Next Article in Journal
Critical Appraisal of Pharmaceutical Therapy in Diabetic Cardiomyopathy—Challenges and Prospectives
Previous Article in Journal
Copper Imparts a New Therapeutic Property to Resveratrol by Generating ROS to Deactivate Cell-Free Chromatin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Mechanisms Underlying Neuroinflammation Intervention with Medicinal Plants: A Critical and Narrative Review of the Current Literature

by
Sandra Maria Barbalho
1,2,
Beatriz Leme Boaro
3,
Jéssica da Silva Camarinha Oliveira
3,
Jiří Patočka
4,5,
Caroline Barbalho Lamas
6,
Masaru Tanaka
7,*,† and
Lucas Fornari Laurindo
1,†
1
Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
2
Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
3
Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil
4
Faculty of Health and Social Studies, Institute of Radiology, Toxicology and Civil Protection, University of South Bohemia Ceske Budejovice, 37005 Ceske Budejovice, Czech Republic
5
Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic
6
Department of Gerontology, School of Gerontology, Universidade Federal de São Carlos (UFSCar), São Carlos 13565-905, São Paulo, Brazil
7
Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos Krt. 113, H-6725 Szeged, Hungary
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2025, 18(1), 133; https://doi.org/10.3390/ph18010133
Submission received: 18 December 2024 / Revised: 16 January 2025 / Accepted: 17 January 2025 / Published: 20 January 2025
(This article belongs to the Section Natural Products)

Abstract

:
Neuroinflammation is a key factor in the progression of neurodegenerative diseases, driven by the dysregulation of molecular pathways and activation of the brain’s immune system, resulting in the release of pro-inflammatory and oxidative molecules. This chronic inflammation is exacerbated by peripheral leukocyte infiltration into the central nervous system. Medicinal plants, with their historical use in traditional medicine, have emerged as promising candidates to mitigate neuroinflammation and offer a sustainable alternative for addressing neurodegenerative conditions in a green healthcare framework. This review evaluates the effects of medicinal plants on neuroinflammation, emphasizing their mechanisms of action, effective dosages, and clinical implications, based on a systematic search of databases such as PubMed, SCOPUS, and Web of Science. The key findings highlight that plants like Cleistocalyx nervosum var. paniala, Curcuma longa, Cannabis sativa, and Dioscorea nipponica reduce pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), inhibit enzymes (COX-2 and iNOS), and activate antioxidant pathways, particularly Nrf2. NF-κB emerged as the primary pro-inflammatory pathway inhibited across studies. While the anti-inflammatory potential of these plants is significant, the variability in dosages and phytochemical compositions limits clinical translation. Here, we highlight that medicinal plants are effective modulators of neuroinflammation, underscoring their therapeutic potential. Future research should focus on animal models, standardized protocols, and safety assessments, integrating advanced methodologies, such as genetic studies and nanotechnology, to enhance their applicability in neurodegenerative disease management.

Graphical Abstract

1. Introduction

The increase in life expectancy has been accompanied by a rise in the prevalence of age-related Neurodegenerative Diseases (NDs). Maintaining neuronal function and cognitive ability depends on the brain’s high energy demands, as it is one of the most energetically active organs despite not performing mechanical work like the skeletal muscles or heart [1,2,3,4]. Typically, the human brain consumes approximately 3.5 mL of oxygen per 100 g of brain tissue per minute [5,6,7]. This high oxygen consumption makes neurons susceptible to Reactive Oxygen Species (ROS) damage and subsequent inflammation [8,9,10,11,12,13,14,15,16].
Neuroinflammation is characterized by activating the brain’s innate immune system in response to inflammatory stimuli. This response involves the activation of microglia and astrocytes, the release of pro-inflammatory cytokines and chemokines, ROS production, and, sometimes, the infiltration of peripheral leukocytes into the Central Nervous System (CNS). While neuroinflammation initially acts as a protective mechanism, sustained or maladaptive inflammation has been identified as a critical factor in the progression of various neurological diseases, including NDs, psychiatric disorders, pain syndromes, stroke, and traumatic brain injury [4,17,18,19,20,21,22]. Substantially, neuroinflammation involves the dysregulation of several signaling pathways, including Nuclear Factor Kappa B (NF-κB), Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2), Nucleotide-Binding and Oligomerization Domain-Like Receptor Family Pyrin Domain-Containing 3 (NLRP3), and Janus Kinase (JAK)/Signal Transducer and Activator of Transcription (STAT), at different proportions. Typically, NF-κB, NLRP3, and JAK/STAT are detrimental, while Nrf2 possesses antioxidant and anti-inflammatory effects [23,24,25,26].
Addressing neuroinflammation to reduce disease severity and improve patient outcomes is a promising strategy against neurodegeneration. From a molecular perspective, there are several conventional drug targets for neuroinflammation, such as enzymes, receptors, and ion channels [22,27,28,29,30,31]. However, the high cost of synthetic drugs presents a challenge, emphasizing the need for alternative approaches [32,33,34,35]. This has heightened interest in naturally occurring medicinal plants known for their antioxidant, anti-inflammatory, and neuroprotective properties. These plants are often more cost-effective and have been safely utilized in treatments for thousands of years [36,37,38].
A substantial body of literature underscores the beneficial effects of dietary phytochemicals and medicinal plants on overall health and longevity [39,40,41,42]. Medicinal-plant-based drug discovery remains a vital and underexplored area of research with substantial potential [43,44,45,46]. Despite pharmaceutical advances, plants have historically been a rich source of therapeutic compounds, yet many of their potential benefits are still untapped [47,48]. For example, resveratrol, a phytochemical widely found in grapes, has the potential to modulate inflammation within the CNS effectively by targeting critical pathways involved in microglia polarization and neuronal cell survival, and research advises incorporating resveratrol sources into dietary routines to decrease the risk or delay the onset of NDs [49,50,51,52,53]. By comprehensively investigating these plants, researchers can uncover novel compounds that could offer significant breakthroughs in treating various conditions. Specifically, this approach could lead to the discovery of new treatments for complex issues such as neuroinflammation and neurodegeneration, which are often challenging to address with current therapies [13,46,54,55].
Despite previous efforts, the existing reviews have yet to fully explore the potential of medicinal plants in regulating neuroinflammation. Tyler et al. [56] offered valuable insights into the therapeutic potential of plants for NDs. However, their review needed a systematic examination of the literature addressing medicinal plants targeting neuroinflammation. Their focus was broader, with a greater emphasis on ethnobotanical aspects rather than exclusively discussing neuroinflammation. In contrast, Suk et al. [57] explored the regulation of neuroinflammation by herbal medicine and its implications for NDs. Their review centered predominantly on traditional medicines and their flavonoid derivatives. Additionally, this review, published in 2005, needs to be updated and would benefit from an update incorporating recent advancements in the field. Due to the limitations of the already published research regarding medicinal plants targeting neuroinflammation, our study aims to not only update the current body of research, integrating cutting-edge recent publications in its findings but also provide a systematic and critical evaluation of how medicinal plants affect signaling pathways and specific molecular targets to modulate neuroinflammation, ultimately giving powerful insights into the molecular aspects of neuroinflammation treatment under green healthcare and advocating for future research endeavors in this field, promoting a future with fewer diseases. This manuscript includes a table (Table 1) that compiles research on medicinal plants’ roles as potential neuroinflammation modulators. Figure 1 illustrates the microglia’s distinct phenotypes, M1 (pro-inflammatory) and M2 (anti-inflammatory).

2. Targeting Molecules and Signaling Pathways in Neuroinflammation Intervention

2.1. Nuclear Factor Kappa B (NF-κB): A Central Player in the Development of Neuroinflammation and Neurodegenerative Conditions

In 1986, Sen and Baltimore discovered that in B lymphocytes, a protein binds to a specific Deoxyribonucleic Acid (DNA) sequence called NF-κB that will act in case of extensive inflammation [76]. NF-κB has the property of transcribing genes, which may be pro-inflammatory, triggering a series of events related to neuroinflammation [77]. The two catalytic subunits, Inhibitory Kappa B (IκB) Kinase Alpha (IKKα) and IκB Kinase Beta (IKKβ), and the regulatory subunit NF-κB Essential Modulator (NEMO) comprise the IκB Kinase Complex (IKK) [78]. Two different pathways activate NF-κB, so the canonical pathway is primarily responsible for inflammation and is stimulated by Toll-Like Receptors (TLRs). First, IκBα captures the p65/p50 dimers in the cytoplasm. Consequently, certain pro-inflammatory stimuli, such as pathogens or cytokines, release p65/p50 dimers through the phosphorylation cascade so that IκBα is degraded. Soon after, the cognate kB motif is linked to p65/p50 translocated in the nucleus, activating the NF-κB gene [77]. The regulatory protein Myeloid Differentiation Primary Response 88 (MyD88) mediates the canonical process [78]. In this sense, there is a negative feedback mechanism since the temporary activation of NF-κB is related to the induction of negative regulators, such as IκBα, A20, and p105 [79]. The non-canonical NF-κB pathway has RelB/p52 as the transcription factor and maintains an intimate relationship with IKKα homodimers, considering that it depends on them to occur [79]. Interestingly, in certain types of cells, some members of NF-κB, such as RELA, associate with p100 and are activated through the induction of p100 processing [80]. In this context, regulating this pathway is only possible through NF-κB-Inducing Kinase (NIK) expression. Thus, NIK recruitment to Tumor Necrosis Factor Receptor-Associated Factor (TRAF) 2 at a steady state is mediated through TRAF3, resulting in the ubiquitination of NIK and its consequent degradation. Therefore, the NF-κB complex remains inactive in the cytoplasm due to low levels of endogenous NIK. Upon activation of the non-canonical NF-κB pathway, TRAF3 proteolysis is induced by TRAF2. This degradation of TRAF3 releases and accumulates NIK. After this, phosphorylation of p100 by IKKα heterodimers is induced by NIK, promoting an incomplete deterioration that makes p52 available. Ultimately, p52-RelB heterodimers translocate to the nucleus, allowing target genes to be transcribed [81].
Since the host may be exposed to pathogens that cause tissue damage and infections, inflammation is a protective response of the body capable of promoting the degradation of pathogens and wound healing [80]. In the case of neuroinflammation, the CNS promotes an immune response, which is sometimes related to the etiology and course of several psychiatric and NDs, such as Alzheimer’s disease, Parkinson’s disease, and Multiple Sclerosis. In this sense, NF-κB in the glial cells has a central role in the regulation of chronic neuroinflammation, which is neurodegenerative [82,83], bearing in mind that various diseases can be exacerbated or intensified in cases of uncontrolled inflammation [84,85].

2.2. Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2): A Novel Approach to Address Oxidative Stress and Neuroinflammation in Neurodegenerative Disorders

It is essential to understand that Nrf2, discovered in 1994 [86], remains low until induced by an oxidative increase inside the cell, resulting in the degradation of this protein and its movement to the nucleus. In principle, Nrf2 is an essential Leucine Zipper Protein (bZIP) that belongs to the Cap ’n’ Collar (CNC) family of transcription factors, which also comprises NRF1, NRF3, and p45 NF-E2 and 605 amino acids [87,88,89]. It is interesting to note that Nrf2 exerts protective action against inflammation by inhibiting NF-κB [90]. While NF-κB is extensively studied for its role in promoting neuroinflammation, its crosstalk with the Nrf2 pathway—a critical regulator of antioxidant responses—remains underexplored in disease-specific contexts. Understanding how these pathways influence each other could unveil novel therapeutic strategies, especially given that Nrf2 activation can suppress NF-κB-driven inflammation through antioxidant mechanisms. Exploring this interplay might be crucial for developing integrated approaches to managing neuroinflammatory conditions. In an oxidative stress scenario, Nrf2 is the most important regulator of phase II genes and binds to Antioxidant Response Elements (AREs). Establishing a Kelch-Like Epichlorohydrin-Related Protein (Keap1)/Nrf2 complex in the cytoplasm makes it possible to affirm that the Nrf2/ARE pathway is organized by the negative regulator Keap1. Nrf2 phosphorylation is induced through Keap1 so that it forms heterodimers (the small musculoaponeurotic fibrosarcoma oncogene homolog Nrf2-Maf) after being translocated to the nucleus [91,92,93]. Some pathways indicate that phosphorylation is occurring, and they are the ones that activate Nfr2: Phosphatidylinositol 3-Kinase (PI3K), Casein Kinase 2 (CK2), Protein Kinase C (PKC), and Mitogen-Activated Protein Kinase (MAPK) [94]. The control of many antioxidant pathways is carried out through Nfr2 target genes. One can mention, as an antioxidant pathway, the synthesis and regeneration of Glutathione (GSH) and the synthesis and recovery of thioredoxin [95].
Given the relevance of Nrf2 for numerous processes in the body, it is crucial to highlight its relationship with neuroinflammation and neurodegeneration. Firstly, it is essential to point out that Nrf2 is expressed in the CNS, which involves cells such as microglia and neurons [96]. Microglial cells play a central role in neuroinflammation, and through two distinct activation pathways, they change from the M0 state (resting state) to the M1 or M2 state (activated state). The M1 state is pro-inflammatory and provides pro-inflammatory cytokines, including Tumor Necrosis Factor-Alpha [TNF-α] and interleukin [IL]-6, increasing the amount of tissue damage through the generation of ROS [84,97,98]. Given this, increased levels of oxidative markers and signaling, in addition to constituent parts of damaged cells, could be seen in patients with diseases such as Parkinson’s disease, Multiple Sclerosis, and Alzheimer’s disease [89,99]. Therefore, activating the Nrf2-ARE pathway induces the action of antioxidant proteins and detoxifying enzymes, which defend the body from inflammatory damage [100]. In this scenario, Nrf2 has activators that can neutralize the injuries caused by ROS that promote neurodegeneration [101].

2.3. Impact of the Nucleotide-Binding and Oligomerization Domain-like Receptor Family Pyrin Domain-Containing 3 (NLRP3) Inflammasome on Neuroinflammation: Exploring a Promising Therapeutic Target for Neuroinflammation

There are three mechanisms of NLRP3 activation: canonical, non-canonical, and alternative pathways. However, before canonical activation, the NLRP3 inflammasome must be prepared to control the expression of components present in it, such as NLRP3 itself, pro-IL-1β, and procaspase-1 [102]. In the first stage, which is called the priming stage or signal one and requires the activation of MyD88 [103], specific Pathogen-Associated Molecular Patterns (PAMPs) and Damage-Associated Molecular Patterns (DAMPs) relate to Pattern-Recognition Receptors (PRRs), such as Nucleotide-Binding Domain and Leucine-Rich Repeat Containing Receptors (NLRs), Nucleotide-Binding Oligomerization Domain-Like Receptors (NODs), or TLRs. It promotes an intense induction and transcription of NF-κB, which causes an increase in the expression of the NLRP3 protein. After this, the deubiquitination of NLRP3 and the ubiquitination and phosphorylation of the Apoptosis-Associated Speck-Like Protein (ASC) occur, which allows the assembly of the inflammasome. Furthermore, this step is responsible for inducing the Post-Transductional Modifications (PTMs) of NLRP3, which make NLRP3 stable in a state without activity and self-suppressed but with an appropriate signal [104,105,106,107,108]. Following the activation step, the NLRP3 oligomer, pro-caspase 1, and ASC are prepared to structure a complex during oligomerization [109]. This phase is known as inflammasome assembly or signal 2, and it is activated by certain molecular and cellular stimuli, such as PAMPs, DAMPs, Adenosine Triphosphate (ATP), and ion influx. In this scenario, the pro-inflammatory cytokines IL-18 and IL-1β and the pyroptosis effector protein, Gasdermin-D (GSDMD), are cleaved by the pro-caspase-1 enzyme so that IL-18 and IL-1β are released extracellularly after the N-terminal fragments of GSDMD create pores in the plasma membrane. It is also important to point out that ionic and osmotic flow occurs through these pores, and the influx of calcium (Ca2+) and efflux of potassium (K+) promote cell turgidity so that the influx of Ca2+ releases cellular vesicles and the efflux of K+ promotes an intensification of inflammasome activation [110,111,112,113,114,115]. Furthermore, the adaptive immune response is induced by IL-18 and IL-1β [116]. In humans, caspase-4 triggers a non-canonical response, which occurs as a reaction after Gram-negative bacteria infect the organism [117]. Unlike the previous pathways, in the alternative path, Lipopolysaccharide (LPS) stimulation induces the maturation and consequent secretion of IL-1, in addition to the activation of caspase-1. Furthermore, this pathway requires Spleen Tyrosine Kinase (Syk) activity, caspase 4/5, and Ca2+ flux influenced by LPS and mediated by the Cluster of Differentiation (CD)14/TLR4 [106].
Given the working mechanism of NLRP3, it is possible to state that it has an intrinsic relationship with neuroinflammation and NDs. The activation of NLRP3 inflammasomes in the CNS is related to misfolded proteins and amyloids where, in pathological situations, these proteins have the role of being a signaling molecule that initiates the transcription and expression of NLRP3 and IL-1β, producing a cascade inflammatory character and positive feedback [118]. Studies have shown that patients with Parkinson’s disease have, in their cerebrospinal fluid, high concentrations of IL-1β and IL-18, in addition to an upregulated gene expression of caspase-1, ACS, and NLRP3, as well as increased protein expression of IL-1β, caspase-1, and NLRP3 in cells with only one nucleus in peripheral blood [119,120]. In this sense, the activation of microglial inflammasomes stimulated by Alpha-Synuclein (α-SYN) and TLR is related to the progression of Parkinson’s disease [121,122]. Alzheimer’s disease is also associated with the high production of caspase-1, IL-18, and IL-1β for the activation of the NLRP3 inflammasome induced by Amyloid Beta (Aβ) in microglia during the positive feedback loop, which is produced during systemic neuroinflammation [123,124,125]. Interestingly, a decrease in the Aβ level and an improvement in the memory impairment of APP/PS1/NLRP3 -/- mice is related to a knockout of NLRP3 [126]. Therefore, the NLRP3 inflammasome has gained attention since its relevance in neurodegenerative and neuroinflammatory diseases gained importance in new research [127,128,129].

2.4. Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT): An Evergreen and Unconventional Pathway in Neuroinflammation and Neurological Dysfunctions

The proliferation and activity of cells present in the brain, such as neurons and astrocytes, are regulated by the JAK/STAT pathway. Neural Stem Cell (NSC) growth does not stop during brain development and can be observed in neurogenic areas, including the olfactory bulb’s Subventricular Zone (SVZ). The JAK/STAT pathway regulates NSC growth so that cytokines, especially IL-15, are expressed by adult NSC in the SVZ, stimulating the action of STAT1, STAT3, and STAT5. Interestingly, blocking JAK prevents NSC multiplication [130].
In this scenario, NDs are also strictly linked to the JAK/STAT pathway. Alzheimer’s disease is related to a dysfunction in the JAK2/STAT3 pathway, in which nicotinic acetylcholine receptors activate this pathway and lower the Aβ neurotoxicity [131,132,133,134]. The pathogenesis of Alzheimer’s disease was also related to the inactivation of STAT3, in which its activated/phosphorylated configuration (p-STAT3) had a considerable decline in the hippocampal neurons of patients with the pathology compared to the control group [135]. Studies have shown that in mice, high amounts of Aβ intensify tyrosine phosphorylation and the transcriptional functioning of Tyrosine Kinase 2 (TYK2), which is related to high phosphorylation of STAT3 in brains with Alzheimer’s disease. Furthermore, when inhibited, JAK/STAT3 signaling blocks microglia and astrocytes’ activation in animal neurodegeneration prototypes [136]. The results of Porro et al. demonstrated that inactivation of the JAK/STAT pathway interrupted the amount of secretion of pro-inflammatory cytokines, promoting a rise in the levels of IL-4 and IL-10, causing a change in the protective phenotype and attenuating the inflammation present in neurodegeneration [137].
Through the activation of the JAK/STAT pathway, the polarization of pro-inflammatory macrophages becomes possible, having an intrinsic relationship with Interferon-Gamma (IFN-γ), so that a deficiency in this interferon, presented by mice, demonstrated a reduction in the amount of neuron loss present in the substantia nigra through the induction of 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine (MPTP), which is also related to the Parkinson’s Disease [138]. In their study, Qin et al. [139] demonstrated the relationship between the JAK/STAT pathway in microglial activation, its vital importance in the recruitment of cytokines and chemokines, and its function in Parkinson’s disease. The research investigated a possible therapy using a JAK/STAT pathway blockade through the JAK1 and JAK2 inhibitor AZD1480. The action of α-SYN activated the JAK/STAT pathway in macrophages and microglia, and the role of AZD1480 was to block the Major Histocompatibility Complex Class II (MHC-II), which is induced by α-SYN and, consequently, decreases the activation of STAT1 and STAT3 due to inflammatory gene inhibition in macrophages and microglia. The results showed that therapy with AZD1480 promoted an interruption of neuroinflammation stimulated by α-SYN, preventing microglia activation, the passage of CD4+ T cells and macrophages, and the formation of pro-inflammatory cytokines and chemokines. Notably, in vivo, the JAK/STAT pathway disruption prevented the deterioration of dopaminergic neurons. Therefore, Qin et al. demonstrated the relationship between neuroinflammation, neurodegeneration, and JAK/STAT pathway suppression. In this context, inhibition of the already activated JAK/STAT pathway may be a positive alternative for treating Parkinson’s disease [140].

3. Neuroinflammation and Microglial Activation: Charting the Path Forward for Alzheimer’s Disease, Parkinson’s Disease, and Multiple Sclerosis

It is crucial to understand that between 5 and 10% of cells in the CNS are microglia, and these have different morphologies closely related to their function and the local environment, such as amoeboid, medusa, rod, and branched [141]. It is essential to understand that in a scenario of damage or injury, microglia begin to have an amoeboid morphology with larger cell bodies and smaller branches [142,143]. Furthermore, microglial cells work with the blood-brain barrier (BBB) to defend the brain against damage and attack pathogens [144].
When installed in the brain, self-renewal is how microglia maintain themselves, requiring the action of transcription factors, such as interferon regulatory factor 8 and PU.1, and cytokines, including Colony-Stimulating Factor (CSF)-1 and IL-34 [145]. For microglia to develop and be preserved, some components are essential, namely, Extracellular Signal-regulated Kinases (ERKs), which allow the multiplication and maintenance of microglia, and the Colony-Stimulating Factor 1 Receptor (CSF1-R), which configures as a tyrosine kinase receptor capable of propagating intracellular signals that have a central role in microglial multiplication, and CSF-1 activates it and IL-34 [146,147]. It is essential to point out that accelerated microglia depletion occurs after a CSF-1R blockade by pharmacological means [142]. What differentiates microglial cells from macrophages in the adult brain is the fact that microglia are capable of expressing Purinergic Receptor P2Y12 (P2RY12)/Purinergic Receptor P2Y13 (P2RY13), Transmembrane Protein 119 (TMEM119), and CD11B, with a low expression of CD45, while circulating macrophages reveal a high expression of CD45 and CD11B [148]. By expressing more than 100 sensory genes, microglia can establish a link with their microenvironment. Among these genes, we can mention complementing system receptors, which help them interpret these signals and act as necessary, in addition to TLR2, P2RY12, and Sialic Acid-Binding Ig-like Lectin H (SiglecH) [149].
There are some mechanisms for regulating microglial activity that play a fundamental role and deserve to be highlighted. PRRs are related to regulating innate immunity and recognizing PAMPs and DAMPs. Cytokine receptors are associated with the production of several cytokines, such as Transforming Growth Factor-Beta (TGF-β), TNF-α, and interleukins, since, in the brain, microglia are the largest producer of cytokines. Chemokine Receptors are linked to G Proteins (GPCRs) called CCL1 receptors (CCR1), CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CXCR1, CXCR2, CXCR3, CXCR4, and CXCR5. Neurotransmitter receptors regulate interactivity between neurons and microglia through the recognition of ATP and its metabolites, and it includes cholinergic receptors, glutamate receptors, adrenergic receptors, purinoceptors, and cannabinoid receptors. There is also expression in the microglia of the Triggering Receptor Expressed on Myeloid Cells 2 (TREM2), which is a receptor present on the surface of cells that is part of the extensive family of Immunoglobulins (Igs) and is triggered through the recognition of pathogens by TLRs so that TREM2 establishes a relationship with the adapter DNAX-protein-12 activation (DAP12) present in the internal part of the cell. In addition to these receptors, others participate in this regulation, such as phosphatidylserine, scavenger, and Fc receptors [150,151,152,153].
Microglial cells are classified by a macrophage-derived system that can sometimes be simplistic: M1 (classical) and M2 (alternative) [154]. First, M1, which is activated by IFN-γ and LPS and has a pro-inflammatory and neurodegenerative profile, is related to the formation of nitric acid, proteases, and pro-inflammatory cytokines (IL-6, IL-1β, TNF-α, and IL-12) through the expression of Nicotinamide Adenine Dinucleotide Phosphate Hydrogen (NADPH) oxidase, which produces Inducible Nitric Oxide Synthase (iNOS), and through the action of the STAT1, which acts via JAK1/JAK2 signaling [155,156]. M2, with an anti-inflammatory and healing profile, has the expression of anti-inflammatory cytokines (IL-10), type 2 helper T cell (Th2) recruiting factor CC chemokine ligands, and others [157]. M2 microglia generate neurotrophic growth factors, such as Brain-Derived Neurotrophic Factor (BDNF) [158]. Interestingly, some microglia, as they synthesize pro-inflammatory and anti-inflammatory markers, have an intermediate phenotype. In this context, IL-4 and IL-13 stimulate M2a-type microglia, which incites CD206, arginase, and chitinase 3-like 3. On the other hand, the M2b phenotype is characterized as an intermediate state of microglia due to the secretion of restorative and inflammatory markers. Furthermore, M2c encourages tissue remodeling and repair once the inflammatory issue is resolved, forming CD206, TGF-β, and CD163 [159,160].
Also noteworthy is the ability of microglia to express CD11b, which is known as a “priming” marker, such as MHC-II [161], TMEM119, Sal-like protein 1 (SALL1), P2RY12, and P2RY13, involved in the immune response of the adult brain. Furthermore, it is interesting to note that microglia constantly investigate the CNS to have a relationship with processes such as protein clustering, phagocytosis, the secretion of soluble factors, and the refinement of the neural circuit. In CNS damage, microglia are activated in stages and are complex, responding through the action of signaling molecules, such as chemokines and cytokines, to repair tissue [151,152,162].
It is essential to understand that to protect the vigilant phenotype of microglia and regulate their activity, the CD200-CD200R and CX3CL1-CX3CR1 axes are vital. However, as a result of aging, there is a reduction in communicability through these axes, a fact that is related to the decrease in the expression of CX3CR1 and the neural ligands CX3CL1 and CD200, creating a pro-inflammatory scenario [149,163,164]. Following this line of reasoning, in the brain of an older adult, chronic inflammation promotes immunosuppression that damages the distribution pattern of microglia and causes a severe reduction in branching [164]. Interestingly, only in the white matter are the number of microglia positive for Mac-2, an indicator of subpopulations of microglia that perform phagocytosis, positively regulated, which does not occur in the gray matter. Therefore, certain elderly people called “Superagers,” have fewer microglia in the cortical white matter and, consequently, have better cognitive function [165].
Microglia have an intrinsic relationship with Alzheimer’s disease, as can be analyzed in numerous studies, including that by Sun et al. [166], who observed 1542 genes that were differentially expressed in Alzheimer’s disease, covering changes characteristic of the state of microglia and the Alzheimer’s disease phase, which relate to 12 microglial transcriptional states that have been observed in 443 individuals. By investigating post-mortem brain samples at an advanced age from 217 patients with Alzheimer’s disease and 226 individuals considered controls, the transcriptome of 152,459 unique microglial nuclei was analyzed in six brain areas. The first step was to collect 174,420 immune cells to perform in silico brain screening of Single-Nucleus RNA Sequencing (snRNA-seq) datasets using previously known marker genes (STAR Methods), which formed 16 clusters. Among these, 12 clusters were microglia (0–8 and 10,012). CD74, CSF-1R, and C3 marked them, so they were determined as different microglial states based on their molecular signatures and functionalities. The homeostatic microglia (MG0) and its intense expression of homeostatic markers CX3CR1 and P2RY12 were subject to annotation and observation, as well as the neuron surveillance microglia (MG1), which has a fierce expression of neurotransmitter receptors, and MG3, which demonstrated significant improvement in ribosome biogenesis. Furthermore, three inflammatory states were described (MG2, MG8, and MG10), with an intense expression of both cytokines and cytokine receptors (IL-1B, IL-4R, IL-17R, IL-15, IL-10RA, and CCL3), which are closely related to immune responses, highlighting signaling through the action of cytokines, and the structuring of the inflammatory reaction. From this, the study analyzed 423 individuals considered controls, with early and late Alzheimer’s disease in the prefrontal cortex, evaluating the statistical significance of the divergences in cellular fractions. It was demonstrated that in Alzheimer’s disease, MG4 and MG8 presented fractions with a considerable increase, unlike MG1, which presented a decrease in the fraction. Furthermore, using Ionized Calcium-Binding Adaptor Molecule 1(Iba1) pan-macrophage markers used in immunohistochemistry, it was possible to observe that individuals with Alzheimer’s disease have nuclei that express P2RY12 with comparatively smaller fractions. With the analysis of the increased expression of Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) evidenced in snRNA-seq data, an increased amount was observed in the fraction of PPARγ+P2RY12+ cells in Alzheimer’s disease patients. Linked to this is an enrichment of Forkhead Box P1 (FOXP1)+ Leucine-Rich Repeat Kinase 2 (LRRK2)+ microglia in this group, demonstrating an MG8 inflammatory state. The discoveries made through in situ hybridization confirm that inflammatory states and lipid processing states establish a correlation with diseases.
In Parkinson’s disease, dopaminergic neurons present in the Substantia Nigra Pars Compacta (SNpc) degenerate gradually and for a long time, and the filamentous protein α-SYN makes up the protein inclusions that are called Lewy bodies [167,168]. It is essential to understand that α-SYN can be presented as β-sheet-containing fibrils and plays the secondary role of a seed element for the aggregation of α-SYN, with self-aggregation being a pathological function. In Parkinson’s disease, a decrease in α-SYN clearance is related to chaperone-mediated macroautophagy and autophagy, favoring the formation of α-SYN inclusions. In cases where neurons overexpress α-SYN, exosomes are released, which can provoke a premature reaction from microglia [169,170]. Furthermore, via the activation of inflammasomes, especially NLRP3, the secretion of pro-inflammatory cytokines, such as IL-18 and IL-1β, are stimulated by α-SYN aggregates, a fact that establishes a negative correlation with the development of Parkinson’s disease [171,172]. α-SYN, in physiological situations, is also responsible for regulating the storage of dopamine in synaptic vesicles and, consequently, establishes a relationship with the expression and functioning of the Vesicular Monoamine Transporter 2 (VMAT2) in neurons in the substantia nigra, corroborating its relationship with Parkinson’s disease. In the interaction between α-SYN aggregates and VMAT2, a decrease in dopamine vesicular storage space would lead to a deficiency in synaptic transmission and, as an effect, the death of neurons [172,173].
It is necessary to point out that an increase in the functioning of the NLRP3 inflammasome causes a deficit in microglial autophagy in Parkinson’s disease, leading to a change to a neurotoxic condition in microglia [174]. Yan et al. [175] explored the relationship between the microglial NLRP3 inflammasome, the PARKIN gene, and Parkinson’s disease. Through the neuronal expression of NLRP3, PARKIN polyubiquitination occurs and triggers neurodegeneration, so the objective of the research was to understand the function of PARKIN in the activation of NLRP3 using the overexpression of PARKIN in microglia following treatment with LPS. Three tests were performed on PARKIN knockout mice and wild-type mice to qualify motor activity: the open field, rotarod, and pole tests. It was then discovered that in LPS-induced mice, the decrease in PARKIN function caused an abundant assembly of microglial NLRP3, contributing to motor deficiency and a reduction in the number of dopaminergic neurons present in the substantia nigra. Therefore, the evidence suggests that regulating microglial NLRP3 activation by PARKIN via polyubiquitination ameliorates neurodegeneration in Parkinson’s disease.
Multiple Sclerosis is a complex disease of the CNS that is chronic, neurodegenerative, inflammatory, and autoimmune, which affects more than 2 million people worldwide [176,177]. Although the etiology of the pathology still seems uncertain, some conditions are related to a greater risk of developing Multiple Sclerosis, including biological sex, genetic constitution, and geographic factors [178]. Ren et al. [179] demonstrated the relationship between Quaking (Qki), a Ribonucleic Acid (RNA)-binding protein, and the control of phagocytosis carried out by microglia in the process of demyelination and remyelination. The results indicate an intensification in the neuroprotective activity of microglia in cases of CNS injuries and neurodegenerative pathologies, such as Multiple Sclerosis, a fact related to more significant phagocytosis by Qki. Therefore, the essential role of microglia in remyelination was confirmed [180]. Moreover, disruption of the BBB depends on microglial activation. This injury to the BBB is characterized as a recurrent and fundamental aspect of the progression of Multiple Sclerosis, demonstrated by a study by Yu et al. [181], further highlighting a possible manipulation of microglia to protect the BBB and ensure an improvement in inflammatory demyelination.

4. Exploring Medicinal Plants in Neuroinflammation: Comprehensive Insights on Effects, Dosage, Mechanisms, and Clinical Applications

The studies on various medicinal plants and their phytochemicals present a compelling overview of potential therapeutic agents against neuroinflammation. For instance, the research on Cleistocalyx nervosum var. paniala indicates that the extract significantly reduces pro-inflammatory markers, such as Cyclooxygenase-2 (COX-2) and iNOS, while inhibiting the activation of NF-κB and its downstream effects. The effective doses, ranging from 5 to 100 μg/mL, reveal its capability to modulate the inflammatory response in TNF-α- and LPS-stimulated BV-2 cells [58,59]. These findings underscore the potential of this plant as a source of new anti-inflammatory agents, particularly for NDs. However, future research should focus on in vivo models to corroborate these results and rigorous safety assessments to establish a comprehensive profile in clinical settings [182]. The role of the BBB in regulating neuroinflammation and its impact on the efficacy of medicinal plants is a critical but underexplored area. Understanding whether and how bioactive compounds in these plants can cross the BBB is essential to evaluating their potential for treating central nervous system disorders. Although preclinical findings provide valuable mechanistic insights, the absence of a detailed analysis of clinical trials or real-world applications limits the translational potential of these findings. Data from human studies, such as safety, efficacy, and practical implementation, must be considered to bridge the gap between experimental research and clinical practice.
The efficacy of Curcuma longa is well-documented in the context of neuroinflammation, mainly through the action of curcumin and its derivatives. The included studies demonstrate a marked decrease in inflammatory cytokines and a reduction in NF-κB activation in LPS-stimulated BV-2 cells. The doses tested ranged from 12.5 to 200 μg/mL, highlighting the potential for Curcuma longa as a natural alternative for treating neuroinflammation and oxidative stress-related disorders [60,61]. Despite its promise, the variability in effective dosing observed across studies indicates a need for more standardized protocols and clinical trials encompassing diverse populations to comprehensively evaluate its efficacy [183]. The substantial divergence in reported dosages, ranging from 10 to 200 μg/mL in vitro and 50 to 200 mg/kg in vivo, underscores the challenge of translating preclinical findings into clinical applications. Addressing this requires systematic dose-response studies and modeling to establish scalable and clinically relevant dosing protocols. The therapeutic efficacy of medicinal plants is highly influenced by variability in their bioactive compounds, which can arise from environmental factors such as soil quality, climate, and agricultural practices, as well as genetic differences among plant species or cultivars. Critically evaluating these variations is essential to standardize dosages and ensure the reproducibility of results in clinical applications. The use and efficacy of medicinal plants are often influenced by cultural practices and geographic factors, which can impact both the availability and the perception of these remedies. For instance, the legal status and cultural acceptance of Cannabis sativa vary significantly across regions, affecting research, accessibility, and its integration into therapeutic protocols. Understanding these cultural and geographic variations is essential for developing globally applicable guidelines for medicinal plant usage.
Cannabis sativa has garnered attention due to its diverse compounds, including cannabinoids that exhibit anti-inflammatory properties in LPS-stimulated BV-2 cells. The included study indicated significant reductions in pro-inflammatory cytokines while also increasing levels of endogenous cannabinoids, suggesting a dual role in inflammation modulation and pain management [62]. While these findings open avenues for novel treatments targeting neuroinflammation and chronic pain, the study’s limitations, such as sparse clinical evidence and the challenges of standardizing cannabis strains, necessitate further research that focuses on specific cannabinoid profiles and their therapeutic implications in clinical settings. Barbalace et al. [63] used Cannabis essential oils to mitigate neuroinflammation in BV-2 cells. Their results were interesting in modulating NF-κB, NLRP3, and other pro-inflammatory proteins among neuronal cells, and their results are promising since using natural oils may offer new avenues for administration than oral. Jiang et al. [64] explored the effects of phenylpropionamides (PHS) from Cannabis sativa against an animal model of Parkinson’s. The treatment demonstrated efficacy in counteracting α-SYN deposition via enhanced autophagy, and the commercialization of the plant before the foliage stage can be effective in countering legislative deviations associated with Cannabis usage as a narcotic. Borgonetti et al. [65] studied the effects of Cannabis sativa oils in mitigating neuroinflammation during traumatic injuries of the spinal cord. The results were positive, and this could be a significant strategy to counteract neuroinflammatory traumatic injuries without synthetic medications, therefore favoring a more cost-effective and reduced-side-effect treatment. Zhou et al. [66] demonstrated that Cannabis sativa’s effects on memory and learning damage, improving neuroinflammatory mediator levels, could be explored as candidate therapies against Alzheimer’s or other dementias. However, the study possesses limitations since it does not address the signaling pathways associated with Cannabis’s effects against neuroinflammation and brain defects in the animal model. Shin et al. [67] went beyond this. They demonstrated the effects of Cannabis sativa’s inflorescence against neuroinflammation in deep cervical lymph nodes and the dura mater and depression of an animal model. Their results are promising in evaluating the plant’s effects against neuroinflammation progression involving organs beyond the brain. Effectively, Cannabis reduced depressive-like behaviors and improved the pro-inflammatory cytokines profile of the animals. Mast cells were inhibited from degranulation. However, the signaling pathways involved in the anti-inflammatory and antidepressant effects of the plant remain unknown. Therefore, whether the effects were associated with NF-κB or PPARγ intervention was not fully explicated.
The study on Dioscorea nipponica also reveals a promising mechanism for regulating neuroinflammation through the modulation of iNOS and COX-2 expressions in both in vitro and in vivo models. The effective doses were from 10 to 100 μg/mL for in vitro studies and 60 mg/kg for in vivo applications [68]. Moreover, an increased expression of BDNF and phosphorylated Cyclic AMP Response Element-Binding Protein (pCREB) in animal models indicates potential cognitive benefits. This highlights the need for further research into the long-term effects and safety of Dioscorea nipponica and its primary compound, Dioscin, particularly regarding their impact on human mood disorders.
Research involving Centipeda minima has highlighted its complex phytochemical composition, demonstrating significant anti-inflammatory effects through inhibiting various pathways, including NF-κB and iNOS expression. Li et al. observed a reduction in inflammatory mediators in both in vitro and in vivo settings, reinforcing its potential to develop comprehensive anti-inflammatory therapies against neuroinflammation [69]. However, the variability in phytochemical compositions underscores the need for future studies that systematically evaluate the contributions of individual components, which would enhance the reproducibility and standardization of therapeutic applications.
Atractylodis rhizoma Alba has effectively suppressed inflammatory markers in LPS-stimulated BV-2 cells. The mechanisms involved include the significant inhibition of NF-κB and MAPK signaling pathways critical in the inflammatory response [70]. The promising results call for integrative approaches in clinical applications. Still, the lack of long-term studies means that further investigations are necessary to determine the potential side effects and interactions with other therapeutic agents.
The potential of Vaccinium bracteatum in modulating neuroinflammation is also noteworthy, with studies showing significant reductions in pro-inflammatory cytokines and oxidative stress markers. The effective doses ranged from 2.5 to 20 μg/mL, illustrating its promise as an anti-inflammatory and antioxidant agent [71]. However, variability across studies necessitates additional research to clarify the underlying mechanisms and confirm its efficacy across different biological models and clinical scenarios. Lonicera japonica also exhibits multifaceted anti-inflammatory properties, significantly reducing markers of inflammation in vitro [72]. However, the complex composition of this medicinal plant necessitates further exploration into which specific compounds contribute most significantly to its therapeutic effects. Future studies should aim to isolate these components, evaluate their respective contributions to enhancing anti-inflammatory responses, and determine the optimal dosages for effective therapeutic outcomes.
In Eom et al.’s study, Bambusae caulis demonstrated promising reductions in key inflammatory markers and oxidative stress levels. The observed alterations in Heme Oxygenase-1 (HO-1) and Nrf2 signaling pathways suggest that this plant may hold potential for developing new treatments for neuroinflammation [73]. However, the need for additional studies to identify specific active components and optimize therapeutic protocols remains a priority for future research.
Zingiberis rhizome demonstrates substantial anti-inflammatory effects in vitro on LPS-stimulated BV-2 cells. The administration of 1 or 10 μg/mL of ginger extract for 24 h resulted in a marked reduction of Nitric Oxide (NO) and Prostaglandin E2 (PGE2) production, alongside decreased expression of COX-2 Messenger Ribonucleic Acid (mRNA). Additionally, the treatment led to lower levels of pro-inflammatory cytokines and downregulated the phosphorylation of MAPK signaling molecules (ERK1/2, p38 MAPK, and c-Jun N-terminal kinase [JNK]). Notably, there was also a reduction in NF-κB nuclear translocation, highlighting its potential in modulating neuroinflammatory pathways [74]. Given these promising findings, Zingiberis rhizome may be a valuable candidate for developing neuroinflammation interventions. However, further research is critical to optimize dosing strategies and clarify the underlying mechanisms of action to realize its therapeutic potential.
Lastly, rice is commonly utilized worldwide for cooking. In this scenario, Subedi et al. [75] used genetically engineered resveratrol-enriched rice to counteract neuroinflammation in vitro. The results demonstrated positive alterations in the pro-inflammatory biomarkers and modulation of pro-inflammatory signaling pathways from enriched rice usage. Still, these results are auspicious due to rice’s ubiquity. If future clinical trials demonstrate the effectiveness of this product, its commercialization worldwide could be a reality, and preventing neuroinflammatory disorders and diseases would be easily amplified to the general public.
Overall, while these studies highlight the potential of medicinal plants in regulating neuroinflammation, further research efforts are essential to validate findings across different biological systems, establish optimal dosages, and explore long-term effects in clinical populations. Future research directions could significantly benefit from integrating various advanced methodologies. For instance, genetic studies can be employed to investigate the molecular pathways affected by phytochemicals found in these plants. By utilizing gene expression profiling and RNA sequencing techniques, researchers could elucidate how specific phytochemicals modulate inflammatory pathways at the genomic level, providing insights into their efficacy and mechanism of action.
Immunological assays are also crucial for comprehensively understanding the immune responses elicited by these medicinal plants. Flow cytometry and Enzyme-Linked Immunosorbent Assay (ELISA) could quantify cytokine levels and other immune markers in response to treatment with various plant extracts. These essays could help identify the most effective extracts and dosages that reduce neuroinflammation and enhance neuroprotection. Additionally, investigating the immunomodulatory effects of these plants on various immune cell types, including microglia and astrocytes, could reveal how they influence the neuroinflammatory milieu in the CNS.
Molecular docking studies based on plants’ phytochemical profiles could also provide valuable insights into their potential interactions with key molecular targets involved in neuroinflammation. Using computational modeling, researchers can predict how phytochemicals bind to receptors or the enzymes involved in inflammatory pathways. Such studies could facilitate the identification of promising compounds for further development and help optimize their structures to enhance efficacy and reduce potential side effects.
Moreover, exploring different extracts, such as aqueous, alcoholic, or supercritical fluid extracts, may reveal varying levels of bioactive compounds and their corresponding effects on neuroinflammation. Comparative studies on the efficacy of these extraction methods could uncover the most effective approaches to maximize the therapeutic potential of medicinal plants. Investigating the influence of specific extraction techniques on the stability and bioavailability of phytochemicals will also be necessary, as this could directly impact their clinical applicability.
Finally, integrating nanomedicine and nanocarriers into the research framework presents an exciting avenue for enhancing the delivery of medicinal plant extracts to treat neuroinflammation. Nanocarriers have shown great promise for improving the delivery of plant-based therapies to the CNS. For instance, curcumin-loaded nanoparticles have improved permeability across the blood-brain barrier, enhancing their efficacy against neuroinflammation. Similarly, cannabinoid nanocarriers have shown potential in targeting specific CNS regions to modulate inflammatory pathways. Integrating such examples would enrich discussions on their clinical applicability. Researchers could improve phytochemicals’ solubility, stability, and bioavailability by employing nanoparticles as carriers. Formulating nanocarriers targeting neuroinflammatory sites in the CNS could significantly enhance therapeutic outcomes while minimizing systemic side effects. Furthermore, studies exploring the synergistic effects of combining multiple phytochemicals within nanocarriers could lead to more effective multi-targeted therapies for neuroinflammation.
A multifaceted research approach combining genetic studies, immunological assays, molecular docking, extraction method evaluations, and nanomedicine strategies will advance our understanding of medicinal plants for neuroinflammation. Such efforts can lead to the development of optimized therapeutic protocols that not only leverage the unique properties of each plant but also ensure their safe and practical application in clinical settings. By addressing these areas, future research can provide a more comprehensive understanding of the role of medicinal plants in mitigating neuroinflammation, ultimately contributing to innovative treatment strategies for related neurological disorders.

5. Conclusions and Future Research Directions

Exploring medicinal plants in neuroinflammation has yielded promising insights, revealing their potential as valuable therapeutic agents. Studies on Cleistocalyx nervosum var. paniala, Curcuma longa, and Cannabis sativa demonstrate their ability to modulate inflammatory pathways and provide symptomatic relief for neurodegenerative conditions. However, the included studies present several significant limitations, which must be listed to maintain the transparency of our findings. Firstly, it is worth noting that most of the findings lack sufficient background in animal trials, animal validations, and in-depth safety profiles for the included medicinal plants. The included studies also did not establish essential mechanistic details in some of the included studies, especially with the BBB analysis of the medicinal plants’ component crossing. The included studies reported a variety of effective doses in diverse populations, complicating standardization and reproducibility. In addition, discrepancies between the in vitro and in vivo findings occurred, reflecting inconsistent methodologies in the research. Also, each plant exhibits unique mechanisms of action, from suppressing pro-inflammatory markers to enhancing neuroprotective factors, underscoring their multifaceted therapeutic potential. Figure 2 illustrates the primary mechanisms by which medicinal plants affect neuroinflammation and associated neuroinflammatory disorders.
Despite these encouraging findings, challenges remain. Variability in effective doses, the need for standardized protocols, and the complexity of phytochemical compositions necessitate further research. Standardizing the phytochemical content in plant extracts remains a critical issue, as variability across studies can hinder reproducibility and the clinical translation of findings. Developing universally accepted protocols for extraction and quantification will be pivotal in addressing these inconsistencies and advancing therapeutic applications. Rigorous in vivo studies, comprehensive safety assessments, and standardized clinical trials are essential to validate these findings and establish these plants’ clinical efficacy and safety profiles. Despite the promising therapeutic potential of medicinal plants, their safety profiles, including possible adverse effects and toxicity at varying dosages, remain inadequately addressed. Comprehensive assessments of these factors are crucial to ensure their clinical translation and minimize risks associated with long-term use or interactions with conventional therapies.
Future research should integrate personalized medicine approaches, tailoring treatments based on individual genetic, environmental, and lifestyle factors to enhance therapeutic efficacy and minimize adverse effects. Emerging areas such as epigenetic regulation and gut-brain interactions offer promising insights into neuroinflammation mechanisms. Epigenetic modifications, including DNA methylation and histone acetylation, can influence the expression of key inflammatory genes. Similarly, the gut microbiome impacts neuroinflammation via the gut-brain axis, with microbial metabolites, such as short-chain fatty acids, playing a crucial role in modulating immune responses. Additionally, the principles of green health care—emphasizing sustainability, ecological balance, and the use of natural therapies—should guide the development and application of medicinal plant-based treatments. For example, plants known as adaptogens exhibit complex and nonspecific effects on human health but with an increasing ability to adapt, develop, and survive under stress conditions. This is of particular interest because they can turn nearly ubiquitous, and recent research has addressed these plants as anti-neuroinflammatory, affecting NF-κB signaling and pro-inflammatory cytokines production, including prostaglandins and leukotrienes, therefore, suppressing or delaying their onset NDs and fighting against depression, anxiety, stroke, and even CNS infections [184]. Employing advanced methodologies, including genetic studies to elucidate molecular pathways, immunological assays to evaluate immune responses, and molecular docking studies to predict interactions with key inflammatory targets, will further advance this field. Artificial intelligence (AI) presents innovative tools for drug discovery, particularly in identifying bioactive compounds within medicinal plants. Machine learning algorithms and neural networks can accelerate the identification of phytochemicals with therapeutic potential, optimizing the screening process for anti-inflammatory and neuroprotective agents. Optimizing extraction methods and exploring nanomedicine for improved delivery could also enhance the therapeutic potential of these plants. Comparing the cost-effectiveness and mechanisms of medicinal plants with synthetic drugs is vital for guiding healthcare decisions. Medicinal plants often offer sustainable and affordable alternatives with unique mechanisms, such as turmeric’s anti-inflammatory properties, which may complement or improve synthetic options. In conclusion, a holistic and multifaceted research approach incorporating personalized medicine and green healthcare principles is crucial for advancing our understanding of medicinal plants in neuroinflammation.

Author Contributions

Conceptualization, S.M.B., B.L.B., J.d.S.C.O., J.P., C.B.L., M.T. and L.F.L.; methodology, S.M.B., B.L.B., J.d.S.C.O., J.P., C.B.L., M.T. and L.F.L.; software, S.M.B., B.L.B., J.d.S.C.O., J.P., C.B.L., M.T. and L.F.L.; validation, S.M.B., M.T. and L.F.L.; formal analysis, S.M.B., B.L.B., J.d.S.C.O., J.P., C.B.L., M.T. and L.F.L.; investigation, S.M.B., B.L.B., J.d.S.C.O., J.P., C.B.L., M.T. and L.F.L.; resources, S.M.B., M.T. and L.F.L.; data curation, S.M.B., B.L.B., J.d.S.C.O., J.P., C.B.L., M.T. and L.F.L.; writing—original draft preparation, S.M.B., B.L.B., J.d.S.C.O., J.P., C.B.L., M.T. and L.F.L.; writing—review and editing, S.M.B., B.L.B., J.d.S.C.O., J.P., C.B.L., M.T. and L.F.L.; visualization, S.M.B., M.T. and L.F.L.; supervision, S.M.B., M.T. and L.F.L.; project administration, S.M.B., M.T. and L.F.L.; funding acquisition, S.M.B., M.T. and L.F.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the HUN-REN Hungarian Research Network to M. Tanaka.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We would like to express our sincere gratitude to Servier Medical Art for providing the medical figures used to produce the images for this article. Their high-quality illustrations significantly enhanced the clarity and visual appeal of our work. Servier Medical Art is licensed under CC BY 4.0, allowing for their use with proper attribution. We appreciate their support and contribution to the advancement of scientific communication.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Davinelli, S.; Maes, M.; Corbi, G.; Zarrelli, A.; Willcox, D.C.; Scapagnini, G. Dietary phytochemicals and neuro-inflammaging: From mechanistic insights to translational challenges. Immun. Ageing 2016, 13, 16. [Google Scholar] [CrossRef] [PubMed]
  2. Pan, T.; Xiao, Q.; Fan, H.J.; Xu, L.; Qin, S.C.; Yang, L.X.; Jin, X.M.; Xiao, B.G.; Zhang, B.; Ma, C.G.; et al. Wuzi Yanzong Pill relieves MPTP-induced motor dysfunction and neuron loss by inhibiting NLRP3 inflammasome-mediated neuroinflammation. Metab. Brain Dis. 2023, 38, 2211–2222. [Google Scholar] [CrossRef]
  3. Tanaka, M.; Vécsei, L. A Decade of Dedication: Pioneering Perspectives on Neurological Diseases and Mental Illnesses. Biomedicines 2024, 12, 1083. [Google Scholar] [CrossRef]
  4. Tanaka, M.; Vécsei, L. Revolutionizing our understanding of Parkinson’s disease: Dr. Heinz Reichmann’s pioneering research and future research direction. J. Neural Transm. 2024, 131, 1367–1387. [Google Scholar] [CrossRef]
  5. Rink, C.; Khanna, S. Significance of brain tissue oxygenation and the arachidonic acid cascade in stroke. Antioxid. Redox Signal. 2011, 14, 1889–1903. [Google Scholar] [CrossRef] [PubMed]
  6. Morozumi, T.; Preziosa, P.; Meani, A.; Albergoni, M.; Margoni, M.; Pagani, E.; Filippi, M.; Rocca, M.A. Influence of cardiorespiratory fitness and MRI measures of neuroinflammation on hippocampal volume in multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2024, 95, 29–36. [Google Scholar] [CrossRef] [PubMed]
  7. Abadin, X.; de Dios, C.; Zubillaga, M.; Ivars, E.; Puigròs, M.; Marí, M.; Morales, A.; Vizuete, M.; Vitorica, J.; Trullas, R.; et al. Neuroinflammation in Age-Related Neurodegenerative Diseases: Role of Mitochondrial Oxidative Stress. Antioxidants 2024, 13, 1440. [Google Scholar] [CrossRef]
  8. Solleiro-Villavicencio, H.; Rivas-Arancibia, S. Effect of Chronic Oxidative Stress on Neuroinflammatory Response Mediated by CD4(+)T Cells in Neurodegenerative Diseases. Front. Cell. Neurosci. 2018, 12, 114. [Google Scholar] [CrossRef]
  9. Direito, R.; Barbalho, S.M.; Figueira, M.E.; Minniti, G.; de Carvalho, G.M.; de Oliveira Zanuso, B.; de Oliveira Dos Santos, A.R.; de Góes Corrêa, N.; Rodrigues, V.D.; de Alvares Goulart, R.; et al. Medicinal Plants, Phytochemicals and Regulation of the NLRP3 Inflammasome in Inflammatory Bowel Diseases: A Comprehensive Review. Metabolites 2023, 13, 728. [Google Scholar] [CrossRef]
  10. Teleanu, D.M.; Niculescu, A.G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef]
  11. de Lima, E.P.; Moretti, R.C., Jr.; Torres Pomini, K.; Laurindo, L.F.; Sloan, K.P.; Sloan, L.A.; Castro, M.V.M.; Baldi, E., Jr.; Ferraz, B.F.R.; de Souza Bastos Mazuqueli Pereira, E.; et al. Glycolipid Metabolic Disorders, Metainflammation, Oxidative Stress, and Cardiovascular Diseases: Unraveling Pathways. Biology 2024, 13, 519. [Google Scholar] [CrossRef] [PubMed]
  12. Girotto, O.S.; Furlan, O.O.; Moretti Junior, R.C.; Goulart, R.A.; Baldi Junior, E.; Barbalho-Lamas, C.; Fornari Laurindo, L.; Barbalho, S.M. Effects of apples (Malus domestica) and their derivatives on metabolic conditions related to inflammation and oxidative stress and an overview of by-products use in food processing. Crit. Rev. Food Sci. Nutr. 2024, 1–32. [Google Scholar] [CrossRef] [PubMed]
  13. Valotto Neto, L.J.; Reverete de Araujo, M.; Moretti Junior, R.C.; Mendes Machado, N.; Joshi, R.K.; Dos Santos Buglio, D.; Barbalho Lamas, C.; Direito, R.; Fornari Laurindo, L.; Tanaka, M.; et al. Investigating the Neuroprotective and Cognitive-Enhancing Effects of Bacopa monnieri: A Systematic Review Focused on Inflammation, Oxidative Stress, Mitochondrial Dysfunction, and Apoptosis. Antioxidants 2024, 13, 393. [Google Scholar] [CrossRef] [PubMed]
  14. Silveira Rossi, J.L.; Barbalho, S.M.; Reverete de Araujo, R.; Bechara, M.D.; Sloan, K.P.; Sloan, L.A. Metabolic syndrome and cardiovascular diseases: Going beyond traditional risk factors. Diabetes/Metab. Res. Rev. 2022, 38, e3502. [Google Scholar] [CrossRef]
  15. Tanaka, M.; Szabó, Á.; Vécsei, L. Redefining Roles: A Paradigm Shift in Tryptophan-Kynurenine Metabolism for Innovative Clinical Applications. Int. J. Mol. Sci. 2024, 25, 12767. [Google Scholar] [CrossRef]
  16. de Lima, E.P.; Tanaka, M.; Lamas, C.B.; Quesada, K.; Detregiachi, C.R.P.; Araújo, A.C.; Guiguer, E.L.; Catharin, V.; de Castro, M.V.M.; Junior, E.B.; et al. Vascular Impairment, Muscle Atrophy, and Cognitive Decline: Critical Age-Related Conditions. Biomedicines 2024, 12, 2096. [Google Scholar] [CrossRef] [PubMed]
  17. Simpson, D.S.A.; Oliver, P.L. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef] [PubMed]
  18. Fabisiak, T.; Patel, M. Crosstalk between neuroinflammation and oxidative stress in epilepsy. Front. Cell Dev. Biol. 2022, 10, 976953. [Google Scholar] [CrossRef] [PubMed]
  19. Rodriguez-Lopez, A.; Torres-Paniagua, A.M.; Acero, G.; Díaz, G.; Gevorkian, G. Increased TSPO expression, pyroglutamate-modified amyloid beta (AβN3(pE)) accumulation and transient clustering of microglia in the thalamus of Tg-SwDI mice. J. Neuroimmunol. 2023, 382, 578150. [Google Scholar] [CrossRef]
  20. Laurindo, L.F.; de Carvalho, G.M.; de Oliveira Zanuso, B.; Figueira, M.E.; Direito, R.; de Alvares Goulart, R.; Buglio, D.S.; Barbalho, S.M. Curcumin-Based Nanomedicines in the Treatment of Inflammatory and Immunomodulated Diseases: An Evidence-Based Comprehensive Review. Pharmaceutics 2023, 15, 229. [Google Scholar] [CrossRef]
  21. Tanaka, M.; Battaglia, S.; Giménez-Llort, L.; Chen, C.; Hepsomali, P.; Avenanti, A.; Vécsei, L. Innovation at the Intersection: Emerging Translational Research in Neurology and Psychiatry. Cells 2024, 13, 790. [Google Scholar] [CrossRef]
  22. Tanaka, M.; Chen, C. Editorial: Towards a mechanistic understanding of depression, anxiety, and their comorbidity: Perspectives from cognitive neuroscience. Front. Behav. Neurosci. 2023, 17, 1268156. [Google Scholar] [CrossRef]
  23. 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.; et al. Immunological dimensions of neuroinflammation and microglial activation: Exploring innovative immunomodulatory approaches to mitigate neuroinflammatory progression. Front. Immunol. 2023, 14, 1305933. [Google Scholar] [CrossRef]
  24. Yu, M.; Wang, F.; Han, K. Silencing of SH3BP2 Inhibits Microglia Activation Via the JAK/STAT Signaling in Spinal Cord Injury Models. Inflammation 2024. [Google Scholar] [CrossRef]
  25. Cokdinleyen, M.; Dos Santos, L.C.; de Andrade, C.J.; Kara, H.; Colás-Ruiz, N.R.; Ibañez, E.; Cifuentes, A. A Narrative Review on the Neuroprotective Potential of Brown Macroalgae in Alzheimer’s Disease. Nutrients 2024, 16, 4394. [Google Scholar] [CrossRef] [PubMed]
  26. Battaglia, S.; Avenanti, A.; Vécsei, L.; Tanaka, M. Neurodegeneration in Cognitive Impairment and Mood Disorders for Experimental, Clinical and Translational Neuropsychiatry. Biomedicines 2024, 12, 574. [Google Scholar] [CrossRef] [PubMed]
  27. Hopper, A.T.; Campbell, B.M.; Kao, H.; Pintchovski, S.A.; Staal, R.G.W. Chapter Four—Recent Developments in Targeting Neuroinflammation in Disease. In Annual Reports in Medicinal Chemistry; Desai, M.C., Ed.; Academic Press: Cambridge, MA, USA, 2012; Volume 47, pp. 37–53. [Google Scholar]
  28. Liu, Y.; Yang, H.; Luo, N.; Fu, Y.; Qiu, F.; Pan, Z.; Li, X.; Jian, W.; Yang, X.; Xue, Q.; et al. An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway. J. Transl. Med. 2023, 21, 486. [Google Scholar] [CrossRef]
  29. Bássoli, R.; Audi, D.; Ramalho, B.; Audi, M.; Quesada, K.; Barbalho, S.M. The Effects of Curcumin on Neurodegenerative Diseases: A Systematic Review. J. Herb. Med. 2023, 42, 100771. [Google Scholar] [CrossRef]
  30. Barbalho, S.M.; Direito, R.; Laurindo, L.F.; Marton, L.T.; Guiguer, E.L.; Goulart, R.d.A.; Tofano, R.J.; Carvalho, A.C.; Flato, U.A.P.; Capelluppi Tofano, V.A.; et al. Ginkgo biloba in the aging process: A narrative review. Antioxidants 2022, 11, 525. [Google Scholar] [CrossRef] [PubMed]
  31. de Oliveira Zanuso, B.; Dos Santos, A.R.d.O.; Miola, V.F.B.; Campos, L.M.G.; Spilla, C.S.G.; Barbalho, S.M. Panax ginseng and aging related disorders: A systematic review. Exp. Gerontol. 2022, 161, 111731. [Google Scholar] [CrossRef] [PubMed]
  32. Rangaraju, S.; Dammer, E.B.; Raza, S.A.; Rathakrishnan, P.; Xiao, H.; Gao, T.; Duong, D.M.; Pennington, M.W.; Lah, J.J.; Seyfried, N.T.; et al. Identification and therapeutic modulation of a pro-inflammatory subset of disease-associated-microglia in Alzheimer’s disease. Mol. Neurodegener. 2018, 13, 24. [Google Scholar] [CrossRef]
  33. Blank-Stein, N.; Mass, E. Macrophage and monocyte subsets in response to ischemic stroke. Eur. J. Immunol. 2023, 53, e2250233. [Google Scholar] [CrossRef]
  34. Cotoia, A.; Charitos, I.A.; Corriero, A.; Tamburrano, S.; Cinnella, G. The Role of Macronutrients and Gut Microbiota in Neuroinflammation Post-Traumatic Brain Injury: A Narrative Review. Nutrients 2024, 16, 4359. [Google Scholar] [CrossRef] [PubMed]
  35. Sarsaiya, S.; Jain, A.; Shu, F.; Jia, Q.; Gong, Q.; Wu, Q.; Shi, J.; Chen, J. Unveiling the potential of dendrobine: Insights into bioproduction, bioactivities, safety, circular economy, and future prospects. Crit. Rev. Biotechnol. 2025, 1–19. [Google Scholar] [CrossRef]
  36. Yuan, H.; Ma, Q.; Ye, L.; Piao, G. The Traditional Medicine and Modern Medicine from Natural Products. Molecules 2016, 21, 559. [Google Scholar] [CrossRef] [PubMed]
  37. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [PubMed]
  38. Menezes, A.A.; Shah, Z.A. A Review of the Consequences of Gut Microbiota in Neurodegenerative Disorders and Aging. Brain Sci. 2024, 14, 1224. [Google Scholar] [CrossRef]
  39. Park, K. The Role of Dietary Phytochemicals: Evidence from Epidemiological Studies. Nutrients 2023, 15, 1371. [Google Scholar] [CrossRef] [PubMed]
  40. Buglio, D.S.; Marton, L.T.; Laurindo, L.F.; Guiguer, E.L.; Araújo, A.C.; Buchaim, R.L.; Goulart, R.A.; Rubira, C.J.; Barbalho, S.M. The Role of Resveratrol in Mild Cognitive Impairment and Alzheimer’s Disease: A Systematic Review. J. Med. Food 2022, 25, 797–806. [Google Scholar] [CrossRef]
  41. Barbalho, S.M.; Bueno Ottoboni, A.M.M.; Fiorini, A.M.R.; Guiguer, E.L.; Nicolau, C.C.T.; Goulart, R.d.A.; Flato, U.A.P. Grape juice or wine: Which is the best option? Crit. Rev. Food Sci. Nutr. 2020, 60, 3876–3889. [Google Scholar] [CrossRef] [PubMed]
  42. Laurindo, L.F.; Direito, R.; Bueno Otoboni, A.M.; Goulart, R.A.; Quesada, K.; Barbalho, S.M. Grape processing waste: Effects on inflammatory bowel disease and colorectal cancer. Food Rev. Int. 2024, 40, 336–369. [Google Scholar] [CrossRef]
  43. Sen, T.; Samanta, S.K. Medicinal plants, human health and biodiversity: A broad review. Adv. Biochem. Eng. Biotechnol. 2015, 147, 59–110. [Google Scholar] [CrossRef] [PubMed]
  44. Shen, L.; Tian, Q.; Ran, Q.; Gan, Q.; Hu, Y.; Du, D.; Qin, Z.; Duan, X.; Zhu, X.; Huang, W. Z-Ligustilide: A Potential Therapeutic Agent for Atherosclerosis Complicating Cerebrovascular Disease. Biomolecules 2024, 14, 1623. [Google Scholar] [CrossRef]
  45. Montazeri-Khosh, Z.; Ebrahimpour, A.; Keshavarz, M.; Sheybani-Arani, M.; Samiei, A. Combination therapies and other therapeutic approaches targeting the NLRP3 inflammasome and neuroinflammatory pathways: A promising approach for traumatic brain injury. Immunopharmacol. Immunotoxicol. 2025, 1–17. [Google Scholar] [CrossRef] [PubMed]
  46. Pagotto, G.L.O.; Santos, L.; Osman, N.; Lamas, C.B.; Laurindo, L.F.; Pomini, K.T.; Guissoni, L.M.; Lima, E.P.; Goulart, R.A.; Catharin, V.; et al. Ginkgo biloba: A Leaf of Hope in the Fight against Alzheimer’s Dementia: Clinical Trial Systematic Review. Antioxidants 2024, 13, 651. [Google Scholar] [CrossRef] [PubMed]
  47. Chaachouay, N.; Zidane, L. Plant-Derived Natural Products: A Source for Drug Discovery and Development. Drugs Drug Candidates 2024, 3, 184–207. [Google Scholar] [CrossRef]
  48. Abou Assale, T.; Afrang, N.; Wissfeld, J.; Cuevas-Rios, G.; Klaus, C.; Linnartz-Gerlach, B.; Neumann, H. Neuroprotective role of sialic-acid-binding immunoglobulin-like lectin-11 in humanized transgenic mice. Front. Neurosci. 2024, 18, 1504765. [Google Scholar] [CrossRef]
  49. Kim, Y.; Lim, J.; Oh, J. Taming neuroinflammation in Alzheimer’s disease: The protective role of phytochemicals through the gut-brain axis. Biomed. Pharmacother. 2024, 178, 117277. [Google Scholar] [CrossRef] [PubMed]
  50. Kisioglu, B.; Onal, E.; Karabulut, D.; Onbasilar, I.; Akyol, A. Neuroprotective Roles of Lauric Acid and Resveratrol: Shared Benefits in Neuroinflammation and Anxiety, Distinct Effects on Memory Enhancement. Food Sci. Nutr. 2024, 12, 9735–9748. [Google Scholar] [CrossRef]
  51. Liu, J.; Wang, Y.; Sun, H.; Lei, D.; Liu, J.; Fei, Y.; Wang, C.; Han, C. Resveratrol ameliorates postoperative cognitive dysfunction in aged mice by regulating microglial polarization through CX3CL1/CX3CR1 signaling axis. Neurosci. Lett. 2024, 847, 138089. [Google Scholar] [CrossRef]
  52. Tao, G.; Wang, X.; Wang, J.; Ye, Y.; Zhang, M.; Lang, Y.; Ding, S. Dihydro-resveratrol ameliorates NLRP3 inflammasome-mediated neuroinflammation via Bnip3-dependent mitophagy in Alzheimer’s disease. Br. J. Pharmacol. 2024, 182, 1005–1024. [Google Scholar] [CrossRef] [PubMed]
  53. Hou, B.L.; Wang, C.C.; Liang, Y.; Jiang, M.; Sun, Y.E.; Huang, Y.L.; Ma, Z.L. Analgesic Effect of Dehydrocorydaline on Chronic Constriction Injury-Induced Neuropathic Pain via Alleviating Neuroinflammation. Chin. J. Integr. Med. 2025. [Google Scholar] [CrossRef] [PubMed]
  54. Carles, A.; Freyssin, A.; Guehairia, S.; Reguero, T.; Vignes, M.; Hirbec, H.; Rubinstenn, G.; Maurice, T. Neuroprotection by chronic administration of Fluoroethylnormemantine (FENM) in mouse models of Alzheimer’s disease. Alzheimer’s Res. Ther. 2025, 17, 7. [Google Scholar] [CrossRef]
  55. Battaglia, S.; Avenanti, A.; Vécsei, L.; Tanaka, M. Neural Correlates and Molecular Mechanisms of Memory and Learning. Int. J. Mol. Sci. 2024, 25, 2724. [Google Scholar] [CrossRef] [PubMed]
  56. Tyler, S.E.B.; Tyler, L.D.K. Pathways to healing: Plants with therapeutic potential for neurodegenerative diseases. IBRO Neurosci. Rep. 2023, 14, 210–234. [Google Scholar] [CrossRef]
  57. Suk, K. Regulation of neuroinflammation by herbal medicine and its implications for neurodegenerative diseases. A focus on traditional medicines and flavonoids. Neurosignals 2005, 14, 23–33. [Google Scholar] [CrossRef] [PubMed]
  58. Janpaijit, S.; Sillapachaiyaporn, C.; Theerasri, A.; Charoenkiatkul, S.; Sukprasansap, M.; Tencomnao, T. Cleistocalyx nervosum var. paniala Berry Seed Protects against TNF-α-Stimulated Neuroinflammation by Inducing HO-1 and Suppressing NF-κB Mechanism in BV-2 Microglial Cells. Molecules 2023, 28, 3057. [Google Scholar] [CrossRef]
  59. Janpaijit, S.; Lertpatipanpong, P.; Sillapachaiyaporn, C.; Baek, S.J.; Charoenkiatkul, S.; Tencomnao, T.; Sukprasansap, M. Anti-neuroinflammatory effects of Cleistocalyx nervosum var. paniala berry-seed extract in BV-2 microglial cells via inhibition of MAPKs/NF-κB signaling pathway. Heliyon 2022, 8, e11869. [Google Scholar] [CrossRef] [PubMed]
  60. Kim, K.W.; Lee, Y.S.; Yoon, D.; Kim, G.S.; Lee, D.Y. The ethanolic extract of Curcuma longa grown in Korea exhibits anti-neuroinflammatory effects by activating of nuclear transcription factor erythroid-2-related factor 2/heme oxygenase-1 signaling pathway. BMC Complement. Med. Ther. 2022, 22, 343. [Google Scholar] [CrossRef] [PubMed]
  61. Eun, C.S.; Lim, J.S.; Lee, J.; Lee, S.P.; Yang, S.A. The protective effect of fermented Curcuma longa L. on memory dysfunction in oxidative stress-induced C6 gliomal cells, proinflammatory-activated BV2 microglial cells, and scopolamine-induced amnesia model in mice. BMC Complement. Altern. Med. 2017, 17, 367. [Google Scholar] [CrossRef]
  62. Borgonetti, V.; Benatti, C.; Governa, P.; Isoldi, G.; Pellati, F.; Alboni, S.; Tascedda, F.; Montopoli, M.; Galeotti, N.; Manetti, F.; et al. Non-psychotropic Cannabis sativa L. phytocomplex modulates microglial inflammatory response through CB2 receptors-, endocannabinoids-, and NF-κB-mediated signaling. Phytother. Res. 2022, 36, 2246–2263. [Google Scholar] [CrossRef]
  63. Barbalace, M.C.; Freschi, M.; Rinaldi, I.; Mazzara, E.; Maraldi, T.; Malaguti, M.; Prata, C.; Maggi, F.; Petrelli, R.; Hrelia, S.; et al. Identification of Anti-Neuroinflammatory Bioactive Compounds in Essential Oils and Aqueous Distillation Residues Obtained from Commercial Varieties of Cannabis sativa L. Int. J. Mol. Sci. 2023, 24, 16601. [Google Scholar] [CrossRef]
  64. Jiang, Y.K.; Li, M.M.; Wang, S.Y.; Hao, Z.C.; Meng, X.; Kuang, H.X.; Yang, B.Y.; Liu, Y. Protective effect of phenylpropionamides in the seed of Cannabis sativa L. on Parkinson’s disease through autophagy. Fitoterapia 2024, 175, 105883. [Google Scholar] [CrossRef] [PubMed]
  65. Borgonetti, V.; Anceschi, L.; Brighenti, V.; Corsi, L.; Governa, P.; Manetti, F.; Pellati, F.; Galeotti, N. Cannabidiol-rich non-psychotropic Cannabis sativa L. oils attenuate peripheral neuropathy symptoms by regulation of CB2-mediated microglial neuroinflammation. Phytother. Res. 2023, 37, 1924–1937. [Google Scholar] [CrossRef] [PubMed]
  66. Zhou, Y.; Wang, S.; Ji, J.; Lou, H.; Fan, P. Hemp (Cannabis sativa L.) Seed Phenylpropionamides Composition and Effects on Memory Dysfunction and Biomarkers of Neuroinflammation Induced by Lipopolysaccharide in Mice. ACS Omega 2018, 3, 15988–15995. [Google Scholar] [CrossRef] [PubMed]
  67. Shin, J.; Kim, D.U.; Bae, G.S.; Han, J.Y.; Lim, D.W.; Lee, Y.M.; Kim, E.; Kwon, E.; Han, D.; Kim, S. Antidepressant-like Effects of Cannabis sativa L. Extract in an Lipopolysaccharide Model: Modulation of Mast Cell Activation in Deep Cervical Lymph Nodes and Dura Mater. Pharmaceuticals 2024, 17, 1409. [Google Scholar] [CrossRef] [PubMed]
  68. Azam, S.; Kim, Y.S.; Jakaria, M.; Yu, Y.J.; Ahn, J.Y.; Kim, I.S.; Choi, D.K. Dioscorea nipponica Makino Rhizome Extract and Its Active Compound Dioscin Protect against Neuroinflammation and Scopolamine-Induced Memory Deficits. Int. J. Mol. Sci. 2022, 23, 9923. [Google Scholar] [CrossRef] [PubMed]
  69. Li, S.Y.; Zhou, Y.L.; He, D.H.; Liu, W.; Fan, X.Z.; Wang, Q.; Pan, H.F.; Cheng, Y.X.; Liu, Y.Q. Centipeda minima extract exerts antineuroinflammatory effects via the inhibition of NF-κB signaling pathway. Phytomed. Int. J. Phytother. Phytopharm. 2020, 67, 153164. [Google Scholar] [CrossRef] [PubMed]
  70. Jeong, Y.H.; Li, W.; Go, Y.; Oh, Y.C. Atractylodis Rhizoma Alba Attenuates Neuroinflammation in BV2 Microglia upon LPS Stimulation by Inducing HO-1 Activity and Inhibiting NF-κB and MAPK. Int. J. Mol. Sci. 2019, 20, 4015. [Google Scholar] [CrossRef]
  71. Kwon, S.H.; Ma, S.X.; Ko, Y.H.; Seo, J.Y.; Lee, B.R.; Lee, T.H.; Kim, S.Y.; Lee, S.Y.; Jang, C.G. Vaccinium bracteatum Thunb. Exerts Anti-Inflammatory Activity by Inhibiting NF-κB Activation in BV-2 Microglial Cells. Biomol. Ther. 2016, 24, 543–551. [Google Scholar] [CrossRef]
  72. Kwon, S.H.; Ma, S.X.; Hong, S.I.; Lee, S.Y.; Jang, C.G. Lonicera japonica THUNB. Extract Inhibits Lipopolysaccharide-Stimulated Inflammatory Responses by Suppressing NF-κB Signaling in BV-2 Microglial Cells. J. Med. Food 2015, 18, 762–775. [Google Scholar] [CrossRef] [PubMed]
  73. Eom, H.W.; Park, S.Y.; Kim, Y.H.; Seong, S.J.; Jin, M.L.; Ryu, E.Y.; Kim, M.J.; Lee, S.J. Bambusae Caulis in Taeniam modulates neuroprotective and anti-neuroinflammatory effects in hippocampal and microglial cells via HO-1- and Nrf-2-mediated pathways. Int. J. Mol. Med. 2012, 30, 1512–1520. [Google Scholar] [CrossRef] [PubMed]
  74. Jung, H.W.; Yoon, C.H.; Park, K.M.; Han, H.S.; Park, Y.K. Hexane fraction of Zingiberis Rhizoma Crudus extract inhibits the production of nitric oxide and proinflammatory cytokines in LPS-stimulated BV2 microglial cells via the NF-kappaB pathway. Food Chem. Toxicol. 2009, 47, 1190–1197. [Google Scholar] [CrossRef]
  75. Subedi, L.; Baek, S.H.; Kim, S.Y. Genetically Engineered Resveratrol-Enriched Rice Inhibits Neuroinflammation in Lipopolysaccharide-Activated BV2 Microglia Via Downregulating Mitogen-Activated Protein Kinase-Nuclear Factor Kappa B Signaling Pathway. Oxid. Med. Cell Longev. 2018, 2018, 8092713. [Google Scholar] [CrossRef]
  76. Sen, R.; Baltimore, D. Inducibility of kappa immunoglobulin enhancer-binding protein Nf-kappa B by a posttranslational mechanism. Cell 1986, 47, 921–928. [Google Scholar] [CrossRef]
  77. Sun, E.; Motolani, A.; Campos, L.; Lu, T. The Pivotal Role of NF-kB in the Pathogenesis and Therapeutics of Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 8972. [Google Scholar] [CrossRef] [PubMed]
  78. Nennig, S.E.; Schank, J.R. The Role of NFkB in Drug Addiction: Beyond Inflammation. Alcohol. Alcohol. 2017, 52, 172–179. [Google Scholar] [CrossRef] [PubMed]
  79. Yu, H.; Lin, L.; Zhang, Z.; Zhang, H.; Hu, H. Targeting NF-kappaB pathway for the therapy of diseases: Mechanism and clinical study. Signal Transduct. Target. Ther. 2020, 5, 209. [Google Scholar] [CrossRef] [PubMed]
  80. Sun, S.C. The non-canonical NF-kappaB pathway in immunity and inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef]
  81. Shih, R.H.; Wang, C.Y.; Yang, C.M. NF-kappaB Signaling Pathways in Neurological Inflammation: A Mini Review. Front. Mol. Neurosci. 2015, 8, 77. [Google Scholar] [CrossRef] [PubMed]
  82. Zusso, M.; Lunardi, V.; Franceschini, D.; Pagetta, A.; Lo, R.; Stifani, S.; Frigo, A.C.; Giusti, P.; Moro, S. Ciprofloxacin and levofloxacin attenuate microglia inflammatory response via TLR4/NF-kB pathway. J. Neuroinflamm. 2019, 16, 148. [Google Scholar] [CrossRef] [PubMed]
  83. Shabab, T.; Khanabdali, R.; Moghadamtousi, S.Z.; Kadir, H.A.; Mohan, G. Neuroinflammation pathways: A general review. Int. J. Neurosci. 2017, 127, 624–633. [Google Scholar] [CrossRef] [PubMed]
  84. Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms underlying inflammation in neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef] [PubMed]
  85. Caetano-Silva, M.E.; Rund, L.A.; Vailati-Riboni, M.; Pacheco, M.T.B.; Johnson, R.W. Copper-Binding Peptides Attenuate Microglia Inflammation through Suppression of NF-kB Pathway. Mol. Nutr. Food Res. 2021, 65, e2100153. [Google Scholar] [CrossRef] [PubMed]
  86. Badenetti, L.; Manzoli, R.; Rubin, M.; Cozza, G.; Moro, E. Monitoring Nrf2/ARE Pathway Activity with a New Zebrafish Reporter System. Int. J. Mol. Sci. 2023, 24, 6804. [Google Scholar] [CrossRef]
  87. Sandberg, M.; Patil, J.; D’Angelo, B.; Weber, S.G.; Mallard, C. NRF2-regulation in brain health and disease: Implication of cerebral inflammation. Neuropharmacology 2014, 79, 298–306. [Google Scholar] [CrossRef] [PubMed]
  88. Wei, Z.; Zhao, J.; Zhang, L.; Xia, M. Cell-Based Assays to Identify Modulators of Nrf2/ARE Pathway. Methods Mol. Biol. 2022, 2474, 59–69. [Google Scholar] [CrossRef]
  89. Sivandzade, F.; Prasad, S.; Bhalerao, A.; Cucullo, L. NRF2 and NF-қB interplay in cerebrovascular and neurodegenerative disorders: Molecular mechanisms and possible therapeutic approaches. Redox Biol. 2019, 21, 101059. [Google Scholar] [CrossRef] [PubMed]
  90. Dordoe, C.; Wang, X.; Lin, P.; Wang, Z.; Hu, J.; Wang, D.; Fang, Y.; Liang, F.; Ye, S.; Chen, J.; et al. Non-mitogenic fibroblast growth factor 1 protects against ischemic stroke by regulating microglia/macrophage polarization through Nrf2 and NF-kappaB pathways. Neuropharmacology 2022, 212, 109064. [Google Scholar] [CrossRef]
  91. Zhang, M.; An, C.; Gao, Y.; Leak, R.K.; Chen, J.; Zhang, F. Emerging roles of Nrf2 and phase II antioxidant enzymes in neuroprotection. Prog. Neurobiol. 2013, 100, 30–47. [Google Scholar] [CrossRef]
  92. Zhang, Q.; Liu, J.; Duan, H.; Li, R.; Peng, W.; Wu, C. Activation of Nrf2/HO-1 signaling: An important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress. J. Adv. Res. 2021, 34, 43–63. [Google Scholar] [CrossRef] [PubMed]
  93. Zhou, J.; Zheng, Q.; Chen, Z. The Nrf2 Pathway in Liver Diseases. Front. Cell Dev. Biol. 2022, 10, 826204. [Google Scholar] [CrossRef] [PubMed]
  94. Taguchi, K.; Motohashi, H.; Yamamoto, M. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution. Genes Cells 2011, 16, 123–140. [Google Scholar] [CrossRef]
  95. Dayalan Naidu, S.; Muramatsu, A.; Saito, R.; Asami, S.; Honda, T.; Hosoya, T.; Itoh, K.; Yamamoto, M.; Suzuki, T.; Dinkova-Kostova, A.T. C151 in KEAP1 is the main cysteine sensor for the cyanoenone class of NRF2 activators, irrespective of molecular size or shape. Sci. Rep. 2018, 8, 8037. [Google Scholar] [CrossRef] [PubMed]
  96. Song, X.; Long, D. Nrf2 and Ferroptosis: A New Research Direction for Neurodegenerative Diseases. Front. Neurosci. 2020, 14, 267. [Google Scholar] [CrossRef] [PubMed]
  97. Mohan, S.; Gupta, D. Crosstalk of toll-like receptors signaling and Nrf2 pathway for regulation of inflammation. Biomed. Pharmacother. 2018, 108, 1866–1878. [Google Scholar] [CrossRef] [PubMed]
  98. Cores, Á.; Piquero, M.; Villacampa, M.; León, R.; Menéndez, J.C. NRF2 Regulation Processes as a Source of Potential Drug Targets against Neurodegenerative Diseases. Biomolecules 2020, 10, 904. [Google Scholar] [CrossRef]
  99. Gan, L.; Johnson, J.A. Oxidative damage and the Nrf2-ARE pathway in neurodegenerative diseases. Biochim. Biophys. Acta 2014, 1842, 1208–1218. [Google Scholar] [CrossRef] [PubMed]
  100. He, W.J.; Lv, C.H.; Chen, Z.; Shi, M.; Zeng, C.X.; Hou, D.X.; Qin, S. The Regulatory Effect of Phytochemicals on Chronic Diseases by Targeting Nrf2-ARE Signaling Pathway. Antioxidants 2023, 12, 236. [Google Scholar] [CrossRef] [PubMed]
  101. Amoroso, R.; Maccallini, C.; Bellezza, I. Activators of Nrf2 to Counteract Neurodegenerative Diseases. Antioxidants 2023, 12, 778. [Google Scholar] [CrossRef] [PubMed]
  102. Seok, J.K.; Kang, H.C.; Cho, Y.Y.; Lee, H.S.; Lee, J.Y. Therapeutic regulation of the NLRP3 inflammasome in chronic inflammatory diseases. Arch. Pharm. Res. 2021, 44, 16–35. [Google Scholar] [CrossRef]
  103. Malik, A.; Kanneganti, T.D. Inflammasome activation and assembly at a glance. J. Cell Sci. 2017, 130, 3955–3963. [Google Scholar] [CrossRef] [PubMed]
  104. Swanson, K.V.; Deng, M.; Ting, J.P. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
  105. Guo, H.; Callaway, J.B.; Ting, J.P. Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat. Med. 2015, 21, 677–687. [Google Scholar] [CrossRef] [PubMed]
  106. Bulte, D.; Rigamonti, C.; Romano, A.; Mortellaro, A. Inflammasomes: Mechanisms of Action and Involvement in Human Diseases. Cells 2023, 12, 1766. [Google Scholar] [CrossRef] [PubMed]
  107. Fu, J.; Wu, H. Structural Mechanisms of NLRP3 Inflammasome Assembly and Activation. Annu. Rev. Immunol. 2023, 41, 301–316. [Google Scholar] [CrossRef]
  108. Song, N.; Li, T. Regulation of NLRP3 Inflammasome by Phosphorylation. Front. Immunol. 2018, 9, 2305. [Google Scholar] [CrossRef]
  109. Wu, N.; Zheng, C.; Xu, J.; Ma, S.; Jia, H.; Yan, M.; An, F.; Zhou, Y.; Qi, J.; Bian, H. Race between virus and inflammasomes: Inhibition or escape, intervention and therapy. Front. Cell Infect. Microbiol. 2023, 13, 1173505. [Google Scholar] [CrossRef] [PubMed]
  110. Anderson, F.L.; Biggs, K.E.; Rankin, B.E.; Havrda, M.C. NLRP3 inflammasome in neurodegenerative disease. Transl. Res. 2023, 252, 21–33. [Google Scholar] [CrossRef]
  111. Paik, S.; Kim, J.K.; Silwal, P.; Sasakawa, C.; Jo, E.K. An update on the regulatory mechanisms of NLRP3 inflammasome activation. Cell Mol. Immunol. 2021, 18, 1141–1160. [Google Scholar] [CrossRef]
  112. Zhan, X.; Li, Q.; Xu, G.; Xiao, X.; Bai, Z. The mechanism of NLRP3 inflammasome activation and its pharmacological inhibitors. Front. Immunol. 2022, 13, 1109938. [Google Scholar] [CrossRef]
  113. Biasizzo, M.; Kopitar-Jerala, N. Interplay Between NLRP3 Inflammasome and Autophagy. Front. Immunol. 2020, 11, 591803. [Google Scholar] [CrossRef] [PubMed]
  114. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed]
  115. Pellegrini, C.; Antonioli, L.; Lopez-Castejon, G.; Blandizzi, C.; Fornai, M. Canonical and Non-Canonical Activation of NLRP3 Inflammasome at the Crossroad between Immune Tolerance and Intestinal Inflammation. Front. Immunol. 2017, 8, 36. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, C.; Zhao, W. NLRP3 Inflammasome-A Key Player in Antiviral Responses. Front. Immunol. 2020, 11, 211. [Google Scholar] [CrossRef]
  117. Zito, G.; Buscetta, M.; Cimino, M.; Dino, P.; Bucchieri, F.; Cipollina, C. Cellular Models and Assays to Study NLRP3 Inflammasome Biology. Int. J. Mol. Sci. 2020, 21, 4294. [Google Scholar] [CrossRef]
  118. Lin, S.; Mei, X. Role of NLRP3 Inflammasomes in Neuroinflammation Diseases. Eur. Neurol. 2020, 83, 576–580. [Google Scholar] [CrossRef]
  119. Han, Q.Q.; Le, W. NLRP3 Inflammasome-Mediated Neuroinflammation and Related Mitochondrial Impairment in Parkinson’s Disease. Neurosci. Bull. 2023, 39, 832–844. [Google Scholar] [CrossRef] [PubMed]
  120. Lu, R.; Zhang, L.; Yang, X. Interaction between autophagy and the NLRP3 inflammasome in Alzheimer’s and Parkinson’s disease. Front. Aging Neurosci. 2022, 14, 1018848. [Google Scholar] [CrossRef] [PubMed]
  121. Soraci, L.; Gambuzza, M.E.; Biscetti, L.; Laganà, P.; Lo Russo, C.; Buda, A.; Barresi, G.; Corsonello, A.; Lattanzio, F.; Lorello, G.; et al. Toll-like receptors and NLRP3 inflammasome-dependent pathways in Parkinson’s disease: Mechanisms and therapeutic implications. J. Neurol. 2023, 270, 1346–1360. [Google Scholar] [CrossRef]
  122. Su, Q.; Ng, W.L.; Goh, S.Y.; Gulam, M.Y.; Wang, L.F.; Tan, E.K.; Ahn, M.; Chao, Y.X. Targeting the inflammasome in Parkinson’s disease. Front. Aging Neurosci. 2022, 14, 957705. [Google Scholar] [CrossRef]
  123. Barczuk, J.; Siwecka, N.; Lusa, W.; Rozpedek-Kaminska, W.; Kucharska, E.; Majsterek, I. Targeting NLRP3-Mediated Neuroinflammation in Alzheimer’s Disease Treatment. Int. J. Mol. Sci. 2022, 23, 8979. [Google Scholar] [CrossRef]
  124. Severini, C.; Barbato, C.; Di Certo, M.G.; Gabanella, F.; Petrella, C.; Di Stadio, A.; de Vincentiis, M.; Polimeni, A.; Ralli, M.; Greco, A. Alzheimer’s Disease: New Concepts on the Role of Autoimmunity and NLRP3 Inflammasome in the Pathogenesis of the Disease. Curr. Neuropharmacol. 2021, 19, 498–512. [Google Scholar] [CrossRef] [PubMed]
  125. Liang, T.; Zhang, Y.; Wu, S.; Chen, Q.; Wang, L. The Role of NLRP3 Inflammasome in Alzheimer’s Disease and Potential Therapeutic Targets. Front. Pharmacol. 2022, 13, 845185. [Google Scholar] [CrossRef]
  126. Yang, Z.; Liu, J.; Wei, S.; Deng, J.; Feng, X.; Liu, S.; Liu, M. A novel strategy for bioactive natural products targeting NLRP3 inflammasome in Alzheimer’s disease. Front. Pharmacol. 2022, 13, 1077222. [Google Scholar] [CrossRef]
  127. Deng, C.; Cai, X.; Jin, K.; Wang, Q. Editorial: The NLRP3 inflammasome-mediated neuroinflammation and its related mitochondrial impairment in neurodegeneration. Front. Aging Neurosci. 2022, 14, 1118281. [Google Scholar] [CrossRef] [PubMed]
  128. Zhu, X.; Liu, H.; Wang, D.; Guan, R.; Zou, Y.; Li, M.; Zhang, J.; Chen, J. NLRP3 deficiency protects against hypobaric hypoxia induced neuroinflammation and cognitive dysfunction. Ecotoxicol. Environ. Saf. 2023, 255, 114828. [Google Scholar] [CrossRef] [PubMed]
  129. Yu, Q.; Zhao, T.; Liu, M.; Cao, D.; Li, J.; Li, Y.; Xia, M.; Wang, X.; Zheng, T.; Liu, C.; et al. Targeting NLRP3 Inflammasome in Translational Treatment of Nervous System Diseases: An Update. Front. Pharmacol. 2021, 12, 707696. [Google Scholar] [CrossRef]
  130. Sarapultsev, A.; Gusev, E.; Komelkova, M.; Utepova, I.; Luo, S.; Hu, D. JAK-STAT signaling in inflammation and stress-related diseases: Implications for therapeutic interventions. Mol. Biomed. 2023, 4, 40. [Google Scholar] [CrossRef]
  131. Cardona, K.; Medina, J.; Orrego-Cardozo, M.; Restrepo de Mejía, F.; Elcoroaristizabal, X.; Naranjo Galvis, C.A. Inflammatory gene expression profiling in peripheral blood from patients with Alzheimer’s disease reveals key pathways and hub genes with potential diagnostic utility: A preliminary study. PeerJ 2021, 9, e12016. [Google Scholar] [CrossRef]
  132. Jain, M.; Singh, M.K.; Shyam, H.; Mishra, A.; Kumar, S.; Kumar, A.; Kushwaha, J. Role of JAK/STAT in the Neuroinflammation and its Association with Neurological Disorders. Ann. Neurosci. 2021, 28, 191–200. [Google Scholar] [CrossRef] [PubMed]
  133. Oh, S.L.; Zhou, M.; Chin, E.W.M.; Amarnath, G.; Cheah, C.H.; Ng, K.P.; Kandiah, N.; Goh, E.L.K.; Chiam, K.H. Alzheimer’s Disease Blood Biomarkers Associated With Neuroinflammation as Therapeutic Targets for Early Personalized Intervention. Front. Digit. Health 2022, 4, 875895. [Google Scholar] [CrossRef] [PubMed]
  134. Varma, V.R.; Desai, R.J.; Navakkode, S.; Wong, L.W.; Anerillas, C.; Loeffler, T.; Schilcher, I.; Mahesri, M.; Chin, K.; Horton, D.B.; et al. Hydroxychloroquine lowers Alzheimer’s disease and related dementias risk and rescues molecular phenotypes related to Alzheimer’s disease. Mol. Psychiatry 2023, 28, 1312–1326. [Google Scholar] [CrossRef] [PubMed]
  135. Rusek, M.; Smith, J.; El-Khatib, K.; Aikins, K.; Czuczwar, S.J.; Pluta, R. The Role of the JAK/STAT Signaling Pathway in the Pathogenesis of Alzheimer’s Disease: New Potential Treatment Target. Int. J. Mol. Sci. 2023, 24, 864. [Google Scholar] [CrossRef] [PubMed]
  136. Nevado-Holgado, A.J.; Ribe, E.; Thei, L.; Furlong, L.; Mayer, M.A.; Quan, J.; Richardson, J.C.; Cavanagh, J.; Consortium, N.; Lovestone, S. Genetic and Real-World Clinical Data, Combined with Empirical Validation, Nominate Jak-Stat Signaling as a Target for Alzheimer’s Disease Therapeutic Development. Cells 2019, 8, 425. [Google Scholar] [CrossRef]
  137. Porro, C.; Cianciulli, A.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. Curcumin Regulates Anti-Inflammatory Responses by JAK/STAT/SOCS Signaling Pathway in BV-2 Microglial Cells. Biology 2019, 8, 51. [Google Scholar] [CrossRef]
  138. Yan, Z.; Gibson, S.A.; Buckley, J.A.; Qin, H.; Benveniste, E.N. Role of the JAK/STAT signaling pathway in regulation of innate immunity in neuroinflammatory diseases. Clin. Immunol. 2018, 189, 4–13. [Google Scholar] [CrossRef]
  139. Qin, H.; Buckley, J.A.; Li, X.; Liu, Y.; Fox, T.H., 3rd; Meares, G.P.; Yu, H.; Yan, Z.; Harms, A.S.; Li, Y.; et al. Inhibition of the JAK/STAT Pathway Protects Against α-Synuclein-Induced Neuroinflammation and Dopaminergic Neurodegeneration. J Neurosci. 2016, 36, 5144–5159. [Google Scholar] [CrossRef] [PubMed]
  140. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
  141. Vidal-Itriago, A.; Radford, R.A.; Aramideh, J.A.; Maurel, C.; Scherer, N.M.; Don, E.K.; Lee, A.; Chung, R.S.; Graeber, M.B.; Morsch, M. Microglia morphophysiological diversity and its implications for the CNS. Front. Front. Immunol. Immunol. 2022, 13, 997786. [Google Scholar] [CrossRef] [PubMed]
  142. Gullotta, G.S.; Costantino, G.; Sortino, M.A.; Spampinato, S.F. Microglia and the blood–brain barrier: An external player in acute and chronic neuroinflammatory conditions. Int. J. Mol. Sci. 2023, 24, 9144. [Google Scholar] [CrossRef] [PubMed]
  143. Son, Y.; Yeo, I.-J.; Hong, J.-T.; Eo, S.-K.; Lee, D.; Kim, K. Side-Chain Immune Oxysterols Induce Neuroinflammation by Activating Microglia. Int. J. Mol. Sci. 2023, 24, 15288. [Google Scholar] [CrossRef] [PubMed]
  144. Sun, Y.; Che, J.; Zhang, J. Emerging non-proinflammatory roles of microglia in healthy and diseased brains. Brain Res. Bull. 2023, 199, 110664. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, W.; Jiang, J.; Xu, Z.; Yan, H.; Tang, B.; Liu, C.; Chen, C.; Meng, Q. Microglia-containing human brain organoids for the study of brain development and pathology. Mol. Psychiatry 2023, 28, 96–107. [Google Scholar] [CrossRef] [PubMed]
  146. Colonna, M.; Butovsky, O. Microglia function in the central nervous system during health and neurodegeneration. Annu. Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef] [PubMed]
  147. Matsudaira, T.; Prinz, M. Life and death of microglia: Mechanisms governing microglial states and fates. Immunol. Lett. 2022, 245, 51–60. [Google Scholar] [CrossRef]
  148. Wright-Jin, E.C.; Gutmann, D.H. Microglia as dynamic cellular mediators of brain function. Trends Mol. Med. 2019, 25, 967–979. [Google Scholar] [CrossRef] [PubMed]
  149. Costa, J.; Martins, S.; Ferreira, P.A.; Cardoso, A.M.; Guedes, J.R.; Peça, J.; Cardoso, A.L. The old guard: Age-related changes in microglia and their consequences. Mech. Ageing Dev. 2021, 197, 111512. [Google Scholar] [CrossRef] [PubMed]
  150. Al-Onaizi, M.; Al-Khalifah, A.; Qasem, D.; ElAli, A. Role of microglia in modulating adult neurogenesis in health and neurodegeneration. Int. J. Mol. Sci. 2020, 21, 6875. [Google Scholar] [CrossRef] [PubMed]
  151. D’Alessandro, G.; Marrocco, F.; Limatola, C. Microglial cells: Sensors for neuronal activity and microbiota-derived molecules. Front. Immunol. 2022, 13, 1011129. [Google Scholar] [CrossRef]
  152. Zhang, L.; Wang, Y.; Liu, T.; Mao, Y.; Peng, B. Novel microglia-based therapeutic approaches to neurodegenerative disorders. Neurosci. Bull. 2023, 39, 491–502. [Google Scholar] [CrossRef]
  153. Liu, W.; Taso, O.; Wang, R.; Bayram, S.; Graham, A.C.; Garcia-Reitboeck, P.; Mallach, A.; Andrews, W.D.; Piers, T.M.; Botia, J.A. Trem2 promotes anti-inflammatory responses in microglia and is suppressed under pro-inflammatory conditions. Human. Mol. Genet. 2020, 29, 3224–3248. [Google Scholar] [CrossRef]
  154. Strizova, Z.; Benesova, I.; Bartolini, R.; Novysedlak, R.; Cecrdlova, E.; Foley, L.K.; Striz, I. M1/M2 macrophages and their overlaps—Myth or reality? Clin. Sci. 2023, 137, 1067–1093. [Google Scholar] [CrossRef] [PubMed]
  155. Gao, C.; Jiang, J.; Tan, Y.; Chen, S. Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 359. [Google Scholar] [CrossRef]
  156. Li, J.; Shui, X.; Sun, R.; Wan, L.; Zhang, B.; Xiao, B.; Luo, Z. Microglial Phenotypic Transition: Signaling Pathways and Influencing Modulators Involved in Regulation in Central Nervous System Diseases. Front. Cell. Neurosci. 2021, 15, 736310. [Google Scholar] [CrossRef] [PubMed]
  157. Lis-López, L.; Bauset, C.; Seco-Cervera, M.; Cosín-Roger, J. Is the Macrophage Phenotype Determinant for Fibrosis Development? Biomedicines 2021, 9, 1747. [Google Scholar] [CrossRef]
  158. Gomes, C.; Ferreira, R.; George, J.; Sanches, R.; Rodrigues, D.I.; Gonçalves, N.; Cunha, R.A. Activation of microglial cells triggers a release of brain-derived neurotrophic factor (BDNF) inducing their proliferation in an adenosine A2A receptor-dependent manner: A2A receptor blockade prevents BDNF release and proliferation of microglia. J. Neuroinflamm. 2013, 10, 780. [Google Scholar] [CrossRef] [PubMed]
  159. Russo, C.; Valle, M.S.; Russo, A.; Malaguarnera, L. The interplay between ghrelin and microglia in neuroinflammation: Implications for obesity and neurodegenerative diseases. Int. J. Mol. Sci. 2022, 23, 13432. [Google Scholar] [CrossRef] [PubMed]
  160. Liu, L.; Tong, F.; Li, H.; Bin, Y.; Ding, P.; Peng, L.; Liu, Z.; Dong, X. Maturation, morphology, and function: The decisive role of intestinal flora on microglia: A review. J. Integr. Neurosci. 2023, 22, 70. [Google Scholar] [CrossRef] [PubMed]
  161. Ng, P.Y.; McNeely, T.L.; Baker, D.J. Untangling senescent and damage-associated microglia in the aging and diseased brain. FEBS J. 2023, 290, 1326–1339. [Google Scholar] [CrossRef] [PubMed]
  162. Umpierre, A.D.; Wu, L.J. How microglia sense and regulate neuronal activity. Glia 2021, 69, 1637–1653. [Google Scholar] [CrossRef]
  163. Hristovska, I.; Robert, M.; Combet, K.; Honnorat, J.; Comte, J.; Pascual, O. Sleep decreases neuronal activity control of microglial dynamics in mice. Nat. Commun. 2022, 13, 6273. [Google Scholar] [CrossRef] [PubMed]
  164. Ikegami, A.; Kato, D.; Wake, H. Microglial process dynamics depend on astrocyte and synaptic activity. Nagoya J. Med. Sci. 2023, 85, 772. [Google Scholar]
  165. Ahn, K.; Lee, S.J.; Mook-Jung, I. White matter-associated microglia: New players in brain aging and neurodegenerative diseases. Ageing Res. Rev. 2022, 75, 101574. [Google Scholar] [CrossRef]
  166. Sun, N.; Victor, M.B.; Park, Y.P.; Xiong, X.; Scannail, A.N.; Leary, N.; Prosper, S.; Viswanathan, S.; Luna, X.; Boix, C.A.; et al. Human microglial state dynamics in Alzheimer’s disease progression. Cell 2023, 186, 4386–4403.e29. [Google Scholar] [CrossRef]
  167. Kelly, R.; Joers, V.; Tansey, M.G.; McKernan, D.P.; Dowd, E. Microglial Phenotypes and Their Relationship to the Cannabinoid System: Therapeutic Implications for Parkinson’s Disease. Molecules 2020, 25, 453. [Google Scholar] [CrossRef] [PubMed]
  168. Tanaka, M.; Vécsei, L. Editorial of Special Issue ‘Dissecting Neurological and Neuropsychiatric Diseases: Neurodegeneration and Neuroprotection’. J. Mol. Sci. 2022, 23, 13. [Google Scholar] [CrossRef]
  169. Harry, G.J. Microglia in Neurodegenerative Events-An Initiator or a Significant Other? Int. J. Mol. Sci. 2021, 22, 5818. [Google Scholar] [CrossRef] [PubMed]
  170. Tofaris, G.K. Initiation and progression of α-synuclein pathology in Parkinson’s disease. Cell Mol. Life Sci. 2022, 79, 210. [Google Scholar] [CrossRef] [PubMed]
  171. Toni, M. Special Issue “Neurobiology of Protein Synuclein”. Int. J. Mol. Sci. 2024, 25, 3223. [Google Scholar] [CrossRef]
  172. Badanjak, K.; Fixemer, S.; Smajić, S.; Skupin, A.; Grünewald, A. The Contribution of Microglia to Neuroinflammation in Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 4676. [Google Scholar] [CrossRef] [PubMed]
  173. Basellini, M.J.; Kothuis, J.M.; Comincini, A.; Pezzoli, G.; Cappelletti, G.; Mazzetti, S. Pathological Pathways and Alpha-Synuclein in Parkinson’s Disease: A View from the Periphery. Front. Biosci. 2023, 28, 33. [Google Scholar] [CrossRef] [PubMed]
  174. Xu, Y.; Li, Y.; Wang, C.; Han, T.; Liu, H.; Sun, L.; Hong, J.; Hashimoto, M.; Wei, J. The reciprocal interactions between microglia and T cells in Parkinson’s disease: A double-edged sword. J. Neuroinflamm. 2023, 20, 33. [Google Scholar] [CrossRef] [PubMed]
  175. Yan, Y.Q.; Zheng, R.; Liu, Y.; Ruan, Y.; Lin, Z.H.; Xue, N.J.; Chen, Y.; Zhang, B.R.; Pu, J.L. Parkin regulates microglial NLRP3 and represses neurodegeneration in Parkinson’s disease. Aging Cell 2023, 22, e13834. [Google Scholar] [CrossRef] [PubMed]
  176. Yang, J.; Hamade, M.; Wu, Q.; Wang, Q.; Axtell, R.; Giri, S.; Mao-Draayer, Y. Current and Future Biomarkers in Multiple Sclerosis. Int. J. Mol. Sci. 2022, 23, 5877. [Google Scholar] [CrossRef] [PubMed]
  177. Charabati, M.; Wheeler, M.A.; Weiner, H.L.; Quintana, F.J. Multiple sclerosis: Neuroimmune crosstalk and therapeutic targeting. Cell 2023, 186, 1309–1327. [Google Scholar] [CrossRef] [PubMed]
  178. Mey, G.M.; Mahajan, K.R.; DeSilva, T.M. Neurodegeneration in multiple sclerosis. WIREs Mech. Dis. 2023, 15, e1583. [Google Scholar] [CrossRef]
  179. Ren, J.; Dai, C.; Zhou, X.; Barnes, J.A.; Chen, X.; Wang, Y.; Yuan, L.; Shingu, T.; Heimberger, A.B.; Chen, Y.; et al. Qki is an essential regulator of microglial phagocytosis in demyelination. J. Exp. Med. 2021, 218, e20190348. [Google Scholar] [CrossRef] [PubMed]
  180. Zhang, X.; Chen, F.; Sun, M.; Wu, N.; Liu, B.; Yi, X.; Ge, R.; Fan, X. Microglia in the context of multiple sclerosis. Front. Neurol. 2023, 14, 1157287. [Google Scholar] [CrossRef] [PubMed]
  181. Yu, Z.; Fang, X.; Liu, W.; Sun, R.; Zhou, J.; Pu, Y.; Zhao, M.; Sun, D.; Xiang, Z.; Liu, P.; et al. Microglia Regulate Blood-Brain Barrier Integrity via MiR-126a-5p/MMP9 Axis during Inflammatory Demyelination. Adv. Sci. 2022, 9, e2105442. [Google Scholar] [CrossRef]
  182. Tanaka, M.; Szabó, Á.; Vécsei, L. Preclinical modeling in depression and anxiety: Current challenges and future research directions. Adv. Clin. Exp. Med. Off. Organ Wroc. Med. Univ. 2023, 32, 505–509. [Google Scholar] [CrossRef]
  183. Nunes, Y.C.; Mendes, N.M.; Pereira de Lima, E.; Chehadi, A.C.; Lamas, C.B.; Haber, J.F.S.; Dos Santos Bueno, M.; Araújo, A.C.; Catharin, V.C.S.; Detregiachi, C.R.P.; et al. Curcumin: A Golden Approach to Healthy Aging: A Systematic Review of the Evidence. Nutrients 2024, 16, 2721. [Google Scholar] [CrossRef] [PubMed]
  184. Wróbel-Biedrawa, D.; Podolak, I. Anti-Neuroinflammatory Effects of Adaptogens: A Mini-Review. Molecules 2024, 29, 866. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Microglia have M1 and M2 phenotypes depending on conditions. In a homeostatic state, they support neuronal health through synaptic pruning and clearing misfolded proteins. Under stress, they shift to the pro-inflammatory M1 phenotype, releasing Reactive Oxygen Species (ROS) and cytokines that can damage neurons. When exposed to Transforming Growth Factor Beta (TGF-β), interleukin (IL)-4, IL-10, or IL-13, they switch to the anti-inflammatory M2 phenotype, aiding in phagocytosis, Extracellular Matrix (ECM) rebuilding, and neuronal survival. Abbreviations: ARG, Arginase; BDNF, Brain-Derived Neurotrophic Factor; CCL, Chemokine (C-C motif) Ligand; CD, Cluster of Differentiation; CSF, Colony-Stimulating Factor; FGF, Fibroblast Growth Factor; GNDF, Glial cell line Derived Neurotrophic Factor; IGF, Insulin-like Growth Factor; IFN-γ, Interferon Gamma; iNOS, Inducible Nitric Oxide Synthase; LPS, Lipopolysaccharide; MHC-II, Major Histocompatibility Complex Class II; STAT, Signal Transducer and Activator of Transcription; TNF-α, Tumor Necrosis Factor-Alpha.
Figure 1. Microglia have M1 and M2 phenotypes depending on conditions. In a homeostatic state, they support neuronal health through synaptic pruning and clearing misfolded proteins. Under stress, they shift to the pro-inflammatory M1 phenotype, releasing Reactive Oxygen Species (ROS) and cytokines that can damage neurons. When exposed to Transforming Growth Factor Beta (TGF-β), interleukin (IL)-4, IL-10, or IL-13, they switch to the anti-inflammatory M2 phenotype, aiding in phagocytosis, Extracellular Matrix (ECM) rebuilding, and neuronal survival. Abbreviations: ARG, Arginase; BDNF, Brain-Derived Neurotrophic Factor; CCL, Chemokine (C-C motif) Ligand; CD, Cluster of Differentiation; CSF, Colony-Stimulating Factor; FGF, Fibroblast Growth Factor; GNDF, Glial cell line Derived Neurotrophic Factor; IGF, Insulin-like Growth Factor; IFN-γ, Interferon Gamma; iNOS, Inducible Nitric Oxide Synthase; LPS, Lipopolysaccharide; MHC-II, Major Histocompatibility Complex Class II; STAT, Signal Transducer and Activator of Transcription; TNF-α, Tumor Necrosis Factor-Alpha.
Pharmaceuticals 18 00133 g001
Figure 2. Key actions of medicinal plants in countering neuroinflammation and related disorders. Abbreviations: CREB, Cyclic AMP Response Element-Binding Protein; HO-1, Heme Oxygenase-1; Nrf2, Nuclear Factor Erythroid 2-Related Factor 2.
Figure 2. Key actions of medicinal plants in countering neuroinflammation and related disorders. Abbreviations: CREB, Cyclic AMP Response Element-Binding Protein; HO-1, Heme Oxygenase-1; Nrf2, Nuclear Factor Erythroid 2-Related Factor 2.
Pharmaceuticals 18 00133 g002
Table 1. Medicinal plant effects on neuroinflammation: in vitro and in vivo models, effective dosages, molecular mechanisms, clinical implications, and study limitations.
Table 1. Medicinal plant effects on neuroinflammation: in vitro and in vivo models, effective dosages, molecular mechanisms, clinical implications, and study limitations.
Plant SpeciesOrgan HarvestedMain Bioactive CompoundsModelsEffective Dose(s)/Treatment(s)Mechanisms Clinical ImplicationsRef.
Cleistocalyx nervosum var. panialaBerry seedFerulic acid, aurentiacin, brassitin, ellagic acid, alpinetin, and resveratrolTNF-α-stimulated BV-2 cells in vitro5, 10, or 25 μg/mL CNSE incubated for 24 h in vitro↓ COX-2 activation, ↓ iNOS function, ↓ TNF-α, IL-6 and IL-1β mRNA expression, ↓ p38MAPK and ERK 1/2 phosphorylation, ↓ NF-κB activation, ↓ p65 and IκB phosphorylation, and↑ HO-1 induction in vitroPotential for new anti-inflammatory agents targeting neurodegenerative diseases. Could pave the way for natural, multi-targeted treatments.[58]
Berry seedLPS-stimulated BV-2 cells in vitro1, 5, 10, 25, 50, or 100 μg/mL CNSE incubated for 24 h in vitro↓ NO production, ↓ iNOS mRNA expression, ↓ TNF-α, IL-6, and IL-1β mRNA expression, ↓ MAPK phosphorylation, ↓ p-JNK, p-ERK 1/2 and p-p38 levels, and ↓ NF-κB activation in vitroThis could contribute to developing targeted anti-inflammatory therapies with fewer side effects.[59]
Curcuma longaRhizomeCurcumin, demethoxycurcumin, and bisdemethoxycurcuminLPS-stimulated BV-2 cells in vitro12.5, 25, 50, 100, 150, or 200 μg/mL CLE incubated for 24 h in vitro↓ NO production, ↓ PGE2 production, ↓ iNOS and COX-2 expression, ↓ TNF-α, IL-6 and IL-1β mRNA expression, ↓ NF-κB activation, ↓ IκB-α phosphorylation and degradation, ↓ p65 nuclear translocation, ↓ MAPK (p38, ERK, and JNK) phosphorylation, ↑ HO-1 expression, and ↑ Nrf2 nuclear translocation in vitroIt could enhance treatments for neuroinflammation and oxidative stress-related disorders, offering a natural alternative to synthetic drugs.[60]
RhizomeLPS-stimulated BV-2 cells in vitro and scopolamine-induced male ICR mice in vivo1, 10, 50, 100, or 150 μg/mL FCL incubated for 24 h in vitro and 50, 100, and 200 mg/kg FCL in vivo↓ NO production, ↓ PGE2 production, ↓ iNOS and COX-2 expression, ↑ AChE inhibition in vitro, and ↑ pCREB and BDNF expression in vivoIt may offer new avenues for treating cognitive deficits and memory impairments associated with neurodegenerative conditions.[61]
Cannabis sativaDried inflorescenceCannabidiol, cannabigerol, cannabidiolic acid, tetrahydrocannabinol, β-caryophyllene, caryophyllene-oxide, α-humulene, apigenin, sesquiterpenes (E)-caryophyllene, N-trans-caffeoyloctopamine, N-trans-caffeoyltyramine, cannabisin A, cannabisin B, N-trans-coumaroyltyramine, N-trans-feryroyltyramine, cannabisin C, cannabisin D, cannabisin E, 3,3′-demethyl-grossamide, cannabisin M, isocannabisin N, cannabisin F, and grossamideLPS-stimulated BV-2 cells in vitro1 μg/mL CSE treated for four h in vitro↓ TNF-α, IL-6, and IL-1β production, ↑ AEA and 2-AG expression, ↓ JNK and p38 activation, ↓ NF-κB nuclear translocation, and ↓ ROS production in vitroIt could be a cornerstone for novel treatments targeting neuroinflammation and chronic pain, with potential applications in psychiatric and neurological disorders.[62]
Dried inflorescenceLPS-stimulated BV-2 cells in vitro5 × 10−3 μL/mL GG EO treated for two h in vitro↓ iNOS, NLRP3, and COX-2 protein levels, ↓ NF-κB nuclear translocation, and modulated p38 MAPK and Akt in vitroUsing GG EO from essential oils could be an effective strategy against neuroinflammation targeting via venues other than oral. GG EO from commercial varieties may be attractive for the pharmaceutical industry.[63]
Seeds1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine-induced Parkinson’s disease model in male C57BL/6 J mice in vivo350 or 700 mg/kg PHS from PHS-rich Cannabis sativa seeds in vivoPHS enhanced autophagic biomarkers and improved α-SYN clearance in vivoCannabis sativa seeds can be a valuable strategy to counteract neuroinflammation in Parkinson’s. Cannabis seeds could be a strategy for commercializing the plant before the foliage stage.[64]
Dried inflorescenceLPS-stimulated BV-2 cells in vitro and spared nerve injury CD1 mice model in vivo250 μg/mL treated for four h in vitro and 25 mg/kg cannabidiol-rich non-psychotropic Cannabis sativa in vivo↓ Microglia pro-inflammatory phenotype through HDAC-1 and IκB-α inhibition and ↑ IL-10 expression in vitro and ↓ MAPK in vivoCannabis sativa could be a significant strategy to counteract neuroinflammation in traumatic neuronal injuries.[65]
SeedsLPS-stimulated male Kunming mice in vivo1 and 2 g/kg/day PHS from PHS-rich Cannabis sativa seeds in vivo ↓ IL-1β, IL-6, and TNF-α levels and ↓ hippocampal neuronal damage, and prevented learning and spatial memory damage in vivoCannabis sativa’s effects on memory and learning damages could be explored as candidate therapies against Alzheimer’s or other dementias. [66]
Dried inflorescenceLPS-stimulated male C57BL/6 mice in vivo10, 20, and 30 mg/kg CSL 30 min before LPS administration in vivo ↓ Depressive-like behaviors, ↓ neutrophil-to-lymphocyte ratio, ↓ IL-1β and TNF-α, and inhibited mast cell degranulation in vivoCannabis sativa is effective in treating neuroinflammation beyond the brain and counteracting neuroinflammation in this central organ, preventing depression.[67]
Dioscorea nipponicaRhizomeDioscinLPS-stimulated BV-2 cells in vitro and scopolamine-induced male C57BL mice in vivo10, 20, 50, or 100 μg/mL DNRE and Dioscin 200 and 400 ng/mL treated for two h in vitro and 60 mg/kg Dioscin in vivo↓ iNOS and COX-2 expression, ↓ NO and PGE2 production, ↓ TNF-α, IL-6, and IL-1β mRNA expression, ↓ NF-κB nuclear translocation, ↓ IκB phosphorylation, ↓ p65 nuclear translocation in vitro, and ↑ BDNF and pCREB expression in vivoIt may support treatments to improve cognitive functions and mood disorders by targeting neuroinflammatory pathways.[68]
Centipeda minimaLeavesChlorogenic acid, caffeic acid, rutin, isochlorogenic acid A, isochlorogenic acid B, isochlorogenic acid C, and 6-O-angeloylplenolinLPS-stimulated BV-2 cells in vitro and LPS-stimulated male C57BL/6J mice in vivo2, 4, or 6 μg/mL ECM incubated for 24 h in vitro and 100 and 200 mg/kg ECM in vivo↓ NF-κB nuclear translocation, ↓ IκB phosphorylation, ↓ COX-2 and iNOS expression, ↓ PGE2 production, ↓ NOX proteins in vitro, ↓ PGE2, TNF-α, IL-6, and IL-1β production, ↓ NF-κB nuclear translocation, ↓ iNOS, COX-2, NOX2, and NOX4 expression in vivoPotential to develop comprehensive anti-inflammatory therapies targeting multiple pathways involved in neuroinflammation.[69]
Atractylodis japonica or Atractylodes macrocephala RhizomeAtractylenolide I, atractylenolide III, and atractylodinLPS-stimulated BV-2 cells in vitro10, 50, or 100 μg/mL ARAE incubated for 24 h in vitro↓ NO production, ↓ TNF-α, IL-6, and IL-1β mRNA expression, ↓ iNOS and COX-2 expression, ↑ HO-1 mRNA expression, ↓ NF-κB activity, and ↓ MAPK, p38, ERK, and JNK activation in vitroIt may contribute to integrative approaches for treating neuroinflammation and related conditions.[70]
Vaccinium bracteatumAboveground parts (not specified)Quercetin, chrysin, apigenin, kaempferol, and lutelinLPS-stimulated BV-2 cells in vitro2.5, 5, 10, or 20 µg/mL VBME treated for 30 min in vitro↓ NO and PGE2 production, ↓ iNOS and COX-2 expression, ↓ NF-κB p65 nuclear translocation, ↓ TNF-α, IL-6, and IL-1β levels, and ↓ ROS production in vitroIt may inspire new anti-inflammatory and antioxidant treatments with fewer side effects.[71]
Lonicera japonicaFlower budsChlorogenic acid, caffeic acid, cryptochlorogenic acid, artichoke, isochlorogenic acid A, isochlorogenic acid B, isochlorogenic acid C, rutin, hibisin, and loganinLPS-stimulated BV-2 cells in vitro0.5, 5, 2.5, 5, or 10 µg/mL LJ treated for 30 min in vitro↓ NO and PGE2 production, ↓ iNOS and COX-2 mRNA expression, ↓ TNF-α, IL-1β, MCP-1, and MMP-9 production, ↓ ROS levels, ↓ p38 MAPKs, ERK 1/2, JNK, and PI3K phosphorylation, ↓ JAK1/STAT1/3 phosphorylation, and ↓ NF-κB nuclear translocation in vitroThis could lead to new treatments targeting both neuroinflammation and related oxidative stress.[72]
Phyllostachys nigra var. henonis or Phyllostachys bambusoidesCaulis(-)-7′-epi-lyoniresinol 4,9′-di-O-β-D-glucopyranoside (7), (-)-lyoniresinol 4,9′-di-O-β-D-glucopyranoside (8), and bambulignan ALPS-stimulated BV-2 cells and glutamate-stimulated hippocampal HT22 cells in vitro10, 20, 40, 60, or 80 μg/mL BCE incubated for 24 h in vitro↓ NO, TNF-α, IL-1β, and IL-6 levels, ↓ iNOS and COX-2 expression, ↓ ROS production, ↑ HO-1 mRNA expression, and ↑ Nrf2 nuclear translocation in vitroPotential for advancing treatments against neuroinflammation and oxidative damage through modulation of key inflammatory and oxidative pathways.[73]
Zingiber officinaleRhizomeGingerols and shogaolLPS-stimulated BV-2 cells in vitro1 or 10 μg/mL GHE incubated for 24 h in vitro↓ NO and PGE2 production, ↓ COX-2 mRNA expression, ↓ TNF-α and IL-1β production, ↓ MAPK molecules, ERK1/2, p38 MAPK, and JNK phosphorylation, and ↓ NF-κB nuclear translocation in vitroA promising candidate for developing interventions targeting neuroinflammatory processes and related molecular pathways.[74]
Oryza sativa var. japonicaSeedResveratrolLPS-stimulated BV-2 cells in vitro1–100 μM RR incubated for 24 h in vitro↓ NO, iNOS, and COX-2, MAPK modulation, and ↓ NF-κB translocation in vitroUsing RR could be a strategy to mitigate neuroinflammation widely since this could be a product of world commercialization.[75]
Abbreviations: ↑, Increase; ↓, Decrease; 2-AG, 2-Arachidonoylglycerol; α-SYN, Alpha Synuclein; AChE, Acetylcholinesterase; AEA, Anandamide; Akt, Protein Kinase b; ARAE, Atractylodis rhizoma Alba Ethanolic Extract; BCE, Bambusae caulis in Taeniam Ethyl Acetate; BDNF, Brain-Derived Neurotrophic Factor; CSE, Cannabis sativa Extract; CSL, Cannabis sativa L. inflorescence extract; CLE, Curcuma longa Extract; CNSE, Cleistocalyx nervosum Ethanolic Seed Extract; COX-2, Cyclooxigenase 2; pCREB, cAMP Response Element-Binding protein; DNRE, Dioscorea nipponica Rhizome Ethanol Extract; ECM, Ethanol Extract of C. Minima; ERK, Extracellular Signal-Regulated Kinase; FCL, Fermented Curcuma Longa; GG EO, Gorilla Glue Essential Oil; GHE, Ginger Hexan Extract; HDAC-1, Histone Deacetylase 1; HO-1, Heme Oxygenase 1; IκB, Nuclear Factor of Kappa Light Polypeptide Gene Enhancer in B-Cells Inhibitor; IL-1β, Interleukin 1β; IL-10, Interleukin 10; IL-6, Interleukin 6; iNOS, Inducible Nitric Oxide Synthase; IκB-α, Nuclear Factor of Kappa Light Polypeptide Gene Enhancer in B-Cells Inhibitor Alpha; JAK/STAT, Janus Kinase/Signal Transducer and Activator of Transcription; JNK, C-Jun N-Terminal Kinase; LJ, Lonicera japonica; LPS, Lipopolysaccharide; MAPK, Mitogen-Activated Protein Kinase; MCP-1, Monocyte Chemoattractant Protein-1; MMP-9, Matrix Metalloproteinase 9; mRNA, Messenger Ribonucleic Acid; NLRP3, Nucleotide-Binding and Oligomerization Domain-Like Receptor Family Pyrin Domain-Containing 3; NO, Nitric Oxide; NOX, NADPH Oxidase; Nrf2, Nuclear Factor Erythroid 2-Related Factor 2; NF-κB, Nuclear Factor Kappa B; PGE2, Prostaglandin E2; PHS, Phenylpropionamides; PI3K, Phosphatidylinositol 3-Kinase; RR, Resveratrol-Enriched Rice; ROS, Reactive Oxygen Species; TNF-α, Tumor Necrosis Factor-Alpha; VBME, Vaccinium bracteatum Methanol Extract.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Barbalho, S.M.; Leme Boaro, B.; da Silva Camarinha Oliveira, J.; Patočka, J.; Barbalho Lamas, C.; Tanaka, M.; Laurindo, L.F. Molecular Mechanisms Underlying Neuroinflammation Intervention with Medicinal Plants: A Critical and Narrative Review of the Current Literature. Pharmaceuticals 2025, 18, 133. https://doi.org/10.3390/ph18010133

AMA Style

Barbalho SM, Leme Boaro B, da Silva Camarinha Oliveira J, Patočka J, Barbalho Lamas C, Tanaka M, Laurindo LF. Molecular Mechanisms Underlying Neuroinflammation Intervention with Medicinal Plants: A Critical and Narrative Review of the Current Literature. Pharmaceuticals. 2025; 18(1):133. https://doi.org/10.3390/ph18010133

Chicago/Turabian Style

Barbalho, Sandra Maria, Beatriz Leme Boaro, Jéssica da Silva Camarinha Oliveira, Jiří Patočka, Caroline Barbalho Lamas, Masaru Tanaka, and Lucas Fornari Laurindo. 2025. "Molecular Mechanisms Underlying Neuroinflammation Intervention with Medicinal Plants: A Critical and Narrative Review of the Current Literature" Pharmaceuticals 18, no. 1: 133. https://doi.org/10.3390/ph18010133

APA Style

Barbalho, S. M., Leme Boaro, B., da Silva Camarinha Oliveira, J., Patočka, J., Barbalho Lamas, C., Tanaka, M., & Laurindo, L. F. (2025). Molecular Mechanisms Underlying Neuroinflammation Intervention with Medicinal Plants: A Critical and Narrative Review of the Current Literature. Pharmaceuticals, 18(1), 133. https://doi.org/10.3390/ph18010133

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop