51
|
Lu C, Hyde DR. Cytokines IL-1β and IL-10 are required for Müller glia proliferation following light damage in the adult zebrafish retina. Front Cell Dev Biol 2024; 12:1406330. [PMID: 38938553 PMCID: PMC11208712 DOI: 10.3389/fcell.2024.1406330] [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: 03/24/2024] [Accepted: 05/16/2024] [Indexed: 06/29/2024] Open
Abstract
Zebrafish possess the ability to regenerate dying neurons in response to retinal injury, with both Müller glia and microglia playing integral roles in this response. Resident Müller glia respond to damage by reprogramming and undergoing an asymmetric cell division to generate a neuronal progenitor cell, which continues to proliferate and differentiate into the lost neurons. In contrast, microglia become reactive, phagocytose dying cells, and release inflammatory signals into the surrounding tissue following damage. In recent years, there has been increased attention on elucidating the role that microglia play in regulating retinal regeneration. Here we demonstrate that inflammatory cytokines are differentially expressed during retinal regeneration, with the expression of a subset of pro-inflammatory cytokine genes upregulated shortly after light damage and the expression of a different subset of cytokine genes subsequently increasing. We demonstrate that both cytokine IL-1β and IL-10 are essential for Müller glia proliferation in the light-damaged retina. While IL-1β is sufficient to induce Müller glia proliferation in an undamaged retina, expression of IL-10 in undamaged retinas only induces Müller glia to express gliotic markers. Together, these findings demonstrate the essential role of inflammatory cytokines IL-1β and IL-10 on Müller glia proliferation following light damage in adult zebrafish.
Collapse
Affiliation(s)
| | - David R. Hyde
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, and Center for Zebrafish Research, Galvin Life Sciences Building, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
52
|
Matt SM, Nolan R, Manikandan S, Agarwal Y, Channer B, Oteju O, Daniali M, Canagarajah JA, LuPone T, Mompho K, Runner K, Nickoloff-Bybel E, Li B, Niu M, Schlachetzki JCM, Fox HS, Gaskill PJ. Dopamine-driven Increase in IL-1β in Myeloid Cells is Mediated by Differential Dopamine Receptor Expression and Exacerbated by HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598137. [PMID: 38915663 PMCID: PMC11195146 DOI: 10.1101/2024.06.09.598137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The catecholamine neurotransmitter dopamine is classically known for regulation of central nervous system (CNS) functions such as reward, movement, and cognition. Increasing evidence also indicates that dopamine regulates critical functions in peripheral organs and is an important immunoregulatory factor. We have previously shown that dopamine increases NF-κB activity, inflammasome activation, and the production of inflammatory cytokines such as IL-1β in human macrophages. As myeloid lineage cells are central to the initiation and resolution of acute inflammatory responses, dopamine-mediated dysregulation of these functions could both impair the innate immune response and exacerbate chronic inflammation. However, the exact pathways by which dopamine drives myeloid inflammation are not well defined, and studies in both rodent and human systems indicate that dopamine can impact the production of inflammatory mediators through both D1-like dopamine receptors (DRD1, DRD5) and D2-like dopamine receptors (DRD2, DRD3, and DRD4). Therefore, we hypothesized that dopamine-mediated production of IL-1β in myeloid cells is regulated by the ratio of different dopamine receptors that are activated. Our data in primary human monocyte-derived macrophages (hMDM) indicate that DRD1 expression is necessary for dopamine-mediated increases in IL-1β, and that changes in the expression of DRD2 and other dopamine receptors can alter the magnitude of the dopamine-mediated increase in IL-1β. Mature hMDM have a high D1-like to D2-like receptor ratio, which is different relative to monocytes and peripheral blood mononuclear cells (PBMCs). We further confirm in human microglia cell lines that a high ratio of D1-like to D2-like receptors promotes dopamine-induced increases in IL-1β gene and protein expression using pharmacological inhibition or overexpression of dopamine receptors. RNA-sequencing of dopamine-treated microglia shows that genes encoding functions in IL-1β signaling pathways, microglia activation, and neurotransmission increased with dopamine treatment. Finally, using HIV as an example of a chronic inflammatory disease that is substantively worsened by comorbid substance use disorders (SUDs) that impact dopaminergic signaling, we show increased effects of dopamine on inflammasome activation and IL-1β in the presence of HIV in both human macrophages and microglia. These data suggest that use of addictive substances and dopamine-modulating therapeutics could dysregulate the innate inflammatory response and exacerbate chronic neuroimmunological conditions like HIV. Thus, a detailed understanding of dopamine-mediated changes in inflammation, in particular pathways regulating IL-1β, will be critical to effectively tailor medication regimens.
Collapse
|
53
|
Donahue CC, Resch JE. Concussion and the Sleeping Brain. SPORTS MEDICINE - OPEN 2024; 10:68. [PMID: 38853235 PMCID: PMC11162982 DOI: 10.1186/s40798-024-00736-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/25/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Emerging research has suggested sleep to be a modifier of the trajectory of concussion recovery in adolescent and adult populations. Despite the growing recognition of the relationship between sleep and concussion, the mechanisms and physiological processes governing this association have yet to be established. MAIN BODY Following a concussion, a pathophysiologic cascade of events occurs, characterized by numerous factors including microglia activation, ionic imbalance, and release of excitatory neurotransmitters. Importantly, each of these factors plays a role in the regulation of the sleep-wake cycle. Therefore, dysregulation of sleep following injury may be a function of the diffuse disruption of cerebral functioning in the wake of both axonal damage and secondary physiological events. As the onset of sleep-related symptoms is highly variable following a concussion, clinicians should be aware of when and how these symptoms present. Post-injury changes in sleep have been reported in the acute, sub-acute, and chronic phases of recovery and can prolong symptom resolution, affect neurocognitive performance, and influence mood state. Though these changes support sleep as a modifier of recovery, limited guidance exists for clinicians or their patients in the management of sleep after concussion. This may be attributed to the fact that research has correlated sleep with concussion recovery but has failed to explain why the correlation exists. Sleep is a complex, multifactorial process and the changes seen in sleep that are seen following concussion are the result of interactions amongst numerous processes that regulate the sleep-wake cycle. SHORT CONCLUSION The assessment and management of sleep by identifying and considering the biological, sociological, and psychological interactions of this multifactorial process will allow for clinicians to address the dynamic nature of changes in sleep following concussion.
Collapse
Affiliation(s)
- Catherine C Donahue
- Department of Orthopedics, University of Colorado School of Medicine, Children's Hospital Colorado, 13123 E. 16th Ave, Box 060, 80045, Aurora, CO, USA.
| | - Jacob E Resch
- Department of Kinesiology, University of Virginia, 550 Brandon Ave, Charlottesville, VA, 22908, USA
| |
Collapse
|
54
|
Chamera K, Curzytek K, Kamińska K, Leśkiewicz M, Basta-Kaim A. Prenatal Immune Challenge Differentiates the Effect of Aripiprazole and Risperidone on CD200-CD200R and CX3CL1-CX3CR1 Dyads and Microglial Polarization: A Study in Organotypic Cortical Cultures. Life (Basel) 2024; 14:721. [PMID: 38929704 PMCID: PMC11205240 DOI: 10.3390/life14060721] [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: 02/06/2024] [Revised: 04/20/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Microglia are the primary innate immune cells of the central nervous system and extensively contribute to brain homeostasis. Dysfunctional or excessive activity of microglia may be associated with several neuropsychiatric disorders, including schizophrenia. Therefore, we examined whether aripiprazole and risperidone could influence the expression of the Cd200-Cd200r and Cx3cl1-Cx3cr1 axes, which are crucial for the regulation of microglial activity and interactions of these cells with neurons. Additionally, we evaluated the impact of these drugs on microglial pro- and anti-inflammatory markers (Cd40, Il-1β, Il-6, Cebpb, Cd206, Arg1, Il-10 and Tgf-β) and cytokine release (IL-6, IL-10). The research was executed in organotypic cortical cultures (OCCs) prepared from the offspring of control rats (control OCCs) or those exposed to maternal immune activation (MIA OCCs), which allows for the exploration of schizophrenia-like disturbances in animals. All experiments were performed under basal conditions and after additional stimulation with lipopolysaccharide (LPS), following the "two-hit" hypothesis of schizophrenia. We found that MIA diminished the mRNA level of Cd200r and affected the OCCs' response to additional LPS exposure in terms of this parameter. LPS downregulated the Cx3cr1 expression and profoundly changed the mRNA levels of pro- and anti-inflammatory microglial markers in both types of OCCs. Risperidone increased Cd200 expression in MIA OCCs, while aripiprazole treatment elevated the gene levels of the Cx3cl1-Cx3cr1 dyad in control OCCs. The antipsychotics limited the LPS-generated increase in the expression of proinflammatory factors (Il-1β and Il-6) and enhanced the mRNA levels of anti-inflammatory components (Cd206 and Tgf-β) of microglial polarization, mostly in the absence of the MIA procedure. Finally, we observed a more pronounced modulating impact of aripiprazole on the expression of pro- and anti-inflammatory cytokines when compared to risperidone in MIA OCCs. In conclusion, our data suggest that MIA might influence microglial activation and crosstalk of microglial cells with neurons, whereas aripiprazole and risperidone could beneficially affect these changes in OCCs.
Collapse
Affiliation(s)
| | | | | | | | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| |
Collapse
|
55
|
Pereira-Santos AR, Candeias E, Magalhães JD, Empadinhas N, Esteves AR, Cardoso SM. Neuronal control of microglia through the mitochondria. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167167. [PMID: 38626829 DOI: 10.1016/j.bbadis.2024.167167] [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: 02/23/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
The microbial toxin β-N-methylamino-L-alanine (BMAA), which is derived from cyanobacteria, targets neuronal mitochondria, leading to the activation of neuronal innate immunity and, consequently, neurodegeneration. Although known to modulate brain inflammation, the precise role of aberrant microglial function in the neurodegenerative process remains elusive. To determine if neurons signal microglial cells, we treated primary cortical neurons with BMAA and then co-cultured them with the N9 microglial cell line. Our observations indicate that microglial cell activation requires initial neuronal priming. Contrary to what was observed in cortical neurons, BMAA was not able to activate inflammatory pathways in N9 cells. We observed that microglial activation is dependent on mitochondrial dysfunction signaled by BMAA-treated neurons. In this scenario, the NLRP3 pro-inflammatory pathway is activated due to mitochondrial impairment in N9 cells. These results demonstrate that microglia activation in the presence of BMAA is dependent on neuronal signaling. This study provides evidence that neurons may trigger microglia activation and subsequent neuroinflammation. In addition, we demonstrate that microglial activation may have a protective role in ameliorating neuronal innate immune activation, at least in the initial phase. This work challenges the current understanding of neuroinflammation by assigning the primary role to neurons.
Collapse
Affiliation(s)
- A R Pereira-Santos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - J D Magalhães
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Nuno Empadinhas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - A Raquel Esteves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Sandra M Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
56
|
Freeman C, A S MD, A S P. Unraveling the Intricacies of OPG/RANKL/RANK Biology and Its Implications in Neurological Disorders-A Comprehensive Literature Review. Mol Neurobiol 2024:10.1007/s12035-024-04227-z. [PMID: 38777981 DOI: 10.1007/s12035-024-04227-z] [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: 02/03/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
The OPG/RANKL/RANK framework, along with its specific receptors, plays a crucial role in bone remodeling and the functioning of the central nervous system (CNS) and associated disorders. Recent research and investigations provide evidence that the components of osteoprotegerin (OPG), receptor activator of NF-kB ligand (RANKL), and receptor activator of NF-kB (RANK) are expressed in the CNS. The CNS structure encompasses cells involved in neuroinflammation, including local macrophages, inflammatory cells, and microglia that cross the blood-brain barrier. The OPG/RANKL/RANK trio modulates the neuroinflammatory response based on the molecular context. The levels of OPG/RANKL/RANK components can serve as biomarkers in the blood and cerebrospinal fluid. They act as neuroprotectants following brain injuries and also participate in the regulation of body weight, internal body temperature, brain ischemia, autoimmune encephalopathy, and energy metabolism. Although the OPG/RANKL/RANK system is primarily known for its role in bone remodeling, further exploring deeper into its multifunctional nature can uncover new functions and novel drug targets for diseases not previously associated with OPG/RANKL/RANK signaling.
Collapse
Affiliation(s)
- Chrisanne Freeman
- Department of Biotechnology, Bishop Heber College, Tamil Nadu, Tiruchirappalli, 620017, India.
| | - Merlyn Diana A S
- Department of Biotechnology, Bishop Heber College, Tamil Nadu, Tiruchirappalli, 620017, India
- Department of Zoology and Research Centre, Lady Doak College, Tamil Nadu, Madurai, 625002, India
| | - Priscilla A S
- Department of Zoology and Research Centre, Lady Doak College, Tamil Nadu, Madurai, 625002, India
| |
Collapse
|
57
|
Munan S, Mondal A, Shailja S, Pati S, Samanta A. Unique Synthetic Strategy for Probing in Situ Lysosomal NO for Screening Neuroinflammatory Phenotypes against SARS-CoV-2 RNA in Phagocytotic Microglia. Anal Chem 2024; 96:7479-7486. [PMID: 38689560 DOI: 10.1021/acs.analchem.3c05981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
In the pathogenesis of microglia, brain immune cells promote nitrergic stress by overproducing nitric oxide (NO), leading to neuroinflammation. Furthermore, NO has been linked to COVID-19 progression, which has caused significant morbidity and mortality. SARS-CoV-2 infection activates inflammation by releasing excess NO and causing cell death in human microglial clone 3 (HMC3). In addition, NO regulates lysosomal functions and complex machinery to neutralize pathogens through phagocytosis. Therefore, developing lysosome-specific NO probes to monitor phagocytosis in microglia during the COVID-19 infection would be a significant study. Herein, a unique synthetic strategy was adopted to develop a NO selective fluorescent probe, PDM-NO, which can discriminate activated microglia from their resting state. The nonfluorescent PDM-NO exhibits a turn-on response toward NO only at lysosomal pH (4.5-5.5). Quantum chemical calculations (DFT/TD-DFT/PCM) and photophysical study revealed that the photoinduced electron transfer (PET) process is pivotal in tuning optical properties. PDM-NO demonstrated good biocompatibility and lysosomal specificity in activated HMC3 cells. Moreover, it can effectively map the dynamics of lysosomal NO against SARS-CoV-2 RNA-induced neuroinflammation in HMC3. Thus, PDM-NO is a potential fluorescent marker for detecting RNA virus infection and monitoring phagocytosis in HMC3.
Collapse
Affiliation(s)
- Subrata Munan
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University (SNIoE), Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| | - Abir Mondal
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University (SNIoE), Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| | - Singh Shailja
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Soumya Pati
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University (SNIoE), Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| | - Animesh Samanta
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University (SNIoE), Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| |
Collapse
|
58
|
Guadalupi L, Vanni V, Balletta S, Caioli S, De Vito F, Fresegna D, Sanna K, Nencini M, Donninelli G, Volpe E, Mariani F, Battistini L, Stampanoni Bassi M, Gilio L, Bruno A, Dolcetti E, Buttari F, Mandolesi G, Centonze D, Musella A. Interleukin-9 protects from microglia- and TNF-mediated synaptotoxicity in experimental multiple sclerosis. J Neuroinflammation 2024; 21:128. [PMID: 38745307 PMCID: PMC11092167 DOI: 10.1186/s12974-024-03120-9] [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: 02/09/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a progressive neurodegenerative disease of the central nervous system characterized by inflammation-driven synaptic abnormalities. Interleukin-9 (IL-9) is emerging as a pleiotropic cytokine involved in MS pathophysiology. METHODS Through biochemical, immunohistochemical, and electrophysiological experiments, we investigated the effects of both peripheral and central administration of IL-9 on C57/BL6 female mice with experimental autoimmune encephalomyelitis (EAE), a model of MS. RESULTS We demonstrated that both systemic and local administration of IL-9 significantly improved clinical disability, reduced neuroinflammation, and mitigated synaptic damage in EAE. The results unveil an unrecognized central effect of IL-9 against microglia- and TNF-mediated neuronal excitotoxicity. Two main mechanisms emerged: first, IL-9 modulated microglial inflammatory activity by enhancing the expression of the triggering receptor expressed on myeloid cells-2 (TREM2) and reducing TNF release. Second, IL-9 suppressed neuronal TNF signaling, thereby blocking its synaptotoxic effects. CONCLUSIONS The data presented in this work highlight IL-9 as a critical neuroprotective molecule capable of interfering with inflammatory synaptopathy in EAE. These findings open new avenues for treatments targeting the neurodegenerative damage associated with MS, as well as other inflammatory and neurodegenerative disorders of the central nervous system.
Collapse
Affiliation(s)
- Livia Guadalupi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Sara Balletta
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
| | - Silvia Caioli
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
| | | | - Diego Fresegna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Krizia Sanna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Monica Nencini
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Gloria Donninelli
- Molecular Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, Rome, 00143, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, Rome, 00143, Italy
| | - Fabrizio Mariani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Luca Battistini
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, Rome, 00143, Italy
| | | | - Luana Gilio
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
| | - Antonio Bruno
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
- Ph.D. Program in Neuroscience, Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Ettore Dolcetti
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
- Ph.D. Program in Neuroscience, Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Fabio Buttari
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, 00166, Italy
| | - Diego Centonze
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy.
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy.
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, 00166, Italy
| |
Collapse
|
59
|
Dócs K, Balázs A, Papp I, Szücs P, Hegyi Z. Reactive spinal glia convert 2-AG to prostaglandins to drive aberrant astroglial calcium signaling. Front Cell Neurosci 2024; 18:1382465. [PMID: 38784707 PMCID: PMC11112260 DOI: 10.3389/fncel.2024.1382465] [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: 02/05/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
The endogenous cannabinoid 2-arachidonoylglycerol (2-AG) influences neurotransmission in the central nervous system mainly by activating type 1 cannabinoid receptor (CB1). Following its release, 2-AG is broken down by hydrolases to yield arachidonic acid, which may subsequently be metabolized by cyclooxygenase-2 (COX-2). COX-2 converts arachidonic acid and also 2-AG into prostanoids, well-known inflammatory and pro-nociceptive mediators. Here, using immunohistochemical and biochemical methods and pharmacological manipulations, we found that reactive spinal astrocytes and microglia increase the expression of COX-2 and the production of prostaglandin E2 when exposed to 2-AG. Both 2-AG and PGE2 evoke calcium transients in spinal astrocytes, but PGE2 showed 30% more efficacy and 55 times more potency than 2-AG. Unstimulated spinal dorsal horn astrocytes responded to 2-AG with calcium transients mainly through the activation of CB1. 2-AG induced exaggerated calcium transients in reactive astrocytes, but this increase in the frequency and area under the curve of calcium signals was only partially dependent on CB1. Instead, aberrant calcium transients were almost completely abolished by COX-2 inhibition. Our results suggest that both reactive spinal astrocytes and microglia perform an endocannabinoid-prostanoid switch to produce PGE2 at the expense of 2-AG. PGE2 in turn is responsible for the induction of aberrant astroglial calcium signals which, together with PGE2 production may play role in the development and maintenance of spinal neuroinflammation-associated disturbances such as central sensitization.
Collapse
Affiliation(s)
- Klaudia Dócs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Balázs
- Department of Theoretical and Integrative Health Sciences, Institute of Health Sciences, Faculty of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Ildikó Papp
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Szücs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-DE Neuroscience Research Group, University of Debrecen, Debrecen, Hungary
| | - Zoltán Hegyi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
60
|
Schreiner TG, Schreiner OD, Ciobanu RC. Spinal Cord Injury Management Based on Microglia-Targeting Therapies. J Clin Med 2024; 13:2773. [PMID: 38792314 PMCID: PMC11122315 DOI: 10.3390/jcm13102773] [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: 04/19/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Spinal cord injury is a complicated medical condition both from the clinician's point of view in terms of management and from the patient's perspective in terms of unsatisfactory recovery. Depending on the severity, this disorder can be devastating despite the rapid and appropriate use of modern imaging techniques and convenient surgical spinal cord decompression and stabilization. In this context, there is a mandatory need for novel adjunctive therapeutic approaches to classical treatments to improve rehabilitation chances and clinical outcomes. This review offers a new and original perspective on therapies targeting the microglia, one of the most relevant immune cells implicated in spinal cord disorders. The first part of the manuscript reviews the anatomical and pathophysiological importance of the blood-spinal cord barrier components, including the role of microglia in post-acute neuroinflammation. Subsequently, the authors present the emerging therapies based on microglia modulation, such as cytokines modulators, stem cell, microRNA, and nanoparticle-based treatments that could positively impact spinal cord injury management. Finally, future perspectives and challenges are also highlighted based on the ongoing clinical trials related to medications targeting microglia.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Department of Medical Specialties III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
- First Neurology Clinic, “Prof. Dr. N. Oblu” Clinical Emergency Hospital, 700309 Iasi, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 700050 Iasi, Romania;
| | - Oliver Daniel Schreiner
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 700050 Iasi, Romania;
- Medical Oncology Department, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Romeo Cristian Ciobanu
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 700050 Iasi, Romania;
| |
Collapse
|
61
|
Serrano PDL, Rodrigues TDPV, Pinto LD, Pereira IC, Farias IB, Cavalheiro RBR, Mendes PM, Peixoto KO, Barile JP, Seneor DD, Correa Silva EG, Oliveira ASB, Pinto WBVDR, Sgobbi P. Assessing Chitinases and Neurofilament Light Chain as Biomarkers for Adult-Onset Leukodystrophies. Curr Issues Mol Biol 2024; 46:4309-4323. [PMID: 38785530 PMCID: PMC11120026 DOI: 10.3390/cimb46050262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Leukodystrophies represent a large and complex group of inherited disorders affecting the white matter of the central nervous system. Adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) is a rare leukodystrophy which still needs the proper identification of diagnostic, prognostic, and monitoring biomarkers. The aim of this study was to determine the diagnostic and prognostic value of chitinases and neurofilament light chain as biomarkers for ALSP. A cross-sectional study was performed to analyze cerebrospinal fluid levels of chitinases (chitotriosidase and chitinase 3-like 2) and neurofilament light chain in five different groups: (i) normal health individuals; (ii) patients with definitive diagnosis of ALSP and genetic confirmation; (iii) asymptomatic patients with CSF1R variants; (iv) patients with other adult-onset leukodystrophies; and (v) patients with amyotrophic lateral sclerosis (external control group). Chitinase levels showed a statistical correlation with clinical assessment parameters in ALSP patients. Chitinase levels were also distinct between ALSP and the other leukodystrophies. Significant differences were noted in the levels of chitinases and neurofilament light chain comparing symptomatic (ALSP) and asymptomatic individuals with CSF1R variants. This study is the first to establish chitinases as a potential biomarker for ALSP and confirms neurofilament light chain as a good biomarker for primary microgliopathies.
Collapse
Affiliation(s)
- Paulo de Lima Serrano
- PSEG Centro de Pesquisa Clínica, São Paulo 04038-002, SP, Brazil; (P.d.L.S.); (T.d.P.V.R.); (L.D.P.); (I.C.P.); (E.G.C.S.)
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | | | - Leslyê Donato Pinto
- PSEG Centro de Pesquisa Clínica, São Paulo 04038-002, SP, Brazil; (P.d.L.S.); (T.d.P.V.R.); (L.D.P.); (I.C.P.); (E.G.C.S.)
| | - Indiara Correia Pereira
- PSEG Centro de Pesquisa Clínica, São Paulo 04038-002, SP, Brazil; (P.d.L.S.); (T.d.P.V.R.); (L.D.P.); (I.C.P.); (E.G.C.S.)
| | - Igor Braga Farias
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Renan Brandão Rambaldi Cavalheiro
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Patrícia Marques Mendes
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Kaliny Oliveira Peixoto
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - João Paulo Barile
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Daniel Delgado Seneor
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | | | - Acary Souza Bulle Oliveira
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Wladimir Bocca Vieira de Rezende Pinto
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Paulo Sgobbi
- PSEG Centro de Pesquisa Clínica, São Paulo 04038-002, SP, Brazil; (P.d.L.S.); (T.d.P.V.R.); (L.D.P.); (I.C.P.); (E.G.C.S.)
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| |
Collapse
|
62
|
Mannino F, Urzì Brancati V, Lauro R, Pirrotta I, Rottura M, Irrera N, Cavallini GM, Pallio G, Gitto E, Manti S. Levosimendan and Dobutamin Attenuate LPS-Induced Inflammation in Microglia by Inhibiting the NF-κB Pathway and NLRP3 Inflammasome Activation via Nrf2/HO-1 Signalling. Biomedicines 2024; 12:1009. [PMID: 38790971 PMCID: PMC11117907 DOI: 10.3390/biomedicines12051009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Hypovolemic shock is a circulatory failure, due to a loss in the effective circulating blood volume, that causes tissue hypoperfusion and hypoxia. This condition stimulates reactive oxygen species (ROS) and pro-inflammatory cytokine production in different organs and also in the central nervous system (CNS). Levosimendan, a cardioprotective inodilator, and dobutamine, a β1-adrenergic agonist, are commonly used for the treatment of hypovolemic shock, thanks to their anti-inflammatory and antioxidant effects. For this reason, we aimed at investigating levosimendan and dobutamine's neuroprotective effects in an "in vitro" model of lipopolysaccharide (LPS)-induced neuroinflammation. Human microglial cells (HMC3) were challenged with LPS (0.1 µg/mL) to induce an inflammatory phenotype and then treated with levosimendan (10 µM) or dobutamine (50 µM) for 24 h. Levosimendan and dobutamine significantly reduced the ROS levels and markedly increased Nrf2 and HO-1 protein expression in LPS-challenged cells. Levosimendan and dobutamine also decreased p-NF-κB expression and turned off the NLRP3 inflammasome together with its downstream signals, caspase-1 and IL-1β. Moreover, a reduction in TNF-α and IL-6 expression and an increase in IL-10 levels in LPS-stimulated HMC3 cells was observed following treatment. In conclusion, levosimendan and dobutamine attenuated LPS-induced neuroinflammation through NF-κB pathway inhibition and NLRP3 inflammasome activation via Nrf2/HO-1 signalling, suggesting that these drugs could represent a promising therapeutic approach for the treatment of neuroinflammation consequent to hypovolemic shock.
Collapse
Affiliation(s)
- Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (F.M.); (V.U.B.); (R.L.); (I.P.); (M.R.); (N.I.); (E.G.)
| | - Valentina Urzì Brancati
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (F.M.); (V.U.B.); (R.L.); (I.P.); (M.R.); (N.I.); (E.G.)
| | - Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (F.M.); (V.U.B.); (R.L.); (I.P.); (M.R.); (N.I.); (E.G.)
| | - Igor Pirrotta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (F.M.); (V.U.B.); (R.L.); (I.P.); (M.R.); (N.I.); (E.G.)
| | - Michelangelo Rottura
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (F.M.); (V.U.B.); (R.L.); (I.P.); (M.R.); (N.I.); (E.G.)
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (F.M.); (V.U.B.); (R.L.); (I.P.); (M.R.); (N.I.); (E.G.)
| | - Gian Maria Cavallini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| | - Giovanni Pallio
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Eloisa Gitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (F.M.); (V.U.B.); (R.L.); (I.P.); (M.R.); (N.I.); (E.G.)
| | - Sara Manti
- Department of Human Pathology of Adult and Childhood Gaetano Barresi, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
63
|
Kong W, Frouard J, Xie G, Corley MJ, Helmy E, Zhang G, Schwarzer R, Montano M, Sohn P, Roan NR, Ndhlovu LC, Gan L, Greene WC. Neuroinflammation generated by HIV-infected microglia promotes dysfunction and death of neurons in human brain organoids. PNAS NEXUS 2024; 3:pgae179. [PMID: 38737767 PMCID: PMC11086946 DOI: 10.1093/pnasnexus/pgae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/17/2024] [Indexed: 05/14/2024]
Abstract
Despite the success of combination antiretroviral therapy (ART) for individuals living with HIV, mild forms of HIV-associated neurocognitive disorder (HAND) continue to occur. Brain microglia form the principal target for HIV infection in the brain. It remains unknown how infection of these cells leads to neuroinflammation, neuronal dysfunction, and/or death observed in HAND. Utilizing two different inducible pluripotent stem cell-derived brain organoid models (cerebral and choroid plexus [ChP] organoids) containing microglia, we investigated the pathogenic changes associated with HIV infection. Infection of microglia was associated with a sharp increase in CCL2 and CXCL10 chemokine gene expression and the activation of many type I interferon stimulated genes (MX1, ISG15, ISG20, IFI27, IFITM3 and others). Production of the proinflammatory chemokines persisted at low levels after treatment of the cell cultures with ART, consistent with the persistence of mild HAND following clinical introduction of ART. Expression of multiple members of the S100 family of inflammatory genes sharply increased following HIV infection of microglia measured by single-cell RNA-seq. However, S100 gene expression was not limited to microglia but was also detected more broadly in uninfected stromal cells, mature and immature ChP cells, neural progenitor cells and importantly in bystander neurons suggesting propagation of the inflammatory response to bystander cells. Neurotransmitter transporter expression declined in uninfected neurons, accompanied by increased expression of genes promoting cellular senescence and cell death. Together, these studies underscore how an inflammatory response generated in HIV-infected microglia is propagated to multiple uninfected bystander cells ultimately resulting in the dysfunction and death of bystander neurons.
Collapse
Affiliation(s)
- Weili Kong
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Julie Frouard
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Guorui Xie
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael J Corley
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ekram Helmy
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Gang Zhang
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Roland Schwarzer
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Mauricio Montano
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Peter Sohn
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Nadia R Roan
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lishomwa C Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Li Gan
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Warner C Greene
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
- Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
64
|
Albadrani HM, Chauhan P, Ashique S, Babu MA, Iqbal D, Almutary AG, Abomughaid MM, Kamal M, Paiva-Santos AC, Alsaweed M, Hamed M, Sachdeva P, Dewanjee S, Jha SK, Ojha S, Slama P, Jha NK. Mechanistic insights into the potential role of dietary polyphenols and their nanoformulation in the management of Alzheimer's disease. Biomed Pharmacother 2024; 174:116376. [PMID: 38508080 DOI: 10.1016/j.biopha.2024.116376] [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: 06/22/2023] [Revised: 01/19/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Alzheimer's disease (AD) is a very common neurodegenerative disorder associated with memory loss and a progressive decline in cognitive activity. The two major pathophysiological factors responsible for AD are amyloid plaques (comprising amyloid-beta aggregates) and neurofibrillary tangles (consisting of hyperphosphorylated tau protein). Polyphenols, a class of naturally occurring compounds, are immensely beneficial for the treatment or management of various disorders and illnesses. Naturally occurring sources of polyphenols include plants and plant-based foods, such as fruits, herbs, tea, vegetables, coffee, red wine, and dark chocolate. Polyphenols have unique properties, such as being the major source of anti-oxidants and possessing anti-aging and anti-cancerous properties. Currently, dietary polyphenols have become a potential therapeutic approach for the management of AD, depending on various research findings. Dietary polyphenols can be an effective strategy to tackle multifactorial events that occur with AD. For instance, naturally occurring polyphenols have been reported to exhibit neuroprotection by modulating the Aβ biogenesis pathway in AD. Many nanoformulations have been established to enhance the bioavailability of polyphenols, with nanonization being the most promising. This review comprehensively provides mechanistic insights into the neuroprotective potential of dietary polyphenols in treating AD. It also reviews the usability of dietary polyphenol as nanoformulation for AD treatment.
Collapse
Affiliation(s)
- Hind Muteb Albadrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province 34212, Saudi Arabia
| | - Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanad University, Rohtak, Haryana 124001, India
| | - Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Mohammed Alsaweed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Munerah Hamed
- Department of Pathology, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | | | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura- 140401, Punjab, India.; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, India.
| |
Collapse
|
65
|
Sternberg Z. Neurodegenerative Etiology of Aromatic L-Amino Acid Decarboxylase Deficiency: a Novel Concept for Expanding Treatment Strategies. Mol Neurobiol 2024; 61:2996-3018. [PMID: 37953352 DOI: 10.1007/s12035-023-03684-2] [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: 02/07/2023] [Accepted: 09/29/2023] [Indexed: 11/14/2023]
Abstract
Aromatic l-amino acid decarboxylase deficiency (AADC-DY) is caused by one or more mutations in the DDC gene, resulting in the deficit in catecholamines and serotonin neurotransmitters. The disease has limited therapeutic options with relatively poor clinical outcomes. Accumulated evidence suggests the involvement of neurodegenerative mechanisms in the etiology of AADC-DY. In the absence of neurotransmitters' neuroprotective effects, the accumulation and the chronic presence of several neurotoxic metabolites including 4-dihydroxy-L-phenylalanine, 3-methyldopa, and homocysteine, in the brain of subjects with AADC-DY, promote oxidative stress and reduce the cellular antioxidant and methylation capacities, leading to glial activation and mitochondrial dysfunction, culminating to neuronal injury and death. These pathophysiological processes have the potential to hinder the clinical efficacy of treatments aimed at increasing neurotransmitters' synthesis and or function. This review describes in detail the mechanisms involved in AADC-DY neurodegenerative etiology, highlighting the close similarities with those involved in other neurodegenerative diseases. We then offer novel strategies for the treatment of the disease with the objective to either reduce the level of the metabolites or counteract their prooxidant and neurotoxic effects. These treatment modalities used singly or in combination, early in the course of the disease, will minimize neuronal injury, preserving the functional integrity of neurons, hence improving the clinical outcomes of both conventional and unconventional interventions in AADC-DY. These modalities may not be limited to AADC-DY but also to other metabolic disorders where a specific mutation leads to the accumulation of prooxidant and neurotoxic metabolites.
Collapse
Affiliation(s)
- Zohi Sternberg
- Jacobs School of Medicine and Biomedical Sciences, Buffalo Medical Center, Buffalo, NY, 14203, USA.
| |
Collapse
|
66
|
Khan Z, Mehan S, Gupta GD, Narula AS. Immune System Dysregulation in the Progression of Multiple Sclerosis: Molecular Insights and Therapeutic Implications. Neuroscience 2024; 548:9-26. [PMID: 38692349 DOI: 10.1016/j.neuroscience.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 05/03/2024]
Abstract
Multiple sclerosis (MS), a prevalent neurological disorder, predominantly affects young adults and is characterized by chronic autoimmune activity. The study explores the immune system dysregulation in MS, highlighting the crucial roles of immune and non-neuronal cells in the disease's progression. This review examines the dual role of cytokines, with some like IL-6, TNF-α, and interferon-gamma (IFN-γ) promoting inflammation and CNS tissue injury, and others such as IL-4, IL-10, IL-37, and TGF-β fostering remyelination and protecting against MS. Elevated chemokine levels in the cerebrospinal fluid (CSF), including CCL2, CCL5, CXCL10, CXCL13, and fractalkine, are analyzed for their role in facilitating immune cell migration across the blood-brain barrier (BBB), worsening inflammation and neurodegeneration. The study also delves into the impact of auto-antibodies targeting myelin components like MOG and AQP4, which activate complement cascades leading to further myelin destruction. The article discusses how compromised BBB integrity allows immune cells and inflammatory mediators to infiltrate the CNS, intensifying MS symptoms. It also examines the involvement of astrocytes, microglia, and oligodendrocytes in the disease's progression. Additionally, the effectiveness of immunomodulatory drugs such as IFN-β and CD20-targeting monoclonal antibodies (e.g., rituximab) in modulating immune responses is reviewed, highlighting their potential to reduce relapse rates and delaying MS progression. These insights emphasize the importance of immune system dysfunction in MS development and progression, guiding the development of new therapeutic strategies. The study underscores recent advancements in understanding MS's molecular pathways, opening avenues for more targeted and effective treatments.
Collapse
Affiliation(s)
- Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga 142001, Punjab, India.
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
67
|
Arvanitaki ES, Goulielmaki E, Gkirtzimanaki K, Niotis G, Tsakani E, Nenedaki E, Rouska I, Kefalogianni M, Xydias D, Kalafatakis I, Psilodimitrakopoulos S, Karagogeos D, Schumacher B, Stratakis E, Garinis GA. Microglia-derived extracellular vesicles trigger age-related neurodegeneration upon DNA damage. Proc Natl Acad Sci U S A 2024; 121:e2317402121. [PMID: 38635632 PMCID: PMC11047102 DOI: 10.1073/pnas.2317402121] [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: 10/09/2023] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
DNA damage and neurodegenerative disorders are intimately linked but the underlying mechanism remains elusive. Here, we show that persistent DNA lesions in tissue-resident macrophages carrying an XPF-ERCC1 DNA repair defect trigger neuroinflammation and neuronal cell death in mice. We find that microglia accumulate dsDNAs and chromatin fragments in the cytosol, which are sensed thereby stimulating a viral-like immune response in Er1Cx/- and naturally aged murine brain. Cytosolic DNAs are packaged into extracellular vesicles (EVs) that are released from microglia and discharge their dsDNA cargo into IFN-responsive neurons triggering cell death. To remove cytosolic dsDNAs and prevent inflammation, we developed targeting EVs to deliver recombinant DNase I to Er1Cx/- brain microglia in vivo. We show that EV-mediated elimination of cytosolic dsDNAs is sufficient to prevent neuroinflammation, reduce neuronal apoptosis, and delay the onset of neurodegenerative symptoms in Er1Cx/- mice. Together, our findings unveil a causal mechanism leading to neuroinflammation and provide a rationalized therapeutic strategy against age-related neurodegeneration.
Collapse
Affiliation(s)
- Ermioni S. Arvanitaki
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Evi Goulielmaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Katerina Gkirtzimanaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - George Niotis
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Edisona Tsakani
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Electra Nenedaki
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Iliana Rouska
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Mary Kefalogianni
- Department of Physics, University of Crete, HeraklionGR71003, Crete, Greece
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - Dionysios Xydias
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
- Materials Science and Technology Department, University of Crete, HeraklionGR70013, Crete, Greece
| | - Ilias Kalafatakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
- Medical School, Division of Basic Sciences, University of Crete, HeraklionGR71003, Crete, Greece
| | - Sotiris Psilodimitrakopoulos
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - Domna Karagogeos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
- Medical School, Division of Basic Sciences, University of Crete, HeraklionGR71003, Crete, Greece
| | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University and University Hospital of Cologne, Cologne50931, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne50931, Germany
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - George A. Garinis
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| |
Collapse
|
68
|
Plachokova AS, Gjaltema J, Hagens ERC, Hashemi Z, Knüppe TBA, Kootstra TJM, Visser A, Bloem BR. Periodontitis: A Plausible Modifiable Risk Factor for Neurodegenerative Diseases? A Comprehensive Review. Int J Mol Sci 2024; 25:4504. [PMID: 38674088 PMCID: PMC11050498 DOI: 10.3390/ijms25084504] [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: 02/27/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
The aim of this comprehensive review is to summarize recent literature on associations between periodontitis and neurodegenerative diseases, explore the bidirectionality and provide insights into the plausible pathogenesis. For this purpose, systematic reviews and meta-analyses from PubMed, Medline and EMBASE were considered. Out of 33 retrieved papers, 6 articles complying with the inclusion criteria were selected and discussed. Additional relevant papers for bidirectionality and pathogenesis were included. Results show an association between periodontitis and Alzheimer's disease, with odds ratios of 3 to 5. A bidirectional relationship is suspected. For Parkinson's disease (PD), current evidence for an association appears to be weak, although poor oral health and PD seem to be correlated. A huge knowledge gap was identified. The plausible mechanistic link for the association between periodontitis and neurodegenerative diseases is the interplay between periodontal inflammation and neuroinflammation. Three pathways are hypothesized in the literature, i.e., humoral, neuronal and cellular, with a clear role of periodontal pathogens, such as Porphyromonas gingivalis. Age, gender, race, smoking, alcohol intake, nutrition, physical activity, socioeconomic status, stress, medical comorbidities and genetics were identified as common risk factors for periodontitis and neurodegenerative diseases. Future research with main emphasis on the collaboration between neurologists and dentists is encouraged.
Collapse
Affiliation(s)
- Adelina S. Plachokova
- Department of Dentistry, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands; (J.G.); (E.R.C.H.); (Z.H.); (T.B.A.K.); (T.J.M.K.)
| | - Jolijn Gjaltema
- Department of Dentistry, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands; (J.G.); (E.R.C.H.); (Z.H.); (T.B.A.K.); (T.J.M.K.)
| | - Eliza R. C. Hagens
- Department of Dentistry, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands; (J.G.); (E.R.C.H.); (Z.H.); (T.B.A.K.); (T.J.M.K.)
| | - Zahra Hashemi
- Department of Dentistry, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands; (J.G.); (E.R.C.H.); (Z.H.); (T.B.A.K.); (T.J.M.K.)
| | - Tim B. A. Knüppe
- Department of Dentistry, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands; (J.G.); (E.R.C.H.); (Z.H.); (T.B.A.K.); (T.J.M.K.)
| | - Thomas J. M. Kootstra
- Department of Dentistry, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands; (J.G.); (E.R.C.H.); (Z.H.); (T.B.A.K.); (T.J.M.K.)
| | - Anita Visser
- Department of Dentistry, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands; (J.G.); (E.R.C.H.); (Z.H.); (T.B.A.K.); (T.J.M.K.)
- Department of Gerodontology, Center for Dentistry and Oral Hygiene, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Bastiaan R. Bloem
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behavior, Department of Neurology, Centre of Expertise for Parkinson and Movement Disorders, 6525 GA Nijmegen, The Netherlands;
| |
Collapse
|
69
|
Cogut V, Goris M, Jansma A, van der Staaij M, Henning RH. Hippocampal neuroimmune response in mice undergoing serial daily torpor induced by calorie restriction. Front Neuroanat 2024; 18:1334206. [PMID: 38686173 PMCID: PMC11056553 DOI: 10.3389/fnana.2024.1334206] [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: 11/06/2023] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
Hibernating animals demonstrate a remarkable ability to withstand extreme physiological brain changes without triggering adverse neuroinflammatory responses. While hibernators may offer valuable insights into the neuroprotective mechanisms inherent to hibernation, studies using such species are constrained by the limited availability of molecular tools. Laboratory mice may serve as an alternative, entering states of hypometabolism and hypothermia similar to the torpor observed in hibernation when faced with energy shortage. Notably, prolonged calorie restriction (CR) induces serial daily torpor patterns in mice, comparable to species that utilize daily hibernation. Here, we examined the neuroinflammatory response in the hippocampus of male C57BL/6 mice undergoing serial daily torpor induced by a 30% CR for 4 weeks. During daily torpor episodes, CR mice exhibited transient increases in TNF-α mRNA expression, which normalized upon arousal. Concurrently, the CA1 region of the hippocampus showed persistent morphological changes in microglia, characterized by reduced cell branching, decreased cell complexity and altered shape. Importantly, these morphological changes were not accompanied by evident signs of astrogliosis or oxidative stress, typically associated with detrimental neuroinflammation. Collectively, the adaptive nature of the brain's inflammatory response to CR-induced torpor in mice parallels observations in hibernators, highlighting its value for studying the mechanisms of brain resilience during torpor. Such insights could pave the way for novel therapeutic interventions in stroke and neurodegenerative disorders in humans.
Collapse
Affiliation(s)
- Valeria Cogut
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
| | | | | | | | | |
Collapse
|
70
|
Hedley KE, Cuskelly A, Callister RJ, Horvat JC, Hodgson DM, Tadros MA. The medulla oblongata shows a sex-specific inflammatory response to systemic neonatal lipopolysaccharide. J Neuroimmunol 2024; 389:578316. [PMID: 38394966 DOI: 10.1016/j.jneuroim.2024.578316] [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: 10/19/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
Early life inflammation has been linked to long-term modulation of behavioural outcomes due to the central nervous system, but it is now becoming apparent it is also linked to dysfunction of visceral physiology. The medulla oblongata contains a number of nuclei critical for homeostasis, therefore we utilised the well-established model of neonatal lipopolysaccharide (LPS) exposure to examine the immediate and long-term impacts of systemic inflammation on the medulla oblongata. Wistar rats were injected with LPS or saline on postnatal days 3 and 5, with tissues collected on postnatal days 7 or 90 in order to assess expression of inflammatory mediators and microglial morphology in autonomic regions of the medulla oblongata. We observed a distinct sex-specific response of all measured inflammatory mediators at both ages, as well as significant neonatal sex differences in inflammatory mediators within saline groups. At both ages, microglial morphology had significant changes in branch length and soma size in a sex-specific manner in response to LPS exposure. This data not only highlights the strong sex-specific response of neonates to LPS administration, but also the significant life-long impact on the medulla oblongata and the potential altered control of visceral organs.
Collapse
Affiliation(s)
- Kateleen E Hedley
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Annalisa Cuskelly
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Psychological Sciences, University of Newcastle, NSW, Australia; School of Education, University of Newcastle, NSW, Australia
| | - Robert J Callister
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jay C Horvat
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Deborah M Hodgson
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Psychological Sciences, University of Newcastle, NSW, Australia
| | - Melissa A Tadros
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
71
|
Kim DW, Lee TK, Ahn JH, Yang SR, Shin MC, Cho JH, Won MH, Kang IJ, Park JH. Porphyran Attenuates Neuronal Loss in the Hippocampal CA1 Subregion Induced by Ischemia and Reperfusion in Gerbils by Inhibiting NLRP3 Inflammasome-Mediated Neuroinflammation. Mar Drugs 2024; 22:170. [PMID: 38667787 PMCID: PMC11050983 DOI: 10.3390/md22040170] [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: 03/19/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Porphyran, a sulfated polysaccharide found in various species of marine red algae, has been demonstrated to exhibit diverse bioactivities, including anti-inflammatory effects. However, the protective effects of porphyran against cerebral ischemia and reperfusion (IR) injury have not been investigated. The aim of this study was to examine the neuroprotective effects of porphyran against brain IR injury and its underlying mechanisms using a gerbil model of transient forebrain ischemia (IR in the forebrain), which results in pyramidal cell (principal neuron) loss in the cornu ammonis 1 (CA1) subregion of the hippocampus on day 4 after IR. Porphyran (25 and 50 mg/kg) was orally administered daily for one week prior to IR. Pretreatment with 50 mg/kg of porphyran, but not 25 mg/kg, significantly attenuated locomotor hyperactivity and protected pyramidal cells located in the CA1 area from IR injury. The pretreatment with 50 mg/kg of porphyran significantly suppressed the IR-induced activation and proliferation of microglia in the CA1 subregion. Additionally, the pretreatment significantly inhibited the overexpressions of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing protein-3 (NLRP3) inflammasome complex, and pro-inflammatory cytokines (interleukin 1 beta and interleukin 18) induced by IR in the CA1 subregion. Overall, our findings suggest that porphyran exerts neuroprotective effects against brain IR injury, potentially by reducing the reaction (activation) and proliferation of microglia and reducing NLRP3 inflammasome-mediated neuroinflammation.
Collapse
Affiliation(s)
- Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
| | - Tae-Kyeong Lee
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea;
| | - Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan 50510, Republic of Korea;
| | - Se-Ran Yang
- Department of Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea; (M.C.S.); (J.H.C.); (M.-H.W.)
| | - Jun Hwi Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea; (M.C.S.); (J.H.C.); (M.-H.W.)
| | - Moo-Ho Won
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea; (M.C.S.); (J.H.C.); (M.-H.W.)
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea;
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
| |
Collapse
|
72
|
Holcomb M, Marshall A, Flinn H, Lozano M, Soriano S, Gomez-Pinilla F, Treangen TJ, Villapol S. Probiotic treatment causes sex-specific neuroprotection after traumatic brain injury in mice. RESEARCH SQUARE 2024:rs.3.rs-4196801. [PMID: 38645104 PMCID: PMC11030542 DOI: 10.21203/rs.3.rs-4196801/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background Recent studies have shed light on the potential role of gut dysbiosis in shaping traumatic brain injury (TBI) outcomes. Changes in the levels and types of Lactobacillus bacteria present might impact the immune system disturbances, neuroinflammatory responses, anxiety and depressive-like behaviors, and compromised neuroprotection mechanisms triggered by TBI. Objective This study aimed to investigate the effects of a daily pan-probiotic (PP) mixture in drinking water containing strains of Lactobacillus plantarum, L. reuteri, L. helveticus, L. fermentum, L. rhamnosus, L. gasseri, and L. casei, administered for either two or seven weeks before inducing TBI on both male and female mice. Methods Mice were subjected to controlled cortical impact (CCI) injury. Short-chain fatty acids (SCFAs) analysis was performed for metabolite measurements. The taxonomic profiles of murine fecal samples were evaluated using 16S rRNA V1-V3 sequencing analysis. Histological analyses were used to assess neuroinflammation and gut changes post-TBI, while behavioral tests were conducted to evaluate sensorimotor and cognitive functions. Results Our findings suggest that PP administration modulates the diversity and composition of the microbiome and increases the levels of SCFAs in a sex-dependent manner. We also observed a reduction of lesion volume, cell death, and microglial and macrophage activation after PP treatment following TBI in male mice. Furthermore, PP-treated mice show motor function improvements and decreases in anxiety and depressive-like behaviors. Conclusion Our findings suggest that PP administration can mitigate neuroinflammation and ameliorate motor and anxiety and depressive-like behavior deficits following TBI. These results underscore the potential of probiotic interventions as a viable therapeutic strategy to address TBI-induced impairments, emphasizing the need for gender-specific treatment approaches.
Collapse
|
73
|
Chen MS, Hu CL, Jiang SK, Chong ZY, Chen JC. Modulation of secretory factors by lipofundin contributes to its anti‑neuroinflammatory effects. Exp Ther Med 2024; 27:169. [PMID: 38476917 PMCID: PMC10929000 DOI: 10.3892/etm.2024.12456] [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: 10/02/2023] [Accepted: 01/16/2024] [Indexed: 03/14/2024] Open
Abstract
As the global population ages, the prevalence of neuroinflammatory diseases such as Alzheimer's disease, Parkinson's disease and stroke continues to increase. Therefore, it is necessary to develop preventive and therapeutic methods against neuroinflammatory diseases. Lipofundin is a lipid emulsion commonly used in clinical anesthetic solvents and nutritional supplements. Lipid emulsions have been shown to possess anti-inflammatory properties. However, the potential beneficial effect of lipofundin against neuroinflammation requires elucidation. In the present study, two cell models were used to investigate the efficacy of lipofundin against neuroinflammation. In the first model, BV2 mouse microglial cells were treated with lipopolysaccharide (LPS) to induce nitric oxide (NO) production as a model of neuroinflammation. In the second model, HMC3 human microglial were activated by LPS, and changes in the secretion of factors associated with inflammation were analyzed using Luminex xMAP® technology. Griess assay results revealed that lipofundin significantly prevented and treated LPS-induced NO production. An anti-neuroinflammatory effect was also observed in HMC3 cells, where lipofundin exhibited excellent preventive and therapeutic properties by reducing the LPS-induced expression and secretion of interleukin-1β. Notably, lipofundin also promoted the secretion of certain growth factors, suggesting a potential neuroprotective effect. These results demonstrate that, in addition to its role as a solvent for drugs and nutritional support, lipofundin may also have beneficial effects in alleviating the progression of neuroinflammation. These findings may serve as an important reference for future translational medicine applications.
Collapse
Affiliation(s)
- Ming-Shan Chen
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan, R.O.C
| | - Chia-Lin Hu
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Shin-Kuang Jiang
- Department of Neurology, China Medical University Hospital, Taichung 404332, Taiwan, R.O.C
| | - Zhi-Yong Chong
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan, R.O.C
| | - Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan, R.O.C
| |
Collapse
|
74
|
Umeda T, Shigemori K, Uekado R, Matsuda K, Tomiyama T. Hawaiian native herb Mamaki prevents dementia by ameliorating neuropathology and repairing neurons in four different mouse models of neurodegenerative diseases. GeroScience 2024; 46:1971-1987. [PMID: 37783918 PMCID: PMC10828292 DOI: 10.1007/s11357-023-00950-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023] Open
Abstract
Neurodegenerative diseases including Alzheimer's disease, frontotemporal dementia, and dementia with Lewy bodies are age-related disorders and the main cause of dementia. They are characterized by the cerebral accumulation of Aβ, tau, α-synuclein, and TDP-43. Because the accumulation begins decades before disease onset, treatment should be started in the preclinical stage. Such intervention would be long-lasting, and therefore, prophylactic agents should be safe, non-invasively taken by the patients, and inexpensive. In addition, the agents should be broadly effective against etiologic proteins and capable of repairing neurons damaged by toxic oligomers. These requirements are difficult to meet with single-ingredient pharmaceuticals but may be feasible by taking proper diets composed of multiple ingredients. As a source of such diets, we focused on the Hawaiian native herb Mamaki. From its dried leaves and fruits, we made three preparations: hot water extract of the leaves, non-extracted simple crush powder of the leaves, and simple crush powder of the fruits, and examined their effects on the cognitive function and neuropathologies in four different mouse models of neurodegenerative dementia. Hot water extract of the leaves attenuated neuropathologies, restored synaptophysin levels, suppressed microglial activation, and improved memory when orally administered for 1 month. Simply crushed leaf powder showed a higher efficacy, but simply crushed fruit powder displayed the strongest effects. Moreover, the fruit powder significantly enhanced the levels of brain-derived neurotrophic factor expression and neurogenesis, indicating its ability to repair neurons. These results suggest that crushed Mamaki leaves and fruits are promising sources of dementia-preventive diets.
Collapse
Affiliation(s)
- Tomohiro Umeda
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Osaka, Abeno-ku, 545-8585, Japan
- Cerebro Pharma Inc, 4-5-6-3F Minamikyuhojimachi, Osaka, Chuo-ku, 541-0058, Japan
| | - Keiko Shigemori
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Osaka, Abeno-ku, 545-8585, Japan
| | - Rumi Uekado
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Osaka, Abeno-ku, 545-8585, Japan
| | - Kazunori Matsuda
- Cerebro Pharma Inc, 4-5-6-3F Minamikyuhojimachi, Osaka, Chuo-ku, 541-0058, Japan
| | - Takami Tomiyama
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Osaka, Abeno-ku, 545-8585, Japan.
- Cerebro Pharma Inc, 4-5-6-3F Minamikyuhojimachi, Osaka, Chuo-ku, 541-0058, Japan.
| |
Collapse
|
75
|
Dong H, Zhang X, Duan Y, He Y, Zhao J, Wang Z, Wang J, Li Q, Fan G, Liu Z, Shen C, Zhang Y, Yu M, Fei J, Huang F. Hypoxia inducible factor-1α regulates microglial innate immune memory and the pathology of Parkinson's disease. J Neuroinflammation 2024; 21:80. [PMID: 38555419 PMCID: PMC10981320 DOI: 10.1186/s12974-024-03070-2] [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: 12/18/2023] [Accepted: 03/20/2024] [Indexed: 04/02/2024] Open
Abstract
Neuroinflammation is one of the core pathological features of Parkinson's disease (PD). Innate immune cells play a crucial role in the progression of PD. Microglia, the major innate immune cells in the brain, exhibit innate immune memory effects and are recognized as key regulators of neuroinflammatory responses. Persistent modifications of microglia provoked by the first stimuli are pivotal for innate immune memory, resulting in an enhanced or suppressed immune response to second stimuli, which is known as innate immune training and innate immune tolerance, respectively. In this study, LPS was used to establish in vitro and in vivo models of innate immune memory. Microglia-specific Hif-1α knockout mice were further employed to elucidate the regulatory role of HIF-1α in innate immune memory and MPTP-induced PD pathology. Our results showed that different paradigms of LPS could induce innate immune training or tolerance in the nigrostriatal pathway of mice. We found that innate immune tolerance lasting for one month protected the dopaminergic system in PD mice, whereas the effect of innate immune training was limited. Deficiency of HIF-1α in microglia impeded the formation of innate immune memory and exerted protective effects in MPTP-intoxicated mice by suppressing neuroinflammation. Therefore, HIF-1α is essential for microglial innate immune memory and can promote neuroinflammation associated with PD.
Collapse
Affiliation(s)
- Hongtian Dong
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yufei Duan
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yongtao He
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jiayin Zhao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Zishan Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jinghui Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Qing Li
- School of Life Science and Technology, Tongji University, 1239 Shipping Road, Shanghai, 200092, China
| | - Guangchun Fan
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Zhaolin Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Chenye Shen
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yunhe Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Shipping Road, Shanghai, 200092, China.
- Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai, 201203, China.
| | - Fang Huang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| |
Collapse
|
76
|
Feng T, Du H, Yang C, Wang Y, Hu F. Loss of TMEM106B exacerbates Tau pathology and neurodegeneration in PS19 mice. Acta Neuropathol 2024; 147:62. [PMID: 38526799 DOI: 10.1007/s00401-024-02702-4] [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: 10/02/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/27/2024]
Abstract
TMEM106B, a gene encoding a lysosome membrane protein, is tightly associated with brain aging, hypomyelinating leukodystrophy, and multiple neurodegenerative diseases, including frontotemporal lobar degeneration with TDP-43 aggregates (FTLD-TDP). Recently, TMEM106B polymorphisms have been associated with tauopathy in chronic traumatic encephalopathy (CTE) and FTLD-TDP patients. However, how TMEM106B influences Tau pathology and its associated neurodegeneration, is unclear. Here we show that loss of TMEM106B enhances the accumulation of pathological Tau, especially in the neuronal soma in the hippocampus, resulting in severe neuronal loss in the PS19 Tau transgenic mice. Moreover, Tmem106b-/- PS19 mice develop significantly increased abnormalities in the neuronal cytoskeleton, autophagy-lysosome activities, as well as glial activation, compared with PS19 and Tmem106b-/- mice. Together, our findings demonstrate that loss of TMEM106B drastically exacerbates Tau pathology and its associated disease phenotypes, and provide new insights into the roles of TMEM106B in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tuancheng Feng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA
| | - Huan Du
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA
| | - Cha Yang
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA
| | - Ya Wang
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA.
| |
Collapse
|
77
|
Cresto N, Courret M, Génin A, Martin CMP, Bourret J, Sakkaki S, de Bock F, Janvier A, Polizzi A, Payrastre L, Ellero-Simatos S, Audinat E, Perroy J, Marchi N. Continuous low-level dietary exposure to glyphosate elicits dose and sex-dependent synaptic and microglial adaptations in the rodent brain. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 345:123477. [PMID: 38307239 DOI: 10.1016/j.envpol.2024.123477] [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: 11/10/2023] [Revised: 01/19/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
Prolonged exposure to low levels of dietary contaminants is a context in modern life that could alter organ physiology gradually. Here, we aimed to investigate the impact of continuous exposure to acceptable daily intake (ADI) and non-observable adverse effect level (NOAEL) of glyphosate from gestation to adulthood using C57BL/6J mice and incorporating these levels into their food pellets. From adulthood, we analyzed neurophysiological and neuro-glia cellular adaptations in male and female animals. Using ex-vivo hippocampal slice electrophysiology, we found a reduced efficacy of Schaffer collateral-to-CA1 excitatory synapses in glyphosate-exposed dietary conditions, with ADI and NOAEL dose-dependent effects. Short-term facilitation of excitatory synaptic transmission was specifically increased in NOAEL conditions, with a predominant influence in males, suggesting a reduced probability of neurotransmitter release. Long-term synaptic potentiation (LTP) was decreased in NOAEL-exposed mice. Next, we explore whether these neurophysiological modifications are associated with neuro-glia changes in the somatosensory cortex and hippocampus. High-resolution confocal microscopy analyses unveil a dose-dependent increased density of excitatory Vglut1+ Homer1+ synapses. Microglial Iba1+ cells displayed a shortening of their ramifications, a sign of cellular reactivity that was more pronounced in males at NOAEL levels. The morphology of GFAP+ astrocytes was generally not modified. Finally, we asked whether mouse-specific cross-correlations exist among all data sets generated. This examination included the novel object recognition (NOR) test performed before ex vivo functional and immunohistochemical examinations. We report a negative linear regression between the number of synapses and NOR or LTP maintenance when plotting ADI and NOAEL datasets. These results outline synaptic and microglial cell adaptations resulting from prenatal and continuous dietary low levels of glyphosate, discernible in, but not limited to, adult males exposed to the NOAEL. We discuss the potential significance of these findings to real-world consumer situations and long-term brain resilience.
Collapse
Affiliation(s)
- Noemie Cresto
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Margot Courret
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Athénaïs Génin
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Céline Marie Pauline Martin
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Julie Bourret
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Sophie Sakkaki
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Frederic de Bock
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Alicia Janvier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Arnaud Polizzi
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Laurence Payrastre
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Etienne Audinat
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Perroy
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicola Marchi
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
78
|
Mesik L, Parkins S, Severin D, Grier BD, Ewall G, Kotha S, Wesselborg C, Moreno C, Jaoui Y, Felder A, Huang B, Johnson MB, Harrigan TP, Knight AE, Lani SW, Lemaire T, Kirkwood A, Hwang GM, Lee HK. Transcranial Low-Intensity Focused Ultrasound Stimulation of the Visual Thalamus Produces Long-Term Depression of Thalamocortical Synapses in the Adult Visual Cortex. J Neurosci 2024; 44:e0784232024. [PMID: 38316559 PMCID: PMC10941064 DOI: 10.1523/jneurosci.0784-23.2024] [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: 04/30/2023] [Revised: 12/13/2023] [Accepted: 01/30/2024] [Indexed: 02/07/2024] Open
Abstract
Transcranial focused ultrasound stimulation (tFUS) is a noninvasive neuromodulation technique, which can penetrate deeper and modulate neural activity with a greater spatial resolution (on the order of millimeters) than currently available noninvasive brain stimulation methods, such as transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS). While there are several studies demonstrating the ability of tFUS to modulate neuronal activity, it is unclear whether it can be used for producing long-term plasticity as needed to modify circuit function, especially in adult brain circuits with limited plasticity such as the thalamocortical synapses. Here we demonstrate that transcranial low-intensity focused ultrasound (LIFU) stimulation of the visual thalamus (dorsal lateral geniculate nucleus, dLGN), a deep brain structure, leads to NMDA receptor (NMDAR)-dependent long-term depression of its synaptic transmission onto layer 4 neurons in the primary visual cortex (V1) of adult mice of both sexes. This change is not accompanied by large increases in neuronal activity, as visualized using the cFos Targeted Recombination in Active Populations (cFosTRAP2) mouse line, or activation of microglia, which was assessed with IBA-1 staining. Using a model (SONIC) based on the neuronal intramembrane cavitation excitation (NICE) theory of ultrasound neuromodulation, we find that the predicted activity pattern of dLGN neurons upon sonication is state-dependent with a range of activity that falls within the parameter space conducive for inducing long-term synaptic depression. Our results suggest that noninvasive transcranial LIFU stimulation has a potential for recovering long-term plasticity of thalamocortical synapses in the postcritical period adult brain.
Collapse
Affiliation(s)
- Lukas Mesik
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Samuel Parkins
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Cell Molecular Developmental Biology and Biophysics Graduate Program, Johns Hopkins University, Baltimore, Maryland 21218
| | - Daniel Severin
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Bryce D Grier
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Gabrielle Ewall
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Sumasri Kotha
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Christian Wesselborg
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Cell Molecular Developmental Biology and Biophysics Graduate Program, Johns Hopkins University, Baltimore, Maryland 21218
| | - Cristian Moreno
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Yanis Jaoui
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Adrianna Felder
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Brian Huang
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Marina B Johnson
- Johns Hopkins Applied Physics Laboratory, Johns Hopkins University, Laurel, Maryland 20723
| | - Timothy P Harrigan
- Johns Hopkins Applied Physics Laboratory, Johns Hopkins University, Laurel, Maryland 20723
| | - Anna E Knight
- Johns Hopkins Applied Physics Laboratory, Johns Hopkins University, Laurel, Maryland 20723
| | - Shane W Lani
- Johns Hopkins Applied Physics Laboratory, Johns Hopkins University, Laurel, Maryland 20723
| | - Théo Lemaire
- Neuroscience Institute, New York University Langone Health, New York, New York 10016
| | - Alfredo Kirkwood
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Grace M Hwang
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Johns Hopkins Applied Physics Laboratory, Johns Hopkins University, Laurel, Maryland 20723
| | - Hey-Kyoung Lee
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
79
|
Chen K, Xu B, Xiao X, Long L, Zhao Q, Fang Z, Tu X, Wang J, Xu J, Wang H. Involvement of CKS1B in the anti-inflammatory effects of cannabidiol in experimental stroke models. Exp Neurol 2024; 373:114654. [PMID: 38104887 DOI: 10.1016/j.expneurol.2023.114654] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/16/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
We have previously demonstrated that treatment with cannabidiol (CBD) ameliorates mitochondrial dysfunction and attenuates neuronal injury in rats following cerebral ischemia. However, the role of CBD in the progression of ischemic stroke-induced inflammation and the molecules involved remain unclear. Here, we found that CBD suppressed the production of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), reduced the activation of microglia, ameliorated mitochondrial deficits, and decreased the phosphorylation of nuclear factor κ-B (NF-κB) in BV-2 cells subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). Cyclin-dependent kinase regulatory subunit 1B (CKS1B) expression was decreased in BV-2 cells following OGD/R and this reduction was blocked by treatment with CBD. Knockdown of CKS1B increased the activation of microglia and enhanced the production of IL-1β and TNF-α in BV-2 cells treated with CBD. Moreover, CKS1B knockdown exacerbated mitochondrial deficits and increased NF-κB phosphorylation. CBD treatment also ameliorated brain injury, reduced neuroinflammation, and enhanced the protein levels of mitochondrial transcription factor A and CKS1B in rats following middle cerebral artery occlusion/reperfusion. These data identify CKS1B as a novel regulator of neuroinflammation; and reveals its involvement in the anti-inflammatory effects of CBD. Interventions targeting CKS1B expression are potentially promising for treating in ischemic stroke.
Collapse
Affiliation(s)
- Kechun Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bingtian Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuan Xiao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lu Long
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qian Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zicen Fang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xingxing Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiakang Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiangping Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou 510515, China.
| | - Haitao Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou 510515, China.
| |
Collapse
|
80
|
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
|
81
|
Barak R, Goshtasbi G, Fatehi R, Firouzabadi N. Signaling pathways and genetics of brain Renin angiotensin system in psychiatric disorders: State of the art. Pharmacol Biochem Behav 2024; 236:173706. [PMID: 38176544 DOI: 10.1016/j.pbb.2023.173706] [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: 07/30/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Along the conventional pathways, Renin-angiotensin system (RAS) plays a key role in the physiology of the CNS and pathogenesis of psychiatric diseases. RAS is a complex regulatory pathway which is composed of several peptides and receptors and comprises two counter-regulatory axes. The classical (ACE1/AngII/AT1 receptor) axis and the contemporary (ACE2/Ang (1-7)/Mas receptor) axis. The genes coding for elements of both axes have been broadly studied. Numerous functional polymorphisms on components of RAS have been identified to serve as informative disease and treatment markers. This review summarizes the role of each peptide and receptor in the pathophysiology of psychiatric disorders (depression, bipolar disorders and schizophrenia), followed by a concise look at the role of genetic polymorphism of the RAS in the pathophysiology of these disorders.
Collapse
Affiliation(s)
- Roya Barak
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ghazal Goshtasbi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reihaneh Fatehi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
82
|
Tsakiri M, Tsichlis I, Zivko C, Demetzos C, Mahairaki V. Lipidic Nanoparticles, Extracellular Vesicles and Hybrid Platforms as Advanced Medicinal Products: Future Therapeutic Prospects for Neurodegenerative Diseases. Pharmaceutics 2024; 16:350. [PMID: 38543244 PMCID: PMC10975844 DOI: 10.3390/pharmaceutics16030350] [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: 01/11/2024] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 04/01/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's and Parkinson's, affect a wide variety of the population and pose significant challenges with progressive and irreversible neural cell loss. The limitations of brain-targeting therapies and the unclear molecular mechanisms driving neurodegeneration hamper the possibility of developing successful treatment options. Thus, nanoscale drug delivery platforms offer a promising solution. This paper explores and compares lipidic nanoparticles, extracellular vesicles (EVs), and hybrid liposomal-EV nanoplatforms as advanced approaches for targeted delivery to combat neurodegeneration. Lipidic nanoparticles are well-characterized platforms that allow multi-drug loading and scalable production. Conversely, EVs offer the ability of selectively targeting specific tissues and high biocompatibility. The combination of these two platforms in one could lead to promising results in the treatment of neurodegeneration. However, many issues, such as the regulatory framework, remain to be solved before these novel products are translated into clinical practice.
Collapse
Affiliation(s)
- Maria Tsakiri
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece; (M.T.); (I.T.); (C.D.)
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA;
| | - Ioannis Tsichlis
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece; (M.T.); (I.T.); (C.D.)
| | - Cristina Zivko
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA;
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece; (M.T.); (I.T.); (C.D.)
| | - Vasiliki Mahairaki
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA;
- The Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
83
|
Zhou X, Zhu Y, Gao L, Li Y, Li H, Huang C, Liu Y, Hu A, Ying C, Song Y. Binding of RAGE and RIPK1 induces cognitive deficits in chronic hyperglycemia-derived neuroinflammation. CNS Neurosci Ther 2024; 30:e14449. [PMID: 37665158 PMCID: PMC10916433 DOI: 10.1111/cns.14449] [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: 03/15/2023] [Revised: 07/08/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
AIMS Chronic hyperglycemia-induced inflammation of the hippocampus is an important cause of cognitive deficits in diabetic patients. The receptor for advanced glycation end products (RAGE), which is widely expressed in the hippocampus, is a crucial factor in this inflammation and the associated cognitive deficits. We aimed to reveal the underlying mechanism by which RAGE regulates neuroinflammation in the pathogenesis of diabetes-induced cognitive impairment. METHODS We used db/db mice as a model for type 2 diabetes to investigate whether receptor-interacting serine/threonine protein kinase 1 (RIPK1), which is expressed in microglia in the hippocampal region, is a key protein partner for RAGE. GST pull-down assays and AutoDock Vina simulations were performed to identify the key structural domain in RAGE that binds to RIPK1. Western blotting, co-immunoprecipitation (Co-IP), and immunofluorescence (IF) were used to detect the levels of key proteins or interaction between RAGE and RIPK1. Cognitive deficits in the mice were assessed with the Morris water maze (MWM) and new object recognition (NOR) and fear-conditioning tests. RESULTS RAGE binds directly to RIPK1 via the amino acid sequence (AAs) 362-367, thereby upregulating phosphorylation of RIPK1, which results in activation of the NLRP3 inflammasome in microglia and ultimately leads to cognitive impairments in db/db mice. We mutated RAGE AAs 362-367 to reverse neuroinflammation in the hippocampus and improve cognitive function, suggesting that RAGE AAs 362-367 is a key structural domain that binds directly to RIPK1. These results also indicate that hyperglycemia-induced inflammation in the hippocampus is dependent on direct binding of RAGE and RIPK1. CONCLUSION Direct interaction of RAGE and RIPK1 via AAs 362-367 is an important mechanism for enhanced neuroinflammation in the hyperglycemic environment and is a key node in the development of cognitive deficits in diabetes.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Department of GeneticsXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yandong Zhu
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Lin Gao
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yan Li
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Hui Li
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Chengyu Huang
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yan Liu
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Ankang Hu
- Lab Animal CenterXuzhou Medical UniversityXuzhouChina
| | - Changjiang Ying
- Department of EndocrinologyAffiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Yuanjian Song
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Department of GeneticsXuzhou Medical UniversityXuzhouJiangsuChina
| |
Collapse
|
84
|
Supakul S, Murakami R, Oyama C, Shindo T, Hatakeyama Y, Itsuno M, Bannai H, Shibata S, Maeda S, Okano H. Mutual interaction of neurons and astrocytes derived from iPSCs with APP V717L mutation developed the astrocytic phenotypes of Alzheimer's disease. Inflamm Regen 2024; 44:8. [PMID: 38419091 PMCID: PMC10900748 DOI: 10.1186/s41232-023-00310-5] [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: 09/05/2023] [Accepted: 11/22/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND The development of induced pluripotent stem cells (iPSCs) technology has enabled human cellular disease modeling for inaccessible cell types, such as neural cells in the brain. However, many of the iPSC-derived disease models established to date typically involve only a single cell type. These monoculture models are inadequate for accurately simulating the brain environment, where multiple cell types interact. The limited cell type diversity in monoculture models hinders the accurate recapitulation of disease phenotypes resulting from interactions between different cell types. Therefore, our goal was to create cell models that include multiple interacting cell types to better recapitulate disease phenotypes. METHODS To establish a co-culture model of neurons and astrocytes, we individually induced neurons and astrocytes from the same iPSCs using our novel differentiation methods, and then co-cultured them. We evaluated the effects of co-culture on neurons and astrocytes using immunocytochemistry, immuno-electron microscopy, and Ca2+ imaging. We also developed a co-culture model using iPSCs from a patient with familial Alzheimer's disease (AD) patient (APP V717L mutation) to investigate whether this model would manifest disease phenotypes not seen in the monoculture models. RESULTS The co-culture of the neurons and astrocytes increased the branching of astrocyte processes, the number of GFAP-positive cells, neuronal activities, the number of synapses, and the density of presynaptic vesicles. In addition, immuno-electron microscopy confirmed the formation of a tripartite synaptic structure in the co-culture model, and inhibition of glutamate transporters increased neuronal activity. Compared to the co-culture model of the control iPSCs, the co-culture model of familial AD developed astrogliosis-like phenotype, which was not observed in the monoculture model of astrocytes. CONCLUSIONS Co-culture of iPSC-derived neurons and astrocytes enhanced the morphological changes mimicking the in vivo condition of both cell types. The formation of the functional tripartite synaptic structures in the co-culture model suggested the mutual interaction between the cells. Furthermore, the co-culture model with the APP V717L mutation expressed in neurons exhibited an astrocytic phenotype reminiscent of AD brain pathology. These results suggest that our co-culture model is a valuable tool for disease modeling of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sopak Supakul
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Rei Murakami
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Chisato Oyama
- Department of Electrical Engineering and Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yuki Hatakeyama
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Maika Itsuno
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroko Bannai
- Department of Electrical Engineering and Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Sumihiro Maeda
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
85
|
Laaksonen S, Saraste M, Nylund M, Hinz R, Snellman A, Rinne J, Matilainen M, Airas L. Sex-driven variability in TSPO-expressing microglia in MS patients and healthy individuals. Front Neurol 2024; 15:1352116. [PMID: 38445263 PMCID: PMC10913932 DOI: 10.3389/fneur.2024.1352116] [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: 12/07/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Background Males with multiple sclerosis (MS) have a higher risk for disability progression than females, but the reasons for this are unclear. Objective We hypothesized that potential differences in TSPO-expressing microglia between female and male MS patients could contribute to sex differences in clinical disease progression. Methods The study cohort consisted of 102 MS patients (mean (SD) age 45.3 (9.7) years, median (IQR) disease duration 12.1 (7.0-17.2) years, 72% females, 74% relapsing-remitting MS) and 76 age- and sex-matched healthy controls. TSPO-expressing microglia were measured using the TSPO-binding radioligand [11C](R)-PK11195 and brain positron emission tomography (PET). TSPO-binding was quantified as distribution volume ratio (DVR) in normal-appearing white matter (NAWM), thalamus, whole brain and cortical gray matter (cGM). Results Male MS patients had higher DVRs compared to female patients in the whole brain [1.22 (0.04) vs. 1.20 (0.02), p = 0.002], NAWM [1.24 (0.06) vs. 1.21 (0.05), p = 0.006], thalamus [1.37 (0.08) vs. 1.32 (0.02), p = 0.008] and cGM [1.25 (0.04) vs. 1.23 (0.04), p = 0.028]. Similarly, healthy men had higher DVRs compared to healthy women except for cGM. Of the studied subgroups, secondary progressive male MS patients had the highest DVRs in all regions, while female controls had the lowest DVRs. Conclusion We observed higher TSPO-binding in males compared to females among people with MS and in healthy individuals. This sex-driven inherent variability in TSPO-expressing microglia may predispose male MS patients to greater likelihood of disease progression.
Collapse
Affiliation(s)
- Sini Laaksonen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Maija Saraste
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Marjo Nylund
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, United Kingdom
| | - Anniina Snellman
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Juha Rinne
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Markus Matilainen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Laura Airas
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| |
Collapse
|
86
|
Chiang MC, Yang YP, Nicol CJB, Wang CJ. Gold Nanoparticles in Neurological Diseases: A Review of Neuroprotection. Int J Mol Sci 2024; 25:2360. [PMID: 38397037 PMCID: PMC10888679 DOI: 10.3390/ijms25042360] [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: 01/11/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
This review explores the diverse applications of gold nanoparticles (AuNPs) in neurological diseases, with a specific focus on Alzheimer's disease (AD), Parkinson's disease (PD), and stroke. The introduction highlights the pivotal role of neuroinflammation in these disorders and introduces the unique properties of AuNPs. The review's core examines the mechanisms by which AuNPs exert neuroprotection and anti-neuro-inflammatory effects, elucidating various pathways through which they manifest these properties. The potential therapeutic applications of AuNPs in AD are discussed, shedding light on promising avenues for therapy. This review also explores the prospects of utilizing AuNPs in PD interventions, presenting a hopeful outlook for future treatments. Additionally, the review delves into the potential of AuNPs in providing neuroprotection after strokes, emphasizing their significance in mitigating cerebrovascular accidents' aftermath. Experimental findings from cellular and animal models are consolidated to provide a comprehensive overview of AuNPs' effectiveness, offering insights into their impact at both the cellular and in vivo levels. This review enhances our understanding of AuNPs' applications in neurological diseases and lays the groundwork for innovative therapeutic strategies in neurology.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Christopher J. B. Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, Cancer Biology and Genetics Division, Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Chieh-Ju Wang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
87
|
Peggion C, Calì T, Brini M. Mitochondria Dysfunction and Neuroinflammation in Neurodegeneration: Who Comes First? Antioxidants (Basel) 2024; 13:240. [PMID: 38397838 PMCID: PMC10885966 DOI: 10.3390/antiox13020240] [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: 02/01/2024] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) encompass an assorted array of disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, each characterised by distinct clinical manifestations and underlying pathological mechanisms. While some cases have a genetic basis, many NDs occur sporadically. Despite their differences, these diseases commonly feature chronic neuroinflammation as a hallmark. Consensus has recently been reached on the possibility that mitochondria dysfunction and protein aggregation can mutually contribute to the activation of neuroinflammatory response and thus to the onset and progression of these disorders. In the present review, we discuss the contribution of mitochondria dysfunction and neuroinflammation to the aetiology and progression of NDs, highlighting the possibility that new potential therapeutic targets can be identified to tackle neurodegenerative processes and alleviate the progression of these pathologies.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biology, University of Padova, 35131 Padova, Italy;
| | - Tito Calì
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
| | - Marisa Brini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
88
|
Mahbub NU, Islam MM, Hong ST, Chung HJ. Dysbiosis of the gut microbiota and its effect on α-synuclein and prion protein misfolding: consequences for neurodegeneration. Front Cell Infect Microbiol 2024; 14:1348279. [PMID: 38435303 PMCID: PMC10904658 DOI: 10.3389/fcimb.2024.1348279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
Abnormal behavior of α-synuclein and prion proteins is the hallmark of Parkinson's disease (PD) and prion illnesses, respectively, being complex neurological disorders. A primary cause of protein aggregation, brain injury, and cognitive loss in prion illnesses is the misfolding of normal cellular prion proteins (PrPC) into an infectious form (PrPSc). Aggregation of α-synuclein causes disruptions in cellular processes in Parkinson's disease (PD), leading to loss of dopamine-producing neurons and motor symptoms. Alteration in the composition or activity of gut microbes may weaken the intestinal barrier and make it possible for prions to go from the gut to the brain. The gut-brain axis is linked to neuroinflammation; the metabolites produced by the gut microbiota affect the aggregation of α-synuclein, regulate inflammation and immunological responses, and may influence the course of the disease and neurotoxicity of proteins, even if their primary targets are distinct proteins. This thorough analysis explores the complex interactions that exist between the gut microbiota and neurodegenerative illnesses, particularly Parkinson's disease (PD) and prion disorders. The involvement of the gut microbiota, a complex collection of bacteria, archaea, fungi, viruses etc., in various neurological illnesses is becoming increasingly recognized. The gut microbiome influences neuroinflammation, neurotransmitter synthesis, mitochondrial function, and intestinal barrier integrity through the gut-brain axis, which contributes to the development and progression of disease. The review delves into the molecular mechanisms that underlie these relationships, emphasizing the effects of microbial metabolites such as bacterial lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs) in regulating brain functioning. Additionally, it looks at how environmental influences and dietary decisions affect the gut microbiome and whether they could be risk factors for neurodegenerative illnesses. This study concludes by highlighting the critical role that the gut microbiota plays in the development of Parkinson's disease (PD) and prion disease. It also provides a promising direction for future research and possible treatment approaches. People afflicted by these difficult ailments may find hope in new preventive and therapeutic approaches if the role of the gut microbiota in these diseases is better understood.
Collapse
Affiliation(s)
- Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
89
|
Carvalho FV, Landis HE, Getachew B, Silva VDA, Ribeiro PR, Aschner M, Tizabi Y. Iron toxicity, ferroptosis and microbiota in Parkinson's disease: Implications for novel targets. ADVANCES IN NEUROTOXICOLOGY 2024; 11:105-132. [PMID: 38770370 PMCID: PMC11105119 DOI: 10.1016/bs.ant.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Parkinson's Disease (PD) is a progressive neurodegenerative disease characterized by loss of dopaminergic neurons in substantia nigra pars compacta (SNpc). Iron (Fe)-dependent programmed cell death known as ferroptosis, plays a crucial role in the etiology and progression of PD. Since SNpc is particularly vulnerable to Fe toxicity, a central role for ferroptosis in the etiology and progression of PD is envisioned. Ferroptosis, characterized by reactive oxygen species (ROS)-dependent accumulation of lipid peroxides, is tightly regulated by a variety of intracellular metabolic processes. Moreover, the recently characterized bi-directional interactions between ferroptosis and the gut microbiota, not only provides another window into the mechanistic underpinnings of PD but could also suggest novel interventions in this devastating disease. Here, following a brief discussion of PD, we focus on how our expanding knowledge of Fe-induced ferroptosis and its interaction with the gut microbiota may contribute to the pathophysiology of PD and how this knowledge may be exploited to provide novel interventions in PD.
Collapse
Affiliation(s)
| | - Harold E. Landis
- Integrative Medicine Fellow, University of Arizona, Tucson, AZ, United States
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | | | - Paulo R. Ribeiro
- Metabolomics Research Group, Institute of Chemistry, Federal University of Bahia, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| |
Collapse
|
90
|
Talbot EJ, Joshi L, Thornton P, Dezfouli M, Tsafou K, Perkinton M, Khoronenkova S. cGAS-STING signalling regulates microglial chemotaxis in genome instability. Nucleic Acids Res 2024; 52:1188-1206. [PMID: 38084916 PMCID: PMC10853792 DOI: 10.1093/nar/gkad1184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 02/10/2024] Open
Abstract
Defective DNA damage signalling and repair is a hallmark of age-related and genetic neurodegenerative disease. One mechanism implicated in disease progression is DNA damage-driven neuroinflammation, which is largely mediated by tissue-resident immune cells, microglia. Here, we utilise human microglia-like cell models of persistent DNA damage and ATM kinase deficiency to investigate how genome instability shapes microglial function. We demonstrate that upon DNA damage the cytosolic DNA sensing cGAS-STING axis drives chronic inflammation and a robust chemokine response, exemplified by production of CCL5 and CXCL10. Transcriptomic analyses revealed that cell migratory pathways were highly enriched upon IFN-β treatment of human iPSC-derived microglia, indicating that the chemokine response to DNA damage mirrors type I interferon signalling. Furthermore, we find that STING deletion leads to a defect in microglial chemotaxis under basal conditions and upon ATM kinase loss. Overall, this work provides mechanistic insights into cGAS-STING-dependent neuroinflammatory mechanisms and consequences of genome instability in the central nervous system.
Collapse
Affiliation(s)
- Emily J Talbot
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Lisha Joshi
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Peter Thornton
- Neuroscience, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK
| | - Mahya Dezfouli
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Gothenburg, Sweden
| | - Kalliopi Tsafou
- Department of Data Sciences & Quantitative Biology, AstraZeneca, Cambridge, UK
| | | | | |
Collapse
|
91
|
Thumu SCR, Jain M, Soman S, Das S, Verma V, Nandi A, Gutmann DH, Jayaprakash B, Nair D, Clement JP, Marathe S, Ramanan N. SRF-deficient astrocytes provide neuroprotection in mouse models of excitotoxicity and neurodegeneration. eLife 2024; 13:e95577. [PMID: 38289036 PMCID: PMC10857791 DOI: 10.7554/elife.95577] [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: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Reactive astrogliosis is a common pathological hallmark of CNS injury, infection, and neurodegeneration, where reactive astrocytes can be protective or detrimental to normal brain functions. Currently, the mechanisms regulating neuroprotective astrocytes and the extent of neuroprotection are poorly understood. Here, we report that conditional deletion of serum response factor (SRF) in adult astrocytes causes reactive-like hypertrophic astrocytes throughout the mouse brain. These SrfGFAP-ERCKO astrocytes do not affect neuron survival, synapse numbers, synaptic plasticity or learning and memory. However, the brains of Srf knockout mice exhibited neuroprotection against kainic-acid induced excitotoxic cell death. Relevant to human neurodegenerative diseases, SrfGFAP-ERCKO astrocytes abrogate nigral dopaminergic neuron death and reduce β-amyloid plaques in mouse models of Parkinson's and Alzheimer's disease, respectively. Taken together, these findings establish SRF as a key molecular switch for the generation of reactive astrocytes with neuroprotective functions that attenuate neuronal injury in the setting of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Monika Jain
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - Sumitha Soman
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - Soumen Das
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific ResearchBangaloreIndia
| | - Arnab Nandi
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - David H Gutmann
- Department of Neurology, Washington University School of MedicineSt. LouisUnited States
| | | | - Deepak Nair
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific ResearchBangaloreIndia
| | - Swananda Marathe
- Centre for Neuroscience, Indian Institute of ScienceBangaloreIndia
| | | |
Collapse
|
92
|
Gary JM, Cramer S, Bolon B, Bradley AE, Butt MT. Incidental Gliosis in the Central Nervous System of Control Nonhuman Primates and Rats. Toxicol Pathol 2024; 52:114-122. [PMID: 38828567 DOI: 10.1177/01926233241253255] [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: 06/05/2024]
Abstract
Gliosis, including microgliosis and astrocytosis, can be challenging to interpret in nonclinical studies. Incidences of glial foci in brains and spinal cords of control rats and nonhuman primates (NHPs) were reviewed in the historical control databases from two contract research organizations, including one specializing in neuropathology. In the brain, minimal to mild (grades 1-2) microgliosis was the most common diagnosis, especially in NHPs, although occasional moderate or marked microgliosis (grades 3 and 4) was encountered in both species. Microgliosis was more common in the cerebral cortex, cerebellum, and medulla oblongata in both species and was frequent in the white matter (brain), thalamus, and basal nuclei of NHPs. Gliosis ("not otherwise specified") of minimal severity was diagnosed in similar brain sub-sites for both species and was more common in NHPs compared with rats. Astrocytosis was most prominent in the cerebellum (molecular layer) of NHPs but was otherwise uncommon. In the spinal cord, microgliosis was most common in the lateral white matter tracts in rats and NHPs, and in the dorsal white matter tracts in NHPs. These data indicate that low-grade spontaneous glial responses occur with some frequency in control animals of two common nonclinical species.
Collapse
|
93
|
Zorkina Y, Ushakova V, Ochneva A, Tsurina A, Abramova O, Savenkova V, Goncharova A, Alekseenko I, Morozova I, Riabinina D, Kostyuk G, Morozova A. Lipids in Psychiatric Disorders: Functional and Potential Diagnostic Role as Blood Biomarkers. Metabolites 2024; 14:80. [PMID: 38392971 PMCID: PMC10890164 DOI: 10.3390/metabo14020080] [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: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 02/25/2024] Open
Abstract
Lipids are a crucial component of the human brain, serving important structural and functional roles. They are involved in cell function, myelination of neuronal projections, neurotransmission, neural plasticity, energy metabolism, and neuroinflammation. Despite their significance, the role of lipids in the development of mental disorders has not been well understood. This review focused on the potential use of lipids as blood biomarkers for common mental illnesses, such as major depressive disorder, anxiety disorders, bipolar disorder, and schizophrenia. This review also discussed the impact of commonly used psychiatric medications, such as neuroleptics and antidepressants, on lipid metabolism. The obtained data suggested that lipid biomarkers could be useful for diagnosing psychiatric diseases, but further research is needed to better understand the associations between blood lipids and mental disorders and to identify specific biomarker combinations for each disease.
Collapse
Affiliation(s)
- Yana Zorkina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Valeria Ushakova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Aleksandra Ochneva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Anna Tsurina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Olga Abramova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Valeria Savenkova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Anna Goncharova
- Moscow Center for Healthcare Innovations, 123473 Moscow, Russia;
| | - Irina Alekseenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academi of Science, 142290 Moscow, Russia
- Russia Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, 2, Kurchatov Square, 123182 Moscow, Russia
| | - Irina Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Daria Riabinina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Georgy Kostyuk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Anna Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| |
Collapse
|
94
|
Kim EH, Lee WS, Lee JH, Kwon DR. Microcurrent therapy as the nonpharmacological new protocol against Alzheimer's disease. Front Aging Neurosci 2024; 16:1344072. [PMID: 38304741 PMCID: PMC10833500 DOI: 10.3389/fnagi.2024.1344072] [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: 11/24/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
Introduction Alzheimer's disease (AD) poses an increasing global health challenge and is marked by gradual cognitive deterioration, memory impairment, and neuroinflammation. Innovative therapeutic approaches as non-pharmacological protocol are urgently needed with side effect risk of drugs. Microcurrent therapy, a non-invasive modality involving low-level electrical currents, has emerged as a potential solution to address AD's complex pathogenesis. This study investigates the optimal application of microcurrent therapy as a clinical protocol for AD, utilizing a comprehensive approach that integrates behavioral assessments and neuroinflammation evaluation in a mouse model of dementia. Methods and results The results reveal that microcurrent therapy holds promise in ameliorating memory impairment and reducing neuroinflammation in AD. Behavioral assessments, including the Novel Object Recognition Test (NOR) and Radial Arm Maze Test (RAM), demonstrated improved cognitive function following microcurrent therapy. Furthermore, microcurrent therapy inhibited expression of neuroinflammatory proteins, including ionized calcium binding adaptor molecule 1 (Iba1), and glial fibrillary acidic protein (GFAP) in current-treated group. Mechanistic insights suggest that microcurrent therapy may modulate neuroinflammation through the regulation of MAPK signaling pathways. Conclusion This study emphasizes the prospect of microcurrent therapy as a safe and efficacious non-pharmacological strategy for Alzheimer's disease (AD), providing optimism to the countless individuals impacted by this debilitating ailment. These results contribute to the developments of an innovative clinical protocol for AD and recovery from neurological injury, underscoring the significance of investigating unconventional therapeutic approaches for addressing this complex condition.
Collapse
Affiliation(s)
- Eun Ho Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Won Seok Lee
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Jae Hee Lee
- Department of Rehabilitation Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Dong Rak Kwon
- Department of Rehabilitation Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| |
Collapse
|
95
|
Fakhfouri G, Mijailović NR, Rahimian R. Psychiatric Comorbidities of Inflammatory Bowel Disease: It Is a Matter of Microglia's Gut Feeling. Cells 2024; 13:177. [PMID: 38247868 PMCID: PMC10814793 DOI: 10.3390/cells13020177] [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: 12/07/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Inflammatory bowel disease (IBD), a common term for Crohn's disease and ulcerative colitis, is a chronic, relapse-remitting condition of the gastrointestinal tract that is increasing worldwide. Psychiatric comorbidities, including depression and anxiety, are more prevalent in IBD patients than in healthy individuals. Evidence suggests that varying levels of neuroinflammation might underlie these states in IBD patients. Within this context, microglia are the crucial non-neural cells in the brain responsible for innate immune responses following inflammatory insults. Alterations in microglia's functions, such as secretory profile, phagocytic activity, and synaptic pruning, might play significant roles in mediating psychiatric manifestations of IBD. In this review, we discuss the role played by microglia in IBD-associated comorbidities.
Collapse
Affiliation(s)
- Gohar Fakhfouri
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, QC H4H 1R3, Canada;
| | - Nataša R. Mijailović
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health Institute, McGill University, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| |
Collapse
|
96
|
Tao L, Yu W, Liu Z, Zhao D, Lin S, Szalóki D, Kicsák M, Kurtán T, Zhang H. JE-133 Suppresses LPS-Induced Neuroinflammation Associated with the Regulation of JAK/STAT and Nrf2 Signaling Pathways. ACS Chem Neurosci 2024; 15:258-267. [PMID: 38181172 DOI: 10.1021/acschemneuro.3c00454] [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: 01/07/2024] Open
Abstract
Neuroinflammation plays an important role in the pathogenesis of neurodegenerative diseases, and interrupting the microglial-mediated neuroinflammation has been suggested as a promising strategy to delay or prevent the progression of neurodegeneration. In this study, we investigated the effects of JE-133, an optically active isochroman-2H-chromene conjugate containing a 1,3-disubstituted isochroman unit, on lipopolysaccharide (LPS)-induced microglial neuroinflammation and underlying mechanisms both in vitro and in vivo. First, JE-133 treatment decreased LPS-induced overproduction of interleukin-1 beta (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), nitrite, and nitric oxide synthase (iNOS) in BV2 microglial cells. Further study revealed that JE-133 downregulated the phosphorylation level of JAK/STAT and upregulated the protein level of Nrf2/HO-1 in LPS-stimulated BV2 microglial cells and verified that JE-133 directly bound to Keap1 by a pull-down assay. Next, JE-133 administration also inhibited neuroinflammation in vivo, as indicated by a reduced CD11b protein level and an overexpressed mRNA level of the pro-inflammatory cytokine TNF-α in the hippocampus of LPS-injected mice. Moreover, the regulative effects of JE-133 on the JAK/STAT and Nrf2/HO-1 pathways were also verified in the hippocampus of LPS-injected mice. Taken together, our study for the first time reports that JE-133 exhibits inhibitory effects against LPS-stimulated neuroinflammation both in vitro and in vivo, which might be associated with the simultaneous regulation of the JAK/STAT and Nrf2 pathways. Our findings may provide important clues for the discovery of effective drug leads/candidates against neuroinflammation-associated neurodegeneration.
Collapse
Affiliation(s)
- Lingxue Tao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 200031, China
| | - Weichen Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ziyi Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Nanchang University, Jiangxi 330031, China
| | - Danfeng Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Sijin Lin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dóra Szalóki
- Department of Organic Chemistry, University of Debrecen, Debrecen, P.O. Box 400, Debrecen H-4002, Hungary
| | - Máté Kicsák
- Department of Organic Chemistry, University of Debrecen, Debrecen, P.O. Box 400, Debrecen H-4002, Hungary
| | - Tibor Kurtán
- Department of Organic Chemistry, University of Debrecen, Debrecen, P.O. Box 400, Debrecen H-4002, Hungary
| | - Haiyan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
97
|
Wang Y, Chen Z, Davis B, Lipman W, Xing S, Zhang L, Wang T, Hafiz P, Xie W, Yan Z, Huang Z, Song J, Bai W. Digital automation of transdermal drug delivery with high spatiotemporal resolution. Nat Commun 2024; 15:511. [PMID: 38218967 PMCID: PMC10787768 DOI: 10.1038/s41467-023-44532-0] [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: 08/19/2023] [Accepted: 12/18/2023] [Indexed: 01/15/2024] Open
Abstract
Transdermal drug delivery is of vital importance for medical treatments. However, user adherence to long-term repetitive drug delivery poses a grand challenge. Furthermore, the dynamic and unpredictable disease progression demands a pharmaceutical treatment that can be actively controlled in real-time to ensure medical precision and personalization. Here, we report a spatiotemporal on-demand patch (SOP) that integrates drug-loaded microneedles with biocompatible metallic membranes to enable electrically triggered active control of drug release. Precise control of drug release to targeted locations (<1 mm2), rapid drug release response to electrical triggers (<30 s), and multi-modal operation involving both drug release and electrical stimulation highlight the novelty. Solution-based fabrication ensures high customizability and scalability to tailor the SOP for various pharmaceutical needs. The wireless-powered and digital-controlled SOP demonstrates great promise in achieving full automation of drug delivery, improving user adherence while ensuring medical precision. Based on these characteristics, we utilized SOPs in sleep studies. We revealed that programmed release of exogenous melatonin from SOPs improve sleep of mice, indicating potential values for basic research and clinical treatments.
Collapse
Affiliation(s)
- Yihang Wang
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zeka Chen
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brayden Davis
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Will Lipman
- Department of Psychology and Neuroscience, University of North Carolina at chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sicheng Xing
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Lin Zhang
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Tian Wang
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Priyash Hafiz
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Wanrong Xie
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zijie Yan
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zhili Huang
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Wubin Bai
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
98
|
Zaccai S, Nemirovsky A, Lerner L, Alfahel L, Eremenko E, Israelson A, Monsonego A. CD4 T-cell aging exacerbates neuroinflammation in a late-onset mouse model of amyotrophic lateral sclerosis. J Neuroinflammation 2024; 21:17. [PMID: 38212835 PMCID: PMC10782641 DOI: 10.1186/s12974-023-03007-1] [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: 11/05/2023] [Accepted: 12/26/2023] [Indexed: 01/13/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset progressive neurodegenerative disorder characterized by the loss of upper and lower motor neurons in the brain and spinal cord. Accumulating evidence suggests that ALS is not solely a neuronal cell- or brain tissue-autonomous disease and that neuroinflammation plays a key role in disease progression. Furthermore, whereas both CD4 and CD8 T cells were observed in spinal cords of ALS patients and in mouse models of the disease, their role in the neuroinflammatory process, especially considering their functional changes with age, is not fully explored. In this study, we revealed the structure of the CD4 T-cell compartment during disease progression of early-onset SOD1G93A and late-onset SOD1G37R mouse models of ALS. We show age-related changes in the CD4 T-cell subset organization between these mutant SOD1 mouse models towards increased frequency of effector T cells in spleens of SOD1G37R mice and robust infiltration of CD4 T cells expressing activation markers and the checkpoint molecule PD1 into the spinal cord. The frequency of infiltrating CD4 T cells correlated with the frequency of infiltrating CD8 T cells which displayed a more exhausted phenotype. Moreover, RNA-Seq and immunohistochemistry analyses of spinal cords from SOD1G37R mice with early clinical symptoms demonstrated immunological trajectories reminiscent of a neurotoxic inflammatory response which involved proinflammatory T cells and antigen presentation related pathways. Overall, our findings suggest that age-related changes of the CD4 T cell landscape is indicative of a chronic inflammatory response, which aggravates the disease process and can be therapeutically targeted.
Collapse
Affiliation(s)
- Shir Zaccai
- Department of Physiology and Cell Biology, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Anna Nemirovsky
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Livnat Lerner
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Leenor Alfahel
- Department of Physiology and Cell Biology, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Ekaterina Eremenko
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel.
| | - Alon Monsonego
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel.
| |
Collapse
|
99
|
Liu P, Tang W, Xiang K, Li G. Pterostilbene in the treatment of inflammatory and oncological diseases. Front Pharmacol 2024; 14:1323377. [PMID: 38259272 PMCID: PMC10800393 DOI: 10.3389/fphar.2023.1323377] [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: 10/17/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Pterostilbene (PTS), a naturally occurring analog of resveratrol (RSV), has garnered significant attention due to its potential therapeutic effects in treating inflammatory and oncological diseases. This comprehensive review elucidates the pharmacological properties, mechanisms of action, and therapeutic potential of PTS. Various studies indicate that PTS exhibits anti-inflammatory, antioxidant, and antitumour properties, potentially making it a promising candidate for clinical applications. Its influence on regulatory pathways like NF-κB and PI3K/Akt underscores its diverse strategies in addressing diseases. Additionally, PTS showcases a favorable pharmacokinetic profile with better oral bioavailability compared to other stilbenoids, thus enhancing its therapeutic potential. Given these findings, there is an increased interest in incorporating PTS into treatment regimens for inflammatory and cancer-related conditions. However, more extensive clinical trials are imperative to establish its safety and efficacy in diverse patient populations.
Collapse
Affiliation(s)
- Peijun Liu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Weihua Tang
- Department of Radiology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Kali Xiang
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Guangcai Li
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| |
Collapse
|
100
|
Fornari Laurindo L, Aparecido Dias J, Cressoni Araújo A, Torres Pomini K, Machado Galhardi C, Rucco Penteado Detregiachi C, Santos de Argollo Haber L, Donizeti Roque D, Dib Bechara M, Vialogo Marques de Castro M, de Souza Bastos Mazuqueli Pereira E, José Tofano R, Jasmin Santos German Borgo I, Maria Barbalho S. Immunological dimensions of neuroinflammation and microglial activation: exploring innovative immunomodulatory approaches to mitigate neuroinflammatory progression. Front Immunol 2024; 14:1305933. [PMID: 38259497 PMCID: PMC10800801 DOI: 10.3389/fimmu.2023.1305933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
The increasing life expectancy has led to a higher incidence of age-related neurodegenerative conditions. Within this framework, neuroinflammation emerges as a significant contributing factor. It involves the activation of microglia and astrocytes, leading to the release of pro-inflammatory cytokines and chemokines and the infiltration of peripheral leukocytes into the central nervous system (CNS). These instances result in neuronal damage and neurodegeneration through activated nucleotide-binding domain and leucine-rich repeat containing (NLR) family pyrin domain containing protein 3 (NLRP3) and nuclear factor kappa B (NF-kB) pathways and decreased nuclear factor erythroid 2-related factor 2 (Nrf2) activity. Due to limited effectiveness regarding the inhibition of neuroinflammatory targets using conventional drugs, there is challenging growth in the search for innovative therapies for alleviating neuroinflammation in CNS diseases or even before their onset. Our results indicate that interventions focusing on Interleukin-Driven Immunomodulation, Chemokine (CXC) Receptor Signaling and Expression, Cold Exposure, and Fibrin-Targeted strategies significantly promise to mitigate neuroinflammatory processes. These approaches demonstrate potential anti-neuroinflammatory effects, addressing conditions such as Multiple Sclerosis, Experimental autoimmune encephalomyelitis, Parkinson's Disease, and Alzheimer's Disease. While the findings are promising, immunomodulatory therapies often face limitations due to Immune-Related Adverse Events. Therefore, the conduction of randomized clinical trials in this matter is mandatory, and will pave the way for a promising future in the development of new medicines with specific therapeutic targets.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília, São Paulo, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Jefferson Aparecido Dias
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Karina Torres Pomini
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Department of Anatomy, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Cristiano Machado Galhardi
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Claudia Rucco Penteado Detregiachi
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Luíza Santos de Argollo Haber
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Domingos Donizeti Roque
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Department of Anatomy, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Marcela Vialogo Marques de Castro
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Eliana de Souza Bastos Mazuqueli Pereira
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Ricardo José Tofano
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Iris Jasmin Santos German Borgo
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, Universidade de São Paulo (FOB-USP), Bauru, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília, São Paulo, Brazil
| |
Collapse
|