Introduction

There is a growing public health concern associated with traumatic brain injuries (TBIs) in people under 45 years of age. TBI is predicted to become the third most common cause of death worldwide by 2020, according to the World Health Organization (WHO) [1]. It is estimated that patients who survive severe TBI are permanently disabled in terms of neurological and psychological functioning and suffer a heavy financial burden. According to Wittchen et al., the costs associated with TBI exceed €33 billion annually in Europe alone, making TBI a key research area on many national agendas [2]. Despite advances in prevention and early resuscitation techniques, long-term neurological morbidity and overall neurological recovery remain major health challenges. To the best of our knowledge, there are currently no approved treatments for TBI that have been approved by any of the regulatory agencies in order to prove their effectiveness [3]. TBI is complex and heterogeneous, and conventional methods of diagnosing and treating it have proved inadequate. As a consequence, alternative methods for develo** TBI therapies should be explored [4].

Medicinal plant sand their bioactive ingredients, in particular polyphenols, have been implicated in various biological activities, including, but not limited to, immunomodulation, anti-inflammatory, cardiovascular protection, antioxidant, and anticancer potential [5,6,7]. It is well known that plants produce polyphenols in the form of glycoside esters and free aglycones. The polyphenol family contains more than 8000 structural variants [8]. Fruits and vegetables contain polyphenols, bioactive compounds responsible for their color, flavor, and health benefits. Based on their chemical structures, they can be divided into several different classes: flavonoids such as flavonols, flavones, isoflavones, neoflavonoids, anthocyanidins, chalcones, and proanthocyanidins, phenolic acids, stilbenoids and phenolic amides [9]. Multiple aromatic rings are attached to hydroxyl ligands to form these molecules, which are predominantly plant metabolites. They can be classified according to their chemical structures [10]. Antioxidants derived from polyphenolic compounds are mainly phenolic compounds derived from plants. Some of these compounds may exist as ester derivatives (e.g., epigallocatechin gallate) or ether derivatives (e.g., ferulic acid) [11]. A polyphenolic compound can be divided into two categories based on the type of phenolic compound it belongs to. In addition, these flavonoids have also been found in pharmaceutical preparations. Among those are epigallocatechin gallate, epigallocatechin, catechins, fisetin, luteolin, and quercetin, which are often used as ingredients in pharmaceutical preparations [12]. There is an active ingredient in Quercetin which inhibits the activity of BACE-1, which is a cleaving enzyme of the amyloid precursor protein [13]. In neurodegenerative diseases like Parkinson’s disease (PD), traumatic brain injury (TBI), and Alzheimer’s disease (AD), flavonoids and NSAIDs modulate the nuclear factor-kappa β (NF-κB) signaling pathway [14]. By scavenging free radicals, polyphenols protect against chronic diseases characterized by the involvement of free radicals in their pathogenesis. Some plant species, such as black soybeans, contain polyphenolic phytochemicals that are effective in maintaining human health, particularly in preventing cancer, neurological disorders, cardiovascular disease, and diabetes. However, their effect on the progression of TBI is controversial, perhaps because of the irreversible brain damage following TBI [15]. A review of various in vitro and in vivo studies on the therapeutic benefits of plant-derived phyotchemicals, with curcumin as an example, in TBI has been presented in this review (Fig. 1).

Fig. 1
figure 1

Therapeutic advantages of curcumin, a polyphenol, against traumatic brain injury through interaction with different inflammatory signaling pathways and their effects on levels of cytokines and related biomarkers

Pathogenesis of TBI

There has been a rapid increase in TBIs and they are considered a highly complex condition. Accidents involving motor vehicles, abuse, and concussions are among the leading causes of TBI. According to research, there are three levels of severity associated with this disease, mild, moderate, and severe. During recovery, there is sometimes a debilitating and transient neurological disorder. While injuries to other parts of the body contribute significantly to brain damage in severe cases such as polytrauma, injuries to the brain can cause even more significant damage [16].

There are several injuries that can result from TBI, including primary and secondary injuries. Besides lacerations and contusions, primary injuries can result in hemorrhages and ruptures. In addition to the primary injury caused by the traumatic event itself, the secondary injuries that develop as a result of inflammation, oxidative stress, and glutamate toxicity have been shown to play a significant role in determining the outcome of a traumatic brain injury. Edema and cell death occur in the brain due to calcium imbalance and oxidative stress after a post-TBI injury [17]. Often, secondary injury is observed within a few hours to a few days of the initial injury due to a cascade of events. After the primary injury mechanism is triggered, a second wave of pathological changes, commonly known as secondary injury mechanisms, is initiated, including metabolic changes, neuronal inflammation, vascular complications, cell death of glia and neurons, axonal damage, and mitochondrial dysfunction. Damaged mitochondria produce large amounts of oxygen near the site of injury. Autophagy destroys these mitochondria for relative neuronal protection and reduces oxidative stress [18, 19]. In neurons that have been damaged by cell death proteins such as those associated with mitochondrial apoptosis are triggered when mitochondrial membranes are damaged and permeability increases. Autophagy is one of the processes in the starvation response that is regulated by the organ and is responsible for the destruction and recycling of cellular components, as well as participating in organ circulation and controlling bioenergetics in the body [20]. Furthermore, autophagy prevents mitochondrial apoptosis or neuroinflammation, in addition to its neuroprotective effects. Furthermore, it is also important to note that excessive autophagy can also contribute to the generation of neuronal death when there are ischemic or hypoxic conditions, although its inhibition does the opposite.

Some signaling pathways such as phosphatidylinositol 3-kinase/ protein kinase B (PI3K/AKT), are involved in apoptosis, and induction of this pathway can suppress it post-TBI [21]. The secondary injury that results from TBI is characterized by an array of inflammatory responses. The Toll-like Receptors (TLRs) in the human immune system are located on the surface of the cells and are responsible for activating the immune system by releasing endogenous ligands and recognizing a wide range of pathogen-associated molecular patterns (PAMPs), including lipopolysaccharides (LPS) flagellin, and single-stranded and double-stranded viral RNA. Multiple aspects of CNS homeostasis are affected by these patterns, which release enzymes and cytokines that trigger an inflammatory cascade [22]. There is increasing evidence that TLR4 plays a critical role in the initiation of inflammatory responses after trauma. TLRs activate two distinct pathways leading to the activation of transcription factors that regulate the expression of pro-inflammatory cytokine genes. Myeloid differentiation factor 88 (Myd88) activates the TNF receptor-associated factor 6 (TRAF6), producing pro-inflammatory cytokines. There is an interaction between adapter-inducing interferons (TRIF) that contain TIR domains, which are activated in a pathway independent of Myd88, and induce nuclear factor kappa B (NF-κB) to produce inflammatory mediators [23, 24].

The NF-κB family consists of five proteins, including NF-κB1 (p50), NF-κB2 (p52), p65, RELB, and c-REL. Both endogenous and exogenous ligands activate NF-κB and, when activated, is found in neurons and glia under various \inflammatory conditions. In addition to the canonical pathway, a non-canonical pathway activates NF-κB [25]. The NF-κB pathway is also involved in a wide range of immune responses and various cellular progressions such as apoptosis and proliferation. As a result of TBI, activation of NF-κB releases several inflammatory agents that lead to secondary brain damage [26]. A recent study has shown that TBI exacerbates oxidative stress by producing free radicals [27]. There is increasing evidence that oxidative stress plays an important role in secondary damage in relation to primary damage. The nuclear factor erythroid 2 related factor 2 (Nrf2) is involved in a variety of functions within cells and accumulates in their cytoplasm under normal conditions [28]. In addition to regulating genes involved in oxidative stress, Nrf2 acts as a transcription factor. As a result of this component of Nrf2, antioxidant enzymes such as malondialdehyde (MDA), heme oxygenase-1 (HO1), superoxide dismutase (SOD), quinone oxidoreductase-1 (NQO1), and glutathione peroxidase (GSH-Px) are activated [29]. This enzyme acts by directly regulating the levels of reactive oxygen species (ROS), meaning that it can directly react with free radicals without harming the cells' vital function. The antioxidant response element (ARE) found in the gene promoter allows Nrf2 to promote the expression of antioxidant genes. The Nrf2/ARE signaling pathway modulates several pathological processes, including oxidative stress [29, 30].

Effect of Different Phytochemicals and Polyphenols on TBI Through Inflammatory Signaling Pathways

Polyphenols exert antioxidant activity through various mechanisms, some of which are multi-step mechanisms [31]. However, free radical scavenging is widely recognized as one mechanism by which polyphenols exert their antioxidant effects. The extended conjugation of the unpaired electron is another key property of polyphenolic antioxidant radicals that contributes to their increased stability. By quenching the reactive nitrogen species (RNS) and reactive oxygen species (ROS) produced by free radicals as they pass through a polyphenolic compound, polyphenolic compounds manifest their antioxidant benefits [32].

As a result of free radical oxidation, ROS, RNS, and other compounds are produced. The ROS and RNS commonly observed in free radical-induced oxidative damage include nitric oxide (·NO), hydroxyl radicals (·OH), superoxide radicals (O2), and peroxynitrite anion (O = NOO). Fenton reactions catalyzed by metal ions (e.g., Fe2+ and Cu+) generate ROS and RNS. When ROS and RNS are subjected to high levels of oxidative stress, they are highly reactive and have short half-lives, which, in conjunction with an imbalance between their production and destruction, can cause a significant amount of protein modifications as well as other reaction products to accumulate at the site of inflammation as a result of ROS and RNS-derived proteins. Inflammatory conditions such as cancer, atherosclerosis, AD, and traumatic brain injury may arise as a result of an excessive accumulation of ROS and RNS-derived reaction products [33,34,35]. In addition to polyphenols, citrus flavanones, including hesperetin, hesperidin, and neohesperidin, can cross the blood–brain barrier (BBB). BBB permeability varies between polyphenols. In addition, animal studies has shown that blueberry consumption induces anthocyanin accumulation in the cortex and cerebellum of animal models (rats and pigs) [36]. Furthermore, the valerolactones undergo further metabolism resulting in the formation of phenolics and polyphenols, such as (hydroxyaryl)propanoic acid, (hydroxyaryl) cinmmic acid, (hydroxyaryl)valeric acid, hydroxybenzoic acid, and (hydroxyaryl)acetic acid derivatives. Secondary polyphenolic metabolites have a relatively higher degree of bioavailability and are more permeable to the BBB than flavonoids or dietary flavonoids and may inhibit neuroinflammation. As a result, polyphenols can be used therapeutically in a wide range of neurodegenerative diseases as well as TBI (Table 1 and 2) [37, 38].

Table 1 In vivo interventions
Table 2 In vitro interventions

Curcumin

The recent findings on curcumin demonstrate its remarkable versatility as a molecule that interacts with a variety of molecular targets [39,40,41,42,43,44,45,46,47,48]. There are curcumin compounds in the rhizomes of the turmeric plant (Curcuma longa), a plant belonging to the Zingiberaceae family. As an antioxidant, anti-infection, anti-inflammatory, and anti-tumor compound, curcumin has been approved by the United States Food and Drug Administration as a safe compound [49]. Various CNS disease models have been shown to be susceptible to curcumin's anti-inflammatory properties, including intracerebral hemorrhage, global brain ischemia, and neurodegeneration, all of which are associated with the inflammation of the CNS. Similarly, curcumin exerts neuroprotective effects in mammals when it crosses the blood–brain barrier. An experiment in mice modeling spinal and bulbar muscular atrophy revealed that 5-hydroxy-1,7-bis (3,4-dimethoxyphenyl)-1,4,6-heptatrien-3-one inhibited the aggregation of pathogenic androgen receptors. Curcumin is suspected to possess neuroprotective properties, but few studies have explored this possibility [49]. Curcumin has been shown to reduce cerebral edema, enhance membrane and energy homeostasis, and influence synaptic plasticity following TBI in a few studies. However, curcumin does not appear to have any immunomodulatory properties on inflammatory reactions.

Curcumin has been shown by numerous studies to be effective in reducing inflammation [50, 51]. A lot of research has shown that curcumin suppresses the activation of NF-κB by inhibiting the phosphorylation and degradation of IκB; as a result, curcumin reduces the inflammation caused by NF-κB. The effects of curcumin have been shown to be not only reduced post-TBI neuroinflammation, but also decreased levels of inflammatory mediators that are produced following a TBI [52]. The anti-inflammatory effects of curcumin were demonstrated in an in vitro study using 100 mg/kg of curcumin [

Data Availability

This is a review article and there is no associated primary data.

Abbreviations

TBI:

Traumatic brain injury

WHO:

World Health Organization

BACE-1:

β-Amyloid precursor protein–cleaving enzyme 1

NSAIDs:

Non-steroidal anti-inflammatory agents

NF-κB:

Nuclear factor-kappa β

AD:

Alzheimer’s disease

PD:

Parkinson’s disease

RNS:

Reactive nitrogen species

ROS:

Reactive oxygen species

BBB:

Blood-brain barrier

RSV:

Resveratrol

PQQ:

Pyrroloquinoline quinone

GSK-3β:

Glycogen synthase kinase 3 beta

NF-κB:

Nuclear factor kappa B

TLR4:

Toll-like receptor 4

IL-1β:

Interleukin-1β

NLRP3:

NLR family pyrin domain containing 3

TNF-α:

Tumor necrosis factor-α

iNOS:

Inducible nitric oxide synthase

MDA:

Malondialdehyde

8-oHdG:

8-Hydroxy-2'-deoxyguanosine

SOD:

Superoxide dismutase

GSH:

Glutathione

CAT:

Catalase

Nrf2/HO-1:

Nuclear factor erythroid 2 related factor 2/heme oxygenase 1

Nrf2:

Nuclear factor erythroid 2 related factor 2

HO-1:

Heme oxygenase-1

PI3K/AKT:

Phosphatidylinositol 3-kinase/protein kinase B

Bax:

BCL2-associated X

Bcl-2:

B-cell lymphoma 2

ERK 1/2:

Extracellular signal-regulated kinase

PPAR-γ:

Peroxisome proliferator-activated receptor-γ

PGC-1α:

PPAR-γ coactivator-1α

MMP:

Mitochondrial membrane potential

EGCG:

Epigallocatechin-3-gallate

IKK α/β:

I kappa B kinase alpha/beta

TGF-1β:

Transforming growth factor beta 1

PAMPs:

Pathogen-associated molecular patterns

LPS:

Lipopolysaccharide

Myd88:

Myeloid differentiation factor 88

TRAF6:

TNF receptor-associated factor 6

TRIF:

TIR domain-containing adaptor-inducing interferons

NQO1:

Nicotinamide adenine dinucleotide phosphate: quinone oxidoreductase-1

GSH-Px:

Glutathione peroxidase

ARE:

Antioxidant response element

CNS:

Central nervous system

SD:

Sprague Dawley

WS:

Wistar

SOD:

Superoxide dismutase

CAT:

Catalase

MDA:

Malondialdehyde

EPO:

Erythropoietin

ERK1/2:

Extracellular signal-regulated kinase 1/2

PI3K-AKT:

Phosphatidylinositol-3-kinase and protein kinase B

PGC1a:

Peroxisome proliferator-activated receptor-gamma coactivator

DMSO:

Dimethyl sulfoxide

Nrf2:

Nuclear factor-erythroid factor 2-related factor 2

AMPK:

AMP-activated protein kinase

IκBα:

Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha

NSCs:

Neural stem cells

EGCG:

Epigallocatechin gallate

TGF-β:

Transforming growth factor-β

NF-Κb:

Nuclear factor kappa-light-chain-enhancer of activated B cells

GFAP:

Glial fibrillary acidic protein

iNOS:

Inducible nitric oxide synthase

CCI:

Controlled cortical impact

AICAR:

5-Aminoimidazole-4-carboxamide ribonucleotide

CMC:

Sodium carboxymethylcellulose

ECs:

Endothelial cells

Nrf2/HO-1:

Nuclear factor erythroid 2-related factor 2/heme oxygenase 1

FFW:

Feeney’s falling weight

GSH-Px:

Glutathione peroxidase

SIRT1:

Caspase-1 and sirtuin 1

NLRP3:

NLR family pyrin domain containing 3

ROS:

Reactive oxygen species

RVS:

Resveratrol

GSK-3β:

Glycogen synthase kinase-3β

IL-6:

Interleukin 6

IL-12:

Interleukin 12

8-OHdG:

8-Hydroxy-2'-deoxyguanosine

GFAP:

Glial fibrillary acidic protein

AQP4:

Aquaporin-4

IGF-1:

Insulin-like growth factor 1

CGNs:

Cerebellar granular neurons

p38MAPK:

P38 mitogen-activated protein kinases

TLR4:

Toll-like receptor 4

NF-κB:

Nuclear factor-kappa B

MyD88:

Myeloid differentiation primary response 88

LPS:

Lipopolysaccharide

DMSO:

Dimethyl sulfoxide

Cur:

Curcumin

IL-1β:

Interleukin-1β

RANTES:

Regulated on expression normal T-cell expressed and secreted

TNF-α:

Tumor necrosis factor-alpha

MCP-1:

Monocyte chemoattractant protein-1

IL-6:

Interleukin 6

References

  1. Murray CJ, Lopez AD (1997) Alternative projections of mortality and disability by cause 1990–2020: Global Burden of Disease Study. Lancet 349(9064):1498–1504

    Article  CAS  PubMed  Google Scholar 

  2. Wittchen HU, Jacobi F, Rehm J, Gustavsson A, Svensson M, Jönsson B et al (2011) The size and burden of mental disorders and other disorders of the brain in Europe 2010. Eur Neuropsychopharmacol 21(9):655–679

    Article  CAS  PubMed  Google Scholar 

  3. Taylor CA, Bell JM, Breiding MJ, Xu L (2017) Traumatic brain injury-related emergency department visits, hospitalizations, and deaths - United States, 2007 and 2013. MMWR Surveill Summ 66(9):1–16

    Article  PubMed  PubMed Central  Google Scholar 

  4. Miller GF, Kegler SR, Stone DM (2020) Traumatic brain injury-related deaths from firearm suicide: United States, 2008–2017. Am J Public Health 110(6):897–899

    Article  PubMed  PubMed Central  Google Scholar 

  5. Khanal LN, Sharma KR, Pokharel YR, Kalauni SK (2022) Phytochemical analysis and in vitro antioxidant and antibacterial activity of different solvent extracts of Beilschmiedia roxburghiana Nees Stem Barks. Sci World J 2022:6717012

    Article  Google Scholar 

  6. Ahmadi A, Jamialahmadi T, Sahebkar A (2022) Polyphenols and atherosclerosis: a critical review of clinical effects on LDL oxidation. Pharmacol Res 184:106414

    Article  CAS  PubMed  Google Scholar 

  7. Hosseini SA, Zahedipour F, Sathyapalan T, Jamialahmadi T, Sahebkar A (2021) Pulmonary fibrosis: therapeutic and mechanistic insights into the role of phytochemicals. BioFactors 47(3):250–269

    Article  CAS  PubMed  Google Scholar 

  8. Ganesan K, Xu B (2017) A critical review on polyphenols and health benefits of black soybeans. Nutrients 9(5):455. https://doi.org/10.3390/nu9050455

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mottaghipisheh J, Doustimotlagh AH, Irajie C, Tanideh N, Barzegar A, Iraji A (2022) The promising therapeutic and preventive properties of anthocyanidins/anthocyanins on prostate cancer. Cells 11(7):1070. https://doi.org/10.3390/cells11071070

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Si W, Zhang Y, Li X, Du Y, Xu Q (2021) Understanding the functional activity of polyphenols using omics-based approaches. Nutrients 13(11):3953. https://doi.org/10.3390/nu13113953

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. de Andrade Teles RB, Diniz TC, Costa Pinto TC, de Oliveira Júnior RG, Gama ESM, de Lavor ÉM et al (2018) Flavonoids as therapeutic agents in Alzheimer’s and Parkinson’s diseases: a systematic review of preclinical evidences. Oxid Med Cell Longev 2018:7043213

    Article  PubMed  PubMed Central  Google Scholar 

  12. Jha NK, Jha SK, Kar R, Nand P, Swati K, Goswami VK (2019) Nuclear factor-kappa β as a therapeutic target for Alzheimer’s disease. J Neurochem 150(2):113–137

    Article  CAS  PubMed  Google Scholar 

  13. Scalbert A, Johnson IT, Saltmarsh M (2005) Polyphenols: antioxidants and beyond. Am J Clin Nutr 81(1 Suppl):215s-s217

    Article  CAS  PubMed  Google Scholar 

  14. Obrenovich ME, Nair NG, Beyaz A, Aliev G, Reddy VP (2010) The role of polyphenolic antioxidants in health, disease, and aging. Rejuvenation Res 13(6):631–643

    Article  CAS  PubMed  Google Scholar 

  15. Kempuraj D, Thangavel R, Kempuraj DD, Ahmed ME, Selvakumar GP, Raikwar SP et al (2021) Neuroprotective effects of flavone luteolin in neuroinflammation and neurotrauma. BioFactors 47(2):190–197

    Article  CAS  PubMed  Google Scholar 

  16. Cornelius C, Crupi R, Calabrese V, Graziano A, Milone P, Pennisi G et al (2013) Traumatic brain injury: oxidative stress and neuroprotection. Antioxid Redox Signal 19(8):836–853

    Article  CAS  PubMed  Google Scholar 

  17. Angeloni C, Prata C, Vieceli Dalla Sega F, Piperno R, Hrelia S (2015) Traumatic brain injury and NADPH oxidase: a deep relationship. Oxid Med Cell Longev 2015:370312. https://doi.org/10.1155/2015/370312

  18. Maiuri MC, Criollo A, Kroemer G (2010) Crosstalk between apoptosis and autophagy within the Beclin 1 interactome. EMBO J 29(3):515–516

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gao Y, Zhuang Z, Gao S, Li X, Zhang Z, Ye Z et al (2017) Tetrahydrocurcumin reduces oxidative stress-induced apoptosis via the mitochondrial apoptotic pathway by modulating autophagy in rats after traumatic brain injury. Am J Transl Res. 9(3):887

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Lucas K, Maes M (2013) Role of the toll like receptor (TLR) radical cycle in chronic inflammation: possible treatments targeting the TLR4 pathway. Mol Neurobiol 48(1):190–204

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhang L, Ding K, Wang H, Wu Y, Xu J (2016) Traumatic brain injury-induced neuronal apoptosis is reduced through modulation of PI3K and autophagy pathways in mouse by FTY720. Cell Mol Neurobiol 36(1):131–142

    Article  PubMed  Google Scholar 

  22. Akira S, Takeda K (2004) Toll-like receptor signalling. Nat Rev Immunol 4(7):499–511

    Article  CAS  PubMed  Google Scholar 

  23. Ahmad A, Crupi R, Campolo M, Genovese T, Esposito E, Cuzzocrea S (2013) Absence of TLR4 reduces neurovascular unit and secondary inflammatory process after traumatic brain injury in mice. PLoS ONE 8(3):e57208

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. Li G-Z, Zhang Y, Zhao J-B, Wu G-J, Su X-F, Hang C-H (2011) Expression of myeloid differentiation primary response protein 88 (Myd88) in the cerebral cortex after experimental traumatic brain injury in rats. Brain Res 1396:96–104

    Article  CAS  PubMed  Google Scholar 

  25. Sun SC (2017) The non-canonical NF-kappaB pathway in immunity and inflammation. Nat Rev Immunol 17(9):545–558

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen C-C, Hung T-H, Wang Y-H, Lin C-W, Wang P-Y, Lee C-Y et al (2012) Wogonin improves histological and functional outcomes, and reduces activation of TLR4/NF-κB signaling after experimental traumatic brain injury. PLoS ONE 7(1):e30294

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bayır H, Kochanek PM, Kagan VE (2006) Oxidative stress in immature brain after traumatic brain injury. Dev Neurosci 28(4–5):420–431

    Article  PubMed  Google Scholar 

  28. Zhang L, Wang H (2018) Targeting the NF-E2-related factor 2 pathway: a novel strategy for traumatic brain injury. Mol Neurobiol 55(2):1773–1785

    Article  CAS  PubMed  Google Scholar 

  29. De Vries HE, Witte M, Hondius D, Rozemuller AJ, Drukarch B, Hoozemans J et al (2008) Nrf2-induced antioxidant protection: a promising target to counteract ROS-mediated damage in neurodegenerative disease? Free Radical Biol Med 45(10):1375–1383

    Article  Google Scholar 

  30. Itoh K, Tong KI, Yamamoto M (2004) Molecular mechanism activating Nrf2–Keap1 pathway in regulation of adaptive response to electrophiles. Free Radical Biol Med 36(10):1208–1213

    Article  CAS  Google Scholar 

  31. Potì F, Santi D, Spaggiari G, Zimetti F, Zanotti I (2019) Polyphenol health effects on cardiovascular and neurodegenerative disorders: a review and meta-analysis. Int J Mol Sci 20(2):351. https://doi.org/10.3390/ijms20020351

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Losada-Barreiro S, Bravo-Díaz C (2017) Free radicals and polyphenols: the redox chemistry of neurodegenerative diseases. Eur J Med Chem 133:379–402

    Article  CAS  PubMed  Google Scholar 

  33. Li C, Li Y, Zhao Z, Lv Y, Gu B, Zhao L (2019) Aerobic exercise regulates synaptic transmission and reactive oxygen species production in the paraventricular nucleus of spontaneously hypertensive rats. Brain Res 1712:82–92

    Article  CAS  PubMed  Google Scholar 

  34. Perron NR, Brumaghim JL (2009) A review of the antioxidant mechanisms of polyphenol compounds related to iron binding. Cell Biochem Biophys 53(2):75–100

    Article  CAS  PubMed  Google Scholar 

  35. Pourzand C, Albieri-Borges A, Raczek NN (2022) Shedding a new light on skin aging, iron- and redox-homeostasis and emerging natural antioxidants. Antioxidants (Basel) 11(3):471. https://doi.org/10.3390/antiox11030471

    Article  CAS  PubMed  Google Scholar 

  36. Andres-Lacueva C, Shukitt-Hale B, Galli RL, Jauregui O, Lamuela-Raventos RM, Joseph JA (2005) Anthocyanins in aged blueberry-fed rats are found centrally and may enhance memory. Nutr Neurosci 8(2):111–120

    Article  CAS  PubMed  Google Scholar 

  37. Croft KD (2016) Dietary polyphenols: antioxidants or not? Arch Biochem Biophys 595:120–124

    Article  CAS  PubMed  Google Scholar 

  38. Carregosa D, Carecho R, Figueira ICNS (2020) Low-molecular weight metabolites from polyphenols as effectors for attenuating neuroinflammation. J Agric Food Chem 68(7):1790–807

    Article  CAS  PubMed  Google Scholar 

  39. Cicero AFG, Sahebkar A, Fogacci F, Bove M, Giovannini M, Borghi C (2020) Effects of phytosomal curcumin on anthropometric parameters, insulin resistance, cortisolemia and non-alcoholic fatty liver disease indices: a double-blind, placebo-controlled clinical trial. Eur J Nutr 59(2):477–483

    Article  CAS  PubMed  Google Scholar 

  40. Iranshahi M, Sahebkar A, Takasaki M, Konoshima T, Tokuda H (2009) Cancer chemopreventive activity of the prenylated coumarin, umbelliprenin, in vivo. Eur J Cancer Prev 18(5):412–415

    Article  CAS  PubMed  Google Scholar 

  41. Keihanian F, Saeidinia A, Bagheri RK, Johnston TP, Sahebkar A (2018) Curcumin, hemostasis, thrombosis, and coagulation. J Cell Physiol 233(6):4497–4511

    Article  CAS  PubMed  Google Scholar 

  42. Marjaneh RM, Rahmani F, Hassanian SM, Rezaei N, Hashemzehi M, Bahrami A et al (2018) Phytosomal curcumin inhibits tumor growth in colitis-associated colorectal cancer. J Cell Physiol 233(10):6785–6798

    Article  CAS  PubMed  Google Scholar 

  43. Mohajeri M, Sahebkar A (2018) Protective effects of curcumin against doxorubicin-induced toxicity and resistance: a review. Crit Rev Oncol Hematol 122:30–51

    Article  PubMed  Google Scholar 

  44. Mokhtari-Zaer A, Marefati N, Atkin SL, Butler AE, Sahebkar A (2018) The protective role of curcumin in myocardial ischemia–reperfusion injury. J Cell Physiol 234(1):214–222

    Article  PubMed  Google Scholar 

  45. Momtazi AA, Sahebkar A (2016) Difluorinated curcumin: a promising curcumin analogue with improved anti-tumor activity and pharmacokinetic profile. Curr Pharm Des 22(28):4386–4397

    Article  CAS  PubMed  Google Scholar 

  46. Panahi Y, Fazlolahzadeh O, Atkin SL, Majeed M, Butler AE, Johnston TP et al (2019) Evidence of curcumin and curcumin analogue effects in skin diseases: a narrative review. J Cell Physiol 234(2):1165–1178

    Article  CAS  PubMed  Google Scholar 

  47. Panahi Y, Sahebkar A, Amiri M, Davoudi SM, Beiraghdar F, Hoseininejad SL et al (2012) Improvement of sulphur mustard-induced chronic pruritus, quality of life and antioxidant status by curcumin: results of a randomised, double-blind, placebo-controlled trial. Br J Nutr 108(7):1272–1279

    Article  CAS  PubMed  Google Scholar 

  48. Sahebkar A (2014) Curcuminoids for the management of hypertriglyceridaemia. Nat Rev Cardiol 11(2):123. https://doi.org/10.1038/nrcardio.2013.140-c1

    Article  CAS  PubMed  Google Scholar 

  49. Khayatan D, Razavi SM, Arab ZN, Niknejad AH, Nouri K, Momtaz S et al (2022) Protective effects of curcumin against traumatic brain injury. Biomed Pharmacother 154:113621

    Article  CAS  PubMed  Google Scholar 

  50. Mohammadi A, Blesso CN, Barreto GE, Banach M, Majeed M, Sahebkar A (2019) Macrophage plasticity, polarization and function in response to curcumin, a diet-derived polyphenol, as an immunomodulatory agent. J Nutr Biochem 66:1–16

    Article  CAS  PubMed  Google Scholar 

  51. Ferguson JJA, Abbott KA, Garg ML (2021) Anti-inflammatory effects of oral supplementation with curcumin: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 79(9):1043–1066

    Article  PubMed  Google Scholar 

  52. Sun G, Miao Z, Ye Y, Zhao P, Fan L, Bao Z et al (2020) Curcumin alleviates neuroinflammation, enhances hippocampal neurogenesis, and improves spatial memory after traumatic brain injury. Brain Res Bull 162:84–93

    Article  CAS  PubMed  Google Scholar 

  53. Zhu HT, Bian C, Yuan JC, Chu WH, **ang X, Chen F et al (2014) Curcumin attenuates acute inflammatory injury by inhibiting the TLR4/MyD88/NF-κB signaling pathway in experimental traumatic brain injury. J Neuroinflammation 11:59

    Article  PubMed  PubMed Central  Google Scholar 

  54. Wu A, Ying Z, Schubert D, Gomez-Pinilla F (2011) Brain and spinal cord interaction: a dietary curcumin derivative counteracts locomotor and cognitive deficits after brain trauma. Neurorehabil Neural Repair 25(4):332–342

    Article  PubMed  PubMed Central  Google Scholar 

  55. Dong X (2018) Current Strategies for Brain Drug Delivery. Theranostics 8(6):1481–1493

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Liang DY, Sahbaie P, Sun Y, Irvine KA, Shi X, Meidahl A et al (2017) TBI-induced nociceptive sensitization is regulated by histone acetylation. IBRO Rep 2:14–23

    Article  PubMed  Google Scholar 

  57. Dai W, Wang H, Fang J, Zhu Y, Zhou J, Wang X et al (2018) Curcumin provides neuroprotection in model of traumatic brain injury via the Nrf2-ARE signaling pathway. Brain Res Bull 140:65–71

    Article  CAS  PubMed  Google Scholar 

  58. Wu A, Ying Z, Gomez-Pinilla F (2006) Dietary curcumin counteracts the outcome of traumatic brain injury on oxidative stress, synaptic plasticity, and cognition. Exp Neurol 197(2):309–317

    Article  CAS  PubMed  Google Scholar 

  59. Attari F, Ghadiri T, Hashemi M (2020) Combination of curcumin with autologous transplantation of adult neural stem/progenitor cells leads to more efficient repair of damaged cerebral tissue of rat. Exp Physiol 105(9):1610–1622

    Article  CAS  PubMed  Google Scholar 

  60. Wei G, Chen B, Lin Q, Li Y, Luo L, He H et al (2017) Tetrahydrocurcumin provides neuroprotection in experimental traumatic brain injury and the Nrf2 signaling pathway as a potential mechanism. NeuroImmunoModulation 24(6):348–355

    Article  ADS  CAS  PubMed  Google Scholar 

  61. Wu A, Ying Z, Gomez-Pinilla F (2014) Dietary strategy to repair plasma membrane after brain trauma: implications for plasticity and cognition. Neurorehabil Neural Repair 28(1):75–84

    Article  CAS  PubMed  Google Scholar 

  62. Sharma S, Ying Z, Gomez-Pinilla F (2010) A pyrazole curcumin derivative restores membrane homeostasis disrupted after brain trauma. Exp Neurol 226(1):191–199

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gao Y, Li J, Wu L, Zhou C, Wang Q, Li X et al (2016) Tetrahydrocurcumin provides neuroprotection in rats after traumatic brain injury: autophagy and the PI3K/AKT pathways as a potential mechanism. J Surg Res 206(1):67–76

    Article  CAS  PubMed  Google Scholar 

  64. Razavi SM, Khayatan D, Arab ZN, Momtaz S, Zare K, Jafari RM et al (2021) Licofelone, a potent COX/5-LOX inhibitor and a novel option for treatment of neurological disorders. Prostaglandins Other Lipid Mediat 157:106587

    Article  CAS  PubMed  Google Scholar 

  65. Mirhadi E, Roufogalis BD, Banach M, Barati M, Sahebkar A (2021) Resveratrol: mechanistic and therapeutic perspectives in pulmonary arterial hypertension. Pharmacol Res 163:105287

    Article  CAS  PubMed  Google Scholar 

  66. Omraninava M, Razi B, Aslani S, Imani D, Jamialahmadi T, Sahebkar A (2021) Effect of resveratrol on inflammatory cytokines: a meta-analysis of randomized controlled trials. Eur J Pharmacol 908:174380

    Article  CAS  PubMed  Google Scholar 

  67. Parsamanesh N, Asghari A, Sardari S, Tasbandi A, Jamialahmadi T, Xu S et al (2021) Resveratrol and endothelial function: a literature review. Pharmacol Res 170:105725

    Article  CAS  PubMed  Google Scholar 

  68. Sahebkar A (2013) Effects of resveratrol supplementation on plasma lipids: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 71(12):822–835

    Article  PubMed  Google Scholar 

  69. Sahebkar A, Serban C, Ursoniu S, Wong ND, Muntner P, Graham IM et al (2015) Lack of efficacy of resveratrol on C-reactive protein and selected cardiovascular risk factors - results from a systematic review and meta-analysis of randomized controlled trials. Int J Cardiol 189(1):47–55

    Article  PubMed  Google Scholar 

  70. Rafiee S, Mohammadi H, Ghavami A, Sadeghi E, Safari Z, Askari G (2021) Efficacy of resveratrol supplementation in patients with nonalcoholic fatty liver disease: a systematic review and meta-analysis of clinical trials. Complement Ther Clin Pract 42:101281

    Article  PubMed  Google Scholar 

  71. Visioli F (2014) The resveratrol fiasco. Pharmacol Res 90:87

    Article  PubMed  Google Scholar 

  72. Shanan N, GhasemiGharagoz A, Abdel-Kader R, Breitinger HG (2019) The effect of pyrroloquinoline quinone and resveratrol on the survival and regeneration of cerebellar granular neurons. Neurosci Lett 694:192–197

    Article  CAS  PubMed  Google Scholar 

  73. Taherian M, Norenberg MD, Panickar KS, Shamaladevi N, Ahmad A, Rahman P et al (2020) Additive effect of resveratrol on astrocyte swelling post-exposure to ammonia, ischemia and trauma in vitro. Neurochem Res 45(5):1156–1167

    Article  CAS  PubMed  Google Scholar 

  74. Lin CJ, Chen TH, Yang LY, Shih CM (2014) Resveratrol protects astrocytes against traumatic brain injury through inhibiting apoptotic and autophagic cell death. Cell Death Dis 5(3):e1147

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Feng Y, Cui Y, Gao JL, Li MH, Li R, Jiang XH et al (2016) Resveratrol attenuates neuronal autophagy and inflammatory injury by inhibiting the TLR4/NF-κB signaling pathway in experimental traumatic brain injury. Int J Mol Med 37(4):921–930

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Singleton RH, Yan HQ, Fellows-Mayle W, Dixon CE (2010) Resveratrol attenuates behavioral impairments and reduces cortical and hippocampal loss in a rat controlled cortical impact model of traumatic brain injury. J Neurotrauma 27(6):1091–1099

    Article  PubMed  PubMed Central  Google Scholar 

  77. Feng Y, Cui Y, Gao JL, Li R, Jiang XH, Tian YX et al (2016) Neuroprotective effects of resveratrol against traumatic brain injury in rats: involvement of synaptic proteins and neuronal autophagy. Mol Med Rep 13(6):5248–5254

    Article  CAS  PubMed  Google Scholar 

  78. Salberg S, Yamakawa G, Christensen J, Kolb B, Mychasiuk R (2017) Assessment of a nutritional supplement containing resveratrol, prebiotic fiber, and omega-3 fatty acids for the prevention and treatment of mild traumatic brain injury in rats. Neuroscience 365:146–157

    Article  CAS  PubMed  Google Scholar 

  79. Zou P, Liu X, Li G, Wang Y (2018) Resveratrol pretreatment attenuates traumatic brain injury in rats by suppressing NLRP3 inflammasome activation via SIRT1. Mol Med Rep 17(2):3212–3217

    CAS  PubMed  Google Scholar 

  80. Shi Z, Qiu W, **ao G, Cheng J, Zhang N (2018) Resveratrol attenuates cognitive deficits of traumatic brain injury by activating p38 signaling in the brain. Med Sci Monit 24:1097–1103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Atalay T, Gulsen I, Colcimen N, Alp HH, Sosuncu E, Alaca I et al (2017) Resveratrol treatment prevents hippocampal neurodegeneration in a rodent model of traumatic brain injury. Turk Neurosurg 27(6):924–930

    PubMed  Google Scholar 

  82. Gatson JW, Liu MM, Abdelfattah K, Wigginton JG, Smith S, Wolf S et al (2013) Resveratrol decreases inflammation in the brain of mice with mild traumatic brain injury. J Trauma Acute Care Surg 74(2):470–4

    Article  CAS  PubMed  Google Scholar 

  83. Sönmez U, Sönmez A, Erbil G, Tekmen I, Baykara B (2007) Neuroprotective effects of resveratrol against traumatic brain injury in immature rats. Neurosci Lett 420(2):133–137

    Article  PubMed  Google Scholar 

  84. Iranshahi M, Sahebkar A, Hosseini ST, Takasaki M, Konoshima T, Tokuda H (2010) Cancer chemopreventive activity of diversin from Ferula diversivittata in vitro and in vivo. Phytomedicine 17(3–4):269–273

    Article  CAS  PubMed  Google Scholar 

  85. Zou H, Ye H, Kamaraj R, Zhang T, Zhang J, Pavek P (2021) A review on pharmacological activities and synergistic effect of quercetin with small molecule agents. Phytomedicine 92:153736

    Article  CAS  PubMed  Google Scholar 

  86. Wang G, Wang Y, Yao L, Gu W, Zhao S, Shen Z et al (2022) Pharmacological activity of quercetin: an updated review. Evid Based Complement Alternat Med 2022:3997190

    Article  PubMed  PubMed Central  Google Scholar 

  87. Guan LP, Liu BY (2016) Antidepressant-like effects and mechanisms of flavonoids and related analogues. Eur J Med Chem 121:47–57

    Article  CAS  PubMed  Google Scholar 

  88. Song J, Du G, Wu H, Gao X, Yang Z, Liu B et al (2021) Protective effects of quercetin on traumatic brain injury induced inflammation and oxidative stress in cortex through activating Nrf2/HO-1 pathway. Restor Neurol Neurosci 39(1):73–84

    CAS  PubMed  Google Scholar 

  89. Kalemci O, Aydin HE, Kizmazoglu C, Kaya I, Yılmaz H, Arda NM (2017) Effects of quercetin and mannitol on erythropoietin levels in rats following acute severe traumatic brain injury. J Korean Neurosurg Soc 60(3):355–361

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Yang T, Kong B, Gu JW, Kuang YQ, Cheng L, Yang WT et al (2014) Anti-apoptotic and anti-oxidative roles of quercetin after traumatic brain injury. Cell Mol Neurobiol 34(6):797–804

    Article  CAS  PubMed  Google Scholar 

  91. Du G, Zhao Z, Chen Y, Li Z, Tian Y, Liu Z et al (2016) Quercetin attenuates neuronal autophagy and apoptosis in rat traumatic brain injury model via activation of PI3K/Akt signaling pathway. Neurol Res 38(11):1012–1019

    Article  CAS  PubMed  Google Scholar 

  92. Du G, Zhao Z, Chen Y, Li Z, Tian Y, Liu Z et al (2018) Quercetin protects rat cortical neurons against traumatic brain injury. Mol Med Rep 17(6):7859–7865

    CAS  PubMed  Google Scholar 

  93. Li X, Wang H, Wen G, Li L, Gao Y, Zhuang Z et al (2018) Neuroprotection by quercetin via mitochondrial function adaptation in traumatic brain injury: PGC-1α pathway as a potential mechanism. J Cell Mol Med 22(2):883–891

    Article  CAS  PubMed  Google Scholar 

  94. Li X, Wang H, Gao Y, Li L, Tang C, Wen G et al (2016) Protective effects of quercetin on mitochondrial biogenesis in experimental traumatic brain injury via the Nrf2 signaling pathway. PLoS ONE 11(10):e0164237

    Article  PubMed  PubMed Central  Google Scholar 

  95. Musial C, Kuban-Jankowska A, Gorska-Ponikowska M (2020) Beneficial properties of green tea catechins. Int J Mol Sci 21(5):1744. https://doi.org/10.3390/ijms21051744

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Jiang Z, Zhang J, Cai Y, Huang J, You L (2017) Catechin attenuates traumatic brain injury-induced blood-brain barrier damage and improves longer-term neurological outcomes in rats. Exp Physiol 102(10):1269–1277

    Article  CAS  PubMed  Google Scholar 

  97. Wu Y, Cui J (2020) (-)-Epigallocatechin-3-gallate provides neuroprotection via AMPK activation against traumatic brain injury in a mouse model. Naunyn Schmiedebergs Arch Pharmacol 393(11):2209–2220

    Article  CAS  PubMed  Google Scholar 

  98. Itoh T, Imano M, Nishida S, Tsubaki M, Hashimoto S, Ito A et al (2011) (-)-Epigallocatechin-3-gallate protects against neuronal cell death and improves cerebral function after traumatic brain injury in rats. Neuromolecular Med 13(4):300–309

    Article  CAS  PubMed  Google Scholar 

  99. Itoh T, Imano M, Nishida S, Tsubaki M, Mizuguchi N, Hashimoto S et al (2012) (-)-Epigallocatechin-3-gallate increases the number of neural stem cells around the damaged area after rat traumatic brain injury. J Neural Transm (Vienna) 119(8):877–890

    Article  CAS  PubMed  Google Scholar 

  100. Giuliano C, Siani F, Mus L, Ghezzi C, Cerri S, Pacchetti B et al (2020) Neuroprotective effects of lignan 7-hydroxymatairesinol (HMR/lignan) in a rodent model of Parkinson’s disease. Nutrition 69:110494

    Article  CAS  PubMed  Google Scholar 

  101. Liu YL, Xu ZM, Yang GY, Yang DX, Ding J, Chen H et al (2017) Sesamin alleviates blood-brain barrier disruption in mice with experimental traumatic brain injury. Acta Pharmacol Sin 38(11):1445–1455

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Dong L, Zhou S, Yang X, Chen Q, He Y, Huang W (2013) Magnolol protects against oxidative stress-mediated neural cell damage by modulating mitochondrial dysfunction and PI3K/Akt signaling. J Mol Neurosci 50(3):469–481

    Article  CAS  PubMed  Google Scholar 

  103. Wang CC, Lin KC, Lin BS, Chio CC, Kuo JR (2013) Resuscitation from experimental traumatic brain injury by magnolol therapy. J Surg Res 184(2):1045–1052

    Article  CAS  PubMed  Google Scholar 

  104. Talarek S, Listos J, Barreca D, Tellone E, Sureda A, Nabavi SF et al (2017) Neuroprotective effects of honokiol: from chemistry to medicine. BioFactors 43(6):760–769

    Article  CAS  PubMed  Google Scholar 

  105. Li W, Wang S, Zhang H, Li B, Xu L, Li Y et al (2021) Honokiol restores microglial phagocytosis by reversing metabolic reprogramming. J Alzheimers Dis 82(4):1475–1485

    Article  CAS  PubMed  Google Scholar 

  106. Çetin A, Deveci E (2019) Expression of vascular endothelial growth factor and glial fibrillary acidic protein in a rat model of traumatic brain injury treated with honokiol: a biochemical and immunohistochemical study. Folia Morphol (Warsz) 78(4):684–694

    Article  PubMed  Google Scholar 

  107. Wang H, Liao Z, Sun X, Shi Q, Huo G, **e Y et al (2014) Intravenous administration of Honokiol provides neuroprotection and improves functional recovery after traumatic brain injury through cell cycle inhibition. Neuropharmacology 86:9–21

    Article  CAS  PubMed  Google Scholar 

  108. Seibel R, Schneider RH, Gottlieb MGV (2021) Effects of spices (saffron, rosemary, cinnamon, turmeric and ginger) in Alzheimer’s disease. Curr Alzheimer Res 18(4):347–357

    Article  CAS  PubMed  Google Scholar 

  109. Sadeghi S, Davoodvandi A, Pourhanifeh MH, Sharifi N, ArefNezhad R, Sahebnasagh R et al (2019) Anti-cancer effects of cinnamon: insights into its apoptosis effects. Eur J Med Chem 178:131–140

    Article  CAS  PubMed  Google Scholar 

  110. Yulug B, Kilic E, Altunay S, Ersavas C, Orhan C, Dalay A et al (2018) Cinnamon polyphenol extract exerts neuroprotective activity in traumatic brain injury in male mice. CNS Neurol Disord Drug Targets 17(6):439–447

    Article  CAS  PubMed  Google Scholar 

  111. Rangasamy SB, Raha S, Dasarathy S, Pahan K (2021) Sodium benzoate, a metabolite of cinnamon and a food additive, improves cognitive functions in mice after controlled cortical impact injury. Int J Mol Sci 23(1):192. https://doi.org/10.3390/ijms23010192

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Li X, Wang H, Gao Y, Li L, Tang C, Wen G et al (2016) Quercetin induces mitochondrial biogenesis in experimental traumatic brain injury via the PGC-1α signaling pathway. Am J Transl Res 8(8):3558–3566

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Yavtushenko IV, Nazarenko SM, Katrushov OV, Kostenko VO (2020) Quercetin limits the progression of oxidative and nitrosative stress in the rats’ tissues after experimental traumatic brain injury. Wiad Lek 73(10):2127–2132

    Article  PubMed  Google Scholar 

  114. Itoh T, Tabuchi M, Mizuguchi N, Imano M, Tsubaki M, Nishida S et al (2013) Neuroprotective effect of (-)-epigallocatechin-3-gallate in rats when administered pre- or post-traumatic brain injury. J Neural Transm (Vienna) 120(5):767–783

    Article  CAS  PubMed  Google Scholar 

  115. Qubty D, Rubovitch V, Benromano T, Ovadia M, Pick CG (2021) Orally administered cinnamon extract attenuates cognitive and neuronal deficits following traumatic brain injury. J Mol Neurosci 71(1):178–186

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Russian Science Foundation (Grant # 22-15-00064).

Funding

Open Access funding provided by the IReL Consortium.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by DK, SMR, MK, ZNA, AS, SM, VNS, and TJ. Supervision: AHA, GEB, and AS. The first draft of the manuscript was written by and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Amir Hossein Abdolghaffari, George E. Barreto or Amirhossein Sahebkar.

Ethics declarations

Ethics Approval

N/A.

Consent to Participate

N/A.

Competing Interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Khayatan, D., Razavi, S.M., Arab, Z.N. et al. Protective Effects of Plant-Derived Compounds Against Traumatic Brain Injury. Mol Neurobiol (2024). https://doi.org/10.1007/s12035-024-04030-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s12035-024-04030-w

Keywords