. [PMID: 37408205 DOI: 10.3390/cells12101372] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 07/07/2023] Open
Abstract
Neurodegenerative diseases are a leading cause of death worldwide with no cures identified. Thus, there is a critical need for preventative measures and treatments as the number of patients is expected to increase. Many neurodegenerative diseases have sex-biased prevalence, indicating a need to examine sex differences when investigating prevention and treatment strategies. Inflammation is a key contributor to many neurodegenerative diseases and is a promising target for prevention since inflammation increases with age, which is known as inflammaging. Here, we analyzed the protein expression levels of cytokines, chemokines, and inflammasome signaling proteins in the cortex of young and aged male and female mice. Our results show an increase in caspase-1, interleukin (IL)-1β, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and ASC specks in females compared to males. Additionally, there was an increase in IL-1α, VEGF-A, CCL3, CXCL1, CCL4, CCL17, and CCL22 in aging females and an increase in IL-8, IL-17a, IL-7, LT-α, and CCL22 in aging males. IL-12/IL-23p40, CCL13, and IL-10 were increased in females compared to males but not with age. These results indicate that there are sex differences in cortical inflammaging and provide potential targets to attenuate inflammation to prevent the development of neurodegenerative disease.
Collapse
Affiliation(s)
- Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Center for Cognitive Neuroscience and Aging, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
61
|
Zambrowicz A, Zabłocka A, Bednarz D, Bobak Ł. Importance for humans of recently discovered protein compounds - yolkin and yolk glycopeptide 40, present in the plasma of hen egg yolk. Poult Sci 2023; 102:102770. [PMID: 37244087 DOI: 10.1016/j.psj.2023.102770] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/20/2023] [Accepted: 04/28/2023] [Indexed: 05/29/2023] Open
Abstract
Vitellogenin (Vt) is considered the primary protein precursor of egg yolk, serving as a source of protein- and lipid-rich nutrients for the developing embryo. However, recent research has revealed that the functions of Vt and Vt-derived polypeptides, such as yolkin (Y) and yolk glycopeptide 40 (YGP40), extend beyond their nutritional roles as a source of amino acids. Emerging evidence has demonstrated that both Y and YGP40 possess immunomodulatory properties and can contribute to host immune defenses. Additionally, Y polypeptides have been shown to exhibit neuroprotective activity, participating in the modulation of neurons' survival and activity, inhibiting neurodegeneration processes, and improving cognitive functions in rats. These non-nutritional functions not only enhance our understanding of the physiological roles of these molecules during embryonic development but also offer a promising basis for the potential application of these proteins in human health.
Collapse
Affiliation(s)
- Aleksandra Zambrowicz
- Department of Functional Food Products Development, Wroclaw University of Environmental and Life Science, 51-640 Wrocław, Poland.
| | - Agnieszka Zabłocka
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Science, 53-114 Wrocław, Poland
| | - Dominika Bednarz
- Department of Functional Food Products Development, Wroclaw University of Environmental and Life Science, 51-640 Wrocław, Poland
| | - Łukasz Bobak
- Department of Functional Food Products Development, Wroclaw University of Environmental and Life Science, 51-640 Wrocław, Poland
| |
Collapse
|
62
|
Khan AW, Farooq M, Hwang MJ, Haseeb M, Choi S. Autoimmune Neuroinflammatory Diseases: Role of Interleukins. Int J Mol Sci 2023; 24:7960. [PMID: 37175665 PMCID: PMC10178921 DOI: 10.3390/ijms24097960] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Autoimmune neuroinflammatory diseases are a group of disorders resulting from abnormal immune responses in the nervous system, causing inflammation and tissue damage. The interleukin (IL) family of cytokines, especially IL-1, IL-6, and IL-17, plays a critical role in the pathogenesis of these diseases. IL-1 is involved in the activation of immune cells, production of pro-inflammatory cytokines, and promotion of blood-brain barrier breakdown. IL-6 is essential for the differentiation of T cells into Th17 cells and has been implicated in the initiation and progression of neuroinflammation. IL-17 is a potent pro-inflammatory cytokine produced by Th17 cells that plays a crucial role in recruiting immune cells to sites of inflammation. This review summarizes the current understanding of the roles of different interleukins in autoimmune neuroinflammatory diseases, including multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, neuromyelitis optica, and autoimmune encephalitis, and discusses the potential of targeting ILs as a therapeutic strategy against these diseases. We also highlight the need for further research to better understand the roles of ILs in autoimmune neuroinflammatory diseases and to identify new targets for treating these debilitating diseases.
Collapse
Affiliation(s)
- Abdul Waheed Khan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Moon-Jung Hwang
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Muhammad Haseeb
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Republic of Korea
| |
Collapse
|
63
|
La Torre ME, Cianciulli A, Monda V, Monda M, Filannino FM, Antonucci L, Valenzano A, Cibelli G, Porro C, Messina G, Panaro MA, Messina A, Polito R. α-Tocopherol Protects Lipopolysaccharide-Activated BV2 Microglia. Molecules 2023; 28:molecules28083340. [PMID: 37110573 PMCID: PMC10141518 DOI: 10.3390/molecules28083340] [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: 03/16/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Microglia, the resident macrophage-like population in the central nervous system, play a crucial role in the pathogenesis of many neurodegenerative disorders by triggering an inflammatory response that leads to neuronal death. Neuroprotective compounds to treat or prevent neurodegenerative diseases are a new field of study in modern medicine. Microglia are activated in response to inflammatory stimuli. The pathogenesis of various neurodegenerative diseases is closely related to the constant activation of microglia due to their fundamental role as a mediator of inflammation in the brain environment. α-Tocopherol, also known as vitamin E, is reported to possess potent neuroprotective effects. The goal of this study was to investigate the biological effects of vitamin E on BV2 microglial cells, as a possible neuroprotective and anti-inflammatory agent, following stimulation with lipopolysaccharide (LPS). The results showed that the pre-incubation of microglia with α-tocopherol can guarantee neuroprotective effects during microglial activation induced by LPS. α-Tocopherol preserved the branched morphology typical of microglia in a physiological state. It also reduced the migratory capacity; the production of pro-inflammatory and anti-inflammatory cytokines such as TNF-α and IL-10; and the activation of receptors such as TRL4 and CD40, which modulate the PI3K-Akt signaling pathway. The results of this study require further insights and research, but they present new scenarios for the application of vitamin E as an antioxidant for the purpose of greater neuroprotection in vivo for the prevention of possible neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria Ester La Torre
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Antonia Cianciulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | | | - Laura Antonucci
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Rita Polito
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
64
|
Contaldi E, Magistrelli L, Comi C. Disease mechanisms as subtypes: Immune dysfunction in Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:67-93. [PMID: 36803824 DOI: 10.1016/b978-0-323-85555-6.00008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
In recent years, the contraposition between inflammatory and neurodegenerative processes has been increasingly challenged. Inflammation has been emphasized as a key player in the onset and progression of Parkinson disease (PD) and other neurodegenerative disorders. The strongest indicators of the involvement of the immune system derived from evidence of microglial activation, profound imbalance in phenotype and composition of peripheral immune cells, and impaired humoral immune responses. Moreover, peripheral inflammatory mechanisms (e.g., involving the gut-brain axis) and immunogenetic factors are likely to be implicated. Even though several lines of preclinical and clinical studies are supporting and defining the complex relationship between the immune system and PD, the exact mechanisms are currently unknown. Similarly, the temporal and causal connections between innate and adaptive immune responses and neurodegeneration are unsettled, challenging our ambition to define an integrated and holistic model of the disease. Despite these difficulties, current evidence is providing the unique opportunity to develop immune-targeted approaches for PD, thus enriching our therapeutic armamentarium. This chapter aims to provide an extensive overview of past and present studies that explored the implication of the immune system in neurodegeneration, thus paving the road for the concept of disease modification in PD.
Collapse
Affiliation(s)
- Elena Contaldi
- Movement Disorders Centre, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Luca Magistrelli
- Movement Disorders Centre, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Cristoforo Comi
- Neurology Unit, S.Andrea Hospital, Department of Translational Medicine, University of Piemonte Orientale, Vercelli, Italy.
| |
Collapse
|
65
|
Nass SR, Hahn YK, Ohene-Nyako M, McLane VD, Damaj MI, Thacker LR, Knapp PE, Hauser KF. Depressive-like Behavior Is Accompanied by Prefrontal Cortical Innate Immune Fatigue and Dendritic Spine Losses after HIV-1 Tat and Morphine Exposure. Viruses 2023; 15:v15030590. [PMID: 36992299 PMCID: PMC10052300 DOI: 10.3390/v15030590] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/23/2023] Open
Abstract
Opioid use disorder (OUD) and HIV are comorbid epidemics that can increase depression. HIV and the viral protein Tat can directly induce neuronal injury within reward and emotionality brain circuitry, including the prefrontal cortex (PFC). Such damage involves both excitotoxic mechanisms and more indirect pathways through neuroinflammation, both of which can be worsened by opioid co-exposure. To assess whether excitotoxicity and/or neuroinflammation might drive depressive behaviors in persons infected with HIV (PWH) and those who use opioids, male mice were exposed to HIV-1 Tat for eight weeks, given escalating doses of morphine during the last two weeks, and assessed for depressive-like behavior. Tat expression decreased sucrose consumption and adaptability, whereas morphine administration increased chow consumption and exacerbated Tat-induced decreases in nesting and burrowing—activities associated with well-being. Across all treatment groups, depressive-like behavior correlated with increased proinflammatory cytokines in the PFC. Nevertheless, supporting the theory that innate immune responses adapt to chronic Tat exposure, most proinflammatory cytokines were unaffected by Tat or morphine. Further, Tat increased PFC levels of the anti-inflammatory cytokine IL-10, which were exacerbated by morphine administration. Tat, but not morphine, decreased dendritic spine density on layer V pyramidal neurons in the anterior cingulate. Together, our findings suggest that HIV-1 Tat and morphine differentially induce depressive-like behaviors associated with increased neuroinflammation, synaptic losses, and immune fatigue within the PFC.
Collapse
Affiliation(s)
- Sara R. Nass
- Department of Pharmacology and Toxicology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Yun K. Hahn
- Department of Anatomy and Neurobiology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0709, USA
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Virginia D. McLane
- Department of Pharmacology and Toxicology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Leroy R. Thacker
- Department of Biostatistics, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Department of Anatomy and Neurobiology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0709, USA
- Institute for Drug and Alcohol Studies, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Department of Anatomy and Neurobiology, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0709, USA
- Institute for Drug and Alcohol Studies, Medical College of Virginia (MCV) Campus, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
- Correspondence: ; Tel.: +1-804-628-7579; Fax: +1-804-828-0676
| |
Collapse
|
66
|
Kumar S, Mehan S, Narula AS. Therapeutic modulation of JAK-STAT, mTOR, and PPAR-γ signaling in neurological dysfunctions. J Mol Med (Berl) 2023; 101:9-49. [PMID: 36478124 DOI: 10.1007/s00109-022-02272-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade is a pleiotropic pathway that involves receptor subunit multimerization. The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine-threonine kinase that perceives and integrates a variety of intracellular and environmental stimuli to regulate essential activities such as cell development and metabolism. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a prototypical metabolic nuclear receptor involved in neural differentiation and axon polarity. The JAK-STAT, mTOR, and PPARγ signaling pathways serve as a highly conserved signaling hub that coordinates neuronal activity and brain development. Additionally, overactivation of JAK/STAT, mTOR, and inhibition of PPARγ signaling have been linked to various neurocomplications, including neuroinflammation, apoptosis, and oxidative stress. Emerging research suggests that even minor disruptions in these cellular and molecular processes can have significant consequences manifested as neurological and neuropsychiatric diseases. Of interest, target modulators have been proven to alleviate neuronal complications associated with acute and chronic neurological deficits. This research-based review explores the therapeutic role of JAK-STAT, mTOR, and PPARγ signaling modulators in preventing neuronal dysfunctions in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India.
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
67
|
Abrishamdar M, Farbood Y, Sarkaki A, Rashno M, Badavi M. Evaluation of betulinic acid effects on pain, memory, anxiety, catalepsy, and oxidative stress in animal model of Parkinson's disease. Metab Brain Dis 2023; 38:467-482. [PMID: 35708868 DOI: 10.1007/s11011-022-00962-1] [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: 11/13/2021] [Accepted: 03/11/2022] [Indexed: 01/25/2023]
Abstract
Parkinson's disease (PD) is known for motor impairments. Betulinic acid (BA) is a natural compound with antioxidant activity. The present study addresses the question of whether BA affects motor and non-motor dysfunctions and molecular changes in the rat model of PD. The right medial forebrain bundle was lesioned by injection of 6-hydroxydopamine in Male Wistar rats (10-12 weeks old, 270-320 g). Animals were divided into Sham, PD, 3 treated groups with BA (0.5, 5, and 10 mg/kg, IP), and a positive control group received L-dopa (20 mg/kg, P.O) for 7 days. rigidity, anxiety, analgesia, and memory were assessed by bar test, open-field, elevated plus-maze (EPM), tail-flick, and shuttle box. Additionally, the malondialdehyde (MDA), Superoxide dismutase (SOD), glutathione peroxidase (GPx) activity, Brain-derived neurotrophic factor (BDNF) and Interleukin 10 (IL10) levels in the whole brain were measured. BA significantly reversed the 6-hydroxydopamine-induced motor and memory complication in the bar test and shuttle box. It modified anxiety-like behavior neither in open-field nor in EPM. It only decreased the time spent in open arms. Moreover, no significant changes were found in the tail-flick between treatment and sham groups. On the other hand, the level of MDA & IL10 were decreased, while the activity of GPx levels of SOD & BDNF in the rats' brains was increased. Our results showed that BA as a free radical scavenger can account for a possible promise as a good therapeutic agent for motor and non-motor complications in PD however further studies may be needed.
Collapse
Affiliation(s)
- M Abrishamdar
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Department of Physiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoob Farbood
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Physiology, Medicine Faculty and Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - A Sarkaki
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - M Rashno
- Department of Immunulogy, Cellular and Molecular Research Center, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - M Badavi
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
68
|
Neuroinflammation in Alzheimer's Disease: Current Progress in Molecular Signaling and Therapeutics. Inflammation 2023; 46:1-17. [PMID: 35986874 DOI: 10.1007/s10753-022-01721-1] [Citation(s) in RCA: 94] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/07/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease, a neurodegenerative disease with amyloid beta accumulation as a major hallmark, has become a dire global health concern as there is a lack of clear understanding of the causative agent. It is a major cause of dementia which is increasing exponentially with age. Alzheimer's disease is marked by tau hyperphosphorylation and amyloid beta accumulation that robs people of their memories. Amyloid beta deposition initiated a spectrum of microglia-activated neuroinflammation, and microglia and astrocyte activation elicited expressions of various inflammatory and anti-inflammatory cytokines. Neuroinflammation is one of the cardinal features of Alzheimer's disease. Pro-inflammatory cytokine signaling plays multifarious roles in neurodegeneration and neuroprotection. Induction of proinflammatory signaling leads to discharge of immune mediators which affect functions of neurons and cause cell death. Sluggish anti-inflammatory system also contributes to neuroinflammation. Numerous pathways like NFκB, p38 MAPK, Akt/mTOR, caspase, nitric oxide, and COX are involved in triggering brain immune cells like astrocytes and microglia to secrete inflammatory cytokines such as tumor necrosis factor, interleukins, and chemokines and participate in Alzheimer's disease pathology. PPAR-γ agonists tend to boost the phagocytosis of amyloid beta and decrease the inflammatory cytokine IL-1β. Recent findings suggest the cross-link between gut microbiota and neuroinflammation contributing in AD which has been explained in this study. The role of cellular, molecular pathways and involvement of inflammatory mediators in neuroinflammation has also been described; targeting them could be a potential therapeutic strategy for treatment of AD.
Collapse
|
69
|
Anwar MM, Fathi MH. Early approaches of YKL-40 as a biomarker and therapeutic target for Parkinson's disease. Neurodegener Dis Manag 2023; 13:85-99. [PMID: 36644988 DOI: 10.2217/nmt-2022-0010] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aim: To investigate whether the estimation of cerebrospinal fluid (CSF) and brain YKL-40 levels may be used as an efficient biomarker for Parkinson's disease (PD). Methods: Lipopolysaccharides (LPS) was injected into the right substantia nigra pars compacta (SNpc). Rats were divided into: control group, early LPS-induced PD group (14 days), and advanced LPS-induced PD group (28 days). YKL-40 and other related factors were detected in CSF and brain tissue. Results: Increased expression of YKL-40 was observed in brain tissue and CSF of PD-induced rats associated with triggered inflammatory cytokine release. Conclusion: The current study was limited to detecting YKL-40 and other inflammatory factors in brain and CSF. YKL-40 may be considered as an early biomarker and therapeutic target for PD.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control & Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Mohamed H Fathi
- Department of Nucleic Acid & Protein structure, Center of Genomics, Proteomics & Bioinformatics, Agricultural Genetic Engineering Research Institute (AGERI), Cairo, Egypt
| |
Collapse
|
70
|
Matsuzaki S, Pouly JL, Canis M. IL-10 is not anti-fibrotic but pro-fibrotic in endometriosis: IL-10 treatment of endometriotic stromal cells in vitro promotes myofibroblast proliferation and collagen type I protein expression. Hum Reprod 2023; 38:14-29. [PMID: 36413036 DOI: 10.1093/humrep/deac248] [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: 08/01/2022] [Revised: 10/25/2022] [Indexed: 11/23/2022] Open
Abstract
STUDY QUESTION Is interleukin-10 (IL-10) anti-fibrotic in endometriosis? SUMMARY ANSWER IL-10 is not anti-fibrotic but pro-fibrotic in endometriosis, because IL-10 treatment of endometriotic stromal cells in vitro promotes myofibroblast proliferation and collagen type I protein expression. WHAT IS KNOWN ALREADY We previously showed that persistent activation of signal transducer and activator of transcription 3 (STAT3) via IL-6 trans-signaling promotes fibrosis of endometriosis. Studies showed marked anti-fibrotic effects of IL-10 via the STAT3 signaling pathway, which is generally considered to be anti-inflammatory, in various organs. STUDY DESIGN, SIZE, DURATION Endometrial and/or endometriotic samples of 54 patients who had histological evidence of deep endometriosis, and endometrial samples from 30 healthy fertile women were analyzed. PARTICIPANTS/MATERIALS, SETTING, METHODS The effects of IL-10/STAT3 signaling as well as inhibition of STAT3 activation by knockdown of STAT3 gene on the pro-fibrotic phenotype in endometrial and endometriotic stromal cells in vitro were investigated. Then, the effects of various time points of IL-10 treatment in combination with transforming growth factor (TGF)-β1 and/or IL-6/soluble IL-6 receptor (sIL-6R) on the profibrotic phenotype of endometrial and endometriotic stromal cells were investigated. MAIN RESULTS AND THE ROLE OF CHANCE IL-10 induced pro-fibrotic phenotype (cell proliferation, collagen type I synthesis, α-smooth muscle actin positive stress fibers and collagen gel contraction) of endometriotic stromal cells. Knockdown of STAT3 gene decreased the IL-10 induced pro-fibrotic phenotype of endometriotic stromal cells. In contrast, IL-10 had no significant effects on pro-fibrotic phenotype of endometrial stromal cells of healthy women. Sequential IL-10 treatment with or without TGF-β1 and/or IL-6/sIL-6R induced persistent activation of STAT3 and significantly increased proliferation of myofibroblasts (cells with α-smooth muscle actin positive stress fibers) and protein expression of collagen type I in endometriotic stromal cells. TGF-β1 and/or IL-6/sIL6RIL-6/sIL6R treatment significantly increased tissue inhibitor of metalloproteinase 1 (TIMP1) protein expression, whereas IL-10 had no significant effects. Knockdown of STAT3 gene significantly decreased the TGF-β1 and/or IL-6/sIL6R induced TIMP1 protein expression. In contrast, pre-treatment with IL-10 before TGF-β1 and/or IL-6/sIL-6R treatment and sequential IL-10 treatment with or without TGF-β1 and/or IL-6/sIL-6R significantly decreased proliferation of fibroblasts (cells without α-smooth muscle actin positive stress fibers) and collagen type I protein expression in endometrial stromal cells of healthy women. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Given the large number of complex interactions and signaling pathways of pro- and anti-inflammatory mediators that are involved in the pathophysiology of endometriosis, the present study investigated only a very small portion of the whole. Further in vivo studies are required to validate the present findings. WIDER IMPLICATIONS OF THE FINDINGS Inflammatory mediators in the pathophysiology of endometriosis have been extensively investigated as potential therapeutic targets. However, the present study showed that anti-inflammatory signals of IL-10 and IL-6 through persistent STAT3 activation may promote endometriosis fibrosis. Therapeutic strategies, such as suppression of 'inflammation', might dysregulate the cross-regulation of 'pro- and anti-inflammatory mediators', leading to detrimental effects in patients with endometriosis, such as fibrosis. To develop new, but not deleterious, therapeutic strategies, studies are required to investigate whether, how and what 'anti-inflammatory mediators' along with pro-inflammatory mediators are involved in individual patients with endometriosis. STUDY FUNDING/COMPETING INTEREST(S) This study was supported in part by KARL STORZ SE & Co. KG (Tuttlingen, Germany). The authors have no conflict of interest to disclose.
Collapse
Affiliation(s)
- Sachiko Matsuzaki
- CHU Clermont-Ferrand, Chirurgie Gynécologique, Clermont-Ferrand, France.,Université Clermont Auvergne, Institut Pascal, UMR6602, CNRS/UCA/SIGMA, Clermont-Ferrand, France
| | - Jean-Luc Pouly
- CHU Clermont-Ferrand, Chirurgie Gynécologique, Clermont-Ferrand, France.,Université Clermont Auvergne, Institut Pascal, UMR6602, CNRS/UCA/SIGMA, Clermont-Ferrand, France
| | - Michel Canis
- CHU Clermont-Ferrand, Chirurgie Gynécologique, Clermont-Ferrand, France.,Université Clermont Auvergne, Institut Pascal, UMR6602, CNRS/UCA/SIGMA, Clermont-Ferrand, France
| |
Collapse
|
71
|
MORIKAWA TETSURO, UEHARA OSAMU, PAUDEL DURGA, YOSHIDA KOKI, HARADA FUMIYA, HIRAKI DAICHI, SATO JUN, MATSUOKA HIROFUMI, KURAMITSU YASUHIRO, MICHIKAWA MAKOTO, ABIKO YOSHIHIRO. Systemic Administration of Lipopolysaccharide from Porphyromonas gingivalis Decreases Neprilysin Expression in the Mouse Hippocampus. In Vivo 2023; 37:163-172. [PMID: 36593043 PMCID: PMC9843806 DOI: 10.21873/invivo.13065] [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: 11/16/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND/AIM Alzheimer's disease is the most common type of neurodegenerative disorder in elderly individuals worldwide. Increasing evidence suggests that periodontal diseases are involved in the pathogenesis of Alzheimer's disease, and an association between periodontitis and amyloid-β deposition in elderly individuals has been demonstrated. The aim of the present study was to examine the effects of systemic administration of Porphyromonas gingivalis-derived lipopolysaccharide (PG-LPS) on neprilysin expression in the hippocampus of adult and senescence-accelerated mice. MATERIALS AND METHODS PG-LPS diluted in saline was intraperitoneally administered to male C57BL/6J and senescence-accelerated mouse prone 8 (SAMP8) mice at a dose of 5 mg/kg every 3 days for 3 months. Both C57BL/6J and SAMP8 mice administered saline without PG-LPS comprised the control group. The mRNA expression levels of neprilysin and interleukin (IL)-10 were evaluated using the quantitative reverse transcriptase-polymerase chain reaction. The protein levels of neprilysin were assessed using western blotting. Sections of the brain tissues were immunohistochemically stained. RESULTS The serum IL-10 concentration significantly increased in both mouse strains after stimulation with PG-LPS. Neprilysin expression at both mRNA and protein levels was significantly lower in the SAMP8 PG-LPS group than those in the SAMP8 control group; however, they did not differ in PG-LPS-treated or non-treated C57BL/6J mice. Additionally, the immunofluorescence intensity of neprilysin in the CA3 region of the hippocampus in PG-LPS-treated SAMP8 mice was significantly lower than that in control SAMP8 mice. CONCLUSION Porphyromonas gingivalis may reduce the expression of neprilysin in elderly individuals and thus increase amyloid-β deposition.
Collapse
Affiliation(s)
- TETSURO MORIKAWA
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - OSAMU UEHARA
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - DURGA PAUDEL
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - KOKI YOSHIDA
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - FUMIYA HARADA
- Division of Oral and Maxillofacial Surgery, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - DAICHI HIRAKI
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - JUN SATO
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - HIROFUMI MATSUOKA
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - YASUHIRO KURAMITSU
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - MAKOTO MICHIKAWA
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - YOSHIHIRO ABIKO
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| |
Collapse
|
72
|
Brody M, Agronin M, Herskowitz BJ, Bookheimer SY, Small GW, Hitchinson B, Ramdas K, Wishard T, McInerney KF, Vellas B, Sierra F, Jiang Z, Mcclain-Moss L, Perez C, Fuquay A, Rodriguez S, Hare JM, Oliva AA, Baumel B. Results and insights from a phase I clinical trial of Lomecel-B for Alzheimer's disease. Alzheimers Dement 2023; 19:261-273. [PMID: 35357079 PMCID: PMC10084163 DOI: 10.1002/alz.12651] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 01/18/2023]
Abstract
HYPOTHESIS We hypothesized that Lomecel-B, an allogeneic medicinal signaling cell (MSC) therapeutic candidate for Alzheimer's disease (AD), is safe and potentially disease-modifying via pleiotropic mechanisms of action. KEY PREDICTIONS We prospectively tested the predictions that Lomecel-B administration to mild AD patients is safe (primary endpoint) and would provide multiple exploratory indications of potential efficacy in clinical and biomarker domains (prespecified secondary/exploratory endpoints). STRATEGY AND KEY RESULTS Mild AD patient received a single infusion of low- or high-dose Lomecel-B, or placebo, in a double-blind, randomized, phase I trial. The primary safety endpoint was met. Fluid-based and imaging biomarkers indicated significant improvement in the Lomecel-B arms versus placebo. The low-dose Lomecel-B arm showed significant improvements versus placebo on neurocognitive and other assessments. INTERPRETATION Our results support the safety of Lomecel-B for AD, suggest clinical potential, and provide mechanistic insights. This early-stage study provides important exploratory information for larger efficacy-powered clinical trials.
Collapse
Affiliation(s)
- Mark Brody
- Brain Matters Research, Delray Beach, Florida, USA
| | | | | | - Susan Y Bookheimer
- Dept. of Psychiatry and Biobehavioral Sciences, and Semel Institute For Neuroscience and Human Behavior, UCLA David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Gary W Small
- Psychiatry, Hackensack Meridian Health, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | | | | | - Tyler Wishard
- Interdepartmental Program in Neuroscience, UCLA, and Semel Institute For Neuroscience and Human Behavior, UCLA David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | - Bruno Vellas
- Gérontopôle, Department of Geriatric Internal Medicine, University of Toulouse, Toulouse, France
| | - Felipe Sierra
- National Institute of Aging, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Carmen Perez
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ana Fuquay
- Brain Matters Research, Delray Beach, Florida, USA
| | | | - Joshua M Hare
- Longeveron Inc., Miami, Florida, USA.,Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Bernard Baumel
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
73
|
Liu Y, Si ZZ, Zou CJ, Mei X, Li XF, Luo H, Shen Y, Hu J, Li XX, Wu L. Targeting neuroinflammation in Alzheimer’s disease: from mechanisms to clinical applications. Neural Regen Res 2023; 18:708-715. [DOI: 10.4103/1673-5374.353484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
74
|
Ethridge VT, Gargas NM, Sonner MJ, Moore RJ, Romer SH, Hatcher-Solis C, Rohan JG. Effects of transcranial direct current stimulation on brain cytokine levels in rats. Front Neurosci 2022; 16:1069484. [PMID: 36620466 PMCID: PMC9822516 DOI: 10.3389/fnins.2022.1069484] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Transcranial direct current stimulation (tDCS) has shown therapeutic potential to mitigate symptoms of various neurological disorders. Studies from our group and others used rodent models to demonstrate that tDCS modulates synaptic plasticity. We previously showed that 30 min of 0.25 mA tDCS administered to rats induced significant enhancement in the synaptic plasticity of hippocampal neurons. It has also been shown that tDCS induces expression of proteins known to mediate synaptic plasticity. This increase in synaptic plasticity may underly the observed therapeutic benefits of tDCS. However, the anti-inflammatory benefits of tDCS have not been thoroughly elucidated. Here we report that three sessions of tDCS spaced 1-3 weeks apart can significantly reduce levels of several inflammatory cytokines in brains of healthy rats. Rats receiving tDCS experienced enhanced synaptic plasticity without detectable improvement in behavioral tests or significant changes in astrocyte activation. The tDCS-mediated reduction in inflammatory cytokine levels supports the potential use of tDCS as a countermeasure against inflammation and offers additional support for the hypothesis that cytokines contribute to the modulation of synaptic plasticity.
Collapse
Affiliation(s)
- Victoria T. Ethridge
- Naval Medical Research Unit Dayton (NAMRU-D), Wright-Patterson Air Force Base, Dayton, OH, United States,Odyssey Systems Consulting Group, Wakefield, MA, United States,Leidos, Reston, VA, United States
| | - Nathan M. Gargas
- Naval Medical Research Unit Dayton (NAMRU-D), Wright-Patterson Air Force Base, Dayton, OH, United States,Odyssey Systems Consulting Group, Wakefield, MA, United States,Leidos, Reston, VA, United States
| | - Martha J. Sonner
- Naval Medical Research Unit Dayton (NAMRU-D), Wright-Patterson Air Force Base, Dayton, OH, United States,Leidos, Reston, VA, United States,ICON, Hinckley, OH, United States
| | - Raquel J. Moore
- Infoscitex, Dayton, OH, United States,711th HPW/RHBCN, Wright-Patterson Air Force Base, Dayton, OH, United States
| | - Shannon H. Romer
- Naval Medical Research Unit Dayton (NAMRU-D), Wright-Patterson Air Force Base, Dayton, OH, United States,Odyssey Systems Consulting Group, Wakefield, MA, United States,Leidos, Reston, VA, United States
| | | | - Joyce G. Rohan
- Naval Medical Research Unit Dayton (NAMRU-D), Wright-Patterson Air Force Base, Dayton, OH, United States,*Correspondence: Joyce G. Rohan,
| |
Collapse
|
75
|
Role of pro-inflammatory cytokines in Alzheimer's disease and neuroprotective effects of pegylated self-assembled nanoscaffolds. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 4:100149. [PMID: 36593925 PMCID: PMC9804106 DOI: 10.1016/j.crphar.2022.100149] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Neurodegeneration and synaptic loss in Alzheimer's disease (AD) lead to impairment in memory functions. Neuroinflammation causes activation of microglia and astrocytes cells that locally and systemically produces inflammatory cytokines which can serve as early diagnostic markers or therapeutic targets in AD. Pro-inflammatory cytokines (Interleukins (IL-1β, IL-6 and IL-10) and tumor necrosis factor (TNF α)) levels were estimated in serum, cerebral tissue, hepatic tissue, and renal tissue in treatment groups of scopolamine-induced amnesia mice model using ELISA protocol. The results showed that cerebral tissue of AD mice exhibited elevated levels of IL1β, IL6, IL10 and TNFα which indicate contribution of pro-inflammatory cytokines in the progression of AD. A significant reduction in the concentration of IL1β, IL-10 and TNF-α were noticed in serum, cerebral tissue and hepatic tissue of animal group treated with marketed memantine tablet (Admenta), pure memantine drug (MEMp), memantine-poly (lactic-co-glycolic acid) self-assembled nanoscaffolds (MEM-PLGA) SANs, Polyethylene Glycol coated memantine-poly (lactic-co-glycolic acid) self-assembled nanoscaffolds [(PEG-MEM-PLGA) SANs] and Polyethylene Glycol coated memantine-poly [(lactic-co-glycolic acid)] self-assembled nanoscaffolds grafted with Bone Marrow Derived Stem Cell ((PEG-MEM-PLGA) SANs-BMSc), whereas a high level of IL-6 was observed in hepatic tissue, cerebral tissue and renal tissues of normal and AD induced mice which showed the emerging potential of IL-6 cytokines that can trigger either neurons survival after injury or causing neurodegeneration and cell apoptosis. The Neuroregenerative potential of stem cells helps in the proliferation of neuronal cell and thus improves cognition in AD animal model.
Collapse
|
76
|
Tsioufis P, Theofilis P, Tsioufis K, Tousoulis D. The Impact of Cytokines in Coronary Atherosclerotic Plaque: Current Therapeutic Approaches. Int J Mol Sci 2022; 23:ijms232415937. [PMID: 36555579 PMCID: PMC9788180 DOI: 10.3390/ijms232415937] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Coronary atherosclerosis is a chronic pathological process that involves inflammation together with endothelial dysfunction and lipoprotein dysregulation. Experimental studies during the past decades have established the role of inflammatory cytokines in coronary artery disease, namely interleukins (ILs), tumor necrosis factor (TNF)-α, interferon-γ, and chemokines. Moreover, their value as biomarkers in disease development and progression further enhance the validity of this interaction. Recently, cytokine-targeted treatment approaches have emerged as potential tools in the management of atherosclerotic disease. IL-1β, based on the results of the CANTOS trial, remains the most validated option in reducing the residual cardiovascular risk. Along the same line, colchicine was also proven efficacious in preventing major adverse cardiovascular events in large clinical trials of patients with acute and chronic coronary syndrome. Other commercially available agents targeting IL-6 (tocilizumab), TNF-α (etanercept, adalimumab, infliximab), or IL-1 receptor antagonist (anakinra) have mostly been assessed in the setting of other inflammatory diseases and further testing in atherosclerosis is required. In the future, potential targeting of the NLRP3 inflammasome, anti-inflammatory IL-10, or atherogenic chemokines could represent appealing options, provided that patient safety is proven to be of no concern.
Collapse
|
77
|
Design and optimization of metformin-loaded solid lipid nanoparticles for neuroprotective effects in a rat model of diffuse traumatic brain injury: A biochemical, behavioral, and histological study. Eur J Pharm Biopharm 2022; 181:122-135. [PMID: 36307002 DOI: 10.1016/j.ejpb.2022.10.018] [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: 07/05/2022] [Revised: 09/13/2022] [Accepted: 10/18/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Following traumatic brain injury, inflammation, mitochondrial dysfunction, oxidative stress, ischemia, and energy crisis can cause mortality or long-term morbidity. As an activator of AMP-activated protein kinase, metformin reduces the secondary injuries of traumatic brain injury by compensating for the lack of energy in damaged cells. But the blood-brain barrier prevents a hydrophilic drug such as metformin from penetrating the brain tissue. Solid lipid nanoparticles with their lipid nature can cross the blood-brain barrier and solve this challenge. so This study aimed to investigate the effect of metformin-loaded lipid nanoparticles (NanoMet) for drug delivery to the brain and reduce complications from traumatic brain injury. METHOD Different formulations of NanoMet were designed by Box-Behnken, and after formulation, particle size, zeta potential, and entrapment efficiency were investigated. For in vivo study, Male rats were divided into eight groups, and except for the intact and sham groups, the other groups underwent brain trauma by the Marmarou method. After the intervention, the Veterinary Coma Scale, Vestibular Motor function, blood-brain barrier integrity, cerebral edema, level of inflammatory cytokines, and histopathology of brain tissue were assessed. RESULTS The optimal formula had a size of 282.2 ± 9.05 nm, a zeta potential of -1.65 ± 0.33 mV, and entrapment efficiency of 60.61 ± 6.09% which released the drug in 1400 min. Concentrations of 5 and 10 mg/kg of this formula improved the consequences of trauma. CONCLUSION This study showed that nanoparticles could help target drug delivery to the brain and apply the desired result.
Collapse
|
78
|
Ramanzini LG, Camargo LFM, Silveira JOF, Bochi GV. Inflammatory markers and depression in Parkinson's disease: a systematic review. Neurol Sci 2022; 43:6707-6717. [PMID: 36040559 DOI: 10.1007/s10072-022-06363-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 08/20/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Parkinson's disease (PD) patients experience non-motor symptoms (NMS), which may appear before motor manifestations. The most common NMS is depression, affecting about 30-40% of PD patients. Both PD and depression are associated with an increased inflammatory burden, with studies showing elevation of diverse inflammatory markers in patients with both conditions. METHODS A systematic review was conducted in PubMed and PsycINFO databases to investigate what inflammatory markers are associated with PD depression (PDD). Only studies in English that measured inflammatory markers and analyzed against depression scores in PD patients were included. RESULTS A total of 1132 articles were retrieved, and 14 entries were found to be eligible. Twelve were cross-sectional studies, one was a cohort, and one was a non-randomized controlled trial. IL-17A was the only marker strongly associated with PDD, while studies assessing sIL-2R and serum amyloid A found a moderate correlation. C-reactive protein, IL-10, tumor necrosis factor-α, monocyte chemoattractant protein-1, and IL-6 yielded conflicting results. Their possible roles in PDD are discussed. PDD was also related to longer disease duration and other NMS, such as anxiety, fatigue, dementia, REM sleep behavior disorder, and autonomic dysfunction. CONCLUSION We suggest that these markers may be used for distinguishing isolated depression from that related to neurodegeneration, especially in individuals that concurrently present with other known prodromal symptoms of PD and other α-synucleinopathies. However, future prospective studies are warranted to confirm this hypothesis.
Collapse
Affiliation(s)
- Luis Guilherme Ramanzini
- Department of Physiology and Pharmacology, Federal University of Santa Maria, 1000 Roraima Avenue, Building 21, Santa Maria, Rio Grande do Sul, Brazil.
| | - Luís Fernando Muniz Camargo
- Department of Physiology and Pharmacology, Federal University of Santa Maria, 1000 Roraima Avenue, Building 21, Santa Maria, Rio Grande do Sul, Brazil
| | | | - Guilherme Vargas Bochi
- Department of Physiology and Pharmacology, Federal University of Santa Maria, 1000 Roraima Avenue, Building 21, Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
79
|
Stone TW, Clanchy FIL, Huang YS, Chiang NY, Darlington LG, Williams RO. An integrated cytokine and kynurenine network as the basis of neuroimmune communication. Front Neurosci 2022; 16:1002004. [PMID: 36507331 PMCID: PMC9729788 DOI: 10.3389/fnins.2022.1002004] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Two of the molecular families closely associated with mediating communication between the brain and immune system are cytokines and the kynurenine metabolites of tryptophan. Both groups regulate neuron and glial activity in the central nervous system (CNS) and leukocyte function in the immune system, although neither group alone completely explains neuroimmune function, disease occurrence or severity. This essay suggests that the two families perform complementary functions generating an integrated network. The kynurenine pathway determines overall neuronal excitability and plasticity by modulating glutamate receptors and GPR35 activity across the CNS, and regulates general features of immune cell status, surveillance and tolerance which often involves the Aryl Hydrocarbon Receptor (AHR). Equally, cytokines and chemokines define and regulate specific populations of neurons, glia or immune system leukocytes, generating more specific responses within restricted CNS regions or leukocyte populations. In addition, as there is a much larger variety of these compounds, their homing properties enable the superimposition of dynamic variations of cell activity upon local, spatially limited, cell populations. This would in principle allow the targeting of potential treatments to restricted regions of the CNS. The proposed synergistic interface of 'tonic' kynurenine pathway affecting baseline activity and the superimposed 'phasic' cytokine system would constitute an integrated network explaining some features of neuroimmune communication. The concept would broaden the scope for the development of new treatments for disorders involving both the CNS and immune systems, with safer and more effective agents targeted to specific CNS regions.
Collapse
Affiliation(s)
- Trevor W. Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom,*Correspondence: Trevor W. Stone,
| | - Felix I. L. Clanchy
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Yi-Shu Huang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Nien-Yi Chiang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - L. Gail Darlington
- Department of Internal Medicine, Ashtead Hospital, Ashtead, United Kingdom
| | - Richard O. Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
80
|
Fakharaldeen Z, Al-Mudhafar A, Radhi A, Hadi N. Potential protective effects of Azelnidipine against cerebral ischemia-reperfusion injury in male rats. J Med Life 2022; 15:1384-1391. [PMID: 36567842 PMCID: PMC9762371 DOI: 10.25122/jml-2022-0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/11/2022] [Indexed: 12/27/2022] Open
Abstract
This study was performed to evaluate the neuroprotective effect of Azelnidipine in cerebral ischemia/reperfusion and to envisage its mechanisms. Twenty-eight adult male Sprague-Dawley rats weighing 200-300 g were randomized into 4 groups (7 rats in each group). Sham (neck dissection without bilateral common carotid artery occlusion), control (30 minutes of bilateral common carotid artery occlusion and reperfusion for 1 hour), vehicle (identical volume of 0.3% carboxymethylcellulose (CMC) orally every day then bilateral common artery occlusion and reperfusion), and Azelnipine-treated rats (7 days of Azelnidipine pretreatment 3 mg/kg/day followed by bilateral common carotid artery occlusion and reperfusion). In addition to brain infarct volume and histopathological assessment, the brain tissues were harvested to evaluate cerebral IL-6, IL-10, TNF-α, ICAM-1, NF-κB p65, and total antioxidant capacity levels. Cerebral levels of IL-6, IL-10, TNF-α, NF-κB p65, and ICAM-1, besides cerebral infarct volume, were significantly elevated in control and vehicle related to sham groups, while total antioxidant capacity was markedly reduced. Azelnidipine treatment resulted in remarkable upregulation of total antioxidant capacity; meanwhile, IL-6, TNF-α, NF-κB p65, and ICAM-1 showed a considerable reduction. Cerebral IL-10 levels were not affected by Azelnidipine pretreatment. Histologically, control and vehicle rats showed severe ischemic injury, which was greatly reversed by Azelnidipine treatment. The current study disclosed that Azelnidipine could markedly reduce cerebral infarct volume and ameliorate histopathological damage in male rats exposed to cerebral ischemia/reperfusion. The neuroprotective effects of Azelnidipine probably stemmed from its anti-inflammatory and antioxidative properties. Azelnidipine had no effect on cerebral IL-10 levels.
Collapse
Affiliation(s)
- Zainab Fakharaldeen
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq,Corresponding Author: Zainab Fakharaldeen, Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Iraq. E-mail:
| | - Ahmed Al-Mudhafar
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Ali Radhi
- Department of Medicine, Al-Hakeem Hospital, Al-Najaf Al-Ashraf, Iraq
| | - Najah Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| |
Collapse
|
81
|
Ruffolo G, Alfano V, Romagnolo A, Zimmer T, Mills JD, Cifelli P, Gaeta A, Morano A, Anink J, Mühlebner A, Vezzani A, Aronica E, Palma E. GABA A receptor function is enhanced by Interleukin-10 in human epileptogenic gangliogliomas and its effect is counteracted by Interleukin-1β. Sci Rep 2022; 12:17956. [PMID: 36289354 PMCID: PMC9605959 DOI: 10.1038/s41598-022-22806-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/19/2022] [Indexed: 01/24/2023] Open
Abstract
Gangliogliomas (GGs) are low-grade brain tumours that cause intractable focal epilepsy in children and adults. In GG, as in epileptogenic focal malformations (i.e., tuberous sclerosis complex, TSC), there is evidence of sustained neuroinflammation with involvement of the pro-inflammatory cytokine IL-1β. On the other hand, anti-inflammatory mediators are less studied but bear relevance for understanding seizure mechanisms. Therefore, we investigated the effect of the key anti-inflammatory cytokine IL-10 on GABAergic neurotransmission in GG. We assessed the IL-10 dependent signaling by transcriptomic analysis, immunohistochemistry and performed voltage-clamp recordings on Xenopus oocytes microtransplanted with cell membranes from brain specimens, to overcome the limited availability of acute GG slices. We report that IL-10-related mRNAs were up-regulated in GG and slightly in TSC. Moreover, we found IL-10 receptors are expressed by neurons and astroglia. Furthermore, GABA currents were potentiated significantly by IL-10 in GG. This effect was time and dose-dependent and inhibited by blockade of IL-10 signaling. Notably, in the same tissue, IL-1β reduced GABA current amplitude and prevented the IL-10 effect. These results suggest that in epileptogenic tissue, pro-inflammatory mechanisms of hyperexcitability prevail over key anti-inflammatory pathways enhancing GABAergic inhibition. Hence, boosting the effects of specific anti-inflammatory molecules could resolve inflammation and reduce intractable seizures.
Collapse
Affiliation(s)
- Gabriele Ruffolo
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| | - Veronica Alfano
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| | - Alessia Romagnolo
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - Till Zimmer
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - James D. Mills
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.83440.3b0000000121901201Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK ,grid.452379.e0000 0004 0386 7187Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Pierangelo Cifelli
- grid.158820.60000 0004 1757 2611Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandro Gaeta
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
| | - Alessandra Morano
- grid.7841.aDepartment of Human Neuroscience, University of Rome Sapienza, Rome, Italy
| | - Jasper Anink
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.7692.a0000000090126352Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annamaria Vezzani
- grid.4527.40000000106678902Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Eleonora Aronica
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.419298.f0000 0004 0631 9143Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands
| | - Eleonora Palma
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| |
Collapse
|
82
|
Sakurai K, Toshimitsu T, Okada E, Anzai S, Shiraishi I, Inamura N, Kobayashi S, Sashihara T, Hisatsune T. Effects of Lactiplantibacillus plantarum OLL2712 on Memory Function in Older Adults with Declining Memory: A Randomized Placebo-Controlled Trial. Nutrients 2022; 14:nu14204300. [PMID: 36296983 PMCID: PMC9610166 DOI: 10.3390/nu14204300] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The use of probiotics is expected to be an intervention in neurodegenerative conditions that cause dementia owing to their ability to modulate neuroinflammatory responses via the microbiome-gut–brain axis. Therefore, we selected Lactiplantibacillus plantarum OLL2712 (OLL2712), the optimal anti-inflammatory lactic acid bacteria strain with high IL-10-inducing activity in immune cells, and aimed to verify its protective effects on memory function in older adults. A 12-week, randomized, double-blind, placebo-controlled trial was performed with older adults over the age of 65 years with declining memory. The participants consumed either powder containing heat-treated OLL2712 cells or placebo. Memory function was assessed using a computer-assisted cognitive test, Cognitrax. Daily dietary nutrient intake was assessed using the Brief-type Self-administered Diet History Questionnaire (BDHQ). The composition of the gut microbiota was analyzed by fecal DNA extraction and 16S rDNA sequencing. Data from 78 participants who completed the entire procedure were analyzed, and significant improvements in composite memory and visual memory scores were observed in the active group, after accounting for the effect of daily nutritional intake (p = 0.044 and p = 0.021, respectively). In addition, the active group had a lower abundance ratio of Lachnoclostridium, Monoglobus, and Oscillibacter genera, which have been reported to be involved in inflammation. The present study suggests that OLL2712 ingestion has protective effects against memory function decline in older adults.
Collapse
Affiliation(s)
- Keisuke Sakurai
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan
| | - Takayuki Toshimitsu
- Food Microbiology Research Laboratories, Applied Microbiology Research Department, Division of Research and Development, Meiji Co., Ltd., Hachiouji 192-0919, Japan
| | - Erika Okada
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan
| | - Saya Anzai
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan
| | - Izumi Shiraishi
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan
| | - Noriko Inamura
- Urban Design Center Kashiwanoha (UDCK), Kashiwa 277-0871, Japan
- Community Health Promotion Laboratory, Mitsui Fudosan, Co., Ltd., Kashiwa 277-8519, Japan
| | - Satoru Kobayashi
- Community Health Promotion Laboratory, Mitsui Fudosan, Co., Ltd., Kashiwa 277-8519, Japan
| | - Toshihiro Sashihara
- Food Microbiology Research Laboratories, Applied Microbiology Research Department, Division of Research and Development, Meiji Co., Ltd., Hachiouji 192-0919, Japan
| | - Tatsuhiro Hisatsune
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan
- Correspondence: ; Tel.: +81-04-7136-3632
| |
Collapse
|
83
|
Singh V, Kaur K, Kaur S, Shri R, Singh TG, Singh M. Trimethoxyflavones from Ocimum basilicum L. leaves improve long term memory in mice by modulating multiple pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115438. [PMID: 35671863 DOI: 10.1016/j.jep.2022.115438] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/24/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditionally, Ocimum basilicum L. leaves (OB) are recommended for various brain disorders. AIM OF THE STUDY Scientific evidence highlights the cognition improvement capacity of Ocimum basilicum L. leave extract (OBE), however, the compound(s) responsible for this effect and the associated mechanism was not reported. The present study was, thus, designed to isolate and identify the compound responsible for memory improvement effects of OB and to delineate the associated mechanism of action. MATERIALS AND METHODS In-vitro acetylcholinesterase (AChE) inhibitory (Ellman method) and antioxidant (DPPH scavenging) assays guided fractionation was employed to isolate the bioactive compounds from OBE. The isolated compounds were characterised using spectroscopic techniques (FTIR, NMR and MS). In-silico and in-vivo [mouse model of scopolamine (SCOP) induced amnesia] investigations were used to substantiate the memory improvement effects of isolated compounds and to understand their mechanism of action. RESULTS AChE and DPPH assays guided fractionation of OBE lead to isolation of two pure compounds namely, 5,7-dihydroxy-3',4',5'-trimethoxyflavone (S1) and 3-hydroxy-3',4',5'-trimethoxyflavone (S2). Both S1 and S2 mitigated the cognitive impairment due to SCOP in mice by reducing brain AChE activity, TBARS, TNF-α, IL-1β, IL-6 and caspase-3 concentrations and elevating reduced glutathione and IL-10 levels; together with amelioration of brain hippocampus histopathological aberration (H and E staining). Moreover, the molecular docking of S1 and S2 at the active pockets of AChE and caspase-3 has shown good interactions with vital amino acid residues. CONCLUSIONS Our findings show that trimethoxy flavones are responsible for the memory improvement effect of OBE due to their anticholinergic, antioxidant, anti-inflammatory and anti-apoptotic properties. These maybe developed as valuable alternatives for management of cognitive disorders.
Collapse
Affiliation(s)
- Varinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Kiranpreet Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India.
| | - Sanimardeep Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India.
| | - Richa Shri
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India.
| | | | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
84
|
Cai L, He C, Liu Y, Sun Y, He L, Baranova A. Inflammation and immunity connect hypertension with adverse COVID-19 outcomes. Front Genet 2022; 13:933148. [PMID: 36160003 PMCID: PMC9493274 DOI: 10.3389/fgene.2022.933148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/08/2022] [Indexed: 11/23/2022] Open
Abstract
Objectives: To explore the connection of hypertension and severe COVID-19 outcomes. Methods: A total of 68 observational studies recording mortality and/or general severity of COVID-19 were pooled for meta-analyses of the relationship of severe COVID-19 outcomes with hypertension as well as systolic and diastolic blood pressure. Genome-wide cross-trait meta-analysis (GWCTM) was performed to explore the genes linking between hypertension and COVID-19 severity. Results: The results of meta-analysis with the random effect model indicated that pooled risk ratios of hypertension on mortality and severity of COVID-19 were 1.80 [95% confidence interval (CI) 1.54-2.1] and 1.78 (95% confidence interval 1.56-2.04), respectively, although the apparent heterogeneity of the included studies was detected. In subgroup analysis, cohorts of severe and mild patients of COVID-19 assessed in Europe had a significant pooled weighted mean difference of 6.61 mmHg (95% CI 3.66-9.55) with no heterogeneity found (p = 0.26). The genes in the shared signature of hypertension and the COVID-19 severity were mostly expressed in lungs. Analysis of molecular networks commonly affected both by hypertension and by severe COVID-19 highlighted CCR1/CCR5 and IL10RB signaling, as well as Th1 and Th2 activation pathways, and also a potential for a shared regulation with multiple sclerosis. Conclusion: Hypertension is significantly associated with the severe course of COVID-19. Genetic variants within inflammation- and immunity-related genes may affect their expression in lungs and confer liability to both elevated blood pressure and to severe COVID-19.
Collapse
Affiliation(s)
- Lei Cai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai, China
| | - Chuan He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai, China
| | - Yonglin Liu
- Sanya Women and Children’s Hospital managed by Shanghai Children’s Medical Center, Sanya, China
| | - Yanlan Sun
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Genetics and Development, Shanghai Mental Health Center, Shanghai Jiaotong University, Shanghai, China
- Shanghai Center for Women and Children’s Health, Shanghai, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Fairfax, VA, United States
| |
Collapse
|
85
|
Rahbaran M, Zekiy AO, Bahramali M, Jahangir M, Mardasi M, Sakhaei D, Thangavelu L, Shomali N, Zamani M, Mohammadi A, Rahnama N. Therapeutic utility of mesenchymal stromal cell (MSC)-based approaches in chronic neurodegeneration: a glimpse into underlying mechanisms, current status, and prospects. Cell Mol Biol Lett 2022; 27:56. [PMID: 35842587 PMCID: PMC9287902 DOI: 10.1186/s11658-022-00359-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/30/2022] [Indexed: 12/11/2022] Open
Abstract
Recently, mesenchymal stromal cell (MSC)-based therapy has become an appreciated therapeutic approach in the context of neurodegenerative disease therapy. Accordingly, a myriad of studies in animal models and also some clinical trials have evinced the safety, feasibility, and efficacy of MSC transplantation in neurodegenerative conditions, most importantly in Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington’s disease (HD). The MSC-mediated desired effect is mainly a result of secretion of immunomodulatory factors in association with release of various neurotrophic factors (NTFs), such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF). Thanks to the secretion of protein-degrading molecules, MSC therapy mainly brings about the degradation of pathogenic protein aggregates, which is a typical appearance of chronic neurodegenerative disease. Such molecules, in turn, diminish neuroinflammation and simultaneously enable neuroprotection, thereby alleviating disease pathological symptoms and leading to cognitive and functional recovery. Also, MSC differentiation into neural-like cells in vivo has partially been evidenced. Herein, we focus on the therapeutic merits of MSCs and also their derivative exosome as an innovative cell-free approach in AD, HD, PD, and ALS conditions. Also, we give a brief glimpse into novel approaches to potentiate MSC-induced therapeutic merits in such disorders, most importantly, administration of preconditioned MSCs.
Collapse
Affiliation(s)
- Mohaddeseh Rahbaran
- Biotechnology Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Mahta Bahramali
- Biotechnology Department, University of Tehran, Tehran, Iran
| | | | - Mahsa Mardasi
- Biotechnology Department, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Delaram Sakhaei
- School of Medicine, Sari Branch, Islamic Azad University, Sari, Iran
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Zamani
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Ali Mohammadi
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran.
| | - Negin Rahnama
- Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
86
|
Hur HJ, Lee JY, Kim DH, Cho MS, Lee S, Kim HS, Kim DW. Conditioned Medium of Human Pluripotent Stem Cell-Derived Neural Precursor Cells Exerts Neurorestorative Effects against Ischemic Stroke Model. Int J Mol Sci 2022; 23:7787. [PMID: 35887140 PMCID: PMC9319001 DOI: 10.3390/ijms23147787] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023] Open
Abstract
Previous studies have shown that early therapeutic events of neural precursor cells (NPCs) transplantation to animals with acute ischemic stroke readily protected neuronal cell damage and improved behavioral recovery through paracrine mechanisms. In this study, we tested the hypothesis that administration of conditioned medium from NPCs (NPC-CMs) could recapitulate the beneficial effects of cell transplantation. Rats with permanent middle cerebral artery occlusion (pMCAO) were randomly assigned to one of the following groups: PBS control, Vehicle (medium) controls, single (NPC-CM(S)) or multiple injections of NPC-CM(NPC-CM(M)) groups. A single intravenous injection of NPC-CM exhibited strong neuroregenerative potential to induce behavioral recovery, and multiple injections enhanced this activity further by suppressing inflammatory damage and inducing endogenous neurogenesis leading to histopathological and functional recovery. Proteome analysis of NPC-CM identified a number of proteins that are known to be associated with nervous system development, neurogenesis, and angiogenesis. In addition, transcriptome analysis revealed the importance of the inflammatory response during stroke recovery and some of the key hub genes in the interaction network were validated. Thus, our findings demonstrated that NPC-CM promoted functional recovery and reduced cerebral infarct and inflammation with enhanced endogenous neurogenesis, and the results highlighted the potency of NPC-CM in stroke therapy.
Collapse
Affiliation(s)
- Hye-Jin Hur
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-J.H.); (D.-H.K.)
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji Yong Lee
- Research Institute of Hyperbaric Medicine and Science, Yonsei University Wonju College of Medicine, Wonju-si 26426, Korea;
| | - Do-Hun Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-J.H.); (D.-H.K.)
- S. Biomedics Co., Ltd., Seoul 04979, Korea;
| | | | - Sangsik Lee
- Department of Biomedical Engineering, College of Medical Convergence, Catholic Kwandong University, Gangneung-si 25601, Korea;
| | - Han-Soo Kim
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si 25601, Korea
| | - Dong-Wook Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-J.H.); (D.-H.K.)
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
87
|
Rashno M, Sarkaki A, Ghaderi S, Khoshnam SE. Sesamin: Insights into its protective effects against lead-induced learning and memory deficits in rats. J Trace Elem Med Biol 2022; 72:126993. [PMID: 35550983 DOI: 10.1016/j.jtemb.2022.126993] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/03/2022] [Accepted: 05/03/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Lead (Pb) is one of the most hazardous pollutants that induce a wide spectrum of neurological changes such as learning and memory deficits. Sesamin, a phytonutrient of the lignan class, exhibits anti-inflammatory, anti-apoptotic, and neuroprotective properties. The present study was designed to investigate the effects of sesamin against Pb-induced learning and memory deficits, disruption of hippocampal theta and gamma rhythms, inflammatory response, inhibition of blood δ-aminolevulinic acid dehydratase (δ-ALA-D) activity, Pb accumulation, and neuronal loss in rats. METHODS Sesamin treatment (30 mg/kg/day; P.O.) was started simultaneously with Pb acetate exposure (500 ppm in standard drinking water) in rats, and they continued for eight consecutive weeks. RESULTS The results showed that chronic exposure to Pb disrupted the learning and memory functions in both passive-avoidance and water-maze tests, which was accompanied by increase in spectral theta power and theta/gamma ratio, and a decrease in spectral gamma power in the hippocampus. Additionally, Pb exposure resulted in an enhanced tumor necrosis factor-alpha (TNF-α) content, decreased interleukin-10 (IL-10) production, inhibited blood δ-ALA-D activity, increased Pb accumulation, and neuronal loss of rats. In contrast, sesamin treatment improved all the above-mentioned Pb-induced pathological changes. CONCLUSION This data suggests that sesamin could improve Pb-induced learning and memory deficits, possibly through amelioration of hippocampal theta and gamma rhythms, modulation of inflammatory status, restoration of the blood δ-ALA-D activity, reduction of Pb accumulation in the blood and the brain tissues, and prevention of neuronal loss.
Collapse
Affiliation(s)
| | - Alireza Sarkaki
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahab Ghaderi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
88
|
Paulina MS, Małgorzata C, Zbigniew W, Jaroslaw M, Anna S. High-intensity interval training modulates inflammatory response in Parkinson's disease. Aging Clin Exp Res 2022; 34:2165-2176. [PMID: 35699838 PMCID: PMC9192928 DOI: 10.1007/s40520-022-02153-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/08/2022] [Indexed: 11/29/2022]
Abstract
Background Recent discoveries show that high-intensity interval training (HIIT) can bring many positive effects such as decreases in fat tissue, lower blood sugar levels, improved learning and memory, and lower risk of cardiac disease. Parkinson’s disease (PD) is a neurodegenerative disorder characterized by loss of the dopaminergic neurons, accompanied by chronic inflammation and neuroinflammation. Previous research shows that interval training can bring a beneficial effect on the inflammation and neuroplasticity in PD. Objectives The objective of this study was to investigate the effect of 12 weeks of HIIT on the inflammation levels and antioxidant capacity in the serum of PD patients. Methods Twenty-eight people diagnosed with PD were enrolled in this study. Fifteen PD patients performed 12 weeks of HIIT on a cycloergometer. Thirteen non-exercised PD patients constitute the control group. Concentrations of inflammation markers and antioxidants’ capacity in the serum were measured at 3 sampling points (a week before, a week after, and 3 months after the HIIT). Results Twelve weeks of HIIT decreases the level of TNF-α (p = 0.034) and increases the level of IL-10 (p = 0.024). Those changes were accompanied by a decreased level of neutrophils (p = 0.03), neutrophil/lymphocyte ratio (p = 0.048) and neutrophil/monocyte ratio (p = 0.0049) with increases in superoxide dismutase levels (p = 0.04). Conclusions Twelve weeks of HIIT can decrease systemic inflammation in PD patients and improve the antioxidant capacity in their serum, which can slow down the progression of the disease.
Collapse
Affiliation(s)
- Malczynska-Sims Paulina
- Department of Genetics, Institute of Psychiatry and Neurology, 9 Sobieskiego St, 02-957, Warsaw, Poland.
| | - Chalimoniuk Małgorzata
- Department of Physical Education and Health in Biała Podlaska, Józef Piłsudski University of Physical Education in Warsaw, 2 Akademicka St, 21-500, Biała Podlaska, Poland
| | - Wronski Zbigniew
- Department of Rehabilitation, Medical Faculty, Medical University of Warsaw, 61 Żwirki i Wigury St., 02-091, Warsaw, Poland
| | - Marusiak Jaroslaw
- Department of Kinesiology, University School of Physical Education in Wroclaw, Wroclaw, Poland
| | - Sulek Anna
- Department of Genetics, Institute of Psychiatry and Neurology, 9 Sobieskiego St, 02-957, Warsaw, Poland
| |
Collapse
|
89
|
Ponce-Regalado MD, Salazar-Juárez A, Oscar RE, Contis-Montes de Oca A, Hurtado-Alvarado G, Arce-Paredes P, Pérez-Sánchez G, Pavón L, Girón-Pérez MI, Hernández-Pando R, Alvarez-Sánchez ME, Enrique BV. Development of Anxiolytic and Depression-like Behavior in Mice Infected with Mycobacterium lepraemurium. Neuroscience 2022; 493:15-30. [PMID: 35447197 DOI: 10.1016/j.neuroscience.2022.03.021] [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: 05/06/2021] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022]
Abstract
Murine leprosy is a systemic infectious disease of mice caused by Mycobacterium lepraemurium (MLM) in which the central nervous system (CNS) is not infected; nevertheless, diseased animals show measurable cognitive alterations. For this reason, in this study, we explored the neurobehavioral changes in mice chronically infected with MLM. BALB/c mice were infected with MLM, and 120 days later, the alterations in mice were evaluated based on immunologic, histologic, endocrine, neurochemical, and behavioral traits. We found increases in the levels of IL-4 and IL-10 associated with high bacillary loads. We also found increase in the serum levels of corticosterone, epinephrine, and norepinephrine in the adrenal gland, suggesting neuroendocrine deregulation. Mice exhibited depression-like behavior in the tail suspension and forced swimming tests and anxiolytic behavior in the open field and elevated plus maze tests. The neurobehavioral alterations of mice were correlated with the histologic damage in the prefrontal cortex, ventral hippocampus, and amygdala, as well as with a blood-brain barrier disruption in the hippocampus. These results reveal an interrelated response of the neuroimmune-endocrinological axis in unresolved chronic infections that result in neurocognitive deterioration.
Collapse
Affiliation(s)
- M D Ponce-Regalado
- Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara, Carretera a Yahualica, Km. 7.5 Tepatitlán de Morelos, Jalisco 47600, Mexico
| | - A Salazar-Juárez
- Branch Clinical Research. Laboratory of Molecular Neurobiology and Neurochemistry of Addiction, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - Rojas-Espinosa Oscar
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala, Colonia Santo Tomás, 11340, Ciudad de México, Mexico.
| | - A Contis-Montes de Oca
- Sección de estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Diaz Miron y Plan de San Luis S/N, Miguel Hidalgo, 11340 Mexico City, Mexico
| | - G Hurtado-Alvarado
- Area of Neurosciences, Department of Biology of Reproduction, Area of Neurosciences, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - P Arce-Paredes
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
| | - G Pérez-Sánchez
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - L Pavón
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - M I Girón-Pérez
- Laboratorio de Inmunotoxicología, Secretaría de Investigación y Posgrado Universidad Autónoma de Nayarit, Boulevard Tepic-Xalisco s/n. Cd, de la Cultura Amado Nervo, C.P. 63000 Tepic, Nayarit, México
| | - R Hernández-Pando
- Experimental Pathology Section, Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Colonia Belisario Dominguez Seccion XVI, 14080, Deleg. Tlalpan, México City, Mexico
| | - M E Alvarez-Sánchez
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo # 290, Col. Del Valle, CP 03100 México City, México
| | - Becerril-Villanueva Enrique
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico.
| |
Collapse
|
90
|
Mann CN, Devi SS, Kersting CT, Bleem AV, Karch CM, Holtzman DM, Gallardo G. Astrocytic α2-Na +/K + ATPase inhibition suppresses astrocyte reactivity and reduces neurodegeneration in a tauopathy mouse model. Sci Transl Med 2022; 14:eabm4107. [PMID: 35171651 DOI: 10.1126/scitranslmed.abm4107] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most dominant form of dementia characterized by the deposition of extracellular amyloid plaques and intracellular neurofibrillary tau tangles (NFTs). In addition to these pathologies, an emerging pathophysiological mechanism that influences AD is neuroinflammation. Astrocytes are a vital type of glial cell that contribute to neuroinflammation, and reactive astrocytes, or astrogliosis, are a well-known pathological feature of AD. However, the mechanisms by which astrocytes contribute to the neurodegenerative process in AD have not been fully elucidated. Here, we showed that astrocytic α2-Na+/K+ adenosine triphosphatase (α2-NKA) is elevated in postmortem human brain tissue from AD and progressive nuclear palsy, a primary tauopathy. The increased astrocytic α2-NKA was also recapitulated in a mouse model of tauopathy. Pharmacological inhibition of α2-NKA robustly suppressed neuroinflammation and reduced brain atrophy. In addition, α2-NKA knockdown in tauopathy mice halted the accumulation of tau pathology. We also demonstrated that α2-NKA promoted tauopathy, in part, by regulating the proinflammatory protein lipocalin-2 (Lcn2). Overexpression of Lcn2 in tauopathy mice increased tau pathology, and prolonged Lcn2 exposure to primary neurons promoted tau uptake in vitro. These studies collectively highlight the contribution of reactive astrocytes to tau pathogenesis in mice and define α2-NKA as a major regulator of astrocytic-dependent neuroinflammation.
Collapse
Affiliation(s)
- Carolyn N Mann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Shamulailatpam Shreedarshanee Devi
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Corey T Kersting
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Amber V Bleem
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Celeste M Karch
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - Gilbert Gallardo
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
91
|
Benameur T, Panaro MA, Porro C. Exosomes and their Cargo as a New Avenue for Brain and Treatment of CNS-Related Diseases. Open Neurol J 2022. [DOI: 10.2174/1874205x-v16-e2201190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Extracellular Vesicles (EVs), which belong to nanoscale vesicles, including microvesicles (MVs) and exosomes, are now considered a new important tool for intercellular neuronal communication in the Central Nervous System (CNS) under physiological and pathological conditions. EVs are shed into blood, peripheral body fluids and cerebrospinal fluid (CSF) by a large variety of cells.
EVs can act locally on neighboring and distant cells. EVs represent the fingerprints of the originating cells and can carry a variety of molecular constituents of their cell of origin, including protein, lipids, DNA and microRNAs (miRNAs).
The most studied EVs are the exosomes because they are ubiquitous and have the capacity to transfer cell-derived components and bioactive molecules to target cells. In this minireview, we focused on cell-cell communication in CNS mediated by exosomes and their important cargo as an innovative way to treat or follow up with CNS diseases.
Collapse
|
92
|
Jiang X, He H, Mo L, Liu Q, Yang F, Zhou Y, Li L, Su D, Yi S, Zhang J. Mapping the Plasticity of Morphology, Molecular Properties and Function in Mouse Primary Microglia. Front Cell Neurosci 2022; 15:811061. [PMID: 35153675 PMCID: PMC8825496 DOI: 10.3389/fncel.2021.811061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Microglia exert diverse functions by responding in diverse ways to different stimuli, yet little is known about the plasticity of various phenotypes that microglia display. We used interferon (IFN)-γ, interleukin (IL)-4 and IL-10 to induce different phenotypes in mouse primary microglia. RNA sequencing was used to identify genes differentially expressed in response to stimulation, and the different stimulated populations were compared in terms of morphology, proliferative capacity, phagocytic ability and neurotoxicity. IFN-γ induced an “immunodefensive” phenotype characterizing both induction of filopodia and upregulation of inducible nitric oxide synthase (iNOS) and tumor necrosis factor α. Microglia with this phenotype mediated an acute inflammatory response accompanied by excellent proliferative capacity and neurotoxicity, and remained susceptible to remodeling for up to 48 h after initial stimulation. IL-4 induced an enduring “neuroimmunoregulatory” phenotype involving induction of lamellipodium and persistent upregulation of arginase (Arg)-1 and YM-1 expression. Microglia with this phenotype remained susceptible to remodeling for up to 24 h after initial stimulation. IL-10 induced an “immunosuppressive” phenotype involving induction of ameba-like morphology and upregulation of transforming growth factor β and IL-10 as well as inhibition of inflammation. This phenotype was accompanied by inhibition of self-proliferation, while its morphology, molecular properties and function were the least susceptible to remodeling. IFN-γ, IL-4, or IL-10 appear to induce substantially different phenotypes in microglia. The immunodefensive microglia induced by IFN-γ showed remarkable plasticity, which may help repair CNS inflammation damage under pathological condition. Chronic activation with IL-10 decreases microglial plasticity, which may help protect the brain form the immune response. Our research justifies and guides further studies into the molecular pathways that operate in each phenotype to help multitasking microglia regulate homeostasis in the brain.
Collapse
Affiliation(s)
- Xue Jiang
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hui He
- School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Mo
- School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Qin Liu
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Fan Yang
- Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ying Zhou
- Guizhou Provincial People’s Hospital, Guiyang, China
| | - Liangyuan Li
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dapeng Su
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Saini Yi
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jinqiang Zhang
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- *Correspondence: Jinqiang Zhang,
| |
Collapse
|
93
|
Dabravolski SA, Nikiforov NG, Zhuravlev AD, Orekhov NA, Grechko AV, Orekhov AN. Role of the mtDNA Mutations and Mitophagy in Inflammaging. Int J Mol Sci 2022; 23:ijms23031323. [PMID: 35163247 PMCID: PMC8836173 DOI: 10.3390/ijms23031323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Ageing is an unavoidable multi-factorial process, characterised by a gradual decrease in physiological functionality and increasing vulnerability of the organism to environmental factors and pathogens, ending, eventually, in death. One of the most elaborated ageing theories implies a direct connection between ROS-mediated mtDNA damage and mutations. In this review, we focus on the role of mitochondrial metabolism, mitochondria generated ROS, mitochondrial dynamics and mitophagy in normal ageing and pathological conditions, such as inflammation. Also, a chronic form of inflammation, which could change the long-term status of the immune system in an age-dependent way, is discussed. Finally, the role of inflammaging in the most common neurodegenerative diseases, such as Alzheimer’s and Parkinson’s, is also discussed.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Clinical Diagnostics, Vitebsk State Academy of Veterinary Medicine [UO VGAVM], 7/11 Dovatora Str., 210026 Vitebsk, Belarus
- Correspondence:
| | - Nikita G. Nikiforov
- AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia; (N.G.N.); (A.D.Z.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Alexander D. Zhuravlev
- AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia; (N.G.N.); (A.D.Z.)
| | - Nikolay A. Orekhov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia; (N.A.O.); (A.N.O.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia; (N.A.O.); (A.N.O.)
| |
Collapse
|
94
|
Yoo TJ. Anti-Inflammatory Gene Therapy Improves Spatial Memory Performance in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2021; 85:1001-1008. [PMID: 34897091 PMCID: PMC8925118 DOI: 10.3233/jad-215270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system plays a critical role in neurodegenerative processes involved in Alzheimer’s disease (AD). In this study, a gene-based immunotherapeutic method examined the effects of anti-inflammatory cellular immune response elements (CIREs) in the amyloid-β protein precursor (AβPP) mouse model. Bi-monthly intramuscular administration, beginning at either 4 or 6 months, and examined at 7.5 through 16 months, with plasmids encoding Interleukin (IL)-10, IL-4, TGF-β polynucleotides, or a combination thereof, into AβPP mice improved spatial memory performance. This work demonstrates an efficient gene therapy strategy to downregulate neuroinflammation, and possibly prevent or delay cognitive decline in AD.
Collapse
Affiliation(s)
- Tai June Yoo
- Korea Allergy Clinic, KangNam Gu, Seoul, South Korea.,University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
95
|
Wang Y, Wu Z, Wang D, Huang C, Xu J, Liu C, Yang C. Muscle-brain communication in pain: The key role of myokines. Brain Res Bull 2021; 179:25-35. [PMID: 34871710 DOI: 10.1016/j.brainresbull.2021.11.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/24/2022]
Abstract
Pain is the most common reason for a physician visit, which accounts for a considerable proportion of the global burden of disease and greatly affects patients' quality of life. Therefore, there is an urgent need to identify new therapeutic targets involved in pain. Exercise-induced hypoalgesia (EIH) is a well known phenomenon observed worldwide. However, the available evidence demonstrates that the mechanisms of EIH remain unclear. One of the most accepted hypotheses has been the activation of several endogenous systems in the brain. Recently, the concept that the muscle acts as a secretory organ has attracted increasing attention. Proteins secreted by the muscle are called myokines, playing a critical role in communicating with other organs, such as the brain. This review will focus on several myokines and discuss their roles in EIH.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chaoli Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Jiali Xu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
96
|
Doroszkiewicz J, Mroczko P, Kulczyńska-Przybik A. Inflammation in the CNS - understanding various aspects of the pathogenesis of Alzheimer's disease. Curr Alzheimer Res 2021; 19:16-31. [PMID: 34856902 PMCID: PMC9127729 DOI: 10.2174/1567205018666211202143935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease is a progressive and deadly neurodegenerative disorder, and one of the most common causes of dementia in the world. Current, insufficiently sensitive and specific methods of early diagnosis and monitoring of this disease prompt a search for new tools. Numerous literature data indicate that the pathogenesis of Alzheimer's disease (AD) is not limited to the neuronal compartment, but involves various immunological mechanisms. Neuroinflammation has been recognized as a very important process in AD pathology. It seems to play pleiotropic roles, both neuroprotective as well as neurodegenerative, in the development of cognitive impairment depending on the stage of the disease. Mounting evidence demonstrates that inflammatory proteins could be considered biomarkers of disease progression. Therefore, the present review summarizes the role of some inflammatory molecules and their potential utility in the detection and monitoring of dementia severity. The paper also provides a valuable insight into new mechanisms leading to the development of dementia, which might be useful in discovering possible anti-inflammatory treatment.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok. Poland
| | - Piotr Mroczko
- Department of Criminal Law and Criminology, Faculty of Law, University of Bialystok, Bialystok. Poland
| | | |
Collapse
|
97
|
Linden T, Hanses F, Domingo-Fernández D, DeLong LN, Kodamullil AT, Schneider J, Vehreschild MJGT, Lanznaster J, Ruethrich MM, Borgmann S, Hower M, Wille K, Feldt T, Rieg S, Hertenstein B, Wyen C, Roemmele C, Vehreschild JJ, Jakob CEM, Stecher M, Kuzikov M, Zaliani A, Fröhlich H. Machine Learning Based Prediction of COVID-19 Mortality Suggests Repositioning of Anticancer Drug for Treating Severe Cases. ARTIFICIAL INTELLIGENCE IN THE LIFE SCIENCES 2021; 1:100020. [PMID: 34988543 PMCID: PMC8677630 DOI: 10.1016/j.ailsci.2021.100020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 02/08/2023]
Abstract
Despite available vaccinations COVID-19 case numbers around the world are still growing, and effective medications against severe cases are lacking. In this work, we developed a machine learning model which predicts mortality for COVID-19 patients using data from the multi-center 'Lean European Open Survey on SARS-CoV-2-infected patients' (LEOSS) observational study (>100 active sites in Europe, primarily in Germany), resulting into an AUC of almost 80%. We showed that molecular mechanisms related to dementia, one of the relevant predictors in our model, intersect with those associated to COVID-19. Most notably, among these molecules was tyrosine kinase 2 (TYK2), a protein that has been patented as drug target in Alzheimer's Disease but also genetically associated with severe COVID-19 outcomes. We experimentally verified that anti-cancer drugs Sorafenib and Regorafenib showed a clear anti-cytopathic effect in Caco2 and VERO-E6 cells and can thus be regarded as potential treatments against COVID-19. Altogether, our work demonstrates that interpretation of machine learning based risk models can point towards drug targets and new treatment options, which are strongly needed for COVID-19.
Collapse
Affiliation(s)
- Thomas Linden
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- University of Bonn, Bonn-Aachen International Center for IT, Friedrich Hirzebruch-Allee 6, 53115 Bonn, Germany
| | - Frank Hanses
- Emergency Department, University Hospital Regensburg, 93053 Regensburg, Germany
- Department for Infectious Diseases and Infection Control, University Hospital Regensburg, Germany
| | - Daniel Domingo-Fernández
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
| | - Lauren Nicole DeLong
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- University of Bonn, Bonn-Aachen International Center for IT, Friedrich Hirzebruch-Allee 6, 53115 Bonn, Germany
| | - Alpha Tom Kodamullil
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
| | - Jochen Schneider
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, 81675 Munich, Germany
| | - Maria J G T Vehreschild
- Department II of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Julia Lanznaster
- Department of Internal Medicine II, Hospital Passau, Innstraße 76, 94032 Passau, Germany
| | - Maria Madeleine Ruethrich
- Institute for Infection Medicine and Hospital Hygiene, University Hospital Jena, 07743 Jena, Germany
| | - Stefan Borgmann
- Department of Infectious Diseases and Infection Control, Hospital Ingolstadt, 85049 Ingolstadt, Germany
| | - Martin Hower
- Department of Pneumology, Infectious Diseases and Intensive Care, Klinikum Dortmund gGmbH, Hospital of University Witten / Herdecke, 44137 Dortmund, Germany
| | - Kai Wille
- University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Centre Minden, 32429 Minden, Germany
| | - Torsten Feldt
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Siegbert Rieg
- Department of Medicine II, University Hospital Freiburg, 79110 Freiburg, Germany
| | - Bernd Hertenstein
- Department of Medicine II, University Hospital Freiburg, 79110 Freiburg, Germany
| | - Christoph Wyen
- Christoph Wyen, Praxis am Ebertplatz Cologne, 50668 Cologne, Germany
| | - Christoph Roemmele
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Jörg Janne Vehreschild
- Department II of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Carolin E M Jakob
- Department I for Internal Medicine, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Melanie Stecher
- Fraunhofer Institute for Translational Medicine and Pharmacologie (ITMP), VolksparkLabs, Schnackenburgallee 114, 22535 Hamburg, Germany
| | - Maria Kuzikov
- Department for Infectious Diseases and Infection Control, University Hospital Regensburg, Germany
| | - Andrea Zaliani
- Department for Infectious Diseases and Infection Control, University Hospital Regensburg, Germany
| | - Holger Fröhlich
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- University of Bonn, Bonn-Aachen International Center for IT, Friedrich Hirzebruch-Allee 6, 53115 Bonn, Germany
| |
Collapse
|
98
|
Ahmadi F, Salmasi Z, Mojarad M, Eslahi A, Tayarani-Najaran Z. G-CSF augments the neuroprotective effect of conditioned medium of dental pulp stem cells against hypoxic neural injury in SH-SY5Y cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1743-1752. [PMID: 35432810 PMCID: PMC8976909 DOI: 10.22038/ijbms.2021.60217.13344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/24/2021] [Indexed: 11/25/2022]
Abstract
Objective(s): Dental pulp stem cells (DPSCs) can differentiate into functional neurons and have the potential for cell therapy in neurological diseases. Granulocyte colony-stimulating factor (G-CSF) is a glycoprotein family shown neuroprotective effect in models of nerve damage. we evaluated the protective effects of G-CSF, conditioned media from DPSCs (DPSCs-CM) and conditioned media from transfected DPSCs with plasmid encoding G-CSF (DPSC-CMT) on SH-SY5Y exposed to CoCl2 as a model of hypoxia-induced neural damage. Materials and Methods: SH-SY5Y exposed to CoCl2 were treated with DPSCs-CM, G-CSF, simultaneous combination of DPSCs-CM and G-CSF and finally DPSC-CMT. Cell viability and apoptosis were determined by resazurin (or lactate dehydrogenase (LDH) assay alternatively) and propidium iodide (PI) staining. Western blot analysis was performed to detect changes in apoptotic protein levels. The interleukin-6 and interleukin-10 IL6/IL10 levels were measured with Enzyme-Linked Immunosorbent Assay (ELISA). Results: DPSCs-CM and G-CSF were able to significantly protect SH-SY5Y against neural cell damage caused by CoCl2 according to resazurin and LDH analysis. Also, the percentage of apoptotic cells decreased when SH-SY5Y were treated with DPSCs-CM and G-CSF simultaneously. After transfection of DPSCs with G-CSF plasmid, DPSC-CMT could significantly improve the protection. The amount of β-catenin, cleaved PARP and caspase-3 were significantly decreased and the expression of survivin was considerably increased when hypoxic SH-SY5Y treated with DPSCs-CM plus G-CSF according to Western blot. Decreased level of IL-6/IL-10, which exposed to CoCl2, after treatment with DPSCs-CM indicated the suppression of inflammatory mediators. Conclusion: Combination therapy of G-CSF and DPSCs-CM improved the protective activity.
Collapse
Affiliation(s)
- Farahnaz Ahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarad
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atieh Eslahi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran,Corresponding author: Zahra Tayarani-Najaran. Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Mashhad, Iran. Tel: +98-51-31801178;
| |
Collapse
|
99
|
Methyl acetate arrests Th1 in peripheral immune system and alleviates CNS inflammation in EAE. Int Immunopharmacol 2021; 101:108291. [PMID: 34799286 DOI: 10.1016/j.intimp.2021.108291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/04/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory autoimmune disease of the central nervous system (CNS) mediated by immune cells. The pathogenesis of most autoimmune diseases has some degree of similarity to that of MS, and therefore the study of MS has clinical and scientific significance for other autoimmune diseases as well. As a widely used organic solvent, Methyl Acetate (MA) has a similar structure to acetate which has been shown to be therapeutic in the mouse model of multiple sclerosis. Here we found that MA was effective in reducing the disease severity of Experimental Autoimmune Encephalomyelitis (EAE). Pathological sections showed that MA reduced inflammatory cell infiltration in the CNS and attenuated demyelination in the spinal cord. MA increases the proportion of Th1 cells in the periphery of EAE mice. Further mechanistic studies have demonstrated that MA treatment induces Th1 retention in the peripheral immune system by increasing the expression levels of peripheral Th1-related chemokines CXCR3. CXCL9, CXCL10. In addition, we observed that MA alleviated intestinal inflammation in EAE mice. The data showed that this phenomenon is achieved by enhancing IL-10 and inhibiting IL-6 secretion. Our data indicates that MA might have therapeutic implications for autoimmune diseases such as MS.
Collapse
|
100
|
Liu H, Zhou YC, Song W. Involvement of IL-10R/STAT3 pathway in amyloid β clearance by microlgia in Alzheimer's disease. Int Immunopharmacol 2021; 101:108263. [PMID: 34710847 DOI: 10.1016/j.intimp.2021.108263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 10/20/2022]
Abstract
Both the total amount and annual growth rate of Alzheimer's disease (AD) patients in China are much higher than in other regions in the world. This trend of rapid growth will be difficult to change in the next few decades, hence the prevention and treatment situation of AD patients in China is more severe. Maintaining the balance between the production and removal pathways of Aβ is an important guarantee for the body to maintain its normal physiological state. The dysfunction of Aβ clearance is an important factor of Aβ accumulation in brain tissue of AD patients causing neurotoxicity of synaptic damage and neuronal death. Based on the literature review, it introduced the important role of microglias in clearing Aβ deposits in the process of Alzheimer's disease. And most of these phagocytic cells were the specific phenotype of disease-related microglia (DAM-I/DAM-II) that induced microglial differentiation after activation. IL-10KO promoted the transformation of microglial phenotype DAM-II, and enhanced its phagocytosis for Aβ oligomers. There is a hypothesis that IL-10R/STAT3 negatively regulates microglial phagocytosis. It was learnt that blocking the IL-10R/STAT3 pathway promoted microglial activation and enhanced phagocytosis. The comprehensive review on the involvement of IL-10R/STAT3 pathway in the process of AD would open up new ideas and discover new targets for the development of new therapeutic drugs.
Collapse
Affiliation(s)
- Hao Liu
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo 315211, China.
| | - Yu-Cong Zhou
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Wei Song
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|