Next Article in Journal
Aberrations of DNA Repair Pathways in Prostate Cancer—The State of the Art
Next Article in Special Issue
The Transcription Factor Nrf2 Mediates the Effects of Antrodia camphorata Extract on Neuropathological Changes in a Mouse Model of Parkinson’s Disease
Previous Article in Journal
Characteristics of the (Auto)Reactive T Cells in Rheumatoid Arthritis According to the Immune Epitope Database
Previous Article in Special Issue
Anti-Inflammatory Activity of 3, 5-Diprenyl-4-hydroxyacetophenone Isolated from Ageratina pazcuarensis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Inflammatory Effects of Flavonoids in Common Neurological Disorders Associated with Aging

by
Hilda Martínez-Coria
1,
Isabel Arrieta-Cruz
2,
Roger Gutiérrez-Juárez
3 and
Héctor Eduardo López-Valdés
1,*
1
Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
2
Department of Basic Research, National Institute of Geriatrics, Ministry of Health, Mexico City 10200, Mexico
3
Department of Biomedical Sciences, School of Medicine, Faculty of Higher Studies Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(5), 4297; https://doi.org/10.3390/ijms24054297
Submission received: 1 January 2023 / Revised: 8 February 2023 / Accepted: 15 February 2023 / Published: 21 February 2023
(This article belongs to the Special Issue Natural Bioactives and Molecular Pathways of Inflammation)

Abstract

:
Aging reduces homeostasis and contributes to increasing the risk of brain diseases and death. Some of the principal characteristics are chronic and low-grade inflammation, a general increase in the secretion of proinflammatory cytokines, and inflammatory markers. Aging-related diseases include focal ischemic stroke and neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Flavonoids are the most common class of polyphenols and are abundantly found in plant-based foods and beverages. A small group of individual flavonoid molecules (e.g., quercetin, epigallocatechin-3-gallate, and myricetin) has been used to explore the anti-inflammatory effect in vitro studies and in animal models of focal ischemic stroke and AD and PD, and the results show that these molecules reduce the activated neuroglia and several proinflammatory cytokines, and also, inactivate inflammation and inflammasome-related transcription factors. However, the evidence from human studies has been limited. In this review article, we highlight the evidence that individual natural molecules can modulate neuroinflammation in diverse studies from in vitro to animal models to clinical studies of focal ischemic stroke and AD and PD, and we discuss future areas of research that can help researchers to develop new therapeutic agents.

1. Introduction

The most common aging-related diseases include focal ischemic stroke and neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Aging is a progressive, irreversible, and inevitable process that involves a distinctive decline in physiological functions and physical appearance, resulting from tissue degeneration and the dysfunction of vital organs [1]. A focal ischemic stroke is caused by the occlusion of a cerebral artery and is the most frequent type of cerebrovascular disease, and aging is its most nonmodifiable risk factor. Neurodegenerative diseases have no cure and are caused by progressive degeneration and/or neuron death to produce debilitating conditions. Neurodegenerative diseases also include Lewy body dementia, vascular dementia, amyotrophic lateral sclerosis, and frontotemporal dementia [2,3]. Meanwhile, the components of the human diet, such as vegetables, cereals, tea, wine, and fruits, contain different compounds including flavonoids, which are a class of polyphenols [4]. Many recent studies have shown that these compounds have a beneficial effect on the primary cell culture of glial and neurons and pre-clinical animal models of human focal ischemic stroke and neurodegenerative diseases. The active compounds and their mechanism of action for most flavonoids in the human diet and herbal medicine are still not well defined because they contain multiple bioactive molecules that can modulate multiple pharmacologic targets. To obtain more significant information, the scientific research has concentrated mainly on known biological activities of purified single compounds to offer an evidence base for the rationale of traditional practice but also to support their integration into modern medical practice [5].
In the sections below, we will describe the basic knowledge of natural flavonoids, neuroinflammation, aging, focal ischemic stroke, AD, and PD, and we will summarize the evidence relating to the anti-inflammatory effects of single flavonoids in different model systems of those diseases and clinical studies, and finally, we will suggest future areas of research to improve our understanding of single flavonoids molecules to help to establish more solid bases to facilitate their future use as a therapeutic alternative.

2. Flavonoids

Flavonoids are present in all vascular plants and are the most common class of polyphenols. In the plant, flavonoids are secondary metabolites that have a wide range of biochemical, physiological, and ecological functions, such as the coloration of petals and flowers, protection against ultraviolet light, and cell growth. Moreover, a single plant often contains many different flavonoids [6]. In nature, flavonoids are broadly distributed in the plant kingdom and are the main phytochemicals found in more than 6000 species of plants, and they also are abundantly found in plant-based foods and beverages, including vegetables, tea, fruits, grains roots, cocoa, and wine [7]. Flavonoids are low-molecular-weight compounds that can be divided into subclasses such as: anthocyanins, chalcones, flavanols (or catechins), flavones, flavanones, flavonols, flavanonols, and isoflavonoids (Figure 1) [7]. Except for catechins, all flavonoids contained in foods and beverages are in the form of glycosides, which must be removed to be absorbed from the small intestine upon ingestion. Flavonoid aglycones undergo conjugation reactions before passing into the bloodstream, and these reactions form sulphates, glucuronides, and/or methylates metabolites (conjugates), which can be subjected to additional phase II metabolism in the liver, and then returned to the circulatory system. Moreover, some conjugates may be exported into the bile duct or excreted by the kidney. Flavonoids not absorbed from the small intestine will be degraded in the colon [8]. Except from catechins, the rest of the flavonoids present in plasma and urine are primarily conjugated forms. Consequently, cells in the body are usually exposed to apparently less active flavonoid metabolites and conjugates, rather than aglycones [9]. However, there is evidence that deconjugation may occur in situ. An in vivo study has shown that glucuronidated quercetin metabolites were deconjugated in their aglycone form in the mesenteric vasculature of rats by the action of β-glucuronidase, and this effect was inhibited when an inhibitor of that enzyme was present [10], which suggests that deconjugation may occur in situ to produce a more effective aglycone form. The absorption and distribution of flavonoids around the body, as well as their excretion in urine, are carried out by members of ATP-binding cassette (ABC) transport systems, which translocate solutes across cell membranes [11]. The physicochemical properties (such as molecular size and lipophilicity, solubility, configuration, and pKa value) of each flavonoid determine the degree of absorption [12]. Flavonoids show great variability in the velocity and magnitude of absorption, plasma half-life, bioavailability, and plasma kinetics, but in general, they show rapid urinary and biliary excretion and low bioavailability, and it has been suggested that after the consumption of 10–100 mg of a single phenolic compound, their plasma concentration rarely exceeds 1 μM [13]. Moreover, Manach et al. [14] analyzed 97 bioavailability studies in humans and found that plasma concentrations of total metabolites ranged from 0 to 4 µmol/L with an intake of 50 mg aglycone equivalents, and the relative urinary excretion ranged from 0.3% to 43% of the ingested dose depending on the specific polyphenol. They also found that among all of the compounds analyzed, isoflavones and gallic acid had the best rate of absorption, followed by catechins, flavanones, and quercetin glycosides, and that proanthocyanidins and anthocyanins had very low bioavailability. Some evidence suggests that some flavonoids and/or their metabolites such as anthocyanins and (−)-epicatechin and some of their metabolites can cross the blood–brain barrier [15,16,17]. Flavonoids have pleiotropic effects that can produce health benefits, as shown in several diseases including cardiovascular diseases, neurological disorders, and cancer, and these benefits are seen in antiviral, antioxidant, and anti-inflammatory mechanisms, among others [6,8].

3. Neuroinflammation

An inflammatory reaction in the central nervous system (CNS) or neuroinflammation are generally induced when innate immune cells detect tissue damage or an infection. This response is critical to isolate damaged tissue from uninjured areas and to repair and clean the extracellular matrix. Acute inflammation is beneficial and promotes regeneration, but chronic and excessive, as well as stable, low-grade, inflammation can produce the onset or exacerbation of cell injury [3]. It is generally accepted that low-grade neuroinflammation is the main factor in the onset and development of several neurological diseases. In aging, the major source for low-grade, chronic inflammation is the accumulation of endogenous host-derived cell debris due to both increased production and impaired elimination, and also immunosenescence, but these alterations are not commonly the initiating factor of neurodegenerative diseases. However, they contribute to amplifying the disease state, which would suggest that neuroinflammation plays an important role in neuronal dysfunction and death [18].
Glial cells in the CNS carry out different activities during neuroinflammation and can promote protection or damage, depending on the particular environments of inflammation and time. All glial cells play a role in the immune response, but the most important ones are the microglia and astrocytes. Microglia is the main immune cell in the CNS. These cells continuously scan the microenvironment of the parenchyma and are the first cells to respond to the occurrence of any damage [18]. In addition to being the most numerous cell type in the CNS, astrocytes have varied homeostatic functions, for example, they control the extracellular pH, antioxidant functions, neurotransmitter uptake, and the recycling of glutamate and GABA, regulate the cerebral blood flow and the blood–brain barrier (BBB), promote synaptogenesis, supply energy metabolites to the neurons, and they form part of the innate immune system of the CNS [19].
In neurological disease, glial cells can recognize endogenous molecules released by damaged or dead cells (damage-associated molecular patterns: DAMPs) and molecules present in pathogens (pathogen-associated molecular patterns: PAMPs) through pattern recognition receptors (PRR), which are formed by several subfamilies including the Toll-like receptors (TLRs) [18,20]. TLRs are type 1 transmembrane glycoproteins that are extensively expressed in microglia and astrocytes, with specific subtypes expressed in neurons and oligodendrocytes [21]. Each TLR subtype recognizes different PAMPs or DAMPs, for example, TLR4 recognizes lipopolysaccharide (LPS), and also, accumulated, misfolded proteins, including Aβ and α-synuclein present in AD and PD, respectively [18]. DAMPs comprise an extensive range of molecules such as uric acid, cytokine IL-1α, ATP, and nuclear and cytoplasmic proteins released during necrosis, and it has also been suggested that some members of the extended IL-1 cytokine family including IL-1β, IL-18, IL-33, IL-36α, IL-36β, and IL-36γ also act as DAMPs and stimulate the sterile inflammation induced by necrosis [22]. The interaction of ligands with the TLR of the host cell activates an intracellular signaling cascade that causes the release of inflammatory cytokines and other immune modulators as a protective mechanism and to repair the damaged tissue. However, excessive TLR activation disrupts immune homeostasis, causing constant proinflammatory cytokine and chemokine production, which contributes to the development and progression of many diseases [21]. After the initial interaction of the ligand and TLR, the latter one activates one of the several signal transduction pathways such as phosphoinositide 3-kinase/protein kinase B (PI3K/AKT), mammalian target of rapamycin (mTOR), or mitogen-activated protein kinase (MAPK), which leads to the activation of different transcription factors such as activator protein 1 (AP-1), nuclear factor kappa-B (NF-κB), nitric oxide synthase (iNOS), interferon regulatory factor 3 (IRF3), and cyclooxygenase-2 (COX-2), which mediate the production of proinflammatory cytokines, chemokines, and inducible enzymes, all of which result in neuroinflammation [23,24]. The activation of the PI3K/AKT/mTOR pathway can activate the transcription factor NF-κB and induce the expression of proinflammatory molecules (e.g., IL-6 and TNF-α), iNOS, and COX-2, while other TLRs can activate the MAPK (p38 MAPK or SAPK/JNK) pathway, which then activates the transcription factor AP-1 and promotes the expression of proinflammatory molecules, including cytokines (e.g., IL-6, iNOS, and COX-2) [24]. The NOD subfamily is another well-known PRR member, which is part of the inflammasome NLRP3, together with the adaptor protein apoptosis-associated speck-like protein comprising a caspase recruitment domain (ASC) and procaspase 1, which together form an oligomer when the stimuli activate the receptor, and thus, induce the active precursors of proinflammatory cytokines, such as IL-1β and IL-18 [25]. Cytokines are the main communication mechanism used by the immune system and consist of polypeptides and glycoproteins synthetized by immune cells such as chemokines, lymphokines, interleukins (IL), tumor necrosis factor (TNF), and interferons (IFN), which can act as pro- and anti-inflammatory molecules [26]. Cytokines can have both anti- and proinflammatory effects. Anti-inflammatory cytokines include: IL-4, IL-6, IL-10, IL-11, IL-13, IL-1 receptor antagonist (IL-1RA), and TGF-β, while the proinflammatory cytokines include IL-1β, IL-6, IL-8, IL-12, TNF-α, and interferons, among others [26]. After secretion to the extracellular space, these molecules interact with cytokine receptors to initiate cytokine intracellular signaling, which modulates a diverse range of biological functions, and most cytokine receptors activate the JAK-STAT pathway [27]. The primary proinflammatory cytokines, such as TNF-α, IL-1α, and IL-6, contribute significantly to inflammaging in healthy elderly people and play a major role in several age-related diseases, including neurodegenerative diseases [22]. Furthermore, TNF-α also induces apoptosis by activating receptors such as tumor necrosis factor receptor 1 (TNFR1), 2 (p75), and CD95 (APO-1/Fas), which contain a homologous cytoplasmic sequence identifying an intracellular death domain [24]. Microglia and astrocytes can release anti- and proinflammatory cytokines, but the type of secretion seems to be related to the specific phenotypes of the glial cells, their interaction with each other, and the specific context. Glial cells show different phenotypes that are the result of transcriptional and functional changes that are generally known as activation or reaction ones. Several different phenotypes have been observed, but M1 for harmful and neurotoxic inflammation functions and M2 for pro-reparative and anti-inflammation functions are considered to be opposite states of reactive microglia, but the microglia can switch between these two phenotypes according to different environments [28]. Similarly, for astrocytes, the A1 phenotype is toxic to neurons and oligodendrocytes, while the A2 phenotype is protective [29,30]. In human neurodegenerative illnesses such as PD and AD, M1 microglia, as well as astrocyte A1, are highly present, and it has been proposed that M1 microglia induces the A1 astrocyte phenotype in these illnesses [31]. M1 microglia and A1 astrocytes secrete proinflammatory cytokines, which can induce apoptosis through the activation of extrinsic pathways and also induce the overproduction of ROS, mitochondrial dysfunction, DNA damage, and the production of more inflammatory mediators that contribute to cell aging, and also, induce the permeabilization of the BBB [32,33]. Most types of CNS diseases have a neuroinflammatory component, and activated microglia and astrocytes have been found [18].

4. Aging

Aging is a complex and inevitable process that results from the interaction of environmental epigenetic and genetic factors that produce a gradual reduction of homeostasis with age. This change is characterized by the progressive degeneration of tissue and functions in several organs, which contributes to increasing the risk of disease and death [34,35]. In the brain, aging affects different cell types and regions differently, and the individual variability is broad, but in general terms, there is a reduction of white and grey matter density, volume loss, cortical thinning, and the atrophy of specific brain regions, including the hippocampus [36]. Age-related diseases or aging-related diseases group different illnesses together, including arthritis, cancer, hypertension, type 2 diabetes, focal ischemic stroke, and many neurodegenerative diseases such as AD and PD. In normal aging, alterations occur in the nervous system to produce functional and structural changes. Functional changes include a decrease in blood flow and a reduction of synapses and neurotransmitter release and several areas, but the area that is most commonly affected is the hippocampus. On the other hand, structural changes include the enlargement of the ventricles, cerebral atrophy, and neuronal loss [37]. It is important to note that endothelial dysfunction is a significant contributor to cerebrovascular aging, which promotes oxidative stress and neuroinflammation in age-related disorders such as dementias and focal ischemic stroke [38]. There is a noticeable change in altered intercellular communication known as ‘inflammaging’, which is characterized by a process of chronic and low-grade inflammation, and a general increase in the secretion of proinflammatory cytokines and inflammatory markers [39]. Additionally, another feature is the presence of cellular senescence in tissues and organs, including the immune system (immunosenescence), producing reduced humoral and cellular responses for both the innate and adaptative immune systems. Senescent cells show increased activity of senescence-associated β-galactosidase (SA-β-GAL), a failure to re-enter the cell cycle in response to mitogenic stimuli, resistance to cell death, and a proinflammatory secretome called senescence-associated secretory phenotype (SASP) [32]. Some alterations such as neuroinflammation, DNA damage, and oxidative stress that are present in neurodegenerative diseases and focal ischemic stroke can induce cellular senescence, and it has been suggested that cellular senescence contributes to the pathophysiology of these disorders [40,41].
Many of the inflammaging characteristics can be found in the blood, for example high levels of proinflammatory cytokines (e.g., IL-1β, IL-6, IL-8, and IL-10) in sera or plasma [26], monocytes and macrophages with impaired phagocytosis and ability to heal injuries, the presence of senescent T cells, impaired macrophage polarization, and antibody production by the activated B cells [42]. In age-related diseases, systemic inflammation may contribute to neuroinflammation because circulating proinflammatory molecules can interact with endothelial cells from cerebral vasculature and induce the release of more cytokines to cause BBB impairment [43]. Oxidative stress is another factor present in neurodegenerative age-related diseases. This alteration is due to the excessive production of unstable molecules that contain oxygen called reactive oxygen species (ROS), and also, because the antioxidant system in the cells is not able to neutralize them. This mitochondrial dysfunction produces DAMPs in the cell, which initiate several inflammatory cascades, causing the activation of the innate immune system and NLRP3 inflammasome, which results in chronic inflammation [18].

5. Basic Physiopathology of Focal Ischemic Stroke, AD, and PD

5.1. Focal Ischemic Stroke

Ischemic strokes or cerebrovascular accidents are the most frequent types of cerebrovascular diseases and are one of the main causes of death and disability worldwide [44]. The main type of strokes is the focal ischemic, which is due to an obstruction of the arterial blood flow to a specific brain region [45]. Aging is considered to be the most important nonmodifiable risk factor, and the risk of a stroke occurring doubles every 10 years after the person reaches the age of 55, and approximately three quarters of all strokes occur in persons aged ≥65 years [46,47]. It is important to highlight that modifiable risk factors such as diabetes and high blood pressure steadily increase with age, and it has been shown that these chronic conditions are very highly prevalent in persons that have suffered strokes [47]. In most countries, the approved treatments for an acute focal ischemic stroke are the endovascular thrombectomy and intravenous recombinant tissue plasminogen activator (rtPA), which focus on removing the occlusion in the artery and saving the penumbra cells to reduce the infarct core enlargement [45], but several conditions must be met to obtain the therapeutic benefits. Endovascular thrombectomy shows benefits when it is applied within the first six hours and rtPA shows benefits when it is applied within the first four and a half hours of stroke onset [48].
The cessation of or decrease in cerebral blood flow may produce different levels of damage depending on several factors such as the time elapsed, cell resistance, and the magnitude of the ischemia, which results in the activation of very complex cascades of cellular and molecular events with a temporal overlapping profile that evolves over minutes, hours, or days, inducing transient-to-irreversible injuries (e.g., cell death) in all cell types and damage. The pathologic cascades produce damage in two different areas: the ischemic core and the penumbra. In the ischemic core, the abrupt decrease in cerebral blood flow leads to permanent damage in the cell and rapid cell death by necrosis. The size of the necrotic area will depend mainly on the location of the stroke, its duration, and its magnitude. The penumbra surrounding the core area is perfused by collateral blood vessels, which help to keep the cell structures intact, but functionally weakened [45]. After a few minutes of cerebral blood vessel occlusion, the first pathological event is activated due to the reduction of oxygen and glucose, which leads to a failure to produce high-energy molecules to maintain cellular homeostasis. This event sets off several mechanisms, which include ionic imbalance, cytotoxic and vasogenic edemas, excitotoxicity, calcium overload, excitotoxicity, oxidative and nitrosative stress, peri-infarct depolarization, blood–brain barrier (BBB) disruption, apoptosis, and inflammation [45,49]. Inflammation responses occur after the damaged cells release DAMPs, which interact with Toll-like receptors in the microglia and astrocytes. After this interaction, the microglia become reactive and accumulate at the lesion core and penumbra. During the first hours after the damage, these reactive microglia have an anti-inflammatory profile, but after this period, they switch to a proinflammatory profile, and several of the proinflammatory mediators (e.g., reactive oxygen species, cytokines, and tumor necrosis factor-α) released can induce astrogliosis from 4 to 24 h after stimulation [50,51,52]. M2 microglia found around the lesion site migrate towards the lesion core and in the penumbra, and following cell death, they begin the phagocytic removal of cell debris [53,54]. Proinflammatory cytokines (e.g., IL-1, IL-6, and TNF-α) released by M1 microglia promote a gradual alteration in the BBB and allow the infiltration of circulating leucocytes, which eventually release proinflammatory cytokines (e.g., interleukin-1β and interferon-γ) to damage the cell structures directly or indirectly and contribute to the enlargement of the lesion [55,56,57]. Cytokines released by M1 microglia and other glial cells and neurons promote astrogliosis, and these reactive astrocytes participate in many protective mechanisms after a stroke (e.g., neurotransmitter uptake, pH regulation, the anti-inflammatory release of cytokines, and glial scar formation), but they also have detrimental effects including the release of several detrimental factors such as proinflammatory cytokines (e.g., TNF-α and IL-1), matrix metalloproteinases (e.g., the degradation of the matrix protein), and proteoglycans (e.g., these cause inhibition of axon regeneration and myelination), which can contribute to expanding the lesion and/or decreased recovery [58,59]. Around 6 days after the injury, glial scar formation starts, and involves a subset of reactive astrocytes and other cells (e.g., reactive microglia and NG2 cells) and is completed between 2 and 4 weeks after the stroke [60,61]. Even after the glial scar, which serves as a protective barrier, has been formed, the cells in the penumbra are still exposed to several deleterious mechanisms, such as vasogenic edema, apoptosis, astrogliosis, and inflammation, which can last many months or even years. However, neurons and other cell types are viable and have a long-term potential for remodeling the tissue and forming new circuits to sustain a functional recovery [45,62], and this also the main target for any therapeutic interventions.

5.2. Alzheimer’s Disease (AD)

AD is a progressive disease that is generally manifested by a loss of memory and difficulties with communicating, behaving correctly, and using problem-solving skills. AD is a neurodegenerative disorder that results in neuronal loss and brain atrophy in extensive areas of the hippocampus and cerebral cortex, synapse loss, and ultimately, death [63]. AD is generally divided into familial AD (FAD) and idiopathic or sporadic AD (SAD). FAD is caused by dominant genetic mutations in presenilin 1 (PSEN1), presenilin 2 (PSEN2), and amyloid-beta A4 precursor protein (APP), and it accounts for 3% of the reported cases of AD. SAD has no single genetic cause, and it represents over 95% of all cases. Since age is considered to be the main risk factor for AD, there is another classification that takes into account the age at which the disease began, and this is divided into early-onset and late-onset types. Early onset occurs before the age of 65, and most cases are FAD ones. Most late-onset cases are SAD one, and it has a mean onset age of 80 years [64,65]. In addition to APP, PSEN1, and PSEN2 genes, the ε4 allele of apolipoprotein E (APOE) is another genetic risk factor for AD. APOE is a protein related to lipid metabolism and is immunochemically colocalized to vascular amyloid deposits, neurofibrillary tangles (NFT), and senile plaques in AD [66]. AD is related to the accumulation of insoluble forms of amyloid-β (Aβ) in plaques and the intraneuronal deposition of neurofibrillary tangles (NFT), which are composed of hyperphosphorylated tau protein. The amyloid hypothesis of AD suggests that alterations to APP metabolism and Aβ accumulation are the main events in AD [67]. On the other hand, the tau hypothesis of AD suggests that aggregates of misfolded and fibrillar hyperphosphorylated NFT accumulate inside the neurons and propagate through cells in a prion-like way, eventually disseminating into the brains of AD patients. The neuropathologic hallmarks of AD are Aβ plaques and NFTs, but generally, they are accompanied by neuronal and synaptic loss, reactive astrocytes, microglial activation, the blood–brain barrier alterations, and brain atrophy [68]. Moreover, it is noteworthy that a high percentage of patients with AD also have cerebral amyloid angiopathy, a condition characterized by an accumulation of amyloids in the cerebral vasculature, which can lead to intracerebral hemorrhages and microbleeds, and these events accelerate AD [64]. In AD, the microglia and astrocytes play an important role in the neuroinflammatory response, and also, in the development of the disease. Aβ can interact with the microglia through the NLRP3 inflammatory complex and CD36-TLR4-TLR6 receptor complex, causing immune responses, cell damage, and the release of inflammation-inducing factors, such as IL-1β and TNF-α. Moreover, high levels of proinflammatory cytokines IL-1β and Il-6 are elevated in the peripheral blood of AD patients [69]. Activated microglia release the proinflammatory cytokines IL-1α, C1q, and TNF-α that can induce the A1 proinflammatory astrocytes, which can produce a secondary inflammatory response [31]. The evidence from postmortem analyses of AD patients shows alterations in microglia morphology such as reduced branching and arborized areas and immunoreactivity to ionized calcium-binding adaptor molecule 1 (Iba 1), which is a microglia/macrophage-specific protein that is upregulated in activated microglia [70]. Activated microglia were observed in the entorhinal, temporoparietal, and cingulate cortices in positron emission tomography (PET) studies in humans. These PET studies use the selective marker 11PK11195, which labels the target translocator protein (TSPO) on the external mitochondrial membrane that increases its density in active microglia and the regions [71]. Additionally, the occurrence of microglia activation was confirmed using different markers alone (e.g., 11PK11195 and 11 PBR28) or in combination with tau (e.g., 18 F-AV145) or Aβ plaques markers (e.g., 18 F-flutemetamol). One study found that AD patients have increased microglial activation and amyloid in the frontal, temporal, parietal, occipital, and cingulate cortices [72], while another study found activated microglia in the occipital lobe in AD patients [73]. Additionally, it was reported that microglial activation, tau aggregation, and amyloid deposition were found in similar areas of the association cortex [74]. A longitudinal PET study (14 months) evaluated the change in microglia activation in AD patients with mild cognitive impairment (MCI), and the results show that patients with MCI demonstrated reduced activated microglia levels, which is in contrast to AD patients with more activated microglia [75]. Another study used PET (11PK11195) in combination with structural magnetic resonance imaging (structural MRI) to investigate brain atrophy, and they found activated microglia in the anterior temporal region, tau in the temporoparietal area, and grey matter atrophy [76]. A similar study found that activated microglia in temporal cortices are related to parieto-occipital atrophy and cortical thinning [77]. All of these studies confirm the presence of activated microglia in patients with AD.

5.3. Parkinson’s Disease (PD)

PD is an age-related neurodegenerative disease with a multifactorial etiology that causes tremors at rest, bradykinesia symptoms, and rigidity, and it shares these characteristics with other clinical syndromes referred to as “Parkinsonism” disorders [78]. The other main characteristics of PD are the degeneration of neurons in the substantia nigra pars compacta (one of the basal ganglia), intraneuronal protein aggregates called Lewy bodies, and Lewy neurites [79]. In addition to those mentioned, PD patients may also present other alterations such as sleep disorders, depression, and dementia [80]. A few cases of PD seem to have a genetic origin, and the majority of them correspond to the idiopathic form, for which the risk factors include aging and behavioral and environmental factors such as a history of melanoma, traumatic brain injury, and exposure to pesticides [79]. The monogenic mutation in several genes that encode different proteins constitute the genetic forms of PD, including the α-synuclein, in which gene duplications and triplications, as well as mutations, have been found, and this comprises the main component of Lewy bodies [79]. In physiological conditions, the neuronal protein α-synuclein participates in several activities, such as dopamine synthesis and vesicle trafficking, and it has two conformations, a soluble unfolded monomer and a multimeric membrane-bound helical α-synuclein. Meanwhile, in pathological conditions, the soluble unfolded monomer forms β-sheet-like oligomers named protofibrils, which transform into amyloid fibrils and ultimately deposit into Lewy bodies. Moreover, the protofibrils and fibrils may propagate by a transcellular mechanism from neuron to neuron [79]. Lewy body inclusions, in addition to being present in neurons of the substantia nigra, also are present in structures such as raphe nuclei, the basal nucleus of Meynert, the neocortex, and the amygdala, and also, in oligodendrocytes from the midbrain and the basal ganglia [81].
The contribution of neuroinflammation to cellular damage in PD has been confirmed in several studies. A positron emission tomography (PET) study in PD patients using a marker for active microglia showed that the rate of the activation of the microglia was increased in the substantia nigra, the caudate nucleus, the pre- and postcentral gyrus, the frontal lobe, and the putamen, which agrees well with the known distribution of neuropathological changes [82]. Additionally, increased proinflammatory mediators (e.g., TNF-β) and active astrocytes and microglia have been found in the substantia nigra in post-mortem studies on PD patient brains [83,84,85]. Moreover, the animal model of PD MPTP (1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine), as well as human brains, exhibited an increase in the proinflammatory molecules (e.g., COX-2) in dopaminergic neurons from the substantia nigra [86]. Furthermore, some evidence suggests that the adaptive immune system also participates in the pathology, for example, the presence of CD4+ and CD8+ T cells in the substantia nigra from postmortem studies of PD patients and animal models [87]. Neuronal cell culture studies also show that IL-1 increases the α-synuclein [88] and that α-synuclein induces microglial activation and increases the production of TNF and IL1β [89]. When it is analyzed together, this information shows that inflammatory responses are involved in the pathophysiology of PD.
Figure 2 show the main characteristic of inflammatory responses that share focal ischemic stroke, AD and PD.

6. Anti-Inflammatory Effects of Flavonoids in Focal Ischemic Stroke, AD, and PD

The general neuroprotective and neuroplastic effects of flavonoids are related to anti-inflammatory activity, which can increase the protection of neurons and glial cells against neurotoxins-induced injury and improve the endogenous mechanism of neuroplasticity to gain CNS functions. The effects of flavonoids in the modulation of molecular pathways involved in neuroinflammation have been mainly described in cell cultures using primary cells exposed to oxygen-glucose deprivation as the main model of ischemic stroke and lipopolysaccharides as a model of neuroinflammation in neurodegenerative disease, but animal models and clinical studies have also provided strong evidence of the beneficial effects of flavonoids. In the next sections, you will find a narrative description of the anti-inflammatory effects of the most used flavonoids on each disease in cellular, animal models, and clinical studies.

6.1. Anti-Inflammatory Effects of Flavonoids in Focal Ischemic Stroke

6.1.1. In Vitro Studies

Oxygen glucose deprivation (OGD) is an in vitro model used for the study of the cellular and molecular pathway associated with stroke, and usually, the cells are incubated in a glucose-free medium in a deoxygenated atmosphere for different durations of exposure to these conditions depending on the preparation. Sometimes, after the OGD, the cells are returned to the pre-deprivation conditions to model the focal ischemic reperfusion injury that occurs after the blood supply is restored, and this variant is called OGD/R [90,91]. Myricetin showed a significant protective effect against inflammation in human brain microvessel endothelial cells (HBMECs) in OGD/R conditions by decreasing the number of proinflammatory cytokines such as TNF-α, IL-1β, and IL-6 [92]. Quercetin inhibits inflammation that is mediated by TLR4/MyD88/NF-κB, signaling BV2 microglial cells in mice [93]. Isoquercetin, a glucoside derivative of quercetin, has neuroprotective effects in rat cortical neurons in OGD/R conditions by inhibiting the protein expression of TLR4 and nuclear NF-κB and the mRNA expression of TNF-α and IL-6 [94]. Moreover, in rat hippocampal neurons in OGD/R conditions, isoquercetin inhibits the activation of Toll-like receptor 4 (TLR4), nuclear factor-kappa B (NF-κB), and caspase-1; the phosphorylation of ERK1/2, JNK1/2, and p38 mitogen-activated protein kinase (MAPK); the secretion of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 [95]. Furthermore, cortical neurons in similar conditions inhibit the protein expression of TLR4 and NF-κB and the mRNA expression of TNF-α and IL-6 [94]. Baicalin could effectively downregulate the expression of the NOD2 receptor (protein associated with inflammatory reactions) and TNFα at both the mRNA and protein levels in BV2 microglial cells in OGD conditions [96], inhibit the NLRP3 inflammasome IL-1β, and IL-18 expression in cortical neurons in OGD/R conditions [97], and also, decrease the secretion of TNF-α, IL-1β, IL-6 and inhibit the NF-κB signaling pathway in the brain microvascular endothelial cells (BMECs) in OGD conditions [98] and the BV2 microglia cell line in OGD/R conditions [99]. Moreover, baicalin inhibits the proinflammatory microglial polarization through the inhibition of the TLR4/NF-κB pathway and the downregulation of phosphorylated STAT1 in a microglia-neuron co-culture system in OGD conditions [100] and in human brain microvascular endothelial cells (HBMEC) in OGD conditions, it inhibits the expression of TLR4, MYD88, and p-NF-κB and decreases the release of inflammatory factors IL-6, IL-1α, IL-1β, IL8, and TNF-α [101,102]. Baicalin also decreases the release of TNF-α, IL-1β, IL-6, and IL-8, and Tlr4 mRNA expression in microglia in OGD conditions [102]. Icariin applied before the OGD/R-reduced protein level expression of IL-1β, IL-6, and TNF-α in OGD/R conditions injured the microglia [103]. Casticin reduced the expression of TLR4, NF-κB p65, and NF-κB p50 in PC12 cells in OGD/R conditions [104]. Pratensein in HT22 cells in OGD/R conditions suppresses NLRP3 inflammasome activation through Nrf2 activation, resulting in reduced inflammatory responses [105]. Tectorigenin inhibited ROS inflammatory cytokines IL-1β, IL-6, and TNF-α production in OGD/R-induced HT-22 cells [106]. Astilbin inhibits NLRP3 inflammasome and decreases the release of IL-1β and IL-18 in PC12 cells in OGD conditions [107]. Anthocyanin significantly reduced the secretion of TNF-α, IL-1β, and IL-6 in SH-SY5Y cells exposed to OGD [108]. In the mouse neuroblastoma cells N2a, tricin, an O-methylated flavone, decreases the expression of TNF-α, IL-6, and IL-1β [109]. Diosmetin inhibits the NLRP3 inflammasome pathway and inflammatory cytokines IL-1β and IL-18 in PC12 cells in OGD/R conditions [110]. Schaftoside inhibits the expression of TLR4, IL-1β, IL-6, and TNF-α in OGD-simulated BV2 microglia [111].

6.1.2. Animal Model

The most commonly used animals in preclinic stroke research are rats and mice. Animal models for ischemic stroke can be divided into global, focal, and multifocal ones. There are several global ischemic stroke models, and the most common ones are cardiac arrest, the four-vessel occlusion model, and systemic hypotension and hypoxia. Animal models for focal ischemic strokes have been developed to induce damage within the territory irrigated by the middle cerebral artery (MCA) region to mimic a common clinical situation. There are different approaches to achieve this, but the most common ones are the intraluminal suture middle cerebral artery occlusion model without reperfusion (MCAO) and with reperfusion (MCAO/R), the photothrombotic model, and the endothelin-1 induced stroke model [112]. In this section, we will analyze only the studies that applied flavonoids after vessel occlusion with or without reperfusion because these models are more similar to clinical strokes. In a rat MCAO/R model, kaempferol-3-O-rutinoside (KRS) and kaempferol-3-O-glucoside (KGS) reduced the neurological deficits and infarct volume and inhibited the proinflammatory mediators (STAT3 and NF-κB) and interleukin 1β [113]. In the same model, kaempferol administered for 7 days after stroke also decreased the NF-κB and pro-inflammatory cytokines such as IL-5, TNF-α, IL-1β, and IL-6, but the reduction of last three cytokines only occurred with high doses [114]. Fisetin applied 3 h after the onset of ischemia to the MCAO/R mice model significantly reduced the infarct size and decreased TNF-α production in the microglia and the infiltration of leukocytes and macrophages [115]. A morin treatment for 7 days after MCAO in rats reduced the neurological deficits and inhibited the proinflammatory cytokine mRNA expression of TNF-α and IL-6 [116], and in the MCAO/R rat model, it decreased the rates of pNF-κB, TNF-α, IL-1β, and TLR4 expression and improved the tight junctions of the BBB by significantly increasing occludin and claudin expression [117]. (−)-Epigallocatechin-3-gallate (EGCG) decreased the infarct volume, TNF-α, IL-1β, and IL-6, and also, inhibited NF-κB/p65 in a MCAO/R rat model when it was applied immediately after reperfusion [118]. Luteolin administered intraperitoneally after MCAO/R in rats suppressed hippocampus inflammation, reduced the infarct volume, and decreased the astrocyte and microglia activation [119]. Luteoloside decreased the infarct volume, TNF-α, and IL-1β when this molecule was intraperitoneally injected immediately and 12 h after MCAO surgery in rats [120]. MCOA/R rats treated with nobiletin after reperfusion resulted in improved neurological deficits and decreased brain swelling and infarct volume [121]. Eriodictyol applied after stroke improves neurological deficits in the MCAO mice model and also decreases infarct volume TNF-α and GFAP expression [122]. Tricin applied by oral administration 2 h, 4 h, and 6 h after MCA/R resulted in decreased serum levels of TNF-α, IL-6, and IL-1β [109]. Eupatilin administered to mice 5 h after MCAO/R showed a reduction of the activated microglia in the peri-ischemic tissue and inhibited the NF-κB pathway [123].

Synergistic Effect of Flavonoids with rtPA

The simultaneous treatment of EGCG and rt-PA 4 h after MCAO significantly reversed the neurobehavioral deficit, brain infarction, cerebral edema, and blood–brain barrier disruption [124].

Preventive Treatment with Flavonoids

Rats treated with naringenin once daily for 21 days, and then subjected to MCOA/R, resulted in a significant decrease in the infarct volume, the expression of NF-κB, TNF-α, IL-1β, and GFAP in the astrocytes, and Iba1 in the microglia, and improved neurologic deficits [125]. Moreover, rats treated with the same molecule for 4 days before MCAO also showed a decrease in NF-κB, the infarct volume, and improvements to the neurologic deficits [126]. Moreover, the same molecule applied 7 days before MCAO/R in rats decreased the expression of TNF-α and IL-6 in the brain tissue [126]. Rats treated with hesperidin for 15 days followed by MCAO showed an improvement of the neurological deficits, infarct volume, and decreased levels of IL-1β [127]. In a rat model of global stroke, a pinocembrin treatment administered daily for 7 days before a stroke decreased the infarct size and NF-κB, TNF-α, and IL-6 in the hippocampal tissue [128]. In an MCAO mice model, a fourteen-day-long genistein treatment before a stroke reduced the infarct volume, improved the neurological deficit, and inhibited NF-κB activation [129]. Sanggenon administered intragastrically in rats seven days before MCAO/R surgery resulted in a decrease in the levels of TNF-α, IL-1β, and IL-6 [130]. Astilbin applied 3 days before the MCAO in rats inhibited NLRP3 inflammasome and decreased the serum concentration of IL-1β and IL-18 [107]. Chrysin applied for 7 days before the MCAO in rats decreased the release of inflammatory cytokines IL-6, IL-1β, and TNF-α [131]. Baicalin applied 4 days before the MCAO in rats decreased the expression level of the NLRP3 inflammasome, IL-1β, and IL-18 [97]. Eupafolin applied for 7 days before the MCAO/R in rats decreased TLR-4, TNF-α, IL-1β, and IL-6 expression [132]. Rats were treated with Biochanin A for 14 days before the MCAO in rats decreased the protein and gene expression of TNF-α and IL-1β [133].

6.1.3. Clinical

Though no clinical studies using flavonoids as a post-stroke treatment could be found, some human studies suggest that flavonoids can be useful in the treatment of this condition. A study using the flavonol fisetin combined with rt-PA in stroke patients shows that the addition of this flavonoid extends the therapeutic window of rt-PA treatment and dramatically improves the neurological deficits evaluated by the National Institutes of Health Stroke Scale (NIHSS) and decreases the plasma levels of C-reactive protein (CRP) and matrix metalloproteinases (MMP)-2 and -9 [134]. In a similar study, EGCG also extends the therapeutic window of the rt-PA treatment and improves the NIHSS scale, while decreasing plasma levels of matrix metalloproteinases (MMP)-2 and -9 [135].

6.2. Anti-Inflammatory Effects of Flavonoids in AD

6.2.1. In Vitro Studies

Numerous in vitro studies have evaluated the effects of different flavonoids in the Aβ oligomer and its assembly into aggregates. EGCG inhibits the fibrillogenesis of Aβ [136], modifies Aβ fibrils into smaller protein aggregates that are nontoxic to mammalian cells [137], and in cultured hippocampal neuronal cells, it has protective effects against Aβ-induced neuronal apoptosis through scavenging reactive oxygen species [138]. Quercetin used as a pretreatment with primary hippocampal cultures significantly decreases Aβ (1–42)-induced cytotoxicity, lipid peroxidation, protein oxidation, and apoptosis [139]. Luteolin and diosmetin decrease Aβ (1–40 and 1–42) in primary neuronal cells and SweAPP N2a cells [140]. Myricetin prevents the fibrillogenesis of Aβ [141]. Cyanidin 3-O-β-glucopyranoside in neuroblastoma SH-SY5Y cells reduces the cytotoxicity of Aβ (25–35) and its aggregation [142]. In the same cells, wogonin reduces Aβ aggregation and phosphorylated Tau [143]. Through different biochemical techniques, it has been found that Baicalein prevents the aggregation of the human tau protein [144,145]. Quercetin and rutin prevent the formation of Aβ fibrils and disaggregate Aβ fibrils in a cell system overexpressing APP Swedish mutation (APPswe) [146].

6.2.2. Animal Model Studies

Animal models for AD include chemically induced ones (e.g., amyloid infusion and streptozotocin), spontaneous ones (e.g., senescence-accelerated mouse), and several transgenic mice and a few transgenic rats that express mutant human genes related to the production of amyloid plaques and neurofibrillary tangles (e.g., 3XTg and 5XFAD, TG2576, and APP/PS1) [147]. These transgenic animals model familial AD and partly recapitulate the idiopathic forms and they express amyloid plaques and neurofibrillary tangles and all of the manifested deficits in memory, but the majority the animals do not present with neurodegeneration, and this is one of the aspects that limits their use in neuroinflammation research [147,148]. Since these animal models do not present with neurodegeneration, the main interest of flavonoids in the research in these animal models has been focused on the effects of the Aβ aggregation and the impairment of the cognition functions. In the chemical mouse model of memory deficits induced by scopolamine, isorhamnetin inhibits learning and memory deficits, and also, induces an increase in brain-derived neurotrophic factor (BDNF) levels in the prefrontal cortex and hippocampus [149], while naringin and rutin improve memory [150]. In another chemical model (streptozotocin), kaempferol increases the density of intact neurons in the CA1 area of the hippocampus and improves memory [151]. Luteolin improves memory [152] and a hesperidin pretreatment decreases inflammatory markers, such as NF-κB, iNOS, COX-2, and astrogliosis, and improves memory [153]. Nobiletin in APP-SL 7-5 transgenic mice reduces the quantity of soluble Aβ (1–40 and 1–42) and Aβ plaques in the hippocampus [154]. In 3XTg mice, diosmin and its bioactive metabolites decrease tau hyperphosphorylation and Aβ generation [155]. In transgenic APP/PS1 mice, hesperidin reduces Aβ plaque in the cortex and the hippocampus, decreases astrogliosis and microglial activation, and restores the ability to perform social interaction [156]. In transgenic h-APPswe, h-Tau P301L, and h-PS1 M146V mice, wogonin improves memory [143]. In the Tg2576 transgenic mouse model, diosmin and luteolin reduce Aβ 1–40 and 1–42 [140]. In APPsw transgenic mice, EGCG reduces the amount of soluble Aβ (1–40 and 1–42) and Aβ deposits in different cortical brain regions and the hippocampus [157], and in the Aβ infusion model, it prevents memory dysfunction and reduces the Aβ (1–42) and alpha-secretase levels and increases beta- and gamma-secretase in both the cortex and hippocampus, and similar results were obtained in the presenilin 2 (PS2) mutant mice [158], while in the APPsw transgenic mouse model, diosmin shows improved memory [159]. Nobiletin reduces Aβ plaques in the hippocampus and improves memory deficits in APP-SL 7-5 transgenic mice [154], and in 3XTg mice, it reversed the damage to memory and decreased the levels of Aβ 1–40 [160]. Nobiletin reverses memory impairment in the hippocampus in senescence-accelerated mice SAMP8 [161]. In 3XTg mice, quercetin reduces the plaques of Aβ and hyperphosphorylated tau in the CA1 area of the hippocampus and improves memory [162]. Cyanidin 3-O-glucoside decreases tau phosphorylation in the hippocampus and reverses memory impairment in Aβ infusion rats [163] and in the APP(swe)/PS1(ΔE9) mouse model, it improved memory and learning [142]. Fisetin in APPswe/PS1dE9 double transgenic mice inhibits the development of memory and learning problems through the modulation of cyclin-dependent kinase 5 (Cdk5), where hyperactivity induces neuroinflammation and neurodegeneration [164]. In two different transgenic mouse models (TG2576 and TG-SwDI), a dihydromyricetin treatment improves exploratory and locomotor activities, decreases anxiety, improves memory, and reverses Aβ accumulation [165]. As you may note, the majority of the studies mentioned focused on the effects of flavonoids on amyloidopathy and cognitive deficits, while studies on tauopathy are scarce.

6.2.3. Clinical

There are no clinical studies with a single flavonoid molecule, but we can illustrate the potential of these molecules using the results obtained with cocoa flavanol, which is a mixture of flavanols, mainly catechin and epicatechin. Cocoa flavanol consumption for 8 weeks improved cognitive functions in patients with mild cognitive impairment [166], and a double-blind study showed improved cognitive functions in aging subjects [167]. Moreover, the consumption of these flavanols by healthy 50–69-year-old subjects over 3 months improves the dentate gyrus functions evaluated by a high-resolution variant of functional magnetic resonance imaging (fMRI) and cognitive testing [168].

6.3. Anti-Inflammatory Effects of Flavonoids in PD

6.3.1. In Vitro Studies

Some in vitro studies have evaluated the effects of different flavonoids on the formation of α-synuclein oligomers and their assembly into aggregates and found that flavonoids inhibit oligomer formation and aggregation. These flavonoids include: apigenin, baicalein myricetin, genistein, morin, quercetin, EGCG, and scutellarein [169,170,171]. In activated microglia induced by the LPS model, nobiletin prevents the release of the proinflammatory cytokines TNF-α and IL-1β [172], and apigenin and luteolin decrease TNF-α and IL-6 [173]. Naringenin inhibits NF-κB, iNOS, and COX-2, and induces the expression of the suppressor of cytokine signaling 3 (SOCS-2), a negative regulator of cytokines in activated microglia [174,175]. Diadzein downregulates the activation of NF-κB and the production of IL-6 and [176], and its metabolite Equol (7-hydroxy-3-(4′-hydroxyphenyl)-chroman) prevents the secretion of TNF-α, IL-6, and NF-κB activation [177]. In PC12 cells exposed to the neurotoxic MPP+, morin reduces cell apoptosis and mortality [178] and decreases the rate of astrogliosis and the nuclear translocation of NF-κB in primary cultured astrocytes exposed to the same neurotoxicity [179]. Butein, butin, fisetin, fustin, and sulfuretin protect the murine hippocampal HT22 cells against glutamate-induced neurotoxicity and reduce the induced nitric oxide (NO) production in BV2 cells microglial cell lines, and also, butein suppresses the expression of iNOS and COX-2 [180]. Genkwanin suppressed the MPP+-induced activation of the TLR4/MyD88/NLRP3 inflammasome pathway in SH-SY5Y cells [181].

6.3.2. Animal Model Studies

Chemically induced animal models of PD are the most widely used ones. Rat or mice models of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA) are two of the most common types. MPTP is the precursor to the neurotoxic 1-methyl-4-phenyl-2,3-dihydropyridinium (MPDP+), which is converted into glial cells. Both MPDP+ and 6-OHDA are neurotoxic to dopaminergic neurons and elicit a motor phenotype [182,183]. Additionally, the rotenone model of PD also shows the main pathological hallmarks of the disease [184]. In this last model, baicalein reduced the formation and accumulation of α-synuclein oligomers and protected dopaminergic neurons [185]. In the MPP+ rat model, this flavonoid attenuates α-synuclein aggregation, inhibits inflammasome activation [186], improves the motor ability, decreases the number of activated microglia and astrocytes, and increases dopamine and serotonin neurotransmitters in the striatum [187,188,189]. Apigenin in the rotenone rat model decreases the expression of NF-κB, increases the expression of dopamine D2 receptor (D2R), and decreases α-synuclein aggregation in the rat rotenone model [190]. Nobiletin in the MPP+ rat model preserved the expression of the glial-cell-line-derived neurotrophic factor (GDNF), inhibited microglial activation [191] and increased the dopamine contents in the striatum and hippocampal CA1 region, and improved the motor deficits [192]. In the MPP+ rat model, naringin increased the GDNF level in the substantia nigra, reduced TNF-α expression [193], and protected the nigrostriatal DA projection [194]. In the MPDP+ mice model, EGCG reduced the dopamine neuronal loss in the substantia nigra [195], and the same flavonoid in the rotenone rat model inhibited TNF-α, IL-1β, and IL-6 in the striatum [196]. Quercetin increased the striatal dopamine level and reduced dopaminergic neuronal loss in the 6-OHDA rat model [197]. The same flavonoid in the MitoPark transgenic mouse models of PD reversed dopaminergic neuronal loss, striatal dopamine depletion, and the behavioral deficits [198]. Quercetin and kaempferol in an MTPT mouse model improved striatal dopamine secretion and motor coordination [199,200]. In the same model, hesperidin reduced the expression of IL-1β, TNF-α, and IL-6 and improved motor coordination [201,202]. In the 6-OHDA rat model, tangeretin protected the striatal dopaminergic neurons [203], rutin protected dopaminergic neurons and improved motor coordination [204], troxerutin reduced neuronal loss, astrogliosis, and striatal lipoperoxidation [205], and myricitrin inhibited the expression of TNF-α and protected the dopaminergic neurons from the substantia nigra [206]. In the MPTP mouse model, a pretreatment with morin reduced dopaminergic neuronal death, behavioral deficits, and striatal dopamine depletion [178]; it reduced the dopaminergic neuronal losses, astrogliosis, and improved motor dysfunction [179]. In the MPTP mouse model, icariin inhibited NLRP3 inflammasome and decreased the IL-1β and TNF-α serum levels [207]. Europinidin in the rotenone rat model decreased the IL-6, IL-1β, and TNF-α levels in the brain tissue [208]. Diosmin in a rotenone rat model decreased the expression of NF-κB and the TNF-α levels [209].

6.3.3. Clinical

We did not find any clinical study using a single flavonoid molecule, but research using flavonoid-rich pure cocoa in patients with PD could illustrate the possible benefits for this disease. A randomized (1:1), double-blind, placebo-controlled feasibility study with 30 patients with PD showed a reduction of fatigue and fatigability [210].
Table 1, Table 2 and Table 3 show the main anti-inflammatory effects in models of focal ischemic stroke, AD, and PD.

7. Conclusions and Future Directions

Flavonoids have pleiotropic effects, which are demonstrated mainly through in vitro and animal models of diverse human diseases. The best-known mechanism is antioxidants, but in the last two decades, our knowledge of their effects on neuroinflammation has grown. Many in vitro and animal model studies highlight the anti-inflammatory effect of flavonoids by decreasing the activated microglia and astrocytes, and also, by decreasing the proinflammatory cytokines such as TNF-α, IL-1β, IL-6, COX-2, and iNOS, either by the indirect or direct inactivation of transcription factors such as NF-κB and AP-1, and also, through the inactivation of the inflammasome (Figure 3). On the other hand, several studies have found that flavonoids can downregulate other pathological pathways, and some human studies show that after consuming flavonoid-rich foods and beverages, there is a significant reduction of the proinflammatory molecules, including C-reactive protein and IL-6 [211,212,213]. Searching the clinical trial data set from ClinicalTrails.gov revealed the interest of the scientific community in the beneficial effects of flavonoids for human health, as several ongoing studies are focused on topics related to ischemic strokes and neurodegenerative diseases, such as cognition performance, cognitive aging, the risk of dementia, and endothelial dysfunction, suggesting a growing interest in translating the preclinical knowledge into clinical trials.
Although there have been substantial achievements in the bioavailability of flavonoids, for example, the cutaneous delivery system and application of the nanoencapsulation of bioactive compounds [214,215], we still need to improve our knowledge about aspects such as metabolic transformation, the identification of active molecules (parent molecule and/or their metabolites), the mechanism to cross the blood-brain barrier, and toxicology to find single flavonoids for the future therapy for neurological disorders associated with aging.

Author Contributions

Conceptualization, H.M.-C. and H.E.L.-V.; data curation, H.M.-C., I.A.-C., H.E.L.-V. and R.G.-J.; writing—original draft preparation, H.M.-C., H.E.L.-V., I.A.-C. and R.G.-J.; writing—review and editing, H.M.-C., I.A.-C., H.E.L.-V. and R.G.-J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kirkwood, T.B.L. Understanding the Odd Science of Aging. Cell 2005, 120, 437–447. [Google Scholar] [CrossRef] [Green Version]
  2. Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef] [Green Version]
  3. López-Valdés, H.E.; Martínez-Coria, H. The Role of Neuroinflammation in Age-Related Dementias. Rev. Investig. Clín. Organo Hosp. Enferm. Nutr. 2016, 68, 40–48. [Google Scholar]
  4. Wen, K.; Fang, X.; Yang, J.; Yao, Y.; Nandakumar, K.S.; Salem, M.L.; Cheng, K. Recent Research on Flavonoids and Their Biomedical Applications. Curr. Med. Chem. 2021, 28, 1042–1066. [Google Scholar] [CrossRef]
  5. Ramzan, I.; Li, G.Q. Phytotherapies—Past, Present, and Future. In Phytotherapies; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2015; pp. 1–17. ISBN 978-1-119-00603-9. [Google Scholar]
  6. Dias, M.C.; Pinto, D.C.G.A.; Silva, A.M.S. Plant Flavonoids: Chemical Characteristics and Biological Activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef]
  7. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An Overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Chen, L.; Cao, H.; Huang, Q.; Xiao, J.; Teng, H. Absorption, Metabolism and Bioavailability of Flavonoids: A Review. Crit. Rev. Food Sci. Nutr. 2022, 62, 7730–7742. [Google Scholar] [CrossRef] [PubMed]
  9. Akhlaghi, M.; Foshati, S. Bioavailability and Metabolism of Flavonoids: A Review. Int. J. Nutr. Sci. 2017, 2, 180–184. [Google Scholar]
  10. Menendez, C.; Dueñas, M.; Galindo, P.; González-Manzano, S.; Jimenez, R.; Moreno, L.; Zarzuelo, M.J.; Rodríguez-Gómez, I.; Duarte, J.; Santos-Buelga, C.; et al. Vascular Deconjugation of Quercetin Glucuronide: The Flavonoid Paradox Revealed? Mol. Nutr. Food Res. 2011, 55, 1780–1790. [Google Scholar] [CrossRef]
  11. Williamson, G.; Kay, C.D.; Crozier, A. The Bioavailability, Transport, and Bioactivity of Dietary Flavonoids: A Review from a Historical Perspective. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1054–1112. [Google Scholar] [CrossRef] [Green Version]
  12. Hollman, P.C.H. Absorption, Bioavailability, and Metabolism of Flavonoids. Pharm. Biol. 2004, 42, 74–83. [Google Scholar] [CrossRef]
  13. Scalbert, A.; Williamson, G. Dietary Intake and Bioavailability of Polyphenols. J. Nutr. 2000, 130, 2073S–2085S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Rémésy, C. Bioavailability and Bioefficacy of Polyphenols in Humans. I. Review of 97 Bioavailability Studies. Am. J. Clin. Nutr. 2005, 81, 230S–242S. [Google Scholar] [CrossRef] [Green Version]
  15. El Mohsen, M.A.; Marks, J.; Kuhnle, G.; Moore, K.; Debnam, E.; Kaila Srai, S.; Rice-Evans, C.; Spencer, J.P.E. Absorption, Tissue Distribution and Excretion of Pelargonidin and Its Metabolites Following Oral Administration to Rats. Br. J. Nutr. 2006, 95, 51–58. [Google Scholar] [CrossRef]
  16. Talavéra, S.; Felgines, C.; Texier, O.; Besson, C.; Gil-Izquierdo, A.; Lamaison, J.-L.; Rémésy, C. Anthocyanin Metabolism in Rats and Their Distribution to Digestive Area, Kidney, and Brain. J. Agric. Food Chem. 2005, 53, 3902–3908. [Google Scholar] [CrossRef]
  17. El Mohsen, M.M.A.; Kuhnle, G.; Rechner, A.R.; Schroeter, H.; Rose, S.; Jenner, P.; Rice-Evans, C.A. Uptake and Metabolism of Epicatechin and Its Access to the Brain after Oral Ingestion. Free Radic. Biol. Med. 2002, 33, 1693–1702. [Google Scholar] [CrossRef] [PubMed]
  18. Mishra, A.; Bandopadhyay, R.; Singh, P.K.; Mishra, P.S.; Sharma, N.; Khurana, N. Neuroinflammation in Neurological Disorders: Pharmacotherapeutic Targets from Bench to Bedside. Metab. Brain Dis. 2021, 36, 1591–1626. [Google Scholar] [CrossRef] [PubMed]
  19. Lee, H.-G.; Wheeler, M.A.; Quintana, F.J. Function and Therapeutic Value of Astrocytes in Neurological Diseases. Nat. Rev. Drug Discov. 2022, 21, 339–358. [Google Scholar] [CrossRef] [PubMed]
  20. Li, D.; Wu, M. Pattern Recognition Receptors in Health and Diseases. Signal Transduct. Target. Ther. 2021, 6, 291. [Google Scholar] [CrossRef] [PubMed]
  21. Pascual, M.; Calvo-Rodriguez, M.; Núñez, L.; Villalobos, C.; Ureña, J.; Guerri, C. Toll-like Receptors in Neuroinflammation, Neurodegeneration, and Alcohol-Induced Brain Damage. IUBMB Life 2021, 73, 900–915. [Google Scholar] [CrossRef] [PubMed]
  22. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef] [PubMed]
  23. El-Zayat, S.R.; Sibaii, H.; Mannaa, F.A. Toll-like Receptors Activation, Signaling, and Targeting: An Overview. Bull. Natl. Res. Cent. 2019, 43, 187. [Google Scholar] [CrossRef] [Green Version]
  24. 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]
  25. Wang, L.; Hauenstein, A.V. The NLRP3 Inflammasome: Mechanism of Action, Role in Disease and Therapies. Mol. Asp. Med. 2020, 76, 100889. [Google Scholar] [CrossRef] [PubMed]
  26. Liu, C.; Chu, D.; Kalantar-Zadeh, K.; George, J.; Young, H.A.; Liu, G. Cytokines: From Clinical Significance to Quantification. Adv. Sci. 2021, 8, 2004433. [Google Scholar] [CrossRef]
  27. Brooks, A.J.; Dehkhoda, F.; Kragelund, B.B. Cytokine Receptors. In Principles of Endocrinology and Hormone Action; Endocrinology; Belfiore, A., LeRoith, D., Eds.; Springer International Publishing: Cham, Switzerland, 2018; pp. 157–185. ISBN 978-3-319-44675-2. [Google Scholar]
  28. Guo, S.; Wang, H.; Yin, Y. Microglia Polarization from M1 to M2 in Neurodegenerative Diseases. Front. Aging Neurosci. 2022, 14, 815347. [Google Scholar] [CrossRef]
  29. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C.; et al. Neurotoxic Reactive Astrocytes Are Induced by Activated Microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [Green Version]
  30. Tang, Y.; Le, W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol. Neurobiol. 2016, 53, 1181–1194. [Google Scholar] [CrossRef]
  31. Kwon, H.S.; Koh, S.-H. Neuroinflammation in Neurodegenerative Disorders: The Roles of Microglia and Astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef]
  32. Sikora, E.; Bielak-Zmijewska, A.; Dudkowska, M.; Krzystyniak, A.; Mosieniak, G.; Wesierska, M.; Wlodarczyk, J. Cellular Senescence in Brain Aging. Front. Aging Neurosci. 2021, 13, 646924. [Google Scholar] [CrossRef]
  33. Vandenbark, A.A.; Offner, H.; Matejuk, S.; Matejuk, A. Microglia and Astrocyte Involvement in Neurodegeneration and Brain Cancer. J. Neuroinflamm. 2021, 18, 298. [Google Scholar] [CrossRef]
  34. MacNee, W.; Rabinovich, R.A.; Choudhury, G. Ageing and the Border between Health and Disease. Eur. Respir. J. 2014, 44, 1332–1352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
  36. Blinkouskaya, Y.; Caçoilo, A.; Gollamudi, T.; Jalalian, S.; Weickenmeier, J. Brain Aging Mechanisms with Mechanical Manifestations. Mech. Ageing Dev. 2021, 200, 111575. [Google Scholar] [CrossRef]
  37. Lee, J.; Kim, H.-J. Normal Aging Induces Changes in the Brain and Neurodegeneration Progress: Review of the Structural, Biochemical, Metabolic, Cellular, and Molecular Changes. Front. Aging Neurosci. 2022, 14, 931536. [Google Scholar] [CrossRef] [PubMed]
  38. Seals, D.R.; Kaplon, R.E.; Gioscia-Ryan, R.A.; LaRocca, T.J. You’re Only as Old as Your Arteries: Translational Strategies for Preserving Vascular Endothelial Function with Aging. Physiology 2014, 29, 250–264. [Google Scholar] [CrossRef] [Green Version]
  39. Fulop, T.; Larbi, A.; Pawelec, G.; Khalil, A.; Cohen, A.A.; Hirokawa, K.; Witkowski, J.M.; Franceschi, C. Immunology of Aging: The Birth of Inflammaging. Clin. Rev. Allergy Immunol. 2021. [Google Scholar] [CrossRef]
  40. Martínez-Cué, C.; Rueda, N. Cellular Senescence in Neurodegenerative Diseases. Front. Cell. Neurosci. 2020, 14, 16. [Google Scholar] [CrossRef]
  41. Torres-Querol, C.; Torres, P.; Vidal, N.; Portero-Otín, M.; Arque, G.; Purroy, F. Acute Ischemic Stroke Triggers a Cellular Senescence-Associated Secretory Phenotype. Sci. Rep. 2021, 11, 15752. [Google Scholar] [CrossRef]
  42. Rodrigues, L.P.; Teixeira, V.R.; Alencar-Silva, T.; Simonassi-Paiva, B.; Pereira, R.W.; Pogue, R.; Carvalho, J.L. Hallmarks of Aging and Immunosenescence: Connecting the Dots. Cytokine Growth Factor Rev. 2021, 59, 9–21. [Google Scholar] [CrossRef] [PubMed]
  43. Zang, X.; Chen, S.; Zhu, J.; Ma, J.; Zhai, Y. The Emerging Role of Central and Peripheral Immune Systems in Neurodegenerative Diseases. Front. Aging Neurosci. 2022, 14, 872134. [Google Scholar] [CrossRef] [PubMed]
  44. Kuriakose, D.; Xiao, Z. Pathophysiology and Treatment of Stroke: Present Status and Future Perspectives. Int. J. Mol. Sci. 2020, 21, 7609. [Google Scholar] [CrossRef] [PubMed]
  45. Martínez-Coria, H.; Arrieta-Cruz, I.; Cruz, M.-E.; López-Valdés, H.E. Physiopathology of Ischemic Stroke and Its Modulation Using Memantine: Evidence from Preclinical Stroke. Neural Regen. Res. 2021, 16, 433–439. [Google Scholar] [CrossRef] [PubMed]
  46. Yousufuddin, M.; Young, N. Aging and Ischemic Stroke. Aging 2019, 11, 2542–2544. [Google Scholar] [CrossRef]
  47. Yousufuddin, M.; Bartley, A.C.; Alsawas, M.; Sheely, H.L.; Shultz, J.; Takahashi, P.Y.; Young, N.P.; Murad, M.H. Impact of Multiple Chronic Conditions in Patients Hospitalized with Stroke and Transient Ischemic Attack. J. Stroke 2017, 26, 1239–1248. [Google Scholar] [CrossRef] [PubMed]
  48. Lyden, S.; Wold, J. Acute Treatment of Ischemic Stroke. Neurol. Clin. 2022, 40, 17–32. [Google Scholar] [CrossRef] [PubMed]
  49. Qin, C.; Yang, S.; Chu, Y.-H.; Zhang, H.; Pang, X.-W.; Chen, L.; Zhou, L.-Q.; Chen, M.; Tian, D.-S.; Wang, W. Signaling Pathways Involved in Ischemic Stroke: Molecular Mechanisms and Therapeutic Interventions. Signal Transduct. Target. Ther. 2022, 7, 215. [Google Scholar] [CrossRef]
  50. Gelderblom, M.; Sobey, C.G.; Kleinschnitz, C.; Magnus, T. Danger Signals in Stroke. Ageing Res. Rev. 2015, 24, 77–82. [Google Scholar] [CrossRef]
  51. Grønberg, N.V.; Johansen, F.F.; Kristiansen, U.; Hasseldam, H. Leukocyte Infiltration in Experimental Stroke. J. Neuroinflamm. 2013, 10, 892. [Google Scholar] [CrossRef] [Green Version]
  52. Hu, X.; Li, P.; Guo, Y.; Wang, H.; Leak, R.K.; Chen, S.; Gao, Y.; Chen, J. Microglia/Macrophage Polarization Dynamics Reveal Novel Mechanism of Injury Expansion after Focal Cerebral Ischemia. Stroke J. Cereb. Circ. 2012, 43, 3063–3070. [Google Scholar] [CrossRef] [Green Version]
  53. Neumann, J.; Gunzer, M.; Gutzeit, H.O.; Ullrich, O.; Reymann, K.G.; Dinkel, K. Microglia Provide Neuroprotection after Ischemia. FASEB J. 2006, 20, 714–716. [Google Scholar] [CrossRef]
  54. Denes, A.; Vidyasagar, R.; Feng, J.; Narvainen, J.; McColl, B.W.; Kauppinen, R.A.; Allan, S.M. Proliferating Resident Microglia after Focal Cerebral Ischaemia in Mice. J. Cereb. Blood Flow Metab. 2007, 27, 1941–1953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ritzel, R.M.; Patel, A.R.; Grenier, J.M.; Crapser, J.; Verma, R.; Jellison, E.R.; McCullough, L.D. Functional Differences between Microglia and Monocytes after Ischemic Stroke. J. Neuroinflamm. 2015, 12, 106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Lambertsen, K.L.; Gregersen, R.; Meldgaard, M.; Clausen, B.H.; Heibøl, E.K.; Ladeby, R.; Knudsen, J.; Frandsen, A.; Owens, T.; Finsen, B. A Role for Interferon-Gamma in Focal Cerebral Ischemia in Mice. J. Neuropathol. Exp. Neurol. 2004, 63, 942–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Allan, S.M.; Tyrrell, P.J.; Rothwell, N.J. Interleukin-1 and Neuronal Injury. Nat. Rev. Immunol. 2005, 5, 629–640. [Google Scholar] [CrossRef]
  58. Sims, N.R.; Yew, W.P. Reactive Astrogliosis in Stroke: Contributions of Astrocytes to Recovery of Neurological Function. Neurochem. Int. 2017, 107, 88–103. [Google Scholar] [CrossRef]
  59. Sofroniew, M.V.; Vinters, H.V. Astrocytes: Biology and Pathology. Acta Neuropathol. 2010, 119, 7–35. [Google Scholar] [CrossRef] [Green Version]
  60. Adams, K.L.; Gallo, V. The Diversity and Disparity of the Glial Scar. Nat. Neurosci. 2018, 21, 9–15. [Google Scholar] [CrossRef]
  61. Ding, S. Dynamic Reactive Astrocytes after Focal Ischemia. Neural Regen. Res. 2014, 9, 2048–2052. [Google Scholar] [CrossRef]
  62. Burda, J.E.; Sofroniew, M.V. Reactive Gliosis and the Multicellular Response to CNS Damage and Disease. Neuron 2014, 81, 229–248. [Google Scholar] [CrossRef] [Green Version]
  63. Polis, B.; Samson, A.O. A New Perspective on Alzheimer’s Disease as a Brain Expression of a Complex Metabolic Disorder. In Alzheimer’s Disease; Wisniewski, T., Ed.; Codon Publications: Brisbane, Australia, 2019; ISBN 978-0-646-80968-7. [Google Scholar]
  64. Pons, V.; Rivest, S. Targeting Systemic Innate Immune Cells as a Therapeutic Avenue for Alzheimer Disease. Pharmacol. Rev. 2022, 74, 1–17. [Google Scholar] [CrossRef] [PubMed]
  65. Alzheimer’s Association. 2019 Alzheimer’s Disease Facts and Figures. Alzheimers Dement. 2019, 15, 321–387. [Google Scholar] [CrossRef]
  66. Golde, T.E. Alzheimer’s Disease—The Journey of a Healthy Brain into Organ Failure. Mol. Neurodegener. 2022, 17, 18. [Google Scholar] [CrossRef] [PubMed]
  67. Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef] [PubMed]
  68. Henstridge, C.M.; Hyman, B.T.; Spires-Jones, T.L. Beyond the Neuron-Cellular Interactions Early in Alzheimer Disease Pathogenesis. Nat. Rev. Neurosci. 2019, 20, 94–108. [Google Scholar] [CrossRef] [PubMed]
  69. De Oliveira, J.; Kucharska, E.; Garcez, M.L.; Rodrigues, M.S.; Quevedo, J.; Moreno-Gonzalez, I.; Budni, J. Inflammatory Cascade in Alzheimer’s Disease Pathogenesis: A Review of Experimental Findings. Cells 2021, 10, 2581. [Google Scholar] [CrossRef]
  70. Davies, D.S.; Ma, J.; Jegathees, T.; Goldsbury, C. Microglia Show Altered Morphology and Reduced Arborization in Human Brain during Aging and Alzheimer’s Disease. Brain Pathol. 2017, 27, 795–808. [Google Scholar] [CrossRef]
  71. Cagnin, A.; Brooks, D.J.; Kennedy, A.M.; Gunn, R.N.; Myers, R.; Turkheimer, F.E.; Jones, T.; Banati, R.B. In-Vivo Measurement of Activated Microglia in Dementia. Lancet 2001, 358, 461–467. [Google Scholar] [CrossRef]
  72. Edison, P.; Archer, H.A.; Gerhard, A.; Hinz, R.; Pavese, N.; Turkheimer, F.E.; Hammers, A.; Tai, Y.F.; Fox, N.; Kennedy, A.; et al. Microglia, Amyloid, and Cognition in Alzheimer’s Disease: An [11C](R)PK11195-PET and [11C]PIB-PET Study. Neurobiol. Dis. 2008, 32, 412–419. [Google Scholar] [CrossRef]
  73. Schuitemaker, A.; Kropholler, M.A.; Boellaard, R.; van der Flier, W.M.; Kloet, R.W.; van der Doef, T.F.; Knol, D.L.; Windhorst, A.D.; Luurtsema, G.; Barkhof, F.; et al. Microglial Activation in Alzheimer’s Disease: An (R)-[11C]PK11195 Positron Emission Tomography Study. Neurobiol. Aging 2013, 34, 128–136. [Google Scholar] [CrossRef] [Green Version]
  74. Dani, M.; Wood, M.; Mizoguchi, R.; Fan, Z.; Walker, Z.; Morgan, R.; Hinz, R.; Biju, M.; Kuruvilla, T.; Brooks, D.J.; et al. Microglial Activation Correlates in Vivo with Both Tau and Amyloid in Alzheimer’s Disease. Brain J. Neurol. 2018, 141, 2740–2754. [Google Scholar] [CrossRef]
  75. Fan, Z.; Brooks, D.J.; Okello, A.; Edison, P. An Early and Late Peak in Microglial Activation in Alzheimer’s Disease Trajectory. Brain J. Neurol. 2017, 140, 792–803. [Google Scholar] [CrossRef]
  76. Malpetti, M.; Kievit, R.A.; Passamonti, L.; Jones, P.S.; Tsvetanov, K.A.; Rittman, T.; Mak, E.; Nicastro, N.; Bevan-Jones, W.R.; Su, L.; et al. Microglial Activation and Tau Burden Predict Cognitive Decline in Alzheimer’s Disease. Brain 2020, 143, 1588–1602. [Google Scholar] [CrossRef]
  77. Nicastro, N.; Malpetti, M.; Mak, E.; Williams, G.B.; Bevan-Jones, W.R.; Carter, S.F.; Passamonti, L.; Fryer, T.D.; Hong, Y.T.; Aigbirhio, F.I.; et al. Gray Matter Changes Related to Microglial Activation in Alzheimer’s Disease. Neurobiol. Aging 2020, 94, 236–242. [Google Scholar] [CrossRef]
  78. Bologna, M.; Truong, D.; Jankovic, J. The Etiopathogenetic and Pathophysiological Spectrum of Parkinsonism. J. Neurol. Sci. 2022, 433, 120012. [Google Scholar] [CrossRef]
  79. Simon, D.K.; Tanner, C.M.; Brundin, P. Parkinson Disease Epidemiology, Pathology, Genetics, and Pathophysiology. Clin. Geriatr. Med. 2020, 36, 1–12. [Google Scholar] [CrossRef] [PubMed]
  80. Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s Disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef] [PubMed]
  81. Dickson, D.W. Neuropathology of Parkinson Disease. Park. Relat. Disord. 2018, 46 (Suppl. S1), S30–S33. [Google Scholar] [CrossRef]
  82. Gerhard, A.; Watts, J.; Trender-Gerhard, I.; Turkheimer, F.; Banati, R.B.; Bhatia, K.; Brooks, D.J. In Vivo Imaging of Microglial Activation with [11C](R)-PK11195 PET in Corticobasal Degeneration. Mov. Disord. 2004, 19, 1221–1226. [Google Scholar] [CrossRef] [PubMed]
  83. Boka, G.; Anglade, P.; Wallach, D.; Javoy-Agid, F.; Agid, Y.; Hirsch, E.C. Immunocytochemical Analysis of Tumor Necrosis Factor and Its Receptors in Parkinson’s Disease. Neurosci. Lett. 1994, 172, 151–154. [Google Scholar] [CrossRef] [PubMed]
  84. Banati, R.B.; Daniel, S.E.; Blunt, S.B. Glial Pathology but Absence of Apoptotic Nigral Neurons in Long-Standing Parkinson’s Disease. Mov. Disord. 1998, 13, 221–227. [Google Scholar] [CrossRef] [PubMed]
  85. Vawter, M.P.; Dillon-Carter, O.; Tourtellotte, W.W.; Carvey, P.; Freed, W.J. TGFβ1 and TGFβ2 Concentrations Are Elevated in Parkinson’s Disease in Ventricular Cerebrospinal Fluid. Exp. Neurol. 1996, 142, 313–322. [Google Scholar] [CrossRef] [PubMed]
  86. Teismann, P.; Tieu, K.; Choi, D.-K.; Wu, D.-C.; Naini, A.; Hunot, S.; Vila, M.; Jackson-Lewis, V.; Przedborski, S. Cyclooxygenase-2 Is Instrumental in Parkinson’s Disease Neurodegeneration. Proc. Natl. Acad. Sci. USA 2003, 100, 5473–5478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Brochard, V.; Combadière, B.; Prigent, A.; Laouar, Y.; Perrin, A.; Beray-Berthat, V.; Bonduelle, O.; Alvarez-Fischer, D.; Callebert, J.; Launay, J.-M.; et al. Infiltration of CD4+ Lymphocytes into the Brain Contributes to Neurodegeneration in a Mouse Model of Parkinson Disease. J. Clin. Investig. 2009, 119, 182–192. [Google Scholar] [CrossRef]
  88. Griffin, W.S.T.; Liu, L.; Li, Y.; Mrak, R.E.; Barger, S.W. Interleukin-1 Mediates Alzheimer and Lewy Body Pathologies. J. Neuroinflamm. 2006, 3, 5. [Google Scholar] [CrossRef] [Green Version]
  89. Klegeris, A.; Pelech, S.; Giasson, B.I.; Maguire, J.; Zhang, H.; McGeer, E.G.; McGeer, P.L. Alpha-Synuclein Activates Stress Signaling Protein Kinases in THP-1 Cells and Microglia. Neurobiol. Aging 2008, 29, 739–752. [Google Scholar] [CrossRef]
  90. Holloway, P.M.; Gavins, F.N.E. Modeling Ischemic Stroke In Vitro: Status Quo and Future Perspectives. Stroke 2016, 47, 561–569. [Google Scholar] [CrossRef] [Green Version]
  91. Tasca, C.I.; Dal-Cim, T.; Cimarosti, H. In Vitro Oxygen-Glucose Deprivation to Study Ischemic Cell Death. In Neuronal Cell Death; Methods in Molecular Biology; Lossi, L., Merighi, A., Eds.; Humana Press: New York, NY, USA, 2015; Volume 1254, pp. 197–210. [Google Scholar] [CrossRef]
  92. Zhang, S.; Hu, X.; Guo, S.; Shi, L.; He, Q.; Zhang, P.; Yu, S.; Zhao, R. Myricetin Ameliorated Ischemia/Reperfusion-Induced Brain Endothelial Permeability by Improvement of ENOS Uncoupling and Activation ENOS/NO. J. Pharmacol. Sci. 2019, 140, 62–72. [Google Scholar] [CrossRef]
  93. Le, K.; Song, Z.; Deng, J.; Peng, X.; Zhang, J.; Wang, L.; Zhou, L.; Bi, H.; Liao, Z.; Feng, Z. Quercetin Alleviates Neonatal Hypoxic-Ischemic Brain Injury by Inhibiting Microglia-Derived Oxidative Stress and TLR4-Mediated Inflammation. Inflamm. Res. 2020, 69, 1201–1213. [Google Scholar] [CrossRef]
  94. Wang, C.-P.; Li, J.-L.; Zhang, L.-Z.; Zhang, X.-C.; Yu, S.; Liang, X.-M.; Ding, F.; Wang, Z.-W. Isoquercetin Protects Cortical Neurons from Oxygen-Glucose Deprivation-Reperfusion Induced Injury via Suppression of TLR4-NF-κB Signal Pathway. Neurochem. Int. 2013, 63, 741–749. [Google Scholar] [CrossRef]
  95. Wang, C.-P.; Shi, Y.-W.; Tang, M.; Zhang, X.-C.; Gu, Y.; Liang, X.-M.; Wang, Z.-W.; Ding, F. Isoquercetin Ameliorates Cerebral Impairment in Focal Ischemia Through Anti-Oxidative, Anti-Inflammatory, and Anti-Apoptotic Effects in Primary Culture of Rat Hippocampal Neurons and Hippocampal CA1 Region of Rats. Mol. Neurobiol. 2017, 54, 2126–2142. [Google Scholar] [CrossRef] [PubMed]
  96. Li, H.; Hu, J.; Ma, L.; Yuan, Z.; Wang, Y.; Wang, X.; Xing, D.; Lei, F.; Du, L. Comprehensive Study of Baicalin Down-Regulating NOD2 Receptor Expression of Neurons with Oxygen-Glucose Deprivation in Vitro and Cerebral Ischemia-Reperfusion In Vivo. Eur. J. Pharmacol. 2010, 649, 92–99. [Google Scholar] [CrossRef]
  97. Zheng, W.-X.; He, W.-Q.; Zhang, Q.-R.; Jia, J.-X.; Zhao, S.; Wu, F.-J.; Cao, X.-L. Baicalin Inhibits NLRP3 Inflammasome Activity via the AMPK Signaling Pathway to Alleviate Cerebral Ischemia-Reperfusion Injury. Inflammation 2021, 44, 2091–2105. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, P.; Hou, J.; Fu, J.; Li, D.; Zhang, C.; Liu, J. Baicalin Protects Rat Brain Microvascular Endothelial Cells Injured by Oxygen-Glucose Deprivation via Anti-Inflammation. Brain Res. Bull. 2013, 97, 8–15. [Google Scholar] [CrossRef] [PubMed]
  99. Li, H.; Wang, Y.; Wang, B.; Li, M.; Liu, J.; Yang, H.; Shi, Y. Baicalin and Geniposide Inhibit Polarization and Inflammatory Injury of OGD/R-Treated Microglia by Suppressing the 5-LOX/LTB4 Pathway. Neurochem. Res. 2021, 46, 1844–1858. [Google Scholar] [CrossRef]
  100. Ran, Y.; Qie, S.; Gao, F.; Ding, Z.; Yang, S.; Tian, G.; Liu, Z.; Xi, J. Baicalein Ameliorates Ischemic Brain Damage through Suppressing Proinflammatory Microglia Polarization via Inhibiting the TLR4/NF-ΚB and STAT1 Pathway. Brain Res. 2021, 1770, 147626. [Google Scholar] [CrossRef]
  101. Ye-Hao, Z.; Lan, M.; Peng, Z.; Guang-Yu, L.; Jian-Xun, L. Effect of baicalin on inflammatory response and TLR4/NF-κB signaling pathway of human brain microvascular endothelial cell after hypoxia-reoxygenation injury. Zhongguo Zhongyao Zazhi China J. Chin. Mater. Med. 2020, 45, 4686–4691. [Google Scholar] [CrossRef]
  102. Hou, J.; Wang, J.; Zhang, P.; Li, D.; Zhang, C.; Zhao, H.; Fu, J.; Wang, B.; Liu, J. Baicalin Attenuates Proinflammatory Cytokine Production in Oxygen-Glucose Deprived Challenged Rat Microglial Cells by Inhibiting TLR4 Signaling Pathway. Int. Immunopharmacol. 2012, 14, 749–757. [Google Scholar] [CrossRef]
  103. Mo, Z.-T.; Zheng, J.; Liao, Y.-L. Icariin Inhibits the Expression of IL-1β, IL-6 and TNF-α Induced by OGD/R through the IRE1/XBP1s Pathway in Microglia. Pharm. Biol. 2021, 59, 1473–1479. [Google Scholar] [CrossRef]
  104. Huang, D.; Zhou, J.; Li, W.; Zhang, L.; Wang, X.; Liu, Q. Casticin Protected against Neuronal Injury and Inhibited the TLR4/NF-ΚB Pathway after Middle Cerebral Artery Occlusion in Rats. Pharmacol. Res. Perspect. 2021, 9, e00752. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, R.; Wei, Y.; Deng, W.; Teng, J. Pratensein Mitigates Oxidative Stress and NLRP3 Inflammasome Activation in OGD/R-Injured HT22 Cells by Activating Nrf2-Anti-Oxidant Signaling. Neurotox. Res. 2022, 40, 384–394. [Google Scholar] [CrossRef]
  106. Yao, L.; Yang, M.; Zhang, J.; Wang, F.; Liu, Q.; Xie, X.; Liu, Z.; Guo, Q.; Su, H.; Zhai, J.; et al. Tectorigenin Attenuates the OGD/R-Induced HT-22 Cell Damage through Regulation of the PI3K/AKT and the PPARγ/NF-ΚB Pathways. Hum. Exp. Toxicol. 2021, 40, 1320–1331. [Google Scholar] [CrossRef] [PubMed]
  107. Li, Y.; Wang, R.; Xue, L.; Yang, Y.; Zhi, F. Astilbin Protects against Cerebral Ischaemia/Reperfusion Injury by Inhibiting Cellular Apoptosis and ROS-NLRP3 Inflammasome Axis Activation. Int. Immunopharmacol. 2020, 84, 106571. [Google Scholar] [CrossRef] [PubMed]
  108. Cai, Y.; Li, X.; Pan, Z.; Zhu, Y.; Tuo, J.; Meng, Q.; Dai, G.; Yang, G.; Pan, Y. Anthocyanin Ameliorates Hypoxia and Ischemia Induced Inflammation and Apoptosis by Increasing Autophagic Flux in SH-SY5Y Cells. Eur. J. Pharmacol. 2020, 883, 173360. [Google Scholar] [CrossRef]
  109. Liu, Y.; Qu, X.; Yan, M.; Li, D.; Zou, R. Tricin Attenuates Cerebral Ischemia/Reperfusion Injury through Inhibiting Nerve Cell Autophagy, Apoptosis and Inflammation by Regulating the PI3K/Akt Pathway. Hum. Exp. Toxicol. 2022, 41, 1–10. [Google Scholar] [CrossRef] [PubMed]
  110. Shi, M.; Wang, J.; Bi, F.; Bai, Z. Diosmetin Alleviates Cerebral Ischemia-Reperfusion Injury through Keap1-Mediated Nrf2/ARE Signaling Pathway Activation and NLRP3 Inflammasome Inhibition. Environ. Toxicol. 2022, 37, 1529–1542. [Google Scholar] [CrossRef] [PubMed]
  111. Zhou, K.; Wu, J.; Chen, J.; Zhou, Y.; Chen, X.; Wu, Q.; Xu, Y.; Tu, W.; Lou, X.; Yang, G.; et al. Schaftoside Ameliorates Oxygen Glucose Deprivation-Induced Inflammation Associated with the TLR4/Myd88/Drp1-Related Mitochondrial Fission in BV2 Microglia Cells. J. Pharmacol. Sci. 2019, 139, 15–22. [Google Scholar] [CrossRef]
  112. Kumar, A.; Aakriti; Gupta, V. A Review on Animal Models of Stroke: An Update. Brain Res. Bull. 2016, 122, 35–44. [Google Scholar] [CrossRef]
  113. Yu, L.; Chen, C.; Wang, L.-F.; Kuang, X.; Liu, K.; Zhang, H.; Du, J.-R. Neuroprotective Effect of Kaempferol Glycosides against Brain Injury and Neuroinflammation by Inhibiting the Activation of NF-ΚB and STAT3 in Transient Focal Stroke. PLoS ONE 2013, 8, e55839. [Google Scholar] [CrossRef] [PubMed]
  114. Li, W.-H.; Cheng, X.; Yang, Y.-L.; Liu, M.; Zhang, S.-S.; Wang, Y.-H.; Du, G.-H. Kaempferol Attenuates Neuroinflammation and Blood Brain Barrier Dysfunction to Improve Neurological Deficits in Cerebral Ischemia/Reperfusion Rats. Brain Res. 2019, 1722, 146361. [Google Scholar] [CrossRef]
  115. Gelderblom, M.; Leypoldt, F.; Lewerenz, J.; Birkenmayer, G.; Orozco, D.; Ludewig, P.; Thundyil, J.; Arumugam, T.V.; Gerloff, C.; Tolosa, E.; et al. The Flavonoid Fisetin Attenuates Postischemic Immune Cell Infiltration, Activation and Infarct Size after Transient Cerebral Middle Artery Occlusion in Mice. J. Cereb. Blood Flow Metab. 2012, 32, 835–843. [Google Scholar] [CrossRef]
  116. Chen, Y.; Li, Y.; Xu, H.; Li, G.; Ma, Y.; Pang, Y.J. Morin Mitigates Oxidative Stress, Apoptosis and Inflammation in Cerebral Ischemic Rats. Afr. J. Tradit. Complement. Altern. Med. AJTCAM 2017, 14, 348–355. [Google Scholar] [CrossRef]
  117. Khamchai, S.; Chumboatong, W.; Hata, J.; Tocharus, C.; Suksamrarn, A.; Tocharus, J. Morin Protects the Blood-Brain Barrier Integrity against Cerebral Ischemia Reperfusion through Anti-Inflammatory Actions in Rats. Sci. Rep. 2020, 10, 13379. [Google Scholar] [CrossRef]
  118. Zhang, F.; Li, N.; Jiang, L.; Chen, L.; Huang, M. Neuroprotective Effects of (−)-Epigallocatechin-3-Gallate Against Focal Cerebral Ischemia/Reperfusion Injury in Rats through Attenuation of Inflammation. Neurochem. Res. 2015, 40, 1691–1698. [Google Scholar] [CrossRef]
  119. Li, L.; Pan, G.; Fan, R.; Li, D.; Guo, L.; Ma, L.; Liang, H.; Qiu, J. Luteolin Alleviates Inflammation and Autophagy of Hippocampus Induced by Cerebral Ischemia/Reperfusion by Activating PPAR Gamma in Rats. BMC Complement. Med. Ther. 2022, 22, 176. [Google Scholar] [CrossRef]
  120. Li, Q.; Tian, Z.; Wang, M.; Kou, J.; Wang, C.; Rong, X.; Li, J.; Xie, X.; Pang, X. Luteoloside Attenuates Neuroinflammation in Focal Cerebral Ischemia in Rats via Regulation of the PPARγ/Nrf2/NF-ΚB Signaling Pathway. Int. Immunopharmacol. 2019, 66, 309–316. [Google Scholar] [CrossRef] [PubMed]
  121. Yasuda, N.; Ishii, T.; Oyama, D.; Fukuta, T.; Agato, Y.; Sato, A.; Shimizu, K.; Asai, T.; Asakawa, T.; Kan, T.; et al. Neuroprotective Effect of Nobiletin on Cerebral Ischemia–Reperfusion Injury in Transient Middle Cerebral Artery-Occluded Rats. Brain Res. 2014, 1559, 46–54. [Google Scholar] [CrossRef] [PubMed]
  122. Ferreira, V.L.; Borba, H.H.L.; de F. Bonetti, A.; Leonart, L.P.; Pontarolo, R. Cytokines and Interferons: Types and Functions. In Autoantibodies and Cytokines; Khan, W.A., Ed.; IntechOpen: London, UK, 2018. [Google Scholar] [CrossRef] [Green Version]
  123. Sapkota, A.; Gaire, B.P.; Cho, K.S.; Jeon, S.J.; Kwon, O.W.; Jang, D.S.; Kim, S.Y.; Ryu, J.H.; Choi, J.W. Eupatilin Exerts Neuroprotective Effects in Mice with Transient Focal Cerebral Ischemia by Reducing Microglial Activation. PLoS ONE 2017, 12, e0171479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. You, Y.-P. Epigallocatechin Gallate Extends the Therapeutic Window of Recombinant Tissue Plasminogen Activator Treatment in Ischemic Rats. J. Stroke Cerebrovasc. Dis. 2016, 25, 990–997. [Google Scholar] [CrossRef]
  125. Raza, S.S.; Khan, M.M.; Ahmad, A.; Ashafaq, M.; Islam, F.; Wagner, A.P.; Safhi, M.M.; Islam, F. Neuroprotective Effect of Naringenin Is Mediated through Suppression of NF-ΚB Signaling Pathway in Experimental Stroke. Neuroscience 2013, 230, 157–171. [Google Scholar] [CrossRef]
  126. Bai, X.; Zhang, X.; Chen, L.; Zhang, J.; Zhang, L.; Zhao, X.; Zhao, T.; Zhao, Y. Protective Effect of Naringenin in Experimental Ischemic Stroke: Down-Regulated NOD2, RIP2, NF-ΚB, MMP-9 and up-Regulated Claudin-5 Expression. Neurochem. Res. 2014, 39, 1405–1415. [Google Scholar] [CrossRef] [PubMed]
  127. Raza, S.S.; Khan, M.M.; Ahmad, A.; Ashafaq, M.; Khuwaja, G.; Tabassum, R.; Javed, H.; Siddiqui, M.S.; Safhi, M.M.; Islam, F. Hesperidin Ameliorates Functional and Histological Outcome and Reduces Neuroinflammation in Experimental Stroke. Brain Res. 2011, 1420, 93–105. [Google Scholar] [CrossRef] [PubMed]
  128. Saad, M.A.; Salam, R.M.A.; Kenawy, S.A.; Attia, A.S. Pinocembrin Attenuates Hippocampal Inflammation, Oxidative Perturbations and Apoptosis in a Rat Model of Global Cerebral Ischemia Reperfusion. Pharmacol. Rep. 2015, 67, 115–122. [Google Scholar] [CrossRef]
  129. Qian, Y.; Guan, T.; Huang, M.; Cao, L.; Li, Y.; Cheng, H.; Jin, H.; Yu, D. Neuroprotection by the Soy Isoflavone, Genistein, via Inhibition of Mitochondria-Dependent Apoptosis Pathways and Reactive Oxygen Induced-NF-ΚB Activation in a Cerebral Ischemia Mouse Model. Neurochem. Int. 2012, 60, 759–767. [Google Scholar] [CrossRef]
  130. Zhao, Y.; Xu, J. Sanggenon C Ameliorates Cerebral Ischemia-Reperfusion Injury by Inhibiting Inflammation and Oxidative Stress through Regulating RhoA-ROCK Signaling. Inflammation 2020, 43, 1476–1487. [Google Scholar] [CrossRef] [Green Version]
  131. Li, T.-F.; Ma, J.; Han, X.-W.; Jia, Y.-X.; Yuan, H.-F.; Shui, S.-F.; Guo, D.; Yan, L. Chrysin Ameliorates Cerebral Ischemia/Reperfusion (I/R) Injury in Rats by Regulating the PI3K/Akt/MTOR Pathway. Neurochem. Int. 2019, 129, 104496. [Google Scholar] [CrossRef]
  132. Chen, X.; Yao, Z.; Peng, X.; Wu, L.; Wu, H.; Ou, Y.; Lai, J. Eupafolin Alleviates Cerebral Ischemia/Reperfusion Injury in Rats via Blocking the TLR4/NF-ΚB Signaling Pathway. Mol. Med. Rep. 2020, 22, 5135–5144. [Google Scholar] [CrossRef]
  133. Wang, W.; Tang, L.; Li, Y.; Wang, Y. Biochanin A Protects against Focal Cerebral Ischemia/Reperfusion in Rats via Inhibition of P38-Mediated Inflammatory Responses. J. Neurol. Sci. 2015, 348, 121–125. [Google Scholar] [CrossRef]
  134. Wang, L.; Cao, D.; Wu, H.; Jia, H.; Yang, C.; Zhang, L. Fisetin Prolongs Therapy Window of Brain Ischemic Stroke Using Tissue Plasminogen Activator: A Double-Blind Randomized Placebo-Controlled Clinical Trial. Clin. Appl. Thromb. Off. J. Int. Acad. Clin. Appl. Thromb. 2019, 25, 1–8. [Google Scholar] [CrossRef]
  135. Wang, X.-H.; You, Y.-P. Epigallocatechin Gallate Extends Therapeutic Window of Recombinant Tissue Plasminogen Activator Treatment for Brain Ischemic Stroke: A Randomized Double-Blind and Placebo-Controlled Trial. Clin. Neuropharmacol. 2017, 40, 24–28. [Google Scholar] [CrossRef] [PubMed]
  136. Ehrnhoefer, D.E.; Bieschke, J.; Boeddrich, A.; Herbst, M.; Masino, L.; Lurz, R.; Engemann, S.; Pastore, A.; Wanker, E.E. EGCG Redirects Amyloidogenic Polypeptides into Unstructured, off-Pathway Oligomers. Nat. Struct. Mol. Biol. 2008, 15, 558–566. [Google Scholar] [CrossRef] [PubMed]
  137. Bieschke, J.; Russ, J.; Friedrich, R.P.; Ehrnhoefer, D.E.; Wobst, H.; Neugebauer, K.; Wanker, E.E. EGCG Remodels Mature α-Synuclein and Amyloid-β Fibrils and Reduces Cellular Toxicity. Proc. Natl. Acad. Sci. USA 2010, 107, 7710–7715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Choi, Y.T.; Jung, C.H.; Lee, S.R.; Bae, J.H.; Baek, W.K.; Suh, M.H.; Park, J.; Park, C.W.; Suh, S.I. The Green Tea Polyphenol (−)-Epigallocatechin Gallate Attenuates Beta-Amyloid-Induced Neurotoxicity in Cultured Hippocampal Neurons. Life Sci. 2001, 70, 603–614. [Google Scholar] [CrossRef] [PubMed]
  139. Ansari, M.A.; Abdul, H.M.; Joshi, G.; Opii, W.O.; Butterfield, D.A. Protective Effect of Quercetin in Primary Neurons against Aβ(1–42): Relevance to Alzheimer’s Disease. J. Nutr. Biochem. 2009, 20, 269–275. [Google Scholar] [CrossRef] [Green Version]
  140. Rezai-Zadeh, K.; Shytle, R.D.; Bai, Y.; Tian, J.; Hou, H.; Mori, T.; Zeng, J.; Obregon, D.; Town, T.; Tan, J. Flavonoid-Mediated Presenilin-1 Phosphorylation Reduces Alzheimer’s Disease β-Amyloid Production. J. Cell. Mol. Med. 2009, 13, 574–588. [Google Scholar] [CrossRef]
  141. Hirohata, M.; Hasegawa, K.; Tsutsumi-Yasuhara, S.; Ohhashi, Y.; Ookoshi, T.; Ono, K.; Yamada, M.; Naiki, H. The Anti-Amyloidogenic Effect Is Exerted against Alzheimer’s Beta-Amyloid Fibrils in Vitro by Preferential and Reversible Binding of Flavonoids to the Amyloid Fibril Structure. Biochemistry 2007, 46, 1888–1899. [Google Scholar] [CrossRef]
  142. Song, N.; Zhang, L.; Chen, W.; Zhu, H.; Deng, W.; Han, Y.; Guo, J.; Qin, C. Cyanidin 3-O-β-Glucopyranoside Activates Peroxisome Proliferator-Activated Receptor-γ and Alleviates Cognitive Impairment in the APP(Swe)/PS1(ΔE9) Mouse Model. Biochim. Biophys. Acta 2016, 1862, 1786–1800. [Google Scholar] [CrossRef]
  143. Huang, D.-S.; Yu, Y.-C.; Wu, C.-H.; Lin, J.-Y. Protective Effects of Wogonin against Alzheimer’s Disease by Inhibition of Amyloidogenic Pathway. Evid. Based Complement. Altern. Med. 2017, 2017, 3545169. [Google Scholar] [CrossRef] [Green Version]
  144. Sonawane, S.K.; Uversky, V.N.; Chinnathambi, S. Baicalein Inhibits Heparin-Induced Tau Aggregation by Initializing Non-Toxic Tau Oligomer Formation. Cell Commun. Signal. 2021, 19, 16. [Google Scholar] [CrossRef]
  145. Sonawane, S.K.; Balmik, A.A.; Boral, D.; Ramasamy, S.; Chinnathambi, S. Baicalein Suppresses Repeat Tau Fibrillization by Sequestering Oligomers. Arch. Biochem. Biophys. 2019, 675, 108119. [Google Scholar] [CrossRef]
  146. Jiménez-Aliaga, K.; Bermejo-Bescós, P.; Benedí, J.; Martín-Aragón, S. Quercetin and Rutin Exhibit Antiamyloidogenic and Fibril-Disaggregating Effects in Vitro and Potent Antioxidant Activity in APPswe Cells. Life Sci. 2011, 89, 939–945. [Google Scholar] [CrossRef]
  147. Mullane, K.; Williams, M. Preclinical Models of Alzheimer’s Disease: Relevance and Translational Validity. Curr. Protoc. Pharmacol. 2019, 84, e57. [Google Scholar] [CrossRef] [Green Version]
  148. Vitek, M.P.; Araujo, J.A.; Fossel, M.; Greenberg, B.D.; Howell, G.R.; Rizzo, S.J.S.; Seyfried, N.T.; Tenner, A.J.; Territo, P.R.; Windisch, M.; et al. Translational Animal Models for Alzheimer’s Disease: An Alzheimer’s Association Business Consortium Think Tank. Alzheimer Dement. Transl. Res. Clin. Interv. 2020, 6, e12114. [Google Scholar] [CrossRef] [PubMed]
  149. Ishola, I.O.; Osele, M.O.; Chijioke, M.C.; Adeyemi, O.O. Isorhamnetin Enhanced Cortico-Hippocampal Learning and Memory Capability in Mice with Scopolamine-Induced Amnesia: Role of Antioxidant Defense, Cholinergic and BDNF Signaling. Brain Res. 2019, 1712, 188–196. [Google Scholar] [CrossRef] [PubMed]
  150. Ramalingayya, G.V.; Nampoothiri, M.; Nayak, P.G.; Kishore, A.; Shenoy, R.R.; Rao, C.M.; Nandakumar, K. Naringin and Rutin Alleviates Episodic Memory Deficits in Two Differentially Challenged Object Recognition Tasks. Pharmacogn. Mag. 2016, 12, S63–S70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Darbandi, N.; Ramezani, M.; Khodagholi, F.; Noori, M. Kaempferol Promotes Memory Retention and Density of Hippocampal CA1 Neurons in Intra-Cerebroventricular STZ-Induced Experimental AD Model in Wistar Rats. Biologija 2016, 62, 157–168. [Google Scholar] [CrossRef] [Green Version]
  152. Wang, H.; Wang, H.; Cheng, H.; Che, Z. Ameliorating Effect of Luteolin on Memory Impairment in an Alzheimer’s Disease Model. Mol. Med. Rep. 2016, 13, 4215–4220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Javed, H.; Vaibhav, K.; Ahmed, M.E.; Khan, A.; Tabassum, R.; Islam, F.; Safhi, M.M.; Islam, F. Effect of Hesperidin on Neurobehavioral, Neuroinflammation, Oxidative Stress and Lipid Alteration in Intracerebroventricular Streptozotocin Induced Cognitive Impairment in Mice. J. Neurol. Sci. 2015, 348, 51–59. [Google Scholar] [CrossRef] [PubMed]
  154. Onozuka, H.; Nakajima, A.; Matsuzaki, K.; Shin, R.-W.; Ogino, K.; Saigusa, D.; Tetsu, N.; Yokosuka, A.; Sashida, Y.; Mimaki, Y.; et al. Nobiletin, a Citrus Flavonoid, Improves Memory Impairment and Aβ Pathology in a Transgenic Mouse Model of Alzheimer’s Disease. J. Pharmacol. Exp. Ther. 2008, 326, 739–744. [Google Scholar] [CrossRef] [PubMed]
  155. Sawmiller, D.; Habib, A.; Li, S.; Darlington, D.; Hou, H.; Tian, J.; Shytle, R.D.; Smith, A.; Giunta, B.; Mori, T.; et al. Diosmin Reduces Cerebral Aβ Levels, Tau Hyperphosphorylation, Neuroinflammation, and Cognitive Impairment in the 3xTg-AD Mice. J. Neuroimmunol. 2016, 299, 98–106. [Google Scholar] [CrossRef] [Green Version]
  156. Li, C.; Zug, C.; Qu, H.; Schluesener, H.; Zhang, Z. Hesperidin Ameliorates Behavioral Impairments and Neuropathology of Transgenic APP/PS1 Mice. Behav. Brain Res. 2015, 281, 32–42. [Google Scholar] [CrossRef] [PubMed]
  157. Rezai-Zadeh, K.; Shytle, D.; Sun, N.; Mori, T.; Hou, H.; Jeanniton, D.; Ehrhart, J.; Townsend, K.; Zeng, J.; Morgan, D.; et al. Green Tea Epigallocatechin-3-Gallate (EGCG) Modulates Amyloid Precursor Protein Cleavage and Reduces Cerebral Amyloidosis in Alzheimer Transgenic Mice. J. Neurosci. 2005, 25, 8807–8814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Lee, J.W.; Lee, Y.K.; Ban, J.O.; Ha, T.Y.; Yun, Y.P.; Han, S.B.; Oh, K.W.; Hong, J.T. Green Tea (−)-Epigallocatechin-3-Gallate Inhibits Beta-Amyloid-Induced Cognitive Dysfunction through Modification of Secretase Activity via Inhibition of ERK and NF-KappaB Pathways in Mice. J. Nutr. 2009, 139, 1987–1993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Rezai-Zadeh, K.; Arendash, G.W.; Hou, H.; Fernandez, F.; Jensen, M.; Runfeldt, M.; Shytle, R.D.; Tan, J. Green Tea Epigallocatechin-3-Gallate (EGCG) Reduces Beta-Amyloid Mediated Cognitive Impairment and Modulates Tau Pathology in Alzheimer Transgenic Mice. Brain Res. 2008, 1214, 177–187. [Google Scholar] [CrossRef]
  160. Nakajima, A.; Aoyama, Y.; Shin, E.-J.; Nam, Y.; Kim, H.-C.; Nagai, T.; Yokosuka, A.; Mimaki, Y.; Yokoi, T.; Ohizumi, Y.; et al. Nobiletin, a Citrus Flavonoid, Improves Cognitive Impairment and Reduces Soluble Aβ Levels in a Triple Transgenic Mouse Model of Alzheimer’s Disease (3XTg-AD). Behav. Brain Res. 2015, 289, 69–77. [Google Scholar] [CrossRef] [PubMed]
  161. Nakajima, A.; Aoyama, Y.; Nguyen, T.-T.L.; Shin, E.-J.; Kim, H.-C.; Yamada, S.; Nakai, T.; Nagai, T.; Yokosuka, A.; Mimaki, Y.; et al. Nobiletin, a Citrus Flavonoid, Ameliorates Cognitive Impairment, Oxidative Burden, and Hyperphosphorylation of Tau in Senescence-Accelerated Mouse. Behav. Brain Res. 2013, 250, 351–360. [Google Scholar] [CrossRef]
  162. Paula, P.-C.; Maria, S.-G.A.; Luis, C.-H.; Patricia, C.-G.G. Preventive Effect of Quercetin in a Triple Transgenic Alzheimer’s Disease Mice Model. Molecules 2019, 24, 2287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Qin, L.; Zhang, J.; Qin, M. Protective Effect of Cyanidin 3-O-Glucoside on Beta-Amyloid Peptide-Induced Cognitive Impairment in Rats. Neurosci. Lett. 2013, 534, 285–288. [Google Scholar] [CrossRef]
  164. Currais, A.; Prior, M.; Dargusch, R.; Armando, A.; Ehren, J.; Schubert, D.; Quehenberger, O.; Maher, P. Modulation of P25 and Inflammatory Pathways by Fisetin Maintains Cognitive Function in Alzheimer’s Disease Transgenic Mice. Aging Cell 2014, 13, 379–390. [Google Scholar] [CrossRef]
  165. Liang, J.; López-Valdés, H.E.; Martínez-Coria, H.; Lindemeyer, A.K.; Shen, Y.; Shao, X.M.; Olsen, R.W. Erratum to: Dihydromyricetin Ameliorates Behavioral Deficits and Reverses Neuropathology of Transgenic Mouse Models of Alzheimer’s Disease. Neurochem. Res. 2014, 39, 1403. [Google Scholar] [CrossRef] [Green Version]
  166. Desideri, G.; Kwik-Uribe, C.; Grassi, D.; Necozione, S.; Ghiadoni, L.; Mastroiacovo, D.; Raffaele, A.; Ferri, L.; Bocale, R.; Lechiara, M.C.; et al. Benefits in Cognitive Function, Blood Pressure, and Insulin Resistance through Cocoa Flavanol Consumption in Elderly Subjects with Mild Cognitive Impairment: The Cocoa, Cognition, and Aging (CoCoA) Study. Hypertension 2012, 60, 794–801. [Google Scholar] [CrossRef] [Green Version]
  167. Mastroiacovo, D.; Kwik-Uribe, C.; Grassi, D.; Necozione, S.; Raffaele, A.; Pistacchio, L.; Righetti, R.; Bocale, R.; Lechiara, M.C.; Marini, C.; et al. Cocoa Flavanol Consumption Improves Cognitive Function, Blood Pressure Control, and Metabolic Profile in Elderly Subjects: The Cocoa, Cognition, and Aging (CoCoA) Study—A Randomized Controlled Trial. Am. J. Clin. Nutr. 2015, 101, 538–548. [Google Scholar] [CrossRef] [Green Version]
  168. Brickman, A.M.; Khan, U.A.; Provenzano, F.A.; Yeung, L.-K.; Suzuki, W.; Schroeter, H.; Wall, M.; Sloan, R.P.; Small, S.A. Enhancing Dentate Gyrus Function with Dietary Flavanols Improves Cognition in Older Adults. Nat. Neurosci. 2014, 17, 1798–1803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Caruana, M.; Neuner, J.; Högen, T.; Schmidt, F.; Kamp, F.; Scerri, C.; Giese, A.; Vassallo, N. Polyphenolic Compounds Are Novel Protective Agents against Lipid Membrane Damage by α-Synuclein Aggregates In Vitro. Biochim. Biophys. Acta 2012, 1818, 2502–2510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Takahashi, R.; Ono, K.; Takamura, Y.; Mizuguchi, M.; Ikeda, T.; Nishijo, H.; Yamada, M. Phenolic Compounds Prevent the Oligomerization of α-Synuclein and Reduce Synaptic Toxicity. J. Neurochem. 2015, 134, 943–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Xu, Y.; Zhang, Y.; Quan, Z.; Wong, W.; Guo, J.; Zhang, R.; Yang, Q.; Dai, R.; McGeer, P.L.; Qing, H. Epigallocatechin Gallate (EGCG) Inhibits Alpha-Synuclein Aggregation: A Potential Agent for Parkinson’s Disease. Neurochem. Res. 2016, 41, 2788–2796. [Google Scholar] [CrossRef]
  172. Cui, Y.; Wu, J.; Jung, S.-C.; Park, D.-B.; Maeng, Y.-H.; Hong, J.Y.; Kim, S.-J.; Lee, S.-R.; Kim, S.-J.; Kim, S.J.; et al. Anti-Neuroinflammatory Activity of Nobiletin on Suppression of Microglial Activation. Biol. Pharm. Bull. 2010, 33, 1814–1821. [Google Scholar] [CrossRef] [Green Version]
  173. Rezai-Zadeh, K.; Ehrhart, J.; Bai, Y.; Sanberg, P.R.; Bickford, P.; Tan, J.; Shytle, R.D. Apigenin and Luteolin Modulate Microglial Activation via Inhibition of STAT1-Induced CD40 Expression. J. Neuroinflamm. 2008, 5, 41. [Google Scholar] [CrossRef] [Green Version]
  174. Wu, L.-H.; Lin, C.; Lin, H.-Y.; Liu, Y.-S.; Wu, C.Y.-J.; Tsai, C.-F.; Chang, P.-C.; Yeh, W.-L.; Lu, D.-Y. Naringenin Suppresses Neuroinflammatory Responses Through Inducing Suppressor of Cytokine Signaling 3 Expression. Mol. Neurobiol. 2016, 53, 1080–1091. [Google Scholar] [CrossRef]
  175. Park, H.Y.; Kim, G.-Y.; Choi, Y.H. Naringenin Attenuates the Release of Pro-Inflammatory Mediators from Lipopolysaccharide-Stimulated BV2 Microglia by Inactivating Nuclear Factor-ΚB and Inhibiting Mitogen-Activated Protein Kinases. Int. J. Mol. Med. 2012, 30, 204–210. [Google Scholar] [CrossRef] [Green Version]
  176. Chinta, S.J.; Ganesan, A.; Reis-Rodrigues, P.; Lithgow, G.J.; Andersen, J.K. Anti-Inflammatory Role of the Isoflavone Diadzein in Lipopolysaccharide-Stimulated Microglia: Implications for Parkinson’s Disease. Neurotox. Res. 2013, 23, 145–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Subedi, L.; Ji, E.; Shin, D.; Jin, J.; Yeo, J.H.; Kim, S.Y. Equol, a Dietary Daidzein Gut Metabolite Attenuates Microglial Activation and Potentiates Neuroprotection In Vitro. Nutrients 2017, 9, 207. [Google Scholar] [CrossRef] [Green Version]
  178. Zhang, Z.; Cao, X.; Xiong, N.; Wang, H.; Huang, J.; Sun, S.; Wang, T. Morin Exerts Neuroprotective Actions in Parkinson Disease Models In Vitro and In Vivo. Acta Pharmacol. Sin. 2010, 31, 900–906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Lee, K.M.; Lee, Y.; Chun, H.J.; Kim, A.H.; Kim, J.Y.; Lee, J.Y.; Ishigami, A.; Lee, J. Neuroprotective and Anti-Inflammatory Effects of Morin in a Murine Model of Parkinson’s Disease. J. Neurosci. Res. 2016, 94, 865–878. [Google Scholar] [CrossRef]
  180. Cho, N.; Choi, J.H.; Yang, H.; Jeong, E.J.; Lee, K.Y.; Kim, Y.C.; Sung, S.H. Neuroprotective and Anti-Inflammatory Effects of Flavonoids Isolated from Rhus Verniciflua in Neuronal HT22 and Microglial BV2 Cell Lines. Food Chem. Toxicol. 2012, 50, 1940–1945. [Google Scholar] [CrossRef]
  181. Li, Q.; Zhang, P.; Cai, Y. Genkwanin Suppresses MPP+-Induced Cytotoxicity by Inhibiting TLR4/MyD88/NLRP3 Inflammasome Pathway in a Cellular Model of Parkinson’s Disease. Neurotoxicology 2021, 87, 62–69. [Google Scholar] [CrossRef]
  182. Blandini, F.; Armentero, M.-T. Animal Models of Parkinson’s Disease. FEBS J. 2012, 279, 1156–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Pingale, T.; Gupta, G.L. Classic and Evolving Animal Models in Parkinson’s Disease. Pharmacol. Biochem. Behav. 2020, 199, 173060. [Google Scholar] [CrossRef] [PubMed]
  184. Johnson, M.E.; Bobrovskaya, L. An Update on the Rotenone Models of Parkinson’s Disease: Their Ability to Reproduce the Features of Clinical Disease and Model Gene-Environment Interactions. Neurotoxicology 2015, 46, 101–116. [Google Scholar] [CrossRef]
  185. Hu, Q.; Uversky, V.N.; Huang, M.; Kang, H.; Xu, F.; Liu, X.; Lian, L.; Liang, Q.; Jiang, H.; Liu, A.; et al. Baicalein Inhibits α-Synuclein Oligomer Formation and Prevents Progression of α-Synuclein Accumulation in a Rotenone Mouse Model of Parkinson’s Disease. Biochim. Biophys. Acta 2016, 1862, 1883–1890. [Google Scholar] [CrossRef] [PubMed]
  186. Hung, K.-C.; Huang, H.-J.; Wang, Y.-T.; Lin, A.M.-Y. Baicalein Attenuates α-Synuclein Aggregation, Inflammasome Activation and Autophagy in the MPP+-Treated Nigrostriatal Dopaminergic System in Vivo. J. Ethnopharmacol. 2016, 194, 522–529. [Google Scholar] [CrossRef]
  187. Lee, E.; Park, H.R.; Ji, S.T.; Lee, Y.; Lee, J. Baicalein Attenuates Astroglial Activation in the 1-Methyl-4-Phenyl-1,2,3,4-Tetrahydropyridine-Induced Parkinson’s Disease Model by Downregulating the Activations of Nuclear Factor-ΚB, ERK, and JNK. J. Neurosci. Res. 2014, 92, 130–139. [Google Scholar] [CrossRef]
  188. Mu, X.; He, G.-R.; Yuan, X.; Li, X.-X.; Du, G.-H. Baicalein Protects the Brain against Neuron Impairments Induced by MPTP in C57BL/6 Mice. Pharmacol. Biochem. Behav. 2011, 98, 286–291. [Google Scholar] [CrossRef] [PubMed]
  189. Cheng, Y.; He, G.; Mu, X.; Zhang, T.; Li, X.; Hu, J.; Xu, B.; Du, G. Neuroprotective Effect of Baicalein against MPTP Neurotoxicity: Behavioral, Biochemical and Immunohistochemical Profile. Neurosci. Lett. 2008, 441, 16–20. [Google Scholar] [CrossRef]
  190. Anusha, C.; Sumathi, T.; Joseph, L.D. Protective Role of Apigenin on Rotenone Induced Rat Model of Parkinson’s Disease: Suppression of Neuroinflammation and Oxidative Stress Mediated Apoptosis. Chem. Biol. Interact. 2017, 269, 67–79. [Google Scholar] [CrossRef] [PubMed]
  191. Jeong, K.H.; Jeon, M.-T.; Kim, H.D.; Jung, U.J.; Jang, M.C.; Chu, J.W.; Yang, S.J.; Choi, I.Y.; Choi, M.-S.; Kim, S.R. Nobiletin Protects Dopaminergic Neurons in the 1-Methyl-4-Phenylpyridinium-Treated Rat Model of Parkinson’s Disease. J. Med. Food 2015, 18, 409–414. [Google Scholar] [CrossRef] [PubMed]
  192. Yabuki, Y.; Ohizumi, Y.; Yokosuka, A.; Mimaki, Y.; Fukunaga, K. Nobiletin Treatment Improves Motor and Cognitive Deficits Seen in MPTP-Induced Parkinson Model Mice. Neuroscience 2014, 259, 126–141. [Google Scholar] [CrossRef]
  193. Leem, E.; Nam, J.H.; Jeon, M.-T.; Shin, W.-H.; Won, S.-Y.; Park, S.-J.; Choi, M.-S.; Jin, B.K.; Jung, U.J.; Kim, S.R. Naringin Protects the Nigrostriatal Dopaminergic Projection through Induction of GDNF in a Neurotoxin Model of Parkinson’s Disease. J. Nutr. Biochem. 2014, 25, 801–806. [Google Scholar] [CrossRef]
  194. Kim, H.D.; Jeong, K.H.; Jung, U.J.; Kim, S.R. Naringin Treatment Induces Neuroprotective Effects in a Mouse Model of Parkinson’s Disease in Vivo, but Not Enough to Restore the Lesioned Dopaminergic System. J. Nutr. Biochem. 2016, 28, 140–146. [Google Scholar] [CrossRef]
  195. Levites, Y.; Weinreb, O.; Maor, G.; Youdim, M.B.; Mandel, S. Green Tea Polyphenol (−)-Epigallocatechin-3-Gallate Prevents N-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Induced Dopaminergic Neurodegeneration. J. Neurochem. 2001, 78, 1073–1082. [Google Scholar] [CrossRef]
  196. Tseng, H.-C.; Wang, M.-H.; Chang, K.-C.; Soung, H.-S.; Fang, C.-H.; Lin, Y.-W.; Li, K.-Y.; Yang, C.-C.; Tsai, C.-C. Protective Effect of (−)Epigallocatechin-3-Gallate on Rotenone-Induced Parkinsonism-like Symptoms in Rats. Neurotox. Res. 2020, 37, 669–682. [Google Scholar] [CrossRef] [PubMed]
  197. Haleagrahara, N.; Siew, C.J.; Mitra, N.K.; Kumari, M. Neuroprotective Effect of Bioflavonoid Quercetin in 6-Hydroxydopamine-Induced Oxidative Stress Biomarkers in the Rat Striatum. Neurosci. Lett. 2011, 500, 139–143. [Google Scholar] [CrossRef] [PubMed]
  198. Ay, M.; Luo, J.; Langley, M.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Molecular Mechanisms Underlying Protective Effects of Quercetin against Mitochondrial Dysfunction and Progressive Dopaminergic Neurodegeneration in Cell Culture and MitoPark Transgenic Mouse Models of Parkinson’s Disease. J. Neurochem. 2017, 141, 766–782. [Google Scholar] [CrossRef] [Green Version]
  199. Li, S.; Pu, X.-P. Neuroprotective Effect of Kaempferol against a 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-Induced Mouse Model of Parkinson’s Disease. Biol. Pharm. Bull. 2011, 34, 1291–1296. [Google Scholar] [CrossRef] [Green Version]
  200. Lv, C.; Hong, T.; Yang, Z.; Zhang, Y.; Wang, L.; Dong, M.; Zhao, J.; Mu, J.; Meng, Y. Effect of Quercetin in the 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-Induced Mouse Model of Parkinson’s Disease. Evid. Based Complement. Altern. Med. 2012, 2012, 928643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Tamilselvam, K.; Nataraj, J.; Janakiraman, U.; Manivasagam, T.; Essa, M.M. Antioxidant and Anti-Inflammatory Potential of Hesperidin against 1-Methyl-4-Phenyl-1, 2, 3, 6-Tetrahydropyridine-Induced Experimental Parkinson’s Disease in Mice. Int. J. Nutr. Pharmacol. Neurol. Dis. 2013, 3, 294. [Google Scholar] [CrossRef]
  202. Baluchnejadmojarad, T.; Roghani, M. The Flavonoid Hesperetin Alleviates Behavioral Abnormality in 6-Hydroxydopamine Rat Model of Hemi-Parkinsonism. Basic Clin. Neurosci. 2010, 2, 20–23. [Google Scholar]
  203. Datla, K.P.; Christidou, M.; Widmer, W.W.; Rooprai, H.K.; Dexter, D.T. Tissue Distribution and Neuroprotective Effects of Citrus Flavonoid Tangeretin in a Rat Model of Parkinson’s Disease. Neuroreport 2001, 12, 3871–3875. [Google Scholar] [CrossRef]
  204. Khan, M.M.; Raza, S.S.; Javed, H.; Ahmad, A.; Khan, A.; Islam, F.; Safhi, M.M.; Islam, F. Rutin Protects Dopaminergic Neurons from Oxidative Stress in an Animal Model of Parkinson’s Disease. Neurotox. Res. 2012, 22, 1–15. [Google Scholar] [CrossRef]
  205. Baluchnejadmojarad, T.; Jamali-Raeufy, N.; Zabihnejad, S.; Rabiee, N.; Roghani, M. Troxerutin Exerts Neuroprotection in 6-Hydroxydopamine Lesion Rat Model of Parkinson’s Disease: Possible Involvement of PI3K/ERβ Signaling. Eur. J. Pharmacol. 2017, 801, 72–78. [Google Scholar] [CrossRef]
  206. Kim, H.D.; Jeong, K.H.; Jung, U.J.; Kim, S.R. Myricitrin Ameliorates 6-Hydroxydopamine-Induced Dopaminergic Neuronal Loss in the Substantia Nigra of Mouse Brain. J. Med. Food 2016, 19, 374–382. [Google Scholar] [CrossRef] [PubMed]
  207. Wu, H.; Liu, X.; Gao, Z.-Y.; Lin, M.; Zhao, X.; Sun, Y.; Pu, X.-P. Icaritin Provides Neuroprotection in Parkinson’s Disease by Attenuating Neuroinflammation, Oxidative Stress, and Energy Deficiency. Antioxidants 2021, 10, 529. [Google Scholar] [CrossRef] [PubMed]
  208. Altharawi, A.; Alharthy, K.M.; Althurwi, H.N.; Albaqami, F.F.; Alzarea, S.I.; Al-Abbasi, F.A.; Nadeem, M.S.; Kazmi, I. Europinidin Inhibits Rotenone-Activated Parkinson’s Disease in Rodents by Decreasing Lipid Peroxidation and Inflammatory Cytokines Pathways. Molecules 2022, 27, 7159. [Google Scholar] [CrossRef]
  209. Habib, C.N.; Mohamed, M.R.; Tadros, M.G.; Tolba, M.F.; Menze, E.T.; Masoud, S.I. The Potential Neuroprotective Effect of Diosmin in Rotenone-Induced Model of Parkinson’s Disease in Rats. Eur. J. Pharmacol. 2022, 914, 174573. [Google Scholar] [CrossRef] [PubMed]
  210. Coe, S.; Andreoli, D.; George, M.; Collett, J.; Reed, A.; Cossington, J.; Izadi, H.; Dixon, A.; Mansoubi, M.; Dawes, H. A Feasibility Study to Determine Whether the Daily Consumption of Flavonoid-Rich Pure Cocoa Has the Potential to Reduce Fatigue and Fatigability in People with Parkinson’s (PwP). Clin. Nutr. ESPEN 2022, 48, 68–73. [Google Scholar] [CrossRef]
  211. Holt, E.M.; Steffen, L.M.; Moran, A.; Basu, S.; Steinberger, J.; Ross, J.A.; Hong, C.-P.; Sinaiko, A.R. Fruit and Vegetable Consumption and Its Relation to Markers of Inflammation and Oxidative Stress in Adolescents. J. Am. Diet. Assoc. 2009, 109, 414–421. [Google Scholar] [CrossRef] [Green Version]
  212. Steptoe, A.; Gibson, E.L.; Vuononvirta, R.; Hamer, M.; Wardle, J.; Rycroft, J.A.; Martin, J.F.; Erusalimsky, J.D. The Effects of Chronic Tea Intake on Platelet Activation and Inflammation: A Double-Blind Placebo Controlled Trial. Atherosclerosis 2007, 193, 277–282. [Google Scholar] [CrossRef]
  213. Widlansky, M.E.; Duffy, S.J.; Hamburg, N.M.; Gokce, N.; Warden, B.A.; Wiseman, S.; Keaney, J.F.; Frei, B.; Vita, J.A. Effects of Black Tea Consumption on Plasma Catechins and Markers of Oxidative Stress and Inflammation in Patients with Coronary Artery Disease. Free Radic. Biol. Med. 2005, 38, 499–506. [Google Scholar] [CrossRef]
  214. Costa, R.; Lima, S.A.C.; Gameiro, P.; Reis, S. On the Development of a Cutaneous Flavonoid Delivery System: Advances and Limitations. Antioxidants 2021, 10, 1376. [Google Scholar] [CrossRef]
  215. Hendawy, O.M. Nano-Delivery Systems for Improving Therapeutic Efficiency of Dietary Polyphenols. Altern. Ther. Health Med. 2021, 27, 162–177. [Google Scholar]
Figure 1. Basic flavonoid share a C6–C3–C6 structure containing two aromatic rings (A and B rings) linked by a three-carbon bridge. Some representative compounds are shown below each flavonoid subgroup.
Figure 1. Basic flavonoid share a C6–C3–C6 structure containing two aromatic rings (A and B rings) linked by a three-carbon bridge. Some representative compounds are shown below each flavonoid subgroup.
Ijms 24 04297 g001
Figure 2. Basic neuroinflammatory responses. In homeostatic conditions, microglia and astrocytes support several brain cell functions, but during aging and in brain diseases, they change their morphology and secretome. Damage-associated molecular patterns (DAMPs) released in pathologic conditions interact with homeostatic microglia, and specific signaling occurs to induce a pro-inflammatory phenotype, which results in a decreases phagocytic effect and the release of several molecules that change homeostatic astrocytes functions towards a proinflammatory state to exacerbate inflammation and worsened the functional recovery.
Figure 2. Basic neuroinflammatory responses. In homeostatic conditions, microglia and astrocytes support several brain cell functions, but during aging and in brain diseases, they change their morphology and secretome. Damage-associated molecular patterns (DAMPs) released in pathologic conditions interact with homeostatic microglia, and specific signaling occurs to induce a pro-inflammatory phenotype, which results in a decreases phagocytic effect and the release of several molecules that change homeostatic astrocytes functions towards a proinflammatory state to exacerbate inflammation and worsened the functional recovery.
Ijms 24 04297 g002
Figure 3. Anti-inflammatory effect of flavonoids. Flavonoids act as inhibitors of the activity of several transcription factors and regulatory enzymes to decrease the synthesis and release of different the pro-inflammatory cytokines.
Figure 3. Anti-inflammatory effect of flavonoids. Flavonoids act as inhibitors of the activity of several transcription factors and regulatory enzymes to decrease the synthesis and release of different the pro-inflammatory cytokines.
Ijms 24 04297 g003
Table 1. Anti-inflammatory effects of flavonoids in focal ischemic stroke models and clinic studies.
Table 1. Anti-inflammatory effects of flavonoids in focal ischemic stroke models and clinic studies.
FlavonoidEffectModel (In Vitro)References
Myricetin↓ TNF-α, IL-1β, and IL-6OGD/R, endothelial cells[92]
Quercetin↓ TLR4/MyD88/NF-κB signalingBV2 microglial cells[93]
Isoquercetin↓ TLR4, NF-κB, TNF-α, and IL-6OGD/R, neurons[94,95]
Baicalin↓ TNFα and NOD2 receptorOGD, BV2 microglial cells[96]
Baicalin↓ NLRP3 inflammasomeOGD/R, cortical neurons[97]
Baicalin↓ TNF-α, IL-1β, and IL-6OGD/R, endothelial cells[98]
Baicalin↓ TNF-α, IL-1β, and IL-6OGD/R, BV2 microglia cell[99]
Baicalin↓ TLR4/NF-κB pathwaymicroglia-neuron co-culture[100]
Baicalin↓ TLR4, MYD88, p-NF-κB expression,
↓ IL-6, IL-1α, IL-1β, IL8, and TNF-α
OGD, endothelial cells[101,102]
Icariin↓ IL-1β, IL-6, and TNF-α expressionOGD/R, microglia[103]
Casticin↓ TLR4, NF-κB p65, NF-κB, and p50 expressionOGD/R, PC12 cells[104]
Pratensein↓ NLRP3 inflammasomeOGD/R, HT22 cells[105]
Tectorigenin↓ IL-1β, IL-6, and TNF-αOGD/R, HT22 cells[106]
Astilbin↓ NLRP3 inflammasome, IL-1β, and IL-18OGD, PC12 cells[107]
Anthocyanin↓ TNF-α, IL-1β, and IL-6OGD, SH-SY5Y cells[108]
Tricin↓ TNF-α and IL-6 and ↓ IL-1β expressionNeuroblastoma cells[109]
Diosmetin↓ NLRP3 inflammasomeOGD/R, PC12 cells[110]
Schaftoside↓ TLR4, IL-1β, IL-6, and ↓ TNFα expressionOGD, BV2 microglia[111]
FlavonoidEffectModel (In Vivo)References
Kaempferol-3-O-rutinoside and 3-O-glucoside↓ infarct volume, STAT3, NF-κB and IL-1βMCAO/R, rat[113]
Kaempferol↓ IL-5, TNF-α, IL-1β, and IL-6MCAO/R, rat[114]
Fisetin↓ infarct size, TNFaMCAO/R, mouse[115]
Morin↓ neurological deficits, TNF-α, and IL-6MCAO/R, rat[116]
Morin↓ pNF-κB, TNF-α, and IL-1β, TLR4 expression,
↑ occluding, claudin expression
MCAO/R, rat[117]
EGCG↓ infarct volume, TNF-α, IL-1β, IL-6, and NF-κB/p65MCAO/R, rat[118]
Luteolin↓ infarct volume, astrocytes and microglia activationMCAO/R, rat[119]
Luteoloside↓ infarct volume, TNF-α, and IL-1βMCAO, rat[120]
Nobiletin↓ infarct volume, brain swelling, and neurological deficitsMCOA/R, rat[121]
Eriodictyol↓ infarct volume, neurological deficits, TNF-α, and GFAP expressionMCAO, mouse[122]
Tricin↓ TNF-α, IL-6, and IL-1β in serumMCAO, mouse[[109]
Eupatilin↓ microglia activation and NF-κB pathwayMCAO, mouse[123]
EGCG + rt-PA↓ neurobehavioral deficit, brain infarction, cerebral edema, and blood-brain barrier disruptionMCAO, rat[124]
FlavonoidEffect (Preventive Treatment)Model (In Vivo)References
Naringenin↓ infarct volume, NF-κB, TNF-α, IL-1β, GFAP, and Iba1MCOA/R, rat[125]
Naringenin↓ infarct volume, neurologic deficits, and NF-κBMCOA/R, rat[126]
Hesperidin↓ IL-1βMCOA, rat[127]
Pinocembrin↓ infarct size and NF-κB, TNF-α, and IL-6Global stroke, rat[128]
Genistein↓ infarct volume, neurological deficit, and NF-κB activationMCAO, mouse[129]
Sanggenon↓ TNF-α, IL-1β, and IL-6MCOA, rat[130]
Astilbin↓ NLRP3 inflammasome, IL-1β, and IL-18MCOA, rat[107]
Chrysin↓ IL-6, IL-1β, and TNF-αMCOA, rat[131]
Eupafolin↓ TLR-4, TNF-α, IL-1β, and IL-6 expressionMCOA/R, rat[132]
Biochanin A↓ TNF-α and IL-1β expressionMCOA, rat[133]
FlavonoidEffectClinical StudiesReferences
fisetin + rt-PA↑ therapeutic window of rt-PA and
↓ neurological deficits, C-reactive protein
Double-blind randomized placebo-controlled[134]
EGCG + rt-PA↑ therapeutic window of rt-PA,
↓ MMP 2, and MMP 9
Double-blind randomized placebo-controlled[135]
↑: significant increase; ↓: significant decrease; TNF-α: tumor necrosis factor α; IL: interleukin; TLR4: Toll-like receptor 4; NF-κB: nuclear factor kappa B; rtPA: recombinant tissue plasminogen activator; OGD: oxygen glucose deprivation; OGD/R: oxygen glucose deprivation with reperfusion; MCAO: middle cerebral artery occlusion; MCAO/R: middle cerebral artery occlusion with reperfusion; MMP: matrix metalloproteinase.
Table 2. Anti-inflammatory effects of flavonoids in AD models and clinical studies.
Table 2. Anti-inflammatory effects of flavonoids in AD models and clinical studies.
FlavonoidEffectModel (In Vitro)References
EGCGPrevents Aβ fibrillogenesis and
↓ cell toxicity
Protein aggregation, PC12 cells[136,137]
EGCGProtective against Aβ toxicityHippocampal neuronal cell culture[138]
Quercetin↓ Aβ cytotoxicity, lipid peroxidation, protein oxidation, and apoptosisHippocampal neuronal cell culture[139]
Luteolin↓ Aβ (1–40 and 1–42)Neuronal cells and SweAPP N2a cells[140]
Diosmetin↓ Aβ (1–40 and 1-42)Neuronal cells and SweAPP N2a cells[140]
MyricetinPrevents Aβ fibrillogenesisCerebral cortices from Tg2576 mouse embryos[141]
Cyanidin 3-O-β-glucopyranoside↓ Aβ (25–35) cytotoxicitySH-SY5Y cells[142]
Wogonin↓ Aβ aggregation and phosphorylated TauSH-SY5Y cells[143]
BaicaleinPrevents tau protein aggregationSeveral biochemical techniques[144,145]
QuercetinPrevent Aβ aggregation and ↑ disaggregateCell system overexpressing APP[146]
RutinPrevent Aβ aggregation and ↑ disaggregateCell system overexpressing APP[146]
FlavonoidEffectModel (In Vivo)References
Isorhamnetin↓ learning and memory deficits and
↑ BDNF in prefrontal cortex and hippocampus
Chemical mouse model[149]
NaringinImprove memoryChemical rat model[150]
RutinImprove memoryChemical rat model[150]
kaempferolImprove memory and ↑ density of neurons in hippocampusChemical rat model[151]
LuteolinImprove memoryChemical rat model[152]
Hesperidinimproves memory and ↓ NF κB, iNOS, COX-2, and astrogliosisChemical mouse model[153]
Nobiletin↓ soluble Aβ (1–40 and 1–42) and Aβ plaques in the hippocampusAPP-SL 7-5 transgenic mouse[154]
Diosmin and its bioactive metabolites↓ tau hyperphosphorylation and Aβ generation3xTg transgenic mouse[155]
Hesperidin↓ Aβ plaque in cortex and hippocampus and ↓ astrocyte and microglial activationTransgenic APP/PS1 mouse[156]
WogoninImprove memoryTransgenic h-APPswe mouse[143]
Diosmin↓ Aβ 1–40 and 1–42Tg2576 transgenic mouse[140]
Luteolin↓ Aβ 1–40 and 1–42Tg2576 transgenic mouse[140]
EGCG↓ soluble Aβ (1–40 and 1–42) and Aβ plaques in cortex and hippocampusAPPsw transgenic mouse[157]
EGCG↓ Aβ (1–42)Aβ infusion model, presenilin 2 mutant mouse[158]
DiosminImprove memoryAPPsw transgenic mouse[159]
Nobiletin↓ Aβ plaques in the hippocampus and
↓ memory deficits
APP-SL 7-5 transgenic mouse[154]
Nobiletin↓ memory impairment, ↓ the levels of Aβ 1–403XTg transgenic mouse[160]
Nobiletin↓ memory impairmentSenescence-accelerated mice SAMP8[161]
Quercetin↑ memory and ↓ plaques of Aβ and hyperphosphorylated tau in hippocampus3XTg transgenic mouse[162]
Cyanidin 3-O-glucoside↓ memory impairment, ↓ hyperphosphorylated tau in hippocampusAβ infusion rats[163]
Fisetin↓ memory and learning problemsAPP(swe)/PS1(ΔE9) mouse[164]
Dihydromyricetin↑ exploratory and locomotor activity, and memory, ↓ anxiety and Aβ accumulationTG2576 and TG-SwDI mouse[165]
FlavonoidEffectClinical StudiesReferences
Cocoa flavanolImproves cognitive functionPatients with mild cognitive impairment[166]
Cocoa flavanolImproved cognitive function in aging subjectsDouble-blind study[167]
Cocoa flavanolImproves dentate gyrus functionsfMRI in healthy 50–69-year-old subjects[168]
↑: significant increase; ↓: significant decrease; TNF-α: tumor necrosis factor α; IL: interleukin; TLR4: Toll-like receptor 4; NF-κB: nuclear factor kappa B; Aβ: amyloid-beta; iNOS: inducible nitric oxide synthase; COX-2: cyclooxygenase 2; fMRI: functional magnetic resonance imaging.
Table 3. Anti-inflammatory effects of flavonoids in PD models and clinical studies.
Table 3. Anti-inflammatory effects of flavonoids in PD models and clinical studies.
FlavonoidEffectModel (In Vitro)References
ApigeninInhibit oligomer formation and aggregation of α-synucleinProtein aggregation[169]
BaicaleinInhibit oligomer formation and aggregation of α-synucleinProtein aggregation[169]
MyricetinInhibit oligomer formation and aggregation of α-synucleinProtein aggregation[170]
MorinInhibit oligomer formation and aggregation of α-synucleinProtein aggregation[169]
GenisteinInhibit oligomer formation and aggregation of α-synucleinProtein aggregation[169]
QuercetinInhibit oligomer formation and aggregation of α-synucleinProtein aggregation[169]
EGCGInhibit oligomer formation and aggregation of α-synucleinProtein aggregation[171]
ScutellareinInhibit oligomer formation and aggregation of α-synucleinProtein aggregation[169]
Nobiletin↓ TNF-α and IL-1βActivated microglia[172]
Apigenin↓ TNF-α and IL-6Activated microglia[173]
Luteolin↓ TNF-α and IL-6Activated microglia[173]
Naringenin↓ NF-κB, iNOS, and COX-2Activated microglia[174,175]
Diadzein↓ NF-κB and IL-6Activated microglia[176]
Equol↓ TNF-α, IL-6, and NF-κBActivated microglia[177]
Morin↓ cell apoptosis and mortalityPC12 cells exposed to MPP+[178]
Morin↓ astrogliosis and NF-κBAstrocytes exposed to MPP+[179]
Butein↓ toxicityHT22 and Microglial BV2 cells exposed to glutamate[180]
Butin↓ toxicityHT22 and Microglial BV2 cells exposed to glutamate[180]
Fisetin↓ toxicityHT22 and Microglial BV2 cells exposed to glutamate[180]
Fustin↓toxicityHT22 and Microglial BV2 cells exposed to glutamate[180]
Sulfuretin↓ toxicityHT22 and Microglial BV2 cells exposed to glutamate[180]
Genkwanin↓ TLR4/MyD88/NLRP3 inflammasome pathwaySH-SY5Y cells[181]
FlavonoidEffectModel (In Vivo)References
Baicalein↓ α-synucleinRotenone[185]
Baicalein↓ α-synuclein and inflammasomeMPP+ rat[186]
Baicalein↑ motor ability, ↓ activated microglia and astrocytes, and
↑ dopamine and serotonin in the striatum
MPP+ rat[187,188,189]
Apigenin↓ α-synuclein and NF-κBRotenone rat[190]
Nobiletin↓ microglial activationMPP+ rat[191]
Nobiletin↑ dopamine in the striatum and hippocampal regionMPP+ rat[192]
Naringin↑ GDNF in the substantia nigraMPP+ rat[193]
NaringinProtects the nigrostriatal DA projectionMPP+ rat[194]
EGCG↓ dopamine neuronal lossMPP+ mouse[195]
EGCG↓ TNF-α, IL-1β, and IL-6 in the striatumMPP+ rat[196]
Quercetin↑ dopamine in the striatum, ↓ dopamine neuronal loss6-OHDA rat[197]
Quercetin↓ dopaminergic neuronal loss and behavioral deficitsMitoPark transgenic mouse[198]
Quercetin↑dopamine and motor coordinationMTPT mouse[199,200]
Kaempferol↑dopamine and motor coordinationMTPT mouse[199,200]
Hesperidin↑ motor coordination and ↓ TNF-α, IL-1β, and IL-6MTPT mouse[201,202]
TangeretinProtects the striatal dopaminergic neurons6-OHDA rat[203]
Rutin↑ motor coordination and dopaminergic neurons6-OHDA rat[204]
Troxerutin↓ neuronal loss and astrogliosis6-Hydroxydopamine lesion, rat[205]
MyricitrinProtects the striatal dopaminergic neurons and ↓ TNF-α6-Hydroxydopamine lesion, rat[206]
Morin↓ neuronal loss and behavioral deficitsMTPT mouse[178]
Morin↑ motor coordination and ↓ dopamine neuronal lossMTPT mouse[179]
Icariin↓ NLRP3 inflammasome, IL-1β, and TNF-α in serumMTPT mouse[207]
Europinidin↓ IL-6, IL-1β, and TNF-αRotenone rat[208]
Diosmin↓ TNF-α and NF-κBRotenone rat[209]
FlavonoidEffectClinical StudiesReferences
Cocoa flavanol↓ fatigue and fatigabilityDouble-blind placebo-controlled[210]
↑: significant increase; ↓: significant decrease; TNF-α: tumor necrosis factor α; IL: interleukin; TLR4: Toll-like receptor 4; NF-κB: nuclear factor kappa B; iNOS: inducible nitric oxide synthase; COX-2: cyclooxygenase 2; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; 6-OHDA: 6-hydroxydopamine: MPDP+: 1-methyl-4-phenyl-2,3-dihydropyridinium.
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

Martínez-Coria, H.; Arrieta-Cruz, I.; Gutiérrez-Juárez, R.; López-Valdés, H.E. Anti-Inflammatory Effects of Flavonoids in Common Neurological Disorders Associated with Aging. Int. J. Mol. Sci. 2023, 24, 4297. https://doi.org/10.3390/ijms24054297

AMA Style

Martínez-Coria H, Arrieta-Cruz I, Gutiérrez-Juárez R, López-Valdés HE. Anti-Inflammatory Effects of Flavonoids in Common Neurological Disorders Associated with Aging. International Journal of Molecular Sciences. 2023; 24(5):4297. https://doi.org/10.3390/ijms24054297

Chicago/Turabian Style

Martínez-Coria, Hilda, Isabel Arrieta-Cruz, Roger Gutiérrez-Juárez, and Héctor Eduardo López-Valdés. 2023. "Anti-Inflammatory Effects of Flavonoids in Common Neurological Disorders Associated with Aging" International Journal of Molecular Sciences 24, no. 5: 4297. https://doi.org/10.3390/ijms24054297

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