1
|
Orciani C, Foret MK, Cuello AC, Do Carmo S. Long-term nucleus basalis cholinergic lesions alter the structure of cortical vasculature, astrocytic density and microglial activity in Wistar rats. Neurobiol Aging 2025; 150:132-145. [PMID: 40121723 DOI: 10.1016/j.neurobiolaging.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Basal forebrain cholinergic neurons (BFCNs) are the sole source of cholinergic innervation to the cerebral cortex and hippocampus in humans and the primary source in rodents. This system undergoes early degeneration in Alzheimer's disease. BFCNs terminal synapses are involved in the regulation of the cerebral blood flow by making classical synaptic contacts with other neurons. Additionally, they are located in proximity to cortical cerebral blood vessels, forming connections with various cell types of the neurovascular unit (NVU), including vascular smooth muscle cells, endothelial cells, and astrocytic end-feet. However, the effects of the BFCNs input on NVU components remain unresolved. To address this issue, we immunolesioned the nucleus basalis by administering bilateral stereotaxic injections of the cholinergic immunotoxin 192-IgG-Saporin in 2.5-month-old Wistar rats. Seven months post-lesion, we observed a significant reduction in cortical vesicular acetylcholine transporter-immunoreactive synapses. This was accompanied by changes in the diameter of cortical capillaries and precapillary arterioles, as well as lower levels of vascular endothelial growth factor A (VEGF-A). Additionally, the cholinergic immunolesion increased the density of cortical astrocytes and microglia in the cortex. At these post-BFCN-lesion stages, astrocytic end-feet exhibited an increased co-localization with arterioles. The number of microglia in the parietal cortex correlated with cholinergic loss and exhibited morphological changes indicative of an intermediate activation state. This was supported by decreased levels of proinflammatory mediators IFN-γ, IL-1β, and KC/GRO (CXCL1), and by increased expression of M2 markers SOCS3, IL4Rα, YM1, ARG1, and Fizz1. Our findings offer a novel insight: that the loss of nucleus basalis cholinergic input negatively impacts cortical blood vessels, NVU components, and microglia phenotype.
Collapse
Affiliation(s)
- Chiara Orciani
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Morgan K Foret
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
| | - A Claudio Cuello
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada; Department of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada; Department of Anatomy & Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada; Department of Pharmacology, Oxford University, Oxford, UK.
| | - Sonia Do Carmo
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
2
|
Bartra C, Vuraić K, Yuan Y, Codony S, Valdés-Quiroz H, Casal C, Slevin M, Máquez-Kisinousky L, Planas AM, Griñán-Ferré C, Pallàs M, Morisseau C, Hammock BD, Vázquez S, Suñol C, Sanfeliu C. Microglial pro-inflammatory mechanisms induced by monomeric C-reactive protein are counteracted by soluble epoxide hydrolase inhibitors. Int Immunopharmacol 2025; 155:114644. [PMID: 40215773 PMCID: PMC12147942 DOI: 10.1016/j.intimp.2025.114644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Monomeric C-reactive protein (mCRP) is a pro-inflammatory molecule generated by the dissociation of native CRP. Clinical and experimental studies suggest that mCRP deposition in the brain induces Alzheimer's disease (AD) pathology and cognitive loss. Pathological neuroinflammation is increasingly suggested as relevant in AD. Innovative therapies against neuroinflammation are desperately needed, and inhibitors of the enzyme soluble epoxide hydrolase (sEH) are a promising new generation of anti-inflammatory drugs. Mouse primary microglia and BV2 cell line cultures were exposed to mCRP to analyze its pro-inflammatory mechanisms. sEH inhibitors, both newly synthesized UB-SCG-55 and UB-SCG-65, and the reference agent TPPU, were tested for their anti-inflammatory action against mCRP. Phenotypic changes were analyzed through cell imaging techniques, as well as molecular analysis of inflammatory mediators and gene activation pathways. Results show that mCRP triggers a pro-inflammatory response through three main inflammatory pathways: iNOS, NLRP3, and COX-2, followed by increased cytokine generation. Polarization of microglia toward a M1-like phenotype was confirmed by morphological analysis. Also, mCRP can bind to and cross the cell membrane, providing further insight into its mechanisms of action. sEH inhibitors were effective against mCRP induction of a reactive microglial phenotype. The first-line compound UB-SCG-55 emerged as the most potent anti-inflammatory against mCRP injury. Therefore, the direct activation of microglia by mCRP provides evidence of its role in triggering and exacerbating neurodegenerative diseases with a neuroinflammatory component, such as AD. Furthermore, the protection given by inhibitors of sEH confirms its potential as innovative drugs against deleterious effects of neuroinflammation.
Collapse
Affiliation(s)
- Clara Bartra
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; PhD Program in Biotechnology, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, 08034 Barcelona, Spain.
| | - Kristijan Vuraić
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Yi Yuan
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Sandra Codony
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Haydeé Valdés-Quiroz
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Carme Casal
- Microscopy Service, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
| | - Mark Slevin
- CCAMF, George Emil Palade Universitatea de Medicina, Farmacie, Stiinte se Technologie, "George Emil Palade" din Targu-Mures, 540142, Tirgu Mures, Romania
| | - Leonardo Máquez-Kisinousky
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Anna M Planas
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Christophe Morisseau
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, United States
| | - Santiago Vázquez
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Cristina Suñol
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
| | - Coral Sanfeliu
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain.
| |
Collapse
|
3
|
Silla A, Punzo A, Caliceti C, Barbalace MC, Hrelia S, Malaguti M. The Role of Antioxidant Compounds from Citrus Waste in Modulating Neuroinflammation: A Sustainable Solution. Antioxidants (Basel) 2025; 14:581. [PMID: 40427463 PMCID: PMC12108332 DOI: 10.3390/antiox14050581] [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: 04/16/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
In normal conditions, neuroinflammation induces microglia and astrocyte activation to maintain brain homeostasis. However, excessive or prolonged neuroinflammation can inflict harmful damage on brain tissue. Numerous factors can trigger chronic neuroinflammation, ultimately leading to neurodegeneration. In this context, considering the pressing need for novel, natural approaches to mitigate neuroinflammatory damage, attention has turned to unconventional sources such as agricultural by-products. Citrus fruits are widely consumed globally, producing substantial waste, including peels, seeds, and pulp. Traditionally regarded as agricultural waste, these by-products are now recognized as valuable reservoirs of bioactive compounds, including flavonoids, carotenoids, terpenoids, and limonoids. Among these, citrus polyphenols-particularly flavanones like hesperidin, naringenin, and eriocitrin-have emerged as potent modulators of neuroinflammatory pathways through their multifaceted interactions with cellular antioxidant systems, pro-inflammatory signaling cascades, neurovascular integrity, and gut-brain axis dynamics. This review aims to characterize the key molecules present in citrus waste and synthesizes preclinical and clinical evidence to elucidate the biochemical mechanisms underlying neuroinflammation in neurodegenerative disorders.
Collapse
Affiliation(s)
- Alessia Silla
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (A.S.); (A.P.); (C.C.)
| | - Angela Punzo
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (A.S.); (A.P.); (C.C.)
| | - Cristiana Caliceti
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (A.S.); (A.P.); (C.C.)
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.C.B.); (M.M.)
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.C.B.); (M.M.)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.C.B.); (M.M.)
| |
Collapse
|
4
|
Jagadeesan SK, Sandarage RV, Mathiyalagan S, Tsai EC. Personalized Stem Cell-Based Regeneration in Spinal Cord Injury Care. Int J Mol Sci 2025; 26:3874. [PMID: 40332538 PMCID: PMC12028285 DOI: 10.3390/ijms26083874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
Spinal cord injury (SCI) remains a major clinical challenge, with limited therapeutic options for restoring lost neurological function. While efforts to mitigate secondary damage have improved early-phase management, achieving sustained neurorepair and functional recovery remains elusive. Advances in stem cell engineering and regenerative medicine have opened new avenues for targeted interventions, particularly through the transplantation of neural stem/progenitor cells (NSPCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs). However, patient-specific factors such as cellular senescence, genetic and epigenetic variability, injury microenvironment, and comorbidities influence the efficacy of stem cell therapies by affecting graft survival and differentiation. Overcoming these challenges necessitates cutting-edge technologies, including single-cell transcriptomics, CRISPR-mediated hypoimmunogenic engineering, and biomaterial-based delivery platforms, which enable personalized and precision-driven SCI repair. Leveraging these advancements may help stem cell therapies overcome translational barriers and establish clinically viable regenerative solutions. This review explores the intersection of patient-specific variability, bioengineering innovations, and transcriptomic-guided precision medicine to define the next frontier in SCI therapy.
Collapse
Affiliation(s)
- Sasi Kumar Jagadeesan
- Department of Neurosciences, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (S.K.J.); (R.V.S.)
- Neuroscience Program, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Ryan Vimukthie Sandarage
- Department of Neurosciences, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (S.K.J.); (R.V.S.)
- Division of Neurosurgery, Department of Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Sathya Mathiyalagan
- Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Eve Chung Tsai
- Department of Neurosciences, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (S.K.J.); (R.V.S.)
- Neuroscience Program, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
- Division of Neurosurgery, Department of Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
5
|
Song W, Giannotti A, Bekiaridou A, Bloom O, Zanos S. Low intensity trans-spinal focused ultrasound reduces mechanical sensitivity and suppresses spinal microglia activation in rats with chronic constriction injury. Bioelectron Med 2025; 11:8. [PMID: 40159475 PMCID: PMC11956222 DOI: 10.1186/s42234-025-00170-z] [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: 11/12/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Low intensity, trans-spinal focused ultrasound (tsFUS) is a noninvasive neuromodulation approach that has been shown to modulate spinal circuit excitability in healthy rats. Here, we evaluated the potential of tsFUS for alleviating neuropathic pain by testing it in a chronic constriction injury (CCI) model. Male rats underwent CCI of the left sciatic nerve and then received tsFUS (2 kHz pulse repetition frequency; 40% duty cycle) or sham stimulation, targeted at spinal segment level L5 for 3 min daily over three days. As expected, CCI causes significant reduction of von Frey Threshold (vFT), a measure of mechanical sensitivity. We found that tsFUS treatment is associated with increased vFT compared to sham; this increase persists beyond the duration of treatment, through days 4 to 23 post-CCI. In spinal cords of tsFUS-treated animals, counts of spinal microglia (Iba1 + cells) and of activated, pro-inflammatory microglia (Iba1 + /CD86 + cells), are reduced compared to sham-treated animals. This reduction in microglia counts is limited to the insonified side of the spinal cord, ipsilateral to CCI. These findings suggest that tsFUS may be a promising approach for treatment of neuropathic pain at early stages, possibly by attenuating the development of microglial-driven inflammation.
Collapse
Affiliation(s)
- Weiguo Song
- Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
| | - Alice Giannotti
- The Biorobotics Institute and Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alexandra Bekiaridou
- Elmezzi Graduate School of Molecular Medicine, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Ona Bloom
- Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Stavros Zanos
- Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
| |
Collapse
|
6
|
Zhuo G, Chen W, Hu Y, Zhang J, Zhu X, Su M, Fu Y, Lin W. Genetic Prediction of the Phosphate-to-Glucose Ratio Mediates the Association Between CXCL5 and Vascular Dementia. Brain Behav 2025; 15:e70378. [PMID: 40135623 PMCID: PMC11938108 DOI: 10.1002/brb3.70378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND AND OBJECTIVES A variety of observational studies suggest a possible connection between C-X-C Motif Chemokine Ligand 5 (CXCL5) and vascular dementia (VaD), though the exact causal relationship is still uncertain. This research aims to investigate the causal connection between CXCL5 and VaD risk through a Mendelian randomization (MR) method and to examine the phosphate-to-glucose ratio as a possible mediator. METHODS Using summary-level data from genome-wide association studies (GWAS), we conducted a two-sample MR analysis to investigate the genetic prediction of CXCL5 and VaD. Horizontal pleiotropy, heterogeneity, and sensitivity analyses were also performed on the MR findings. Additionally, a two-step MR was utilized to quantify the proportion of the effect of CXCL5 on VaD mediated by the phosphate-to-glucose ratio. RESULTS MR analysis identified that higher levels of CXCL5 (IVW: p = 0.022, OR = 1.265, 95% CI = 1.034-1.547) increase the risk of VaD. Tests for horizontal pleiotropy (p > 0.05), heterogeneity (p > 0.05), and sensitivity analyses supported these findings. There is insufficient robust evidence to suggest that genetic predispositions for VaD have any significant impact on CXCL5 (IVW: p = 0.254). The phosphate-to-glucose ratio accounted for 11.1% of increase in the risk of VaD associated with CXCL5 (95% CI = -12.3% to 34.5%). CONCLUSION To conclude, our research confirms a causal link between CXCL5 and VaD and shows that the ratio of phosphate-to-glucose plays a mediating role in a segment of the risk effect of CXCL5 on VaD. However, most of the effects of CXCL5 on VaD are still not well understood. Additional studies are necessary to explore other potential mediators as risk factors. In clinical settings, individuals with abnormally elevated CXCL5 may need to be monitored for an increased risk of developing VaD.
Collapse
Affiliation(s)
- Guifeng Zhuo
- Department of NeurologyThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningChina
- The First Clinical College of MedicineGuangxi University of Chinese MedicineNanningChina
| | - Wei Chen
- Department of NeurologyThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningChina
| | - Yanan Hu
- Ziwei comunity health care service centerThe Second Affiliated HospitalCuhk‐Shenzhen Longgang District People's Hospital of ShenzhenShenzhenChina
| | - Jinzhi Zhang
- Department of NeurologyThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningChina
| | - Xiaomin Zhu
- Department of NeurologyThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningChina
| | - Mingyang Su
- Department of NeurologyThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningChina
| | - Yulan Fu
- Department of NeurologyThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningChina
| | - Wu Lin
- Department of NeurologyThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningChina
- Scientific Laboratorial CentreGuangxi University of Chinese MedicineNanningChina
| |
Collapse
|
7
|
Yang X, Wang J, Jia X, Yang Y, Fang Y, Ying X, Li H, Zhang M, Wei J, Pan Y. Microglial polarization in Alzheimer's disease: Mechanisms, implications, and therapeutic opportunities. J Alzheimers Dis 2025; 104:3-13. [PMID: 39894910 DOI: 10.1177/13872877241313223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-β plaques, neurofibrillary tangles, and chronic neuroinflammation. Microglial cells, the resident immune cells in the central nervous system, play a crucial role in the pathogenesis of AD. Microglia can undergo polarization, shifting between pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes in response to different stimuli. Dysregulation of microglial polarization towards the pro-inflammatory phenotype leads to the release of inflammatory cytokines, oxidative stress, and synaptic dysfunction. These processes contribute to neuronal damage and cognitive decline in AD. However, several challenges remain in this field. The complex molecular mechanisms governing microglial polarization in AD need to be further elucidated. In this review, we discuss the mechanisms underlying microglial polarization in AD and its implications in disease progression.
Collapse
Affiliation(s)
- Xinmao Yang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jie Wang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaotao Jia
- Department of Neurology, The Affifiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, PR China
| | - Yaqian Yang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yan Fang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaoping Ying
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hong Li
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Meiqian Zhang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Wei
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yanfang Pan
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
8
|
Sentyabreva AV, Diatroptova MA, Miroshnichenko EA, Gerasimov AD, Kosyreva AM. Biomolecular and Functional Changes in a Culture of Microglial Cells Caused by Long-Term Exposure to AlCl 3. Bull Exp Biol Med 2025; 178:654-660. [PMID: 40295436 DOI: 10.1007/s10517-025-06392-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Indexed: 04/30/2025]
Abstract
Inflammaging is one of the main risk factors for the development and progression of age-related diseases. The increase in the proinflammatory background and ROS production can lead to persistent activation and dysfunction of microglial cells. The biomolecular and functional changes in microglial cells after long-term exposure to prooxidant aluminum chloride were studied in in vitro experiment. BV2 cells were cultured in the presence of 0.5 and 1 mM AlCl3 for 70 or 140 h. The intensity of ROS production, apoptosis, and M1/M2 phenotype polarization of microglia were assessed by flow cytometry, qPCR-RT, and ELISA. Cells cultured with 0.5 mM AlCl3 showed signs of "healthy aging", while the higher concentration (1 mM) of AlCl3 led to persistent activation of microglia. The data obtained can be used for in vitro modeling of inflammaging and related physiological and pathological processes.
Collapse
Affiliation(s)
- A V Sentyabreva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia.
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia.
| | - M A Diatroptova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - E A Miroshnichenko
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - A D Gerasimov
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - A M Kosyreva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
9
|
Li W, Yang T, Zhang Z, Peng A, Wang Q. Exosomes derived from TNF-α preconditioned bone marrow mesenchymal stem cells alleviate cisplatin-induced ototoxicity in mice. Int J Med Sci 2025; 22:1215-1222. [PMID: 40027193 PMCID: PMC11866538 DOI: 10.7150/ijms.104121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/12/2024] [Indexed: 03/05/2025] Open
Abstract
The polarization of microglia promotes the development of cisplatin-induced ototoxicity, and exosomes (Exo) derived from TNF-α preconditioned mesenchymal stem cells (MSCs) may induce the polarization of macrophage. Mice were intraperitoneally injected with cisplatin to establish the ototoxicity model. Bone marrow MSCs (BMSCs) were preconditioned with TNF-α for 48 h, and the relevant TNF-Exo or Exo was enriched, which were further trans-tympanically administered in the left ear of ototoxic mice. Auditory sensitivity was revealed with auditory brainstem response (ABR) at 8, 16, 24, and 32 kHz. The number of hair cells was detected with Myosin 7a staining. Damaged auditory sensitivity and up-regulated hair cell loss were revealed in cisplatin-exposed mice, which could be reversed by Exo or TNF-Exo treatment. Mechanically, up-regulated Iba1, Cd86, iNOS, Cd206, and Arg1 were detected in cisplatin-exposed cochlea. TNF-Exo or Exo administration further decreased Iba1, Cd86, and iNOS expression, and increased cd206 and Arg1 expression. TNF-Exo or Exo administration inhibited the productin of pro-inflammatory cytokines (IL-1β and IL-6), while enhanced the anti-inflammatory cytokine IL-10 production in the cisplatin-exposed cochlea. Importantly, TNF-Exo administration showed more profound benefits compared with Exo. TNF-α preconditioning might be a new therapeutic option to enhance the capability of BMSCs-derived exosomes against cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
| | | | | | | | - Qin Wang
- Department of Otolaryngology and Head & Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
10
|
Bhardwaj V, Kumari S, Dhapola R, Sharma P, Beura SK, Singh SK, Vellingiri B, HariKrishnaReddy D. Shedding light on microglial dysregulation in Alzheimer's disease: exploring molecular mechanisms and therapeutic avenues. Inflammopharmacology 2025; 33:679-702. [PMID: 39609333 DOI: 10.1007/s10787-024-01598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/26/2024] [Indexed: 11/30/2024]
Abstract
Alzheimer's disease (AD) stands out as the foremost prevalent neurodegenerative disorder, characterized by a complex etiology. Various mechanisms have been proposed to elucidate its onset, encompassing amyloid-beta (Aβ) toxicity, tau hyperphosphorylation, oxidative stress and reactive gliosis. The hallmark of AD comprises Aβ and tau aggregation. These misfolded protein aggregates trigger the activation of glial cells, primarily microglia. Microglial cells serve as a major source of inflammatory mediators and their cytotoxic activation has been implicated in various aspects of AD pathology. Activated microglia can adopt M1 or M2 phenotypes, where M1 promotes inflammation by increasing pro-inflammatory cytokines and M2 suppresses inflammation by boosting anti-inflammatory factors. Overexpressed pro-inflammatory cytokines include interleukin (IL)-1β, IL-6 and tumor necrosis factor-α (TNF-α) in adjacent brain regions. Furthermore, microglial signaling pathways dysregulated in AD are myeloid differentiation primary-response protein 88 (Myd 88), colony-stimulating factor-1 receptor (CSF1R) and dedicator of cytokinesis 2 (DOCK2), which alter the physiology. Despite numerous findings, the causative role of microglia-mediated neuroinflammation in AD remains elusive. This review concisely explores cellular and molecular mechanisms of activated microglia and their correlation with AD pathogenesis. Additionally, it highlights promising therapeutics targeting microglia modulation, currently undergoing preclinical and clinical studies, for developing effective treatment for AD.
Collapse
Affiliation(s)
- Vanshu Bhardwaj
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Sunil Kumar Singh
- Department of Bio-Chemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
11
|
Xie Y, Qin Y, Wang J, Xu Z, Chen L, Kuang Y, Yang R, Huang L. Tinosinenside A inhibits neuroinflammation and protects HT22 cells by suppressing the TLR4/NF-κB/NLRP3 signaling pathway in BV2 cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03828-2. [PMID: 39878812 DOI: 10.1007/s00210-025-03828-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
Microglia-mediated neuroinflammation plays a crucial role in Alzheimer's disease (AD). Tinosinenside A (Tis A) is a novel sesquiterpene glycoside isolated from the dried rattan stem of Tinospora sinensis (Lour.) Merr. Tis A exhibited anti-inflammatory and neuroprotective activities in vitro. However, the mechanism underlying the inhibition of neuroinflammation and protection of nerve cells remains obscure. This study used lipopolysaccharide (LPS)-induced inflammatory response in BV2 cells to simulate a neuroinflammatory model and used Aβ1-42-induced HT22 cells to establish an AD cell model, aiming to investigate the efficacy and mechanism of Tis A through anti-neuroinflammation to protect nerve cells. Tis A had no effect on the proliferation of BV2 and HT22 cells at the tested concentrations. The time- and dose-dependent effects of Tis A on the LPS-induced inflammatory response of BV2 cells demonstrated that the best anti-inflammatory efficacy appeared after 12 h of pretreatment. Tis A inhibited the gene levels of TNF-α, IL-6, IL-1β, iNOS, and IL-10 while enhancing the gene levels of IL-4 and TGF-β. Additionally, Tis A reduced the gene expression levels of CD16 and CD32 and increased the CD36 and CD206 gene expression levels. It also downregulated the protein expression of Iba-1 and iNOS while upregulating CD206. Tis A obviously inhibited NLRP3 gene and protein expression in LPS-stimulated BV2 cells. The inhibitory effect of Tis A on NLRP3 was counteracted by the NLRP3 activator nigericin and overexpression plasmid GV358. Tis A inhibits NLRP3 protein expression to reduce the assembly of NLRP3/ASC/Caspase-1 inflammasome, then regulates the TLR4/NF-κB/NLRP3 signaling pathway. It regulates microglia activation and M1/M2 phenotypic polarization, then inhibits the production of inflammatory factors, and reduces the apoptosis rate of HT22 cells under inflammatory conditions, improving the survival rate of nerve cells to protect neurons.
Collapse
Affiliation(s)
- Yongyan Xie
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yinfang Qin
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jingjing Wang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Ziwei Xu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Liping Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yunxia Kuang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Ruwei Yang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Liping Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China.
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Nanchang, 330004, China.
| |
Collapse
|
12
|
Mohammad ZB, Yudin SCY, Goldberg BJ, Serra KL, Klegeris A. Exploring neuroglial signaling: diversity of molecules implicated in microglia-to-astrocyte neuroimmune communication. Rev Neurosci 2025; 36:91-117. [PMID: 39240134 PMCID: PMC11717358 DOI: 10.1515/revneuro-2024-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024]
Abstract
Effective communication between different cell types is essential for brain health, and dysregulation of this process leads to neuropathologies. Brain glial cells, including microglia and astrocytes, orchestrate immune defense and neuroimmune responses under pathological conditions during which interglial communication is indispensable. Our appreciation of the complexity of these processes is rapidly increasing due to recent advances in molecular biology techniques, which have identified numerous phenotypic states of both microglia and astrocytes. This review focuses on microglia-to-astrocyte communication facilitated by secreted neuroimmune modulators. The combinations of interleukin (IL)-1α, tumor necrosis factor (TNF), plus complement component C1q as well as IL-1β plus TNF are already well-established microglia-derived stimuli that induce reactive phenotypes in astrocytes. However, given the large number of inflammatory mediators secreted by microglia and the rapidly increasing number of distinct functional states recognized in astrocytes, it can be hypothesized that many more intercellular signaling molecules exist. This review identifies the following group of cytokines and gliotransmitters that, while not established as interglial mediators yet, are known to be released by microglia and elicit functional responses in astrocytes: IL-10, IL-12, IL-18, transforming growth factor (TGF)-β, interferon (IFN)-γ, C-C motif chemokine ligand (CCL)5, adenosine triphosphate (ATP), l-glutamate, and prostaglandin E2 (PGE2). The review of molecular mechanisms engaged by these mediators reveals complex, partially overlapping signaling pathways implicated in numerous neuropathologies. Additionally, lack of human-specific studies is identified as a significant knowledge gap. Further research on microglia-to-astrocyte communication is warranted, as it could discover novel interglial signaling-targeted therapies for diverse neurological disorders.
Collapse
Affiliation(s)
- Zainab B. Mohammad
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Samantha C. Y. Yudin
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Benjamin J. Goldberg
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Kursti L. Serra
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Andis Klegeris
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| |
Collapse
|
13
|
Barbalho SM, Leme Boaro B, da Silva Camarinha Oliveira J, Patočka J, Barbalho Lamas C, Tanaka M, Laurindo LF. Molecular Mechanisms Underlying Neuroinflammation Intervention with Medicinal Plants: A Critical and Narrative Review of the Current Literature. Pharmaceuticals (Basel) 2025; 18:133. [PMID: 39861194 PMCID: PMC11768729 DOI: 10.3390/ph18010133] [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: 12/18/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Neuroinflammation is a key factor in the progression of neurodegenerative diseases, driven by the dysregulation of molecular pathways and activation of the brain's immune system, resulting in the release of pro-inflammatory and oxidative molecules. This chronic inflammation is exacerbated by peripheral leukocyte infiltration into the central nervous system. Medicinal plants, with their historical use in traditional medicine, have emerged as promising candidates to mitigate neuroinflammation and offer a sustainable alternative for addressing neurodegenerative conditions in a green healthcare framework. This review evaluates the effects of medicinal plants on neuroinflammation, emphasizing their mechanisms of action, effective dosages, and clinical implications, based on a systematic search of databases such as PubMed, SCOPUS, and Web of Science. The key findings highlight that plants like Cleistocalyx nervosum var. paniala, Curcuma longa, Cannabis sativa, and Dioscorea nipponica reduce pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), inhibit enzymes (COX-2 and iNOS), and activate antioxidant pathways, particularly Nrf2. NF-κB emerged as the primary pro-inflammatory pathway inhibited across studies. While the anti-inflammatory potential of these plants is significant, the variability in dosages and phytochemical compositions limits clinical translation. Here, we highlight that medicinal plants are effective modulators of neuroinflammation, underscoring their therapeutic potential. Future research should focus on animal models, standardized protocols, and safety assessments, integrating advanced methodologies, such as genetic studies and nanotechnology, to enhance their applicability in neurodegenerative disease management.
Collapse
Affiliation(s)
- Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; (S.M.B.); (L.F.L.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Beatriz Leme Boaro
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil
| | - Jéssica da Silva Camarinha Oliveira
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil
| | - Jiří Patočka
- Faculty of Health and Social Studies, Institute of Radiology, Toxicology and Civil Protection, University of South Bohemia Ceske Budejovice, 37005 Ceske Budejovice, Czech Republic
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic
| | - Caroline Barbalho Lamas
- Department of Gerontology, School of Gerontology, Universidade Federal de São Carlos (UFSCar), São Carlos 13565-905, São Paulo, Brazil
| | - Masaru Tanaka
- Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos Krt. 113, H-6725 Szeged, Hungary
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil; (S.M.B.); (L.F.L.)
| |
Collapse
|
14
|
Wang L, Gui J, Ding R, Song H, Tian B, Wang W, Liu J, Jiang L. Identification and verification of key molecules in the epileptogenic process of focal cortical dysplasia. Metab Brain Dis 2024; 40:47. [PMID: 39612062 DOI: 10.1007/s11011-024-01426-4] [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: 07/03/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024]
Abstract
Focal cortical dysplasia (FCD) represents a common developmental malformation associated with drug-resistant epilepsy (DRE) among children. However, the exact molecular mechanisms behind this condition are still unclear. In our study, FCD-associated microarray data from the Gene Expression Omnibus (GEO) database were analyzed. A comprehensive series of bioinformatics analyses were conducted, including screening for differentially expressed genes (DEGs), functional enrichment analysis, weighted gene co-expression network analysis (WGCNA), and protein-protein interaction (PPI) analysis. Subsequently, a freezing lesion (FL) rat model was developed to validate expression levels of hub genes along with the molecular pathways behind FCD epileptogenicity. 320 DEGs were identified, and functional enrichment analysis revealed significant enrichment of these DEGs in "Neuroinflammatory response", "Cytokine production involved in immune response", and "Macrophage activation". Ultimately, 5 potential hub genes (CYBB, ITGAM, FCG3A, LY86, and CD86) were pinpointed. Notably, 4 hub genes (CYBB, ITGAM, FCG3A, and CD86) were validated in in vivo experiments, suggesting possible associations with neuroinflammation triggered by microglia. This underscores the tight relationship between microglia-induced neuroinflammation and the pathological progression of epileptic seizures in FCD. ITGAM, FCG3A, CD86, CYBB, and LY86 may emerge as promising candidate biomarkers, influencing diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Lingman Wang
- Department of Neurology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jianxiong Gui
- Department of Neurology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Ran Ding
- Department of Neurology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Honghong Song
- Department of Neurology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Bing Tian
- Department of Neurology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Wandi Wang
- Department of Neurology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jie Liu
- Department of Neurology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Li Jiang
- Department of Neurology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, 40014, China.
| |
Collapse
|
15
|
Bhatt M, Sharma M, Das B. The Role of Inflammatory Cascade and Reactive Astrogliosis in Glial Scar Formation Post-spinal Cord Injury. Cell Mol Neurobiol 2024; 44:78. [PMID: 39579235 PMCID: PMC11585509 DOI: 10.1007/s10571-024-01519-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
Reactive astrogliosis and inflammation are pathologic hallmarks of spinal cord injury. After injury, dysfunction of glial cells (astrocytes) results in glial scar formation, which limits neuronal regeneration. The blood-spinal cord barrier maintains the structural and functional integrity of the spinal cord and does not allow blood vessel components to leak into the spinal cord microenvironment. After the injury, disruption in the spinal cord barrier causes an imbalance of the immunological microenvironment. This triggers the process of neuroinflammation, facilitated by the actions of microglia, neutrophils, glial cells, and cytokines production. Recent work has revealed two phenotypes of astrocytes, A1 and A2, where A2 has a protective type, and A1 releases neurotoxins, further promoting glial scar formation. Here, we first describe the current understanding of the spinal cord microenvironment, both pre-, and post-injury, and the role of different glial cells in the context of spinal cord injury, which forms the essential update on the cellular and molecular events following injury. We aim to explore in-depth signaling pathways and molecular mediators that trigger astrocyte activation and glial scar formation. This review will discuss the activated signaling pathways in astrocytes and other glial cells and their collaborative role in the development of gliosis through inflammatory responses.
Collapse
Affiliation(s)
- Manini Bhatt
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India
| | - Muskan Sharma
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India.
| |
Collapse
|
16
|
Carnicelli V, De Dominicis N, Scipioni L, Fava M, Fanti F, Cinque B, Leuti A, Angelucci CB, Lizzi AR, Giacominelli-Stuffler R, Flati V, Sergi M, Compagnone D, Sardanelli AM, Tisi A, Oddi S, Maccarrone M. Protective effects of fatty acid amide hydrolase inhibition in UVB-activated microglia. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159524. [PMID: 38857757 DOI: 10.1016/j.bbalip.2024.159524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Neuroinflammation is a hallmark of several neurodegenerative disorders that has been extensively studied in recent years. Microglia, the primary immune cells of the central nervous system (CNS), are key players in this physiological process, demonstrating a remarkable adaptability in responding to various stimuli in the eye and the brain. Within the complex network of neuroinflammatory signals, the fatty acid N-ethanolamines, in particular N-arachidonylethanolamine (anandamide, AEA), emerged as crucial regulators of microglial activity under both physiological and pathological states. In this study, we interrogated for the first time the impact of the signaling of these bioactive lipids on microglial cell responses to a sub-lethal acute UVB radiation, a physical stressor responsible of microglia reactivity in either the retina or the brain. To this end, we developed an in vitro model using mouse microglial BV-2 cells. Upon 24 h of UVB exposure, BV-2 cells showed elevated oxidative stress markers and, cyclooxygenase (COX-2) expression, enhanced phagocytic and chemotactic activities, along with an altered immune profiling. Notably, UVB exposure led to a selective increase in expression and activity of fatty acid amide hydrolase (FAAH), the main enzyme responsible for degradation of fatty acid ethanolamides. Pharmacological FAAH inhibition via URB597 counteracted the effects of UVB exposure, decreasing tumor necrosis factor α (TNF-α) and nitric oxide (NO) release and reverting reactive oxidative species (ROS), interleukin-1β (IL-1β), and interleukin-10 (IL-10) levels to the control levels. Our findings support the potential of enhanced fatty acid amide signaling in mitigating UVB-induced cellular damage, paving the way to further exploration of these lipids in light-induced immune responses.
Collapse
Affiliation(s)
- Veronica Carnicelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Noemi De Dominicis
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy; Department of Physics, University of Trento, 38123 Trento, Italy
| | - Lucia Scipioni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy; European Center for Brain Research/IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Marina Fava
- Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Federico Fanti
- Department of Bioscience and Technology for Agriculture, Food and Environment, Campus Universitario di Coste Sant'Agostino, University of Teramo, Italy
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Alessandro Leuti
- European Center for Brain Research/IRCCS Santa Lucia Foundation, 00143 Rome, Italy; Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | | | - Anna Rita Lizzi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | | | - Vincenzo Flati
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Manuel Sergi
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Dario Compagnone
- Department of Bioscience and Technology for Agriculture, Food and Environment, Campus Universitario di Coste Sant'Agostino, University of Teramo, Italy
| | - Anna Maria Sardanelli
- Department of Translational Biomedicine and Neuroscience 'DiBraiN', University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Sergio Oddi
- European Center for Brain Research/IRCCS Santa Lucia Foundation, 00143 Rome, Italy; Department of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy; European Center for Brain Research/IRCCS Santa Lucia Foundation, 00143 Rome, Italy.
| |
Collapse
|
17
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
18
|
Navabi SP, Badreh F, Khombi Shooshtari M, Hajipour S, Moradi Vastegani S, Khoshnam SE. Microglia-induced neuroinflammation in hippocampal neurogenesis following traumatic brain injury. Heliyon 2024; 10:e35869. [PMID: 39220913 PMCID: PMC11365414 DOI: 10.1016/j.heliyon.2024.e35869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Traumatic brain injury (TBI) is one of the most causes of death and disability among people, leading to a wide range of neurological deficits. The important process of neurogenesis in the hippocampus, which includes the production, maturation and integration of new neurons, is affected by TBI due to microglia activation and the inflammatory response. During brain development, microglia are involved in forming or removing synapses, regulating the number of neurons, and repairing damage. However, in response to injury, activated microglia release a variety of pro-inflammatory cytokines, chemokines and other neurotoxic mediators that exacerbate post-TBI injury. These microglia-related changes can negatively affect hippocampal neurogenesis and disrupt learning and memory processes. To date, the intracellular signaling pathways that trigger microglia activation following TBI, as well as the effects of microglia on hippocampal neurogenesis, are poorly understood. In this review article, we discuss the effects of microglia-induced neuroinflammation on hippocampal neurogenesis following TBI, as well as the intracellular signaling pathways of microglia activation.
Collapse
Affiliation(s)
- Seyedeh Parisa Navabi
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Maryam Khombi Shooshtari
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Hajipour
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sadegh Moradi Vastegani
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
19
|
Lasure VU, Singh Gautam A, Singh RK. Quercetin ameliorates neuroinflammatory and neurodegenerative biomarkers in the brain and improves neurobehavioral parameters in a repeated intranasal amyloid-beta exposed model of Alzheimer's disease. Food Funct 2024; 15:8712-8728. [PMID: 39087409 DOI: 10.1039/d4fo02602k] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Objectives: The aim of the present study was to study the potential therapeutic effects of quercetin in protection against repeated intranasal exposure of an amyloid-beta-induced mouse model. Methods: Mice received intranasal Aβ1-42 (5 μg/10 μL) exposure once daily for seven consecutive days. Quercetin was orally administered to them at 30 mg kg-1 and 100 mg kg-1 doses for one week starting from day five following Aβ1-42 peptide administration. Following this, the animals were evaluated for neurobehavioral parameters using a Morris water maze test and a novel object recognition test. Further to this, the biomarkers for neuroinflammation and neurodegeneration were evaluated in the hippocampus and cortex regions of the brain in these animals. Results: Multiple exposures to intranasal Aβ led to a significant decline in the learning and cognitive memory of the animals, whereas oral treatment with quercetin at dosages of 30 and 100 mg kg-1 alleviated Aβ-induced effects. Quercetin treatment significantly reduced Aβ accumulation, oxidative stress and proinflammatory cytokine biomarkers in the brain. In addition, it also alleviated the activation of astrocytic biomarkers, amyloid precursor protein and phosphorylated-tau proteins in the brain. Conclusion: Quercetin was found to be a potent antioxidant, anti-inflammatory compound with protection against neurodegenerative damage and improved learning and cognitive memory in a repeated Aβ-exposure model of AD.
Collapse
Affiliation(s)
- Vaibhav Uttamrao Lasure
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| |
Collapse
|
20
|
Kaur S, K M, Sharma A, Giridharan VV, Dandekar MP. Brain resident microglia in Alzheimer's disease: foe or friends. Inflammopharmacology 2024:10.1007/s10787-024-01550-8. [PMID: 39167311 DOI: 10.1007/s10787-024-01550-8] [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: 06/27/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024]
Abstract
The neurobiology of Alzheimer's disease (AD) is unclear due to its multifactorial nature. Although a wide range of studies revealed several pathomechanisms of AD, dementia is yet unmanageable with current pharmacotherapies. The recent growing literature illustrates the role of microglia-mediated neuroinflammation in the pathogenesis of AD. Indeed, microglia serve as predominant sentinels of the brain, which diligently monitor the neuroimmune axis by phagocytosis and releasing soluble factors. In the case of AD, microglial cells are involved in synaptic pruning and remodeling by producing inflammatory mediators. The conditional inter-transformation of classical activation (proinflammatory) or alternative activation (anti-inflammatory) microglia is responsible for most brain disorders. In this review, we discussed the role of microglia in neuroinflammatory processes in AD following the accumulation of amyloid-β and tau proteins. We also described the prominent phenotypes of microglia, such as disease-associated microglia (DAM), dark microglia, interferon-responsive microglia (IRMs), human AD microglia (HAMs), and microglial neurodegenerative phenotype (MGnD), which are closely associated with AD incidence. Considering the key role of microglia in AD progression, microglial-based therapeutics may hold promise in mitigating cognitive deficits by addressing the neuroinflammatory responses.
Collapse
Affiliation(s)
- Simranjit Kaur
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, Telangana, India
| | - Malleshwari K
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, Telangana, India
| | - Anamika Sharma
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, Telangana, India
| | - Vijayasree V Giridharan
- Faillace Department of Psychiatry and Behavioural Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Manoj P Dandekar
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
21
|
Hendrix E, Vande Vyver M, Holt M, Smolders I. Regulatory T cells as a possible new target in epilepsy? Epilepsia 2024; 65:2227-2237. [PMID: 38888867 DOI: 10.1111/epi.18038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024]
Abstract
Epilepsy is a complex chronic brain disorder with diverse clinical features that can be caused by various triggering events, such as infections, head trauma, or stroke. During epileptogenesis, various abnormalities are observed, such as altered cellular homeostasis, imbalance of neurotransmitters, tissue changes, and the release of inflammatory mediators, which in combination lead to spontaneous recurrent seizures. Regulatory T cells (Tregs), a subtype of CD4+Foxp3+ T cells, best known for their key function in immune suppression, also seem to play a role in attenuating neurodegeneration and suppressing pathological inflammation in several brain disease states. Considering that epilepsy is also highly associated with neuronal damage and neuroinflammation, modulation of Tregs may be an interesting way to modify the disease course of epilepsy and needs further investigation. In this review, we will describe the currently available information on Tregs in epilepsy.
Collapse
Affiliation(s)
- Evelien Hendrix
- Department of Pharmaceutical Chemistry, Drug Analysis, and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Maxime Vande Vyver
- Department of Pharmaceutical Chemistry, Drug Analysis, and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Neurology and Bru-BRAIN, Universitair Ziekenhuis Brussel, Brussels, Belgium
- NEUR Research Group, Center of Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Matthew Holt
- Instituto de Investigação e Inovação Em Saúde, Porto, Portugal
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis, and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
22
|
Li Y, Dai Y, Chu L. V-ATPase B2 promotes microglial phagocytosis of myelin debris by inactivating the MAPK signaling pathway. Neuropeptides 2024; 106:102436. [PMID: 38733728 DOI: 10.1016/j.npep.2024.102436] [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: 12/22/2023] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Microglial phagocytosis of myelin debris is a crucial process for promoting myelin regeneration in conditions such as multiple sclerosis (MS). Vacuolar-ATPase B2 (V-ATPase B2) has been implicated in various cellular processes, but its role in microglial phagocytosis and its potential impact on MS-related responses remain unclear. In this study, we employed BV-2 murine microglial cells to investigate the influence of V-ATPase B2 on the phagocytosis of myelin debris by microglia. The results revealed that V-ATPase B2 expression increased in response to myelin debris exposure. Overexpression of V-ATPase B2 significantly enhanced BV-2 phagocytosis of myelin debris. Additionally, V-ATPase B2 overexpression shifted microglial polarization towards an anti-inflammatory M2 phenotype, coupled with decreased lysosomal pH and enhanced lysosome degradation capacity. Moreover, endoplasmic reticulum (ER) stress inhibitor, 4-PBA, reversed the effects of V-ATPase B2 silencing on ER stress, M2 polarization, and lysosomal degradation of BV-2 cells. The MAPK pathway was inhibited upon V-ATPase B2 overexpression, contributing to heightened myelin debris clearance by BV-2 cells. Notably, MAPK pathway inhibition partially attenuated the inhibitory effects of V-ATPase B2 knockdown on myelin debris clearance. In conclusion, our findings reveal a pivotal role for V-ATPase B2 in promoting microglial phagocytosis of myelin debris by regulating microglial polarization and lysosomal function via the MAPK signaling pathway, suggesting that targeting V-ATPase B2 may hold therapeutic potential for enhancing myelin debris clearance and modulating microglial responses in MS and related neuroinflammatory disorders.
Collapse
Affiliation(s)
- Yao Li
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Yuhan Dai
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Lan Chu
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
23
|
Duan Z, Yang L, Xu D, Qi Z, Jia W, Wu C. Scutellarin Attenuates Microglia Activation in LPS-Induced BV-2 Microglia via miRNA-7036a/MAPT/PRKCG/ERK Axis. Adv Biol (Weinh) 2024; 8:e2400123. [PMID: 38684459 DOI: 10.1002/adbi.202400123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/08/2024] [Indexed: 05/02/2024]
Abstract
Scutellarin is an herbal agent which can exert anti-neuroinflammatory effects in activated microglia. However, it remains uncertain if it can inhibit microglia-mediated neuroinflammation by regulating miRNAs. This study sought to elucidate the upstream regulatory mechanisms by endogenous microRNAs and its target gene in activated microglia in lipopolysaccharide (LPS)-induced BV-2 microglia. Results show that scutellarin suppressed the expression of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and inducible nitric oxide synthase (iNOS) significantly in LPS-stimulated BV-2 microglia. As with the results of miRNAs function classification in vitro, the expression levels of mir-7036a-5p are upregulated in LPS-activated BV-2 microglia, but are downregulated by scutellarin. Rescue experiments indicated that mir-7036a-5p is a pro-inflammatory factor in activated BV-2 microglia. mir-7036a-5p agomir promoted the expression of phosphorylated tau proteins (p-tau), protein kinase C gamma type (PRKCG), extracellular regulated protein kinases (ERK1/2), but the is reversed by mir-7036a-5p antagomir in vitro. It is shown here that mir-7036a-5p is involved in microglia-mediated inflammation in LPS-induced BV-2 microglia. More important is the novel finding that scutellarin mitigated microglia inflammation by down-regulating the mir-7036a-5p/MAPT/PRKCG/ERK signaling pathway.
Collapse
Affiliation(s)
- Zhaoda Duan
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P. R. China
| | - Li Yang
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P. R. China
| | - Dongyao Xu
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P. R. China
| | - Zhi Qi
- Department of Neurology, No.2 Affiliated Hospital, Kunming Medical University, 374 Dianmian Road, Kunming, 650101, P. R. China
| | - Wenji Jia
- Department of Neurology, No.2 Affiliated Hospital, Kunming Medical University, 374 Dianmian Road, Kunming, 650101, P. R. China
| | - Chunyun Wu
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P. R. China
| |
Collapse
|
24
|
Lee HJ, Shin HK, Kim JH, Choi BT. Transcriptome Analysis of the Striatum of Electroacupuncture-treated Naïve and Ischemic Stroke Mice. J Pharmacopuncture 2024; 27:162-171. [PMID: 38948311 PMCID: PMC11194526 DOI: 10.3831/kpi.2024.27.2.162] [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: 04/26/2024] [Revised: 05/09/2024] [Accepted: 05/23/2024] [Indexed: 07/02/2024] Open
Abstract
Objectives Electroacupuncture (EA) has been demonstrated to aid stroke recovery. However, few investigations have focused on identifying the potent molecular targets of EA by comparing EA stimulation between naïve and disease models. Therefore, this study was undertaken to identify the potent molecular therapeutic mechanisms underlying EA stimulation in ischemic stroke through a comparison of mRNA sequencing data obtained from EA-treated naïve control and ischemic stroke mouse models. Methods Using both naïve control and middle cerebral artery occlusion (MCAO) mouse models, EA stimulation was administered at two acupoints, Baihui (GV20) and Dazhui (GV14), at a frequency of 2 Hz. Comprehensive assessments were conducted, including behavioral evaluations, RNA sequencing to identify differentially expressed genes (DEGs), functional enrichment analysis, protein-protein interaction (PPI) network analysis, and quantitative real-time PCR. Results EA stimulation ameliorated the ischemic insult-induced motor dysfunction in mice with ischemic stroke. Comparative analysis between control vs. MCAO, control vs. control + EA, and MCAO vs. MCAO + EA revealed 4,407, 101, and 82 DEGs, respectively. Of these, 30, 7, and 1 were common across the respective groups. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed upregulated DEGs associated with the regulation of inflammatory immune response in the MCAO vs. MCAO + EA comparison. Conversely, downregulated DEGs in the control vs. control + EA comparison were linked to neuronal development. PPI analysis revealed major clustering related to the regulation of cytokines, such as Cxcl9, Pcp2, Ccl11, and Cxcl13, in the common DEGs of MCAO vs. MCAO + EA, with Esp8l1 identified as the only common downregulated DEG in both EA-treated naïve and ischemic models. Conclusion These findings underscore the diverse potent mechanisms of EA stimulation between naïve and ischemic stroke mice, albeit with few overlaps. However, the potent mechanisms underlying EA treatment in ischemic stroke models were associated with the regulation of inflammatory processes involving cytokines.
Collapse
Affiliation(s)
- Hong Ju Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Ji-Hwan Kim
- Department of Sasang Constitutional Medicine, Division of Clinical Medicine 4, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
25
|
Nusraty S, Boddeti U, Zaghloul KA, Brown DA. Microglia in Glioblastomas: Molecular Insight and Immunotherapeutic Potential. Cancers (Basel) 2024; 16:1972. [PMID: 38893093 PMCID: PMC11171200 DOI: 10.3390/cancers16111972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive and devastating primary brain tumors, with a median survival of 15 months following diagnosis. Despite the intense treatment regimen which routinely includes maximal safe neurosurgical resection followed by adjuvant radio- and chemotherapy, the disease remains uniformly fatal. The poor prognosis associated with GBM is multifactorial owing to factors such as increased proliferation, angiogenesis, and metabolic switching to glycolytic pathways. Critically, GBM-mediated local and systemic immunosuppression result in inadequate immune surveillance and ultimately, tumor-immune escape. Microglia-the resident macrophages of the central nervous system (CNS)-play crucial roles in mediating the local immune response in the brain. Depending on the specific pathological cues, microglia are activated into either a pro-inflammatory, neurotoxic phenotype, known as M1, or an anti-inflammatory, regenerative phenotype, known as M2. In either case, microglia secrete corresponding pro- or anti-inflammatory cytokines and chemokines that either promote or hinder tumor growth. Herein, we review the interplay between GBM cells and resident microglia with a focus on contemporary studies highlighting the effect of GBM on the subtypes of microglia expressed, the associated cytokines/chemokines secreted, and ultimately, their impact on tumor pathogenesis. Finally, we explore how understanding the intricacies of the tumor-immune landscape can inform novel immunotherapeutic strategies against this devastating disease.
Collapse
Affiliation(s)
| | | | | | - Desmond A. Brown
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (S.N.); (U.B.); (K.A.Z.)
| |
Collapse
|
26
|
Paiva IHRD, Maciel LM, Silva RSD, Mendonça IP, Souza JRBD, Peixoto CA. Prebiotics modulate the microbiota-gut-brain axis and ameliorate anxiety and depression-like behavior in HFD-fed mice. Food Res Int 2024; 182:114153. [PMID: 38519181 DOI: 10.1016/j.foodres.2024.114153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 03/24/2024]
Abstract
Previous research has demonstrated that Prebiotics can influence the composition of the gut microbiota, consequently impacting mood regulation. This study aimed to assess the effects of Prebiotics, specifically Fructooligosaccharides (FOS) and Galactooligosaccharides (GOS) on neuroinflammation, depression, and anxiety-like behavior in a mouse model fed a high-fat diet (HFD). Initially, mice were divided into two groups: a control group on a standard diet (n = 15) and a group on an HFD for 18 weeks (n = 45). By the 13th week, the HFD group was further divided into experimental groups: Control (n = 15), HFD (n = 15), HFD receiving Prebiotics (n = 15), and HFD receiving Fluoxetine (n = 15). From the 13th week onward, the HFD + Prebiotics group received both the high-fat diet and a combination of FOS and GOS, while the HFD + Fluoxetine group received Fluoxetine in their drinking water. In the 18th week, all mice underwent tests to evaluate behavior, including the Tail Suspension Test (TST), Forced Swimming Test (FST), Sucrose Preference Test (SPT), and the Plus Maze Test (PMT), after which they were euthanized. Mice on the HFD exhibited increased body weight, abdominal size, blood glucose, triglyceride levels, cholesterol, insulin, HOMA index, and higher serum IL-1β. These obese mice also displayed an increased number of microglia and astrocytes, activation of the TLR4 pathway, and elevated levels of neuroinflammatory markers like TNF-α, IL-1β, and COX-2. Moreover, obese mice showed increased activation of the IDO pathway and decreased levels of NMDA receptors. Additionally, markers of neurogenesis and synaptic plasticity, such as PSD, SAP 102, CREB-p, and BDNF, were lower. Treatment with FOS and GOS reversed symptoms of depression and anxiety in mice subjected to HD. This improvement in behavior resulted from a reduction in dysbiosis with an increase in acetate-producing bacteria (B. acidifaciens and B. dorei) and intestinal permeability, leading to a decrease in chronic peripheral and central inflammation. Furthermore, the modulation of the gut-brain axis by FOS and GOS promoted elevated acetate and GPR43 levels in the brain and a reduction in the levels of pro-inflammatory cytokines, positively impacting signaling pathways of neuronal proliferation and survival in the hippocampus and prefrontal cortex.
Collapse
Affiliation(s)
- Igor Henrique Rodrigues de Paiva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), PE, Brazil; Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil.
| | - Laís Macedo Maciel
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), PE, Brazil
| | - Rodrigo Soares da Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), PE, Brazil; Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - Ingrid Prata Mendonça
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), PE, Brazil; Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | | | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), PE, Brazil; Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Brazil.
| |
Collapse
|
27
|
Wang P, Li Y, Song Y, Gao Y, Hao C, Zhou Y, Bao S, Guo J, Li X. Human umbilical cord mesenchymal stem cells reverse depression in rats induced by chronic unpredictable mild stress combined with lipopolysaccharide. CNS Neurosci Ther 2024; 30:e14644. [PMID: 38433020 PMCID: PMC10909725 DOI: 10.1111/cns.14644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/24/2024] [Accepted: 02/01/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Inflammation and oxidative stress are considered crucial to the pathogenesis of depression. Rat models of depression can be created by combined treatments of chronic unpredictable mild stress (CUMS) and lipopolysaccharide (LPS). Behaviors associated with depression could be improved by treatment with mesenchymal stem cells (MSCs) owing to immunomodulatory functions of the cells. Therapeutic potentials of the MSCs to reverse pro-inflammatory cytokines, proteins, and metabolites were identified by transcriptomic, proteomic, and metabolomic analysis, respectively. METHODS A depression model was established in male SD rats by 2 weeks of CUMS combined with LPS. The models were verified by behavioral tests, namely SPT, OFT, EPM, and qRT-PCR for pro-inflammatory cytokines. Such depressed rats were administered human umbilical cord MSCs (hUC-MSCs) via the tail vein once a week for 2 and 4 weeks. The homing capacity was confirmed by detection of the fluorescent dye on day 7 after the hUC-MSCs were labeled with CM-Dil and administered. The expression of GFAP in astrocytes serves as a biomarker of CNS disorders and IBA1 in microglia serves as a marker of microglia activation were detected by immunohistochemistry at 2 and 4 weeks after final administration of hUC-MSCs. At the same time, transcriptomics of rat hippocampal tissue, proteomic and metabolomic analysis of the serum from the normal, depressed, and treated rats were also compared. RESULTS Reliable models of rat depression were successfully induced by treatments of CUMS combined with LPS. Rat depression behaviors, pro-inflammatory cytokines, and morphological disorders of the hippocampus associated with depression were reversed in 4 weeks by hUC-MSC treatment. hUC-MSCs could reach the hippocampus CA1 region through the blood circulation on day 7 after administration owing to the disruption of blood brain barrier (BBB) by microglial activation from depression. Differentiations of whole-genome expression, protein, and metabolite profiles between the normal and depression-modeled rats, which were analyzed by transcriptomic, proteomics, and metabolomics, further verified the high association with depression behaviors. CONCLUSIONS Rat depression can be reversed or recovered by treatment with hUC-MSCs.
Collapse
Affiliation(s)
- Pengxiang Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland LivestockInner Mongolia UniversityHohhotChina
- Research Center for Animal Genetic Resources of Mongolia PlateauCollege of Life Sciences, Inner Mongolia UniversityHohhotChina
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhotChina
| | - Yunxia Li
- Research Center for Animal Genetic Resources of Mongolia PlateauCollege of Life Sciences, Inner Mongolia UniversityHohhotChina
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic AnimalHohhotChina
| | - Yongli Song
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland LivestockInner Mongolia UniversityHohhotChina
- Research Center for Animal Genetic Resources of Mongolia PlateauCollege of Life Sciences, Inner Mongolia UniversityHohhotChina
| | - Yuan Gao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland LivestockInner Mongolia UniversityHohhotChina
- Research Center for Animal Genetic Resources of Mongolia PlateauCollege of Life Sciences, Inner Mongolia UniversityHohhotChina
| | - Chunxia Hao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland LivestockInner Mongolia UniversityHohhotChina
- Research Center for Animal Genetic Resources of Mongolia PlateauCollege of Life Sciences, Inner Mongolia UniversityHohhotChina
| | - Yang Zhou
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland LivestockInner Mongolia UniversityHohhotChina
- Research Center for Animal Genetic Resources of Mongolia PlateauCollege of Life Sciences, Inner Mongolia UniversityHohhotChina
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhotChina
| | - Siqin Bao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland LivestockInner Mongolia UniversityHohhotChina
- Research Center for Animal Genetic Resources of Mongolia PlateauCollege of Life Sciences, Inner Mongolia UniversityHohhotChina
| | - Jitong Guo
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic AnimalHohhotChina
- Inner Mongolia Yihong Medical Research Co. LtdHohhotChina
| | - Xihe Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland LivestockInner Mongolia UniversityHohhotChina
- Research Center for Animal Genetic Resources of Mongolia PlateauCollege of Life Sciences, Inner Mongolia UniversityHohhotChina
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic AnimalHohhotChina
| |
Collapse
|
28
|
Szlufik S, Kopeć K, Szleszkowski S, Koziorowski D. Glymphatic System Pathology and Neuroinflammation as Two Risk Factors of Neurodegeneration. Cells 2024; 13:286. [PMID: 38334678 PMCID: PMC10855155 DOI: 10.3390/cells13030286] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/26/2024] [Accepted: 02/03/2024] [Indexed: 02/10/2024] Open
Abstract
The key to the effective treatment of neurodegenerative disorders is a thorough understanding of their pathomechanism. Neurodegeneration and neuroinflammation are mutually propelling brain processes. An impairment of glymphatic system function in neurodegeneration contributes to the progression of pathological processes. The question arises as to how neuroinflammation and the glymphatic system are related. This review highlights the direct and indirect influence of these two seemingly independent processes. Protein aggregates, a characteristic feature of neurodegeneration, are correlated with glymphatic clearance and neuroinflammation. Glial cells cannot be overlooked when considering the neuroinflammatory processes. Astrocytes are essential for the effective functioning of the glymphatic system and play a crucial role in the inflammatory responses in the central nervous system. It is imperative to acknowledge the significance of AQP4, a protein that exhibits a high degree of polarization in astrocytes and is crucial for the functioning of the glymphatic system. AQP4 influences inflammatory processes that have not yet been clearly delineated. Another interesting issue is the gut-brain axis and microbiome, which potentially impact the discussed processes. A discussion of the correlation between the functioning of the glymphatic system and neuroinflammation may contribute to exploring the pathomechanism of neurodegeneration.
Collapse
Affiliation(s)
- Stanisław Szlufik
- Department of Neurology, Faculty of Health Science, Medical University of Warsaw, 02-091 Warszawa, Poland; (K.K.)
| | | | | | | |
Collapse
|
29
|
Xu W, Wang X, Hou X, Yang Y, Ma R, Lv R, Yin Q. The role of microglia in the pathogenesis of diabetic-associated cognitive dysfunction. Front Endocrinol (Lausanne) 2024; 14:1246979. [PMID: 38274227 PMCID: PMC10808430 DOI: 10.3389/fendo.2023.1246979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Affiliation(s)
- Wenwen Xu
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xinyu Wang
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xunyao Hou
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Yang
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Rongrong Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Renjun Lv
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qingqing Yin
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
30
|
Chen CA, Li CX, Zhang ZH, Xu WX, Liu SL, Ni WC, Wang XQ, Cheng FF, Wang QG. Qinzhizhudan formula dampens inflammation in microglia polarization of vascular dementia rats by blocking MyD88/NF-κB signaling pathway: Through integrating network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116769. [PMID: 37400007 DOI: 10.1016/j.jep.2023.116769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 07/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qinzhizhudan Formula (QZZD) is composed of Scutellaria baicalensis Georgi (Huang Qin) extract, Gardenia jasminoides (Zhizi) extract and Suis Fellis Pulvis (Zhudanfen) (ratio of 4:5:6). This formula is optimized from Qingkailing (QKL) injection. Regarding brain injury, QZZD is protective. However, the mechanism by which QZZD treats vascular dementia (VD) has not been elucidated. AIM OF THE STUDY To ascertain QZZD's effect on the treatment of VD and further investigate the molecular mechanisms. MATERIALS AND METHODS In this study, we screened the possible components and targets of QZZD against VD and microglia polarization using network pharmacology (NP), then an animal model of bilateral common carotid artery ligation method (2VO) was induced. Afterward, The Morris water maze was employed to evaluate cognitive ability, and pathological alterations in the CA1 area of the hippocampus were detected using HE and Nissl staining. To confirm the affect of QZZD on VD and its molecular mechanism, the contents of inflammatory factors IL-1β, TNF-α, IL-4, and IL-10 were performed to detect by ELISA, the phenotype polarization of microglia cells was detected by immunofluorescence staining, and the expressions of MyD88, p-IκBα and p-NF-κB p65 in brain tissue were detected by western blot. RESULTS A total of 112 active compounds and 363 common targets of QZZD, microglia polarization, and VD were identified, according to the NP analysis. 38 hub targets were screened out from the PPI network. GO analysis and KEGG pathway analysis showed that QZZD may regulate microglia polarization through anti-inflammatory mechanism such as Toll-like receptor signaling pathway and NF-κB signaling pathway. The further results showed that QZZD can alleviate the memory impairment induced by 2VO. QZZD profoundly rescued brain hippocampus neuronal damage and increased the number of neurons. These advantageous outcomes were linked to the control of microglia polarization. QZZD decreased M1 phenotypic marker expression while increasing M2 phenotypic marker expression. QZZD may controll the polarization of the M1 microglia by blocking the core part of Toll-like receptor signaling pathway, that is the MyD88/NF-κB signaling pathway, which reduced the neurotoxic effects of the microglia. CONCLUSION Here, we explored the anti-VD microglial polarization characteristic of QZZD for the first time and clarified its mechanisms. These findings will provide valuable clues for the discovery of anti-VD agents.
Collapse
Affiliation(s)
- Cong-Ai Chen
- Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 100700, China; Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Chang-Xiang Li
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Ze-Han Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wen-Xiu Xu
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shu-Ling Liu
- Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 100700, China; Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wen-Chao Ni
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xue-Qian Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Fa-Feng Cheng
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Qing-Guo Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
31
|
Boyton I, Valenzuela SM, Collins-Praino LE, Care A. Neuronanomedicine for Alzheimer's and Parkinson's disease: Current progress and a guide to improve clinical translation. Brain Behav Immun 2024; 115:631-651. [PMID: 37967664 DOI: 10.1016/j.bbi.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 09/19/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023] Open
Abstract
Neuronanomedicine is an emerging multidisciplinary field that aims to create innovative nanotechnologies to treat major neurodegenerative disorders, such as Alzheimer's (AD) and Parkinson's disease (PD). A key component of neuronanomedicine are nanoparticles, which can improve drug properties and demonstrate enhanced safety and delivery across the blood-brain barrier, a major improvement on existing therapeutic approaches. In this review, we critically analyze the latest nanoparticle-based strategies to modify underlying disease pathology to slow or halt AD/PD progression. We find that a major roadblock for neuronanomedicine translation to date is a poor understanding of how nanoparticles interact with biological systems (i.e., bio-nano interactions), which is partly due to inconsistent reporting in published works. Accordingly, this review makes a set of specific recommendations to help guide researchers to harness the unique properties of nanoparticles and thus realise breakthrough treatments for AD/PD.
Collapse
Affiliation(s)
- India Boyton
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia
| | - Stella M Valenzuela
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia
| | | | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Gadigal Country, NSW 2007, Australia.
| |
Collapse
|
32
|
Tyliszczak M, Wiatrak B, Danielewski M, Szeląg A, Kucharska AZ, Sozański T. Does a pickle a day keep Alzheimer's away? Fermented food in Alzheimer's disease: A review. Exp Gerontol 2023; 184:112332. [PMID: 37967591 DOI: 10.1016/j.exger.2023.112332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023]
Abstract
Fermented food is commonly viewed as healthy, mostly due to its probiotic and digestion-enhancing properties and recently it has been examined with regard to the development of new therapeutic and preventive measures for Alzheimer's disease. Fermented food has been shown to have anti-inflammatory and antioxidant properties and to alter the gut microbiota. However, the exact pathogenesis of Alzheimer's disease is still unknown and its connections to systemic inflammation and gut dysbiosis, as potential targets of fermented food, require further investigation. Therefore, to sum up the current knowledge, this article reviews recent research on the pathogenesis of Alzheimer's disease with emphasis on the role of the gut-brain axis and studies examining the use of fermented foods. The analysis of the fermented food research includes clinical and preclinical in vivo and in vitro studies. The fermented food studies have shown promising effects on amyloid-β metabolism, inflammation, and cognitive impairment in animals and humans. Fermented food has great potential in developing new approaches to Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Michał Tyliszczak
- Department of Pharmacology, Wroclaw Medical University, Wrocław, Poland.
| | - Benita Wiatrak
- Department of Pharmacology, Wroclaw Medical University, Wrocław, Poland
| | | | - Adam Szeląg
- Department of Pharmacology, Wroclaw Medical University, Wrocław, Poland
| | - Alicja Z Kucharska
- Department of Fruit, Vegetable, and Plant Nutraceutical Technology, Wroclaw University of Environmental and Life Sciences, Wrocław, Poland
| | - Tomasz Sozański
- Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Faculty of Medicine, Wroclaw University of Science and Technology, Wrocław, Poland
| |
Collapse
|
33
|
Li Y, Li YJ, Zhu ZQ. To re-examine the intersection of microglial activation and neuroinflammation in neurodegenerative diseases from the perspective of pyroptosis. Front Aging Neurosci 2023; 15:1284214. [PMID: 38020781 PMCID: PMC10665880 DOI: 10.3389/fnagi.2023.1284214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and motor neuron disease, are diseases characterized by neuronal damage and dysfunction. NDs are considered to be a multifactorial disease with diverse etiologies (immune, inflammatory, aging, genetic, etc.) and complex pathophysiological processes. Previous studies have found that neuroinflammation and typical microglial activation are important mechanisms of NDs, leading to neurological dysfunction and disease progression. Pyroptosis is a new mode involved in this process. As a form of programmed cell death, pyroptosis is characterized by the expansion of cells until the cell membrane bursts, resulting in the release of cell contents that activates a strong inflammatory response that promotes NDs by accelerating neuronal dysfunction and abnormal microglial activation. In this case, abnormally activated microglia release various pro-inflammatory factors, leading to the occurrence of neuroinflammation and exacerbating both microglial and neuronal pyroptosis, thus forming a vicious cycle. The recognition of the association between pyroptosis and microglia activation, as well as neuroinflammation, is of significant importance in understanding the pathogenesis of NDs and providing new targets and strategies for their prevention and treatment.
Collapse
Affiliation(s)
- Yuan Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- College of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Ying-Jie Li
- Department of General Surgery, Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| | - Zhao-Qiong Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
34
|
Jia W, Yuan Y, Yang L, Wu C. Scutellarin attenuates microglia activation in experimentally induced hypoxia-ischemia brain damage by down-regulating miRNA-7036a. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1685-1688. [PMID: 37246896 PMCID: PMC10577478 DOI: 10.3724/abbs.2023100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 05/21/2023] [Indexed: 05/30/2023] Open
Affiliation(s)
- Wenji Jia
- Department of Anatomy and Histology/EmbryologyFaculty of Basic Medical SciencesKunming Medical UniversityKunming650500China
- Department of NeurologyNo.2 Affiliated HospitalKunming Medical UniversityKunming650101China
| | - Yun Yuan
- Department of Anatomy and Histology/EmbryologyFaculty of Basic Medical SciencesKunming Medical UniversityKunming650500China
| | - Li Yang
- Department of Anatomy and Histology/EmbryologyFaculty of Basic Medical SciencesKunming Medical UniversityKunming650500China
| | - Chunyun Wu
- Department of Anatomy and Histology/EmbryologyFaculty of Basic Medical SciencesKunming Medical UniversityKunming650500China
| |
Collapse
|
35
|
Biswas K. Microglia mediated neuroinflammation in neurodegenerative diseases: A review on the cell signaling pathways involved in microglial activation. J Neuroimmunol 2023; 383:578180. [PMID: 37672840 DOI: 10.1016/j.jneuroim.2023.578180] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/01/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
Microglia, the immune sentinels of the central nervous system (CNS), have emerged to be the central players in many neurological and neurodegenerative diseases. Recent studies on large genome databases and omics studies in fact provide support to the idea that microglial cells could be the drivers of these diseases. Microglial cells have the capacity to undergo morphological and phenotypic transformations depending on its microenvironment. From the homeostatic ramified state, they can shift their phenotypes between the two extremes, known as the proinflammatory M1 and anti-inflammatory M2 phenotype, with intermediate transitional states, characterized by different transcriptional signature and release of inflammatory mediators. The temporal regulation of the release of the inflammatory factors are critical for damage control and steering the microglia back towards homeostatic conditions. A dysregulation in these can lead to excessive tissue damage and neuronal death. Therefore, targeting the cell signaling pathways that are the underpinnings of microglial modulations are considered to be an important avenue for treatment of various neurodegenerative diseases. In this review we have discussed various signaling pathways that trigger microglial activation from its ramified state and highlight the mechanisms of microglia-mediated neuroinflammation that are associated with various neurodegenerative diseases. Most of the cellular factors that drive microglia towards a proinflammatory phenotype are components of the immune system signaling pathways and cell proliferation, along with certain ion channels. The anti-inflammatory phenotype is mainly elicited by purinoceptors, metabolic receptors and other receptors that primarily suppress the production proinflammatory mediators.
Collapse
Affiliation(s)
- Kaushiki Biswas
- Department of Life Sciences, Presidency University Main campus, 86/1 College Street, Kolkata 700073, India.
| |
Collapse
|
36
|
Fang S, Wu Z, Guo Y, Zhu W, Wan C, Yuan N, Chen J, Hao W, Mo X, Guo X, Fan L, Li X, Chen J. Roles of microglia in adult hippocampal neurogenesis in depression and their therapeutics. Front Immunol 2023; 14:1193053. [PMID: 37881439 PMCID: PMC10597707 DOI: 10.3389/fimmu.2023.1193053] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Adult hippocampal neurogenesis generates functional neurons from neural progenitor cells in the hippocampal dentate gyrus (DG) to complement and repair neurons and neural circuits, thus benefiting the treatment of depression. Increasing evidence has shown that aberrant microglial activity can disrupt the appropriate formation and development of functional properties of neurogenesis, which will play a crucial role in the occurrence and development of depression. However, the mechanisms of the crosstalk between microglia and adult hippocampal neurogenesis in depression are not yet fully understood. Therefore, in this review, we first introduce recent discoveries regarding the roles of microglia and adult hippocampal neurogenesis in the etiology of depression. Then, we systematically discuss the possible mechanisms of how microglia regulate adult hippocampal neurogenesis in depression according to recent studies, which involve toll-like receptors, microglial polarization, fractalkine-C-X3-C motif chemokine receptor 1, hypothalamic-pituitary-adrenal axis, cytokines, brain-derived neurotrophic factor, and the microbiota-gut-brain axis, etc. In addition, we summarize the promising drugs that could improve the adult hippocampal neurogenesis by regulating the microglia. These findings will help us understand the complicated pathological mechanisms of depression and shed light on the development of new treatment strategies for this disease.
Collapse
Affiliation(s)
- Shaoyi Fang
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhibin Wu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yali Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wenjun Zhu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Chunmiao Wan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Shenzhen People’s Hospital, 2Clinical Medical College, Jinan University, Shenzhen, China
| | - Jianbei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenzhi Hao
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaowei Mo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaofang Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lili Fan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaojuan Li
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jiaxu Chen
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
37
|
Stanca S, Rossetti M, Bongioanni P. Astrocytes as Neuroimmunocytes in Alzheimer's Disease: A Biochemical Tool in the Neuron-Glia Crosstalk along the Pathogenetic Pathways. Int J Mol Sci 2023; 24:13880. [PMID: 37762184 PMCID: PMC10531177 DOI: 10.3390/ijms241813880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
This work aimed at assessing Alzheimer's disease (AD) pathogenesis through the investigation of the astrocytic role to transduce the load of amyloid-beta (Aβ) into neuronal death. The backbone of this review is focused on the deepening of the molecular pathways eliciting the activation of astrocytes crucial phenomena in the understanding of AD as an autoimmune pathology. The complex relations among astrocytes, Aβ and tau, together with the role played by the tripartite synapsis are discussed. A review of studies published from 1979 to 2023 on Scopus, PubMed and Google Scholar databases was conducted. The selected papers focused not only on the morphological and metabolic characteristics of astrocytes, but also on the latest notions about their multifunctional involvement in AD pathogenesis. Astrocytes participate in crucial pathways, including pruning and sprouting, by which the AD neurodegeneration evolves from an aggregopathy to neuroinflammation, loss of synapses and neuronal death. A1 astrocytes stimulate the production of pro-inflammatory molecules which have been correlated with the progression of AD cognitive impairment. Further research is needed to "hold back" the A1 polarization and, thus, to slow the worsening of the disease. AD clinical expression is the result of dysfunctional neuronal interactions, but this is only the end of a process involving a plurality of protagonists. One of these is the astrocyte, whose importance this work intends to put under the spotlight in the AD scenario, reflecting the multifaceted nature of this disease in the functional versatility of this glial population.
Collapse
Affiliation(s)
- Stefano Stanca
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy
- NeuroCare Onlus, 56100 Pisa, Italy
| | - Martina Rossetti
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy
- NeuroCare Onlus, 56100 Pisa, Italy
| | - Paolo Bongioanni
- NeuroCare Onlus, 56100 Pisa, Italy
- Medical Specialties Department, Azienda Ospedaliero-Universitaria Pisana, 56100 Pisa, Italy
| |
Collapse
|
38
|
Darwish SF, Elbadry AMM, Elbokhomy AS, Salama GA, Salama RM. The dual face of microglia (M1/M2) as a potential target in the protective effect of nutraceuticals against neurodegenerative diseases. FRONTIERS IN AGING 2023; 4:1231706. [PMID: 37744008 PMCID: PMC10513083 DOI: 10.3389/fragi.2023.1231706] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023]
Abstract
The pathophysiology of different neurodegenerative illnesses is significantly influenced by the polarization regulation of microglia and macrophages. Traditional classifications of macrophage phenotypes include the pro-inflammatory M1 and the anti-inflammatory M2 phenotypes. Numerous studies demonstrated dynamic non-coding RNA modifications, which are catalyzed by microglia-induced neuroinflammation. Different nutraceuticals focus on the polarization of M1/M2 phenotypes of microglia and macrophages, offering a potent defense against neurodegeneration. Caeminaxin A, curcumin, aromatic-turmerone, myricetin, aurantiamide, 3,6'-disinapoylsucrose, and resveratrol reduced M1 microglial inflammatory markers while increased M2 indicators in Alzheimer's disease. Amyloid beta-induced microglial M1 activation was suppressed by andrographolide, sulforaphane, triptolide, xanthoceraside, piperlongumine, and novel plant extracts which also prevented microglia-mediated necroptosis and apoptosis. Asarone, galangin, baicalein, and a-mangostin reduced oxidative stress and pro-inflammatory cytokines, such as interleukin (IL)-1, IL-6, and tumor necrosis factor-alpha in M1-activated microglia in Parkinson's disease. Additionally, myrcene, icariin, and tenuigenin prevented the nod-like receptor family pyrin domain-containing 3 inflammasome and microglial neurotoxicity, while a-cyperone, citronellol, nobiletin, and taurine prevented NADPH oxidase 2 and nuclear factor kappa B activation. Furthermore, other nutraceuticals like plantamajoside, swertiamarin, urolithin A, kurarinone, Daphne genkwa flower, and Boswellia serrata extracts showed promising neuroprotection in treating Parkinson's disease. In Huntington's disease, elderberry, curcumin, iresine celosia, Schisandra chinensis, gintonin, and pomiferin showed promising results against microglial activation and improved patient symptoms. Meanwhile, linolenic acid, resveratrol, Huperzia serrata, icariin, and baicalein protected against activated macrophages and microglia in experimental autoimmune encephalomyelitis and multiple sclerosis. Additionally, emodin, esters of gallic and rosmarinic acids, Agathisflavone, and sinomenine offered promising multiple sclerosis treatments. This review highlights the therapeutic potential of using nutraceuticals to treat neurodegenerative diseases involving microglial-related pathways.
Collapse
Affiliation(s)
- Samar F. Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Abdullah M. M. Elbadry
- Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
- Nanotechnology Research Center (NTRC), The British University in Egypt (BUE), El-Sherouk City, Egypt
| | | | - Ghidaa A. Salama
- Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Rania M. Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| |
Collapse
|
39
|
Kwon YJ, Kwon OI, Hwang HJ, Shin HC, Yang S. Therapeutic effects of phlorotannins in the treatment of neurodegenerative disorders. Front Mol Neurosci 2023; 16:1193590. [PMID: 37305552 PMCID: PMC10249478 DOI: 10.3389/fnmol.2023.1193590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Phlorotannins are natural polyphenolic compounds produced by brown marine algae and are currently found in nutritional supplements. Although they are known to cross the blood-brain barrier, their neuropharmacological actions remain unclear. Here we review the potential therapeutic benefits of phlorotannins in the treatment of neurodegenerative diseases. In mouse models of Alzheimer's disease, ethanol intoxication and fear stress, the phlorotannin monomer phloroglucinol and the compounds eckol, dieckol and phlorofucofuroeckol A have been shown to improve cognitive function. In a mouse model of Parkinson's disease, phloroglucinol treatment led to improved motor performance. Additional neurological benefits associated with phlorotannin intake have been demonstrated in stroke, sleep disorders, and pain response. These effects may stem from the inhibition of disease-inducing plaque synthesis and aggregation, suppression of microglial activation, modulation of pro-inflammatory signaling, reduction of glutamate-induced excitotoxicity, and scavenging of reactive oxygen species. Clinical trials of phlorotannins have not reported significant adverse effects, suggesting these compounds to be promising bioactive agents in the treatment of neurological diseases. We therefore propose a putative biophysical mechanism of phlorotannin action in addition to future directions for phlorotannin research.
Collapse
Affiliation(s)
- Yoon Ji Kwon
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Oh Ig Kwon
- Botamedi Brain Health and Medical Care Company Limited, Central, Hong Kong SAR, China
| | - Hye Jeong Hwang
- Center for Molecular Intelligence, SUNY Korea, Incheon, Republic of Korea
| | - Hyeon-Cheol Shin
- Botamedi Brain Health and Medical Care Company Limited, Central, Hong Kong SAR, China
- Center for Molecular Intelligence, SUNY Korea, Incheon, Republic of Korea
| | - Sungchil Yang
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| |
Collapse
|
40
|
Park E, Ahn SI, Park JS, Shin JH. Shear-induced phenotypic transformation of microglia in vitro. Biophys J 2023; 122:1691-1700. [PMID: 36987391 PMCID: PMC10183375 DOI: 10.1016/j.bpj.2023.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 11/28/2022] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The brain cells are affected by continuous fluid shear stress that is driven by varying hydrostatic and osmotic pressure conditions, depending on the brain's pathophysiological conditions. Although all brain cells are sensitive to the subtle changes in various physicochemical factors in the microenvironment, microglia, the resident brain immune cells, exhibit the most significant morphodynamic transformation. However, little is known about the phenotypic alterations in microglia in response to changes in fluid shear stress. In this study, we established a flow-controlled microenvironment to investigate the effects of shear flow on microglial phenotypes, including morphology, motility, and activation states. We observed two distinct morphologies of microglia in a static condition: bipolar cells that oscillate along their long axis and unipolar cells that migrate persistently. When exposed to flow, a significant fraction of bipolar cells showed unstable oscillation with an increased amplitude of oscillation and a decreased frequency, which consequently led to the phenotypic transformation of oscillating cells into migrating cells. Furthermore, we observed that the level of proinflammatory genes increased in response to shear stress, although there were no significant changes in the level of antiinflammatory genes. Our findings suggest that an interstitial fluid-level stimulus can cause a dramatic phenotypic shift in microglia toward proinflammatory states, shedding light on the pathological outbreaks of severe brain diseases. Given that the fluidic environment in the brain can be locally disrupted in pathological circumstances, the mechanical stimulus by fluid flow should also be considered a crucial element in regulating the immune activities of the microglia in brain diseases.
Collapse
Affiliation(s)
- Eunyoung Park
- School of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Song Ih Ahn
- Department of Mechanical Engineering, Pusan National University, Busan, Korea
| | - Jin-Sung Park
- School of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Jennifer H Shin
- School of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.
| |
Collapse
|
41
|
Luo H, Zhang H, Mao J, Cao H, Tao Y, Zhao G, Zhang Z, Zhang N, Liu Z, Zhang J, Luo P, Xia Y, Cheng Y, Xie Z, Cheng Q, Liu G. Exosome-based nanoimmunotherapy targeting TAMs, a promising strategy for glioma. Cell Death Dis 2023; 14:235. [PMID: 37012233 PMCID: PMC10070666 DOI: 10.1038/s41419-023-05753-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 04/05/2023]
Abstract
Exosomes, the cell-derived small extracellular vehicles, play a vital role in intracellular communication by reciprocally transporting DNA, RNA, bioactive protein, chains of glucose, and metabolites. With great potential to be developed as targeted drug carriers, cancer vaccines and noninvasive biomarkers for diagnosis, treatment response evaluation, prognosis prediction, exosomes show extensive advantages of relatively high drug loading capacity, adjustable therapeutic agents release, enhanced permeation and retention effect, striking biodegradability, excellent biocompatibility, low toxicity, etc. With the rapid progression of basic exosome research, exosome-based therapeutics are gaining increasing attention in recent years. Glioma, the standard primary central nervous system (CNS) tumor, is still up against significant challenges as current traditional therapies of surgery resection combined with radiotherapy and chemotherapy and numerous efforts into new drugs showed little clinical curative effect. The emerging immunotherapy strategy presents convincing results in many tumors and is driving researchers to exert its potential in glioma. As the crucial component of the glioma microenvironment, tumor-associated macrophages (TAMs) significantly contribute to the immunosuppressive microenvironment and strongly influence glioma progression via various signaling molecules, simultaneously providing new insight into therapeutic strategies. Exosomes would substantially assist the TAMs-centered treatment as drug delivery vehicles and liquid biopsy biomarkers. Here we review the current potential exosome-mediated immunotherapeutics targeting TAMs in glioma and conclude the recent investigation on the fundamental mechanisms of diversiform molecular signaling events by TAMs that promote glioma progression.
Collapse
Affiliation(s)
- Hong Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jinning Mao
- Health management center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Brain Hospital of Hunan Province, The Second People's Hospital of Hunan Province, Changsha, China
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhiwen Zhang
- School of Pharmacy, Fudan University, Shanghai, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yuguo Xia
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zongyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
42
|
Liu YL, Huang HJ, Sheu SY, Liu YC, Lee IJ, Chiang SC, Lin AMY. Oral ellagic acid attenuated LPS-induced neuroinflammation in rat brain: MEK1 interaction and M2 microglial polarization. Exp Biol Med (Maywood) 2023; 248:656-664. [PMID: 37340785 PMCID: PMC10350794 DOI: 10.1177/15353702231182230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 03/20/2023] [Indexed: 06/22/2023] Open
Abstract
Ellagic acid, the marker component of peels of Punica granatum L., is known traditionally to treat traumatic hemorrhage. In this study, the cellular mechanism underlying ellagic acid-induced anti-inflammation was investigated using lipopolysaccharides (LPSs) as a neuroinflammation inducer. Our in vitro data showed that LPS (1 μg/mL) consistently phosphorylated ERK and induced neuroinflammation, such as elevation in tumor necrosis factor-α (TNF-α) and nitric oxide production in treated BV-2 cells. Incubation of ellagic acid significantly inhibited LPS-induced ERK phosphorylation and subsequent neuroinflammation in treated BV-2 cells. Furthermore, our in vivo study of neuroinflammation employed an intranigral infusion of LPS that resulted in a time-dependent elevation in phosphorylated ERK levels in the infused substantia nigra (SN). Oral administration of ellagic acid (100 mg/kg) significantly attenuated LPS-induced ERK phosphorylation. A four-day treatment of ellagic acid did not alter LPS-induced ED-1 elevation but ameliorated LPS-induced reduction in CD206 and arginase-1 (two biomarkers of M2 microglia). A seven-day treatment of ellagic acid abolished LPS-induced increases in heme-oxygenase-1, cyclo-oxygenase 2, and α-synuclein trimer levels (a pathological hallmark) in the infused SN. At the same time, ellagic acid attenuated LPS-induced increases in active caspase 3 and receptor-interacting protein kinase-3 levels (respective biomarkers of apoptosis and necroptosis) as well as reduction in tyrosine hydroxylase-positive cells in the infused SN. In silico analysis showed that ellagic acid binds to the catalytic site of MEK1. Our data suggest that ellagic acid is capable of inhibiting MEK1-ERK signaling and then attenuated LPS-induced neuroinflammation, protein aggregation, and programmed cell deaths. Moreover, M2 microglial polarization is suggested as a novel antineuroinflammatory mechanism in the ellagic acid-induced neuroprotection.
Collapse
Affiliation(s)
- Yu-Ling Liu
- Department of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112
| | - Hui-Ju Huang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112
| | - Sheh-Yi Sheu
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 112
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112
| | - Yu-Cheng Liu
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 112
| | - I-Jung Lee
- Pharmaceutical Botany Research Laboratory, Yokohama University of Pharmacy, Yokohama 245-0066, Japan
| | - Shao-Chin Chiang
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112
- Department of Pharmacy, Koo Foundation Sun Yat-Sen Cancer center, Taipei, Taiwan
| | - Anya Maan-Yuh Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112
| |
Collapse
|
43
|
The Pathological Activation of Microglia Is Modulated by Sexually Dimorphic Pathways. Int J Mol Sci 2023; 24:ijms24054739. [PMID: 36902168 PMCID: PMC10003784 DOI: 10.3390/ijms24054739] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Microglia are the primary immunocompetent cells of the central nervous system (CNS). Their ability to survey, assess and respond to perturbations in their local environment is critical in their role of maintaining CNS homeostasis in health and disease. Microglia also have the capability of functioning in a heterogeneous manner depending on the nature of their local cues, as they can become activated on a spectrum from pro-inflammatory neurotoxic responses to anti-inflammatory protective responses. This review seeks to define the developmental and environmental cues that support microglial polarization towards these phenotypes, as well as discuss sexually dimorphic factors that can influence this process. Further, we describe a variety of CNS disorders including autoimmune disease, infection, and cancer that demonstrate disparities in disease severity or diagnosis rates between males and females, and posit that microglial sexual dimorphism underlies these differences. Understanding the mechanism behind differential CNS disease outcomes between men and women is crucial in the development of more effective targeted therapies.
Collapse
|
44
|
Angeloni C, Malaguti M, Prata C, Freschi M, Barbalace MC, Hrelia S. Mechanisms Underlying Neurodegenerative Disorders and Potential Neuroprotective Activity of Agrifood By-Products. Antioxidants (Basel) 2022; 12:94. [PMID: 36670956 PMCID: PMC9854890 DOI: 10.3390/antiox12010094] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
Neurodegenerative diseases, characterized by progressive loss in selected areas of the nervous system, are becoming increasingly prevalent worldwide due to an aging population. Despite their diverse clinical manifestations, neurodegenerative diseases are multifactorial disorders with standard features and mechanisms such as abnormal protein aggregation, mitochondrial dysfunction, oxidative stress and inflammation. As there are no effective treatments to counteract neurodegenerative diseases, increasing interest has been directed to the potential neuroprotective activities of plant-derived compounds found abundantly in food and in agrifood by-products. Food waste has an extremely negative impact on the environment, and recycling is needed to promote their disposal and overcome this problem. Many studies have been carried out to develop green and effective strategies to extract bioactive compounds from food by-products, such as peel, leaves, seeds, bran, kernel, pomace, and oil cake, and to investigate their biological activity. In this review, we focused on the potential neuroprotective activity of agrifood wastes obtained by common products widely produced and consumed in Italy, such as grapes, coffee, tomatoes, olives, chestnuts, onions, apples, and pomegranates.
Collapse
Affiliation(s)
- Cristina Angeloni
- Department for Life Quality Studies, Alma Mater Studiorum–University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum–University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum–University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Michela Freschi
- Department for Life Quality Studies, Alma Mater Studiorum–University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum–University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum–University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy
| |
Collapse
|
45
|
Bloomingdale P, Karelina T, Ramakrishnan V, Bakshi S, Véronneau‐Veilleux F, Moye M, Sekiguchi K, Meno‐Tetang G, Mohan A, Maithreye R, Thomas VA, Gibbons F, Cabal A, Bouteiller J, Geerts H. Hallmarks of neurodegenerative disease: A systems pharmacology perspective. CPT Pharmacometrics Syst Pharmacol 2022; 11:1399-1429. [PMID: 35894182 PMCID: PMC9662204 DOI: 10.1002/psp4.12852] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
Age-related central neurodegenerative diseases, such as Alzheimer's and Parkinson's disease, are a rising public health concern and have been plagued by repeated drug development failures. The complex nature and poor mechanistic understanding of the etiology of neurodegenerative diseases has hindered the discovery and development of effective disease-modifying therapeutics. Quantitative systems pharmacology models of neurodegeneration diseases may be useful tools to enhance the understanding of pharmacological intervention strategies and to reduce drug attrition rates. Due to the similarities in pathophysiological mechanisms across neurodegenerative diseases, especially at the cellular and molecular levels, we envision the possibility of structural components that are conserved across models of neurodegenerative diseases. Conserved structural submodels can be viewed as building blocks that are pieced together alongside unique disease components to construct quantitative systems pharmacology (QSP) models of neurodegenerative diseases. Model parameterization would likely be different between the different types of neurodegenerative diseases as well as individual patients. Formulating our mechanistic understanding of neurodegenerative pathophysiology as a mathematical model could aid in the identification and prioritization of drug targets and combinatorial treatment strategies, evaluate the role of patient characteristics on disease progression and therapeutic response, and serve as a central repository of knowledge. Here, we provide a background on neurodegenerative diseases, highlight hallmarks of neurodegeneration, and summarize previous QSP models of neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter Bloomingdale
- Quantitative Pharmacology and PharmacometricsMerck & Co., Inc.BostonMassachusettsUSA
| | | | | | - Suruchi Bakshi
- Certara QSPOssThe Netherlands,Certara QSPPrincetonNew JerseyUSA
| | | | - Matthew Moye
- Quantitative Pharmacology and PharmacometricsMerck & Co., Inc.BostonMassachusettsUSA
| | - Kazutaka Sekiguchi
- Shionogi & Co., Ltd.OsakaJapan,SUNY Downstate Medical CenterNew YorkNew YorkUSA
| | | | | | | | | | - Frank Gibbons
- Clinical Pharmacology and PharmacometricsBiogenCambridgeMassachusettsUSA
| | | | - Jean‐Marie Bouteiller
- Center for Neural EngineeringDepartment of Biomedical Engineering at the Viterbi School of EngineeringLos AngelesCaliforniaUSA,Institute for Technology and Medical Systems Innovation, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | |
Collapse
|
46
|
Marinelli S, Marrone MC, Di Domenico M, Marinelli S. Endocannabinoid signaling in microglia. Glia 2022; 71:71-90. [PMID: 36222019 DOI: 10.1002/glia.24281] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
Microglia, the innate immune cells of the central nervous system (CNS), execute their sentinel, housekeeping and defense functions through a panoply of genes, receptors and released cytokines, chemokines and neurotrophic factors. Moreover, microglia functions are closely linked to the constant communication with other cell types, among them neurons. Depending on the signaling pathway and type of stimuli involved, the outcome of microglia operation can be neuroprotective or neurodegenerative. Accordingly, microglia are increasingly becoming considered cellular targets for therapeutic intervention. Among signals controlling microglia activity, the endocannabinoid (EC) system has been shown to exert a neuroprotective role in many neurological diseases. Like neurons, microglia express functional EC receptors and can produce and degrade ECs. Interestingly, boosting EC signaling leads to an anti-inflammatory and neuroprotective microglia phenotype. Nonetheless, little evidence is available on the microglia-mediated therapeutic effects of EC compounds. This review focuses on the EC signals acting on the CNS microglia in physiological and pathological conditions, namely on the CB1R, CB2R and TRPV1-mediated regulation of microglia properties. It also provides new evidence, which strengthens the understanding of mechanisms underlying the control of microglia functions by ECs. Given the broad expression of the EC system in glial and neuronal cells, the resulting picture is the need for in vivo studies in transgenic mouse models to dissect the contribution of EC microglia signaling in the neuroprotective effects of EC-derived compounds.
Collapse
Affiliation(s)
- Sara Marinelli
- CNR-National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Maria Cristina Marrone
- EBRI-Fondazione Rita Levi Montalcini, Rome, Italy.,Ministry of University and Research, Mission Unity for Recovery and Resilience Plan, Rome, Italy
| | - Marina Di Domenico
- EBRI-Fondazione Rita Levi Montalcini, Rome, Italy.,Bio@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| | | |
Collapse
|
47
|
Martínez-Gil N, Maneu V, Kutsyr O, Fernández-Sánchez L, Sánchez-Sáez X, Sánchez-Castillo C, Campello L, Lax P, Pinilla I, Cuenca N. Cellular and molecular alterations in neurons and glial cells in inherited retinal degeneration. Front Neuroanat 2022; 16:984052. [PMID: 36225228 PMCID: PMC9548552 DOI: 10.3389/fnana.2022.984052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple gene mutations have been associated with inherited retinal dystrophies (IRDs). Despite the spectrum of phenotypes caused by the distinct mutations, IRDs display common physiopathology features. Cell death is accompanied by inflammation and oxidative stress. The vertebrate retina has several attributes that make this tissue vulnerable to oxidative and nitrosative imbalance. The high energy demands and active metabolism in retinal cells, as well as their continuous exposure to high oxygen levels and light-induced stress, reveal the importance of tightly regulated homeostatic processes to maintain retinal function, which are compromised in pathological conditions. In addition, the subsequent microglial activation and gliosis, which triggers the secretion of pro-inflammatory cytokines, chemokines, trophic factors, and other molecules, further worsen the degenerative process. As the disease evolves, retinal cells change their morphology and function. In disease stages where photoreceptors are lost, the remaining neurons of the retina to preserve their function seek out for new synaptic partners, which leads to a cascade of morphological alterations in retinal cells that results in a complete remodeling of the tissue. In this review, we describe important molecular and morphological changes in retinal cells that occur in response to oxidative stress and the inflammatory processes underlying IRDs.
Collapse
Affiliation(s)
- Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Isabel Pinilla
- Aragón Institute for Health Research (IIS Aragón), Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa University Hospital, Zaragoza, Spain
- Department of Surgery, University of Zaragoza, Zaragoza, Spain
- Isabel Pinilla,
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
- *Correspondence: Nicolás Cuenca,
| |
Collapse
|
48
|
Oh Y, Jung HJ, Hong S, Cho Y, Park J, Cho D, Kim TS. Aminoacyl transfer ribonucleic acid synthetase complex-interacting multifunctional protein 1 induces microglial activation and M1 polarization via the mitogen-activated protein kinase/nuclear factor-kappa B signaling pathway. Front Cell Neurosci 2022; 16:977205. [PMID: 36159396 PMCID: PMC9491728 DOI: 10.3389/fncel.2022.977205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Activation of microglia, which is the primary immune cell of the central nervous system, plays an important role in neuroinflammation associated with several neuronal diseases. Aminoacyl tRNA synthetase (ARS) complex-interacting multifunctional protein 1 (AIMP1), a structural component of the multienzyme ARS complex, is secreted to trigger a pro-inflammatory function and has been associated with several inflammatory diseases. However, the effect of AIMP1 on microglial activation remains unknown. AIMP1 elevated the expression levels of activation-related cell surface markers and pro-inflammatory cytokines in primary and BV-2 microglial cells. In addition to the AIMP1-mediated increase in the expression levels of M1 markers [interleukin (IL)-6, tumor necrosis factor-α, and IL-1β], the expression levels of CD68, an M1 cell surface molecule, were also increased in AIMP-1-treated microglial cells, while those of CD206, an M2 cell surface molecule, were not, indicating that AIMP1 triggers the polarization of microglial cells into the M1 state but not the M2 state. AIMP1 treatment induced the phosphorylation of mitogen-activated protein kinases (MAPKs), while MAPK inhibitors suppressed the AIMP1-induced microglial cell activation. AIMP1 also induced the phosphorylation of the nuclear factor-kappa B (NF-κB) components and nuclear translocation of the NF-κB p65 subunit in microglial cells. Furthermore, c-Jun N-terminal kinase (JNK) and p38 inhibitors markedly suppressed the AIMP1-induced phosphorylation of NF-κB components as well as the nuclear translocation of NF-κB p65 subunit, suggesting the involvement of JNK and p38 as upstream regulators of NF-κB in AIMP1-induced microglial cell activation. The NF-κB inhibitor suppressed the AIMP1-induced M1 polarization of the microglial cells. Taken together, AIMP1 effectively induces M1 microglial activation via the JNK and p38/NF-κB-dependent pathways. These results suggest that AIMP1 released under stress conditions may be a pathological factor that induces neuroinflammation.
Collapse
Affiliation(s)
- Yebin Oh
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Hak-Jun Jung
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Seungwon Hong
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Yerim Cho
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Jiyeong Park
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Daeho Cho
- Institute of Convergence Science, Korea University, Seoul, South Korea
| | - Tae Sung Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
- *Correspondence: Tae Sung Kim,
| |
Collapse
|
49
|
Morató X, Pytel V, Jofresa S, Ruiz A, Boada M. Symptomatic and Disease-Modifying Therapy Pipeline for Alzheimer's Disease: Towards a Personalized Polypharmacology Patient-Centered Approach. Int J Mol Sci 2022; 23:9305. [PMID: 36012569 PMCID: PMC9409252 DOI: 10.3390/ijms23169305] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 02/07/2023] Open
Abstract
Since 1906, when Dr. Alois Alzheimer first described in a patient "a peculiar severe disease process of the cerebral cortex", people suffering from this pathology have been waiting for a breakthrough therapy. Alzheimer's disease (AD) is an irreversible, progressive neurodegenerative brain disorder and the most common form of dementia in the elderly with a long presymptomatic phase. Worldwide, approximately 50 million people are living with dementia, with AD comprising 60-70% of cases. Pathologically, AD is characterized by the deposition of amyloid β-peptide (Aβ) in the neuropil (neuritic plaques) and blood vessels (amyloid angiopathy), and by the accumulation of hyperphosphorylated tau in neurons (neurofibrillary tangles) in the brain, with associated loss of synapses and neurons, together with glial activation, and neuroinflammation, resulting in cognitive deficits and eventually dementia. The current competitive landscape in AD consists of symptomatic treatments, of which there are currently six approved medications: three AChEIs (donepezil, rivastigmine, and galantamine), one NMDA-R antagonist (memantine), one combination therapy (memantine/donepezil), and GV-971 (sodium oligomannate, a mixture of oligosaccharides derived from algae) only approved in China. Improvements to the approved therapies, such as easier routes of administration and reduced dosing frequencies, along with the developments of new strategies and combined treatments are expected to occur within the next decade and will positively impact the way the disease is managed. Recently, Aducanumab, the first disease-modifying therapy (DMT) has been approved for AD, and several DMTs are in advanced stages of clinical development or regulatory review. Small molecules, mAbs, or multimodal strategies showing promise in animal studies have not confirmed that promise in the clinic (where small to moderate changes in clinical efficacy have been observed), and therefore, there is a significant unmet need for a better understanding of the AD pathogenesis and the exploration of alternative etiologies and therapeutic effective disease-modifying therapies strategies for AD. Therefore, a critical review of the disease-modifying therapy pipeline for Alzheimer's disease is needed.
Collapse
Affiliation(s)
- Xavier Morató
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
| | - Vanesa Pytel
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
| | - Sara Jofresa
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
50
|
Ramírez Hernández E, Alanis Olvera B, Carmona González D, Guerrero Marín O, Pantoja Mercado D, Valencia Gil L, Hernández-Zimbrón LF, Sánchez Salgado JL, Limón ID, Zenteno E. Neuroinflammation and galectins: a key relationship in neurodegenerative diseases. Glycoconj J 2022; 39:685-699. [PMID: 35653015 DOI: 10.1007/s10719-022-10064-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/16/2022]
Abstract
Neurodegeneration is a pathological condition that is associated with the loss of neuronal function and structure. In neurodegenerative diseases, mounting evidence indicates that neuroinflammation is a common factor that contributes to neuronal damage and neurodegeneration. Neuroinflammation is characterized by the activation of microglia, the neuroimmune cells of the central nervous system (CNS), which have been implicated as active contributors to neuronal damage. Glycan structure modification is defining the outcome of neuroinflammation and neuronal regeneration; moreover, the expression of galectins, a group of lectins that specifically recognize β-galactosides, has been proposed as a key factor in neuronal regeneration and modulation of the inflammatory response. Of the different galectins identified, galectin-1 stimulates the secretion of neurotrophic factors in astrocytes and promotes neuronal regeneration, whereas galectin-3 induces the proliferation of microglial cells and modulates cell apoptosis. Galectin-8 emerged as a neuroprotective factor, which, in addition to its immunosuppressive function, could generate a neuroprotective environment in the brain. This review describes the role of galectins in the activation and modulation of astrocytes and microglia and their anti- and proinflammatory functions within the context of neuroinflammation. Furthermore, it discusses the potential use of galectins as a therapeutic target for the inflammatory response and remodeling in damaged tissues in the central nervous system.
Collapse
Affiliation(s)
- Eleazar Ramírez Hernández
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | - Beatriz Alanis Olvera
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Daniela Carmona González
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Oscar Guerrero Marín
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Denisse Pantoja Mercado
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lucero Valencia Gil
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis F Hernández-Zimbrón
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Luis Sánchez Salgado
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - I Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de México, Mexico City, Mexico
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|