The Interplay Between Melatonin and Nitric Oxide: Mechanisms and Implications in Stroke Pathophysiology
Abstract
:1. Introduction
1.1. Overview of Melatonin
- Antioxidant mechanisms: Melatonin is a potent antioxidant. It acts as a direct scavenger of a wide range of reactive oxygen species (ROS), such as the hydroxyl radical (•OH) and the superoxide anion O2•−, and reactive nitrogen species (RNS), including peroxynitrite (ONOO−) [7]. Importantly, its main metabolites, such as N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK) and N1-acetyl-5-methoxykynuramine (AMK), also possess notable antioxidant activity, which amplifies and prolongs its protective effect [6]. In addition to direct scavenging, melatonin exerts indirect antioxidant effects by stimulating the activity of key endogenous antioxidant enzymes, such as superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR), and catalase [7]. It also enhances the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, a master transcription factor that regulates the expression of multiple antioxidant defense and detoxification genes [8].
- Anti-inflammatory actions: Melatonin exerts potent anti-inflammatory effects. It reduces the production and release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-1 beta, and interleukin-6 (IL-6) [6]. It modulates the activation of microglia, the brain’s resident immune cells, potentially promoting a shift from a pro-inflammatory (M1) phenotype to an anti-inflammatory and pro-repair (M2) phenotype [8]. It also inhibits key pro-inflammatory signaling pathways, such as that of nuclear factor kappa B (NF-κB), and reduces the expression of cell adhesion molecules involved in leukocyte infiltration into damaged brain tissue [9].
- Anti-apoptotic effects: Melatonin interferes with the apoptotic cascade at multiple levels. One of its crucial sites of action is the mitochondria, where it prevents the opening of the mitochondrial permeability transition pore (mPTP), an event that leads to the dissipation of the mitochondrial membrane potential and the release of pro-apoptotic factors [7]. Consequently, melatonin reduces the release of cytochrome c from the mitochondria to the cytosol and decreases the activation of caspases, the executioner enzymes of apoptosis [7]. Furthermore, it modulates the expression of Bcl-2 family proteins, increasing the levels of anti-apoptotic proteins (e.g., Bcl-2) and decreasing those of pro-apoptotic proteins (e.g., Bax) [7]. It also activates pro-survival signaling pathways, such as the PI3K/Akt pathway [10].
1.2. Overview of Nitric Oxide (NO)
1.3. Significance of Melatonin–NO Interactions in the Central Nervous System
1.4. Rationale and Scope
2. Melatonin and Nitric Oxide Signaling in the Animal CNS
2.1. Melatonin in the CNS
2.1.1. Synthesis and Distribution
2.1.2. Receptors
2.1.3. Key CNS Functions (Beyond Circadian)
2.2. Nitric Oxide in the CNS
2.2.1. Synthesis (NOS Isoforms)
2.2.2. Signaling
2.2.3. Physiological Roles
2.3. Molecular Crosstalk Between Melatonin and NO/NOS in the CNS
2.3.1. Regulation of NOS Expression and Activity
2.3.2. Impact on NO Bioavailability and Downstream Signaling
2.3.3. Direct Scavenging of NO and Reactive Nitrogen Species
3. The Melatonin–NO Axis in Stroke Pathophysiology and Neuroprotection
3.1. Overview of Stroke Pathophysiology
- Excitotoxicity: Excessive release of excitatory neurotransmitters, primarily glutamate, overstimulates receptors (especially NMDA receptors), leading to massive Ca2+ influx [60].
- Ionic imbalance and calcium overload: Energy failure disrupts ion pumps, leading to loss of ionic gradients and toxic intracellular Ca2+ accumulation [61].
- Oxidative and nitrosative stress: I/R dramatically increases the production of ROS and RNS from various sources (mitochondria, NADPH oxidases, uncoupled NOS, xanthine oxidase), overwhelming antioxidant defenses and causing widespread molecular damage [8].
- Neuroinflammation: Rapid activation of resident microglia and astrocytes, followed by infiltration of peripheral immune cells (neutrophils, macrophages), releases pro-inflammatory cytokines (TNF-α, IL-1β, IL-6), chemokines, and matrix metalloproteinases (MMPs), further propagating tissue injury [8].
- Mitochondrial dysfunction: Mitochondria are central players, suffering damage from Ca2+ overload and oxidative/nitrosative stress, leading to impaired ATP production, increased ROS generation, opening of mPTP, and release of pro-apoptotic factors [61].
- Cell death: Neurons and glial cells die via various mechanisms, including necrosis (primarily in the core), apoptosis (prominent in the penumbra), and potentially other regulated cell death pathways like pyroptosis and necroptosis, particularly exacerbated in conditions like obesity [8]. New research has identified ferroptosis as an additional mechanism of cell death in ischemic stroke, and melatonin may play a role in its modulation [62].
- Blood–brain barrier (BBB) breakdown: Increased permeability of the BBB allows influx of fluid and peripheral immune cells, contributing to cerebral edema and inflammation [8].
3.2. The Complex Role of NO and NOS Isoforms in Stroke
3.2.1. Detrimental Roles:
- iNOS: Upregulation of iNOS in microglia, macrophages, and astrocytes, driven by the inflammatory response to ischemia, leads to sustained high-level NO production [48]. This excessive NO contributes significantly to cytotoxicity, neuroinflammation, and oxidative/nitrosative stress [32]. High NO concentrations can directly inhibit mitochondrial respiration and react with superoxide to form the highly damaging ONOO− [36]. Studies often show increased iNOS expression correlating with larger infarct volumes and worse outcomes [49].
- nNOS: While nNOS has physiological roles, its overactivation during the excitotoxic phase of stroke, triggered by Ca2+ influx via NMDA receptors, can also contribute to neuronal damage [32]. nNOS-derived NO can contribute to ONOO− formation and downstream neurotoxicity. Increased nNOS expression or activity is often observed in the peri-infarct regions following ischemia [35].
- Peroxynitrite formation: The reaction between NO (from any source, but particularly high levels from iNOS/nNOS) and superoxide (abundantly produced during I/R) generates ONOO−, a potent oxidant and nitrating agent that damages lipids, proteins, and DNA, contributing significantly to cell death and BBB breakdown [63].
3.2.2. Protective Roles:
- eNOS: NO produced by eNOS in the cerebrovascular endothelium plays a generally protective role by promoting vasodilation, which helps maintain collateral blood flow to the ischemic penumbra, potentially limiting infarct expansion [32]. eNOS-derived NO also has anti-platelet and anti-leukocyte adhesion properties, further supporting microvascular perfusion [31]. However, during I/R, eNOS function can become impaired or “uncoupled” due to cofactor (BH4) oxidation, leading to reduced NO production and increased superoxide generation, thus losing its protective capacity [64]. Some studies report decreased eNOS levels or activity after ischemia [49].
3.3. Melatonin as a Neuroprotective Agent in Animal Stroke Models
- Reduced infarct volume: Melatonin treatment consistently leads to a significant reduction in the size of the brain infarct in both cortical and subcortical regions [8].
- Decreased cerebral edema: Melatonin helps alleviate brain swelling, a dangerous complication of stroke [8].
- Improved neurological function: Animals treated with melatonin exhibit better performance on various sensorimotor and neurological deficit scales, indicating functional recovery [8].
- Attenuated BBB breakdown: Melatonin often reduces the increased permeability of the BBB following I/R, limiting edema and immune cell infiltration [8]. However, one study using a specific pre-treatment regimen did not observe an effect on Evans blue extravasation at 3 h post-reperfusion, suggesting timing and specific endpoints might influence this outcome [65].
- Increased cell survival: melatonin promotes the survival of neurons and potentially other brain cells within the ischemic territory [66].
3.4. Cellular and Molecular Mechanisms of Melatonin–NO Interaction in Stroke Neuroprotection
- Modulation of NOS isoforms: As discussed previously, melatonin exerts a beneficial regulatory effect on NOS isoforms in the context of ischemia. It consistently suppresses the detrimental upregulation of iNOS induced by I/R injury [48]. This action directly reduces the production of large amounts of cytotoxic NO associated with neuroinflammation. Simultaneously, melatonin often prevents the ischemia-induced downregulation or dysfunction of the protective eNOS isoform, helping to maintain beneficial endothelial function and potentially improving microvascular perfusion in the penumbra [49]. The effect on nNOS appears less critical or more variable in stroke models treated with melatonin [48]. By differentially regulating these isoforms, melatonin shifts the balance of NO production away from excessive, damaging levels towards a profile more conducive to tissue survival.
- Attenuation of oxidative and nitrosative stress: This is a cornerstone of melatonin’s neuroprotection. By reducing excessive NO production (mainly via iNOS inhibition) [65] and potently scavenging the highly toxic ONOO− formed from the reaction of NO with superoxide [60], melatonin significantly mitigates nitrosative stress. This is evidenced by reduced levels of nitrotyrosine, a footprint of ONOO− damage, in melatonin-treated animals post-stroke [68]. This action prevents downstream damage, including lipid peroxidation [58] and protein oxidation/nitration [36]. These effects are synergistic with melatonin’s well-established broader antioxidant actions, which include direct scavenging of various ROS (e.g., •OH, H2O2) and indirect enhancement of endogenous antioxidant defenses by stimulating enzymes like SOD, catalase (CAT), and GPx, and maintaining glutathione (GSH) levels [19].
- Anti-inflammatory effects: Neuroinflammation is a major driver of secondary injury after stroke, and melatonin effectively dampens this response [8]. A key mechanism involves modulating the activation state of microglia and infiltrating macrophages. Melatonin promotes a shift from the detrimental, pro-inflammatory M1 phenotype (characterized by production of TNF-α, IL-1β, IL-6, and often high iNOS expression) towards the beneficial, anti-inflammatory and tissue-reparative M2 phenotype [69]. This phenotypic switch leads to reduced levels of pro-inflammatory cytokines and MMPs, and potentially increased levels of anti-inflammatory mediators like IL-10 [70]. The signaling pathways mediating this effect include STAT3, which is enhanced by melatonin in microglia exposed to ischemic conditions, and the JAK2-STAT3 pathway [71]. Blockade of STAT3 diminishes melatonin’s ability to induce this phenotype shift [72]. Furthermore, melatonin’s inhibition of the NF-κB pathway contributes to reduced expression of multiple inflammatory genes, including iNOS [9]. Melatonin also reduces the infiltration of peripheral immune cells into the ischemic brain tissue [9].
- Anti-apoptotic mechanisms: Melatonin significantly reduces neuronal apoptosis, a major form of cell death in the ischemic penumbra [73]. Central to this effect is the protection of mitochondria [61]. Melatonin preserves mitochondrial membrane potential (ΔΨm), inhibits the opening of the mPTP (a critical event leading to cell death), reduces the release of pro-apoptotic factors like cytochrome c from mitochondria into the cytosol, maintains the function of electron transport chain (ETC) complexes (particularly complex I and IV), and decreases mitochondrial ROS/RNS generation [61]. It achieves this partly through its direct antioxidant actions within the mitochondria and by modulating signaling pathways. Melatonin also regulates the balance of Bcl-2 family proteins, increasing the expression of anti-apoptotic members like Bcl-2 and Bcl-xL while decreasing pro-apoptotic members like Bax [7]. Consequently, melatonin inhibits the activation of downstream executioner caspases, such as caspase-3 [8]. Additionally, melatonin has been shown to protect against endoplasmic reticulum (ER) stress, another pathway contributing to cell death after ischemia [61].
- Regulation of specific signaling pathways: The neuroprotective effects of melatonin in stroke, including its interplay with NO, are mediated via the modulation of several key intracellular signaling pathways that act as convergent hubs:
- ○
- Akt/PI3K pathway: Frequently activated by melatonin, this pathway promotes cell survival by phosphorylating and inactivating pro-apoptotic proteins (like Bad and caspase-9) and activating transcription factors involved in survival and antioxidant responses. Its activation is linked to reduced apoptosis and improved mitochondrial function in stroke models treated with melatonin [8]. Melatonin may activate Akt via MT2 receptors [8] or by suppressing PTEN activity [69]. The Akt-SIRT3-SOD2 axis is specifically implicated in melatonin’s protection against mitochondrial impairment in diabetic stroke models [66].
- ○
- ○
- Nrf2/HO-1 pathway: Melatonin activates the transcription factor Nrf2, which induces the expression of antioxidant enzymes like heme oxygenase-1 (HO-1), SOD, and GPx, bolstering cellular defenses against oxidative stress [74].
- ○
- AMPK/mTOR pathway: Melatonin modulates this pathway, which regulates cellular energy homeostasis, autophagy, and protein synthesis. In neonatal hypoxia-ischemia, melatonin’s neuroprotection was associated with regulation of AMPK/mTOR/autophagy signaling [75]. Melatonin may also enhance autophagy via PI3K/Akt/mTOR in other injury models [69].
- ○
- NF-κB pathway: As mentioned, melatonin inhibits NF-κB activation, a critical step in reducing the expression of pro-inflammatory genes, including iNOS, COX-2, and various cytokines, thereby limiting the inflammatory cascade after stroke [9].
- ○
4. Conclusions
4.1. Main Findings
4.2. Recap of Key Mechanisms
- Differential NOS isoform regulation: Suppression of detrimental iNOS, preservation/enhancement of protective eNOS, and variable modulation of nNOS shifts the NO balance towards neuroprotection.
- Direct RNS scavenging: Potent neutralization of NO and, critically, ONOO−, prevents downstream molecular damage.
- Mitochondrial protection: Melatonin safeguards mitochondria from NO/ONOO−-mediated damage, preserving energy production, reducing ROS leakage, and inhibiting apoptotic pathways.
- Signaling pathway modulation: Activation of pro-survival pathways (e.g., Akt/PI3K and Nrf2/HO-1) and inhibition of pro-inflammatory/pro-death pathways (e.g., NF-κB and p38 MAPK) integrate melatonin and NO signals.
- Anti-inflammation via microglia polarization: Melatonin promotes a shift from pro-inflammatory M1 to anti-inflammatory M2 microglia/macrophages, partly via STAT3 signaling, dampening the damaging inflammatory response.
5. Future Perspectives
5.1. Therapeutic Potential
5.2. Knowledge Gaps and Future Directions
- Mechanistic precision: Further studies are needed to dissect the precise molecular mechanisms by which melatonin differentially regulates nNOS and eNOS activity and expression in specific neuronal, glial, and endothelial cell types within the CNS, particularly during different phases of stroke evolution (acute, subacute, and chronic).
- Receptor vs. non-receptor roles: The relative contributions of melatonin’s receptor-mediated actions (via MT1/MT2) versus its direct, receptor-independent scavenging and mitochondrial effects in mediating specific aspects of NO modulation and neuroprotection in stroke need further clarification. Studies utilizing specific receptor antagonists (like luzindole [25]) or agonists, and potentially receptor knockout models (while considering endogenous melatonin levels), are warranted.
- Signaling network integration: A deeper understanding of how signaling pathways like Akt, STAT3, NF-κB, Nrf2, and AMPK/mTOR integrate signals from both melatonin and the NO system to orchestrate the overall cellular response to ischemic injury is required.
- Translational considerations: Rigorous clinical trials are essential to confirm the efficacy, optimal dosing, administration route, and therapeutic window for melatonin in human stroke patients [23]. Potential species differences in melatonin metabolism, receptor pharmacology [25], and NOS regulation [77] must be carefully considered during translation. The issue of endogenous melatonin deficiency in common laboratory mouse strains also highlights the need for appropriate model selection and interpretation [78]. The development of controlled-release or brain-targeted melatonin formulations could enhance its therapeutic efficacy and minimize side effects.
- Role of metabolites: The contribution of melatonin metabolites, such as AFMK and AMK, which possess significant antioxidant properties, to the overall neuroprotective effects observed in the CNS and stroke models warrants further investigation [19].
- Endogenous vs. exogenous melatonin: Distinguishing the protective roles of basal endogenous melatonin levels versus the effects of pharmacological administration is important, particularly given the observed decline in melatonin with age and in certain disease states, including potentially stroke [73].
Funding
Acknowledgments
Conflicts of Interest
References
- Barrenetxe, J.; Delagrange, P.; Martinez, J.A. Physiological and metabolic functions of melatonin. J. Physiol. Biochem. 2004, 60, 61–72. [Google Scholar] [CrossRef] [PubMed]
- Pevet, P. Melatonin in animal models. Dialogues Clin. Neurosci. 2003, 5, 343–352. [Google Scholar] [CrossRef] [PubMed]
- Zhao, D.; Yu, Y.; Shen, Y.; Liu, Q.; Zhao, Z.; Sharma, R.; Reiter, R.J. Melatonin Synthesis and Function: Evolutionary History in Animals and Plants. Front. Endocrinol. 2019, 10, 249. [Google Scholar] [CrossRef]
- Agathokleous, E.; Kitao, M.; Calabrese, E.J. New insights into the role of melatonin in plants and animals. Chem. Biol. Interact. 2019, 299, 163–167. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Candia, A.; Candia, A.A.; Figueroa, E.G.; Feixes, E.; Gonzalez-Candia, C.; Aguilar, S.A.; Ebensperger, G.; Reyes, R.V.; Llanos, A.J.; Herrera, E.A. Melatonin long-lasting beneficial effects on pulmonary vascular reactivity and redox balance in chronic hypoxic ovine neonates. J. Pineal Res. 2020, 68, e12613. [Google Scholar] [CrossRef]
- Alghamdi, B.S. The neuroprotective role of melatonin in neurological disorders. J. Neurosci. Res. 2018, 96, 1136–1149. [Google Scholar] [CrossRef]
- Andrabi, S.S.; Parvez, S.; Tabassum, H. Melatonin and Ischemic Stroke: Mechanistic Roles and Action. Adv. Pharmacol. Sci. 2015, 2015, 384750. [Google Scholar] [CrossRef]
- Zhang, C.; Ma, Y.; Zhao, Y.; Guo, N.; Han, C.; Wu, Q.; Mu, C.; Zhang, Y.; Tan, S.; Zhang, J.; et al. Systematic review of melatonin in cerebral ischemia-reperfusion injury: Critical role and therapeutic opportunities. Front. Pharmacol. 2024, 15, 1356112. [Google Scholar] [CrossRef]
- Lin, H.W.; Lee, E.J. Effects of melatonin in experimental stroke models in acute, sub-acute, and chronic stages. Neuropsychiatr. Dis. Treat. 2009, 5, 157–162. [Google Scholar] [CrossRef]
- Tan, H.Y.; Ng, K.Y.; Koh, R.Y.; Chye, S.M. Pharmacological Effects of Melatonin as Neuroprotectant in Rodent Model: A Review on the Current Biological Evidence. Cell Mol. Neurobiol. 2020, 40, 25–51. [Google Scholar] [CrossRef]
- Wendehenne, D.; Pugin, A.; Klessig, D.F.; Durner, J. Nitric oxide: Comparative synthesis and signaling in animal and plant cells. Trends Plant Sci. 2001, 6, 177–183. [Google Scholar] [CrossRef] [PubMed]
- Dai, Z.; Wu, Z.; Yang, Y.; Wang, J.; Satterfield, M.C.; Meininger, C.J.; Bazer, F.W.; Wu, G. Nitric oxide and energy metabolism in mammals. Biofactors 2013, 39, 383–391. [Google Scholar] [CrossRef]
- Esplugues, J.V. NO as a signalling molecule in the nervous system. Br. J. Pharmacol. 2002, 135, 1079–1095. [Google Scholar] [CrossRef]
- Sadanandan, N.; Cozene, B.; Cho, J.; Park, Y.J.; Saft, M.; Gonzales-Portillo, B.; Borlongan, C.V. Melatonin-A Potent Therapeutic for Stroke and Stroke-Related Dementia. Antioxidants 2020, 9, 672. [Google Scholar] [CrossRef]
- Lachowicz, S.; Swieca, M.; Pejcz, E. Improvement of Health-Promoting Functionality of Rye Bread by Fortification with Free and Microencapsulated Powders from Amelanchier alnifolia Nutt. Antioxidants 2020, 9, 614. [Google Scholar] [CrossRef]
- Schomerus, C.; Korf, H.W. Mechanisms regulating melatonin synthesis in the mammalian pineal organ. Ann. N. Y Acad. Sci. 2005, 1057, 372–383. [Google Scholar] [CrossRef]
- Falcon, J.; Besseau, L.; Fuentes, M.; Sauzet, S.; Magnanou, E.; Boeuf, G. Structural and functional evolution of the pineal melatonin system in vertebrates. Ann. N. Y Acad. Sci. 2009, 1163, 101–111. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Li, W.; Wang, J.; Baranov, S.V.; Heath, B.E.; Jia, J.; Suofu, Y.; Baranova, O.V.; Wang, X.; Larkin, T.M.; et al. Biosynthesis of neuroprotective melatonin is dysregulated in Huntington's disease. J. Pineal Res. 2023, 75, e12909. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.G.; Woo, Y.S.; Park, S.W.; Seog, D.H.; Seo, M.K.; Bahk, W.M. The Neuroprotective Effects of Melatonin: Possible Role in the Pathophysiology of Neuropsychiatric Disease. Brain Sci. 2019, 9, 285. [Google Scholar] [CrossRef]
- Hattori, A.; Suzuki, N. Receptor-Mediated and Receptor-Independent Actions of Melatonin in Vertebrates. Zoolog Sci. 2024, 41, 105–116. [Google Scholar] [CrossRef]
- Ahmadi, S.; Taghizadieh, M.; Mehdizadehfar, E.; Hasani, A.; Khalili Fard, J.; Feizi, H.; Hamishehkar, H.; Ansarin, M.; Yekani, M.; Memar, M.Y. Gut microbiota in neurological diseases: Melatonin plays an important regulatory role. Biomed. Pharmacother. 2024, 174, 116487. [Google Scholar] [CrossRef] [PubMed]
- Leclercq, B.; Hicks, D.; Laurent, V. Photoperiod integration in C3H rd1 mice. J. Pineal Res. 2021, 71, e12711. [Google Scholar] [CrossRef] [PubMed]
- Esposito, E.; Cuzzocrea, S. Antiinflammatory activity of melatonin in central nervous system. Curr. Neuropharmacol. 2010, 8, 228–242. [Google Scholar] [CrossRef]
- Pevet, P.; Klosen, P.; Felder-Schmittbuhl, M.P. The hormone melatonin: Animal studies. Best. Pract. Res. Clin. Endocrinol. Metab. 2017, 31, 547–559. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Gao, S.; Lenahan, C.; Gu, Y.; Wang, X.; Fang, Y.; Xu, W.; Wu, H.; Pan, Y.; Shao, A.; et al. Melatonin as an Antioxidant Agent in Stroke: An Updated Review. Aging Dis. 2022, 13, 1823–1844. [Google Scholar] [CrossRef]
- Camberos-Barraza, J.; Camacho-Zamora, A.; Batiz-Beltran, J.C.; Osuna-Ramos, J.F.; Rabago-Monzon, A.R.; Valdez-Flores, M.A.; Angulo-Rojo, C.E.; Guadron-Llanos, A.M.; Picos-Cardenas, V.J.; Calderon-Zamora, L.; et al. Sleep, Glial Function, and the Endocannabinoid System: Implications for Neuroinflammation and Sleep Disorders. Int. J. Mol. Sci. 2024, 25, 3160. [Google Scholar] [CrossRef]
- Reddi, S.; Thakker-Varia, S.; Alder, J.; Giarratana, A.O. Status of precision medicine approaches to traumatic brain injury. Neural Regen. Res. 2022, 17, 2166–2171. [Google Scholar] [CrossRef]
- Zhang, W.; Chen, S.J.; Guo, L.Y.; Zhang, Z.; Zhang, J.B.; Wang, X.M.; Meng, X.B.; Zhang, M.Y.; Zhang, K.K.; Chen, L.L.; et al. Nitric oxide synthase and its function in animal reproduction: An update. Front. Physiol. 2023, 14, 1288669. [Google Scholar] [CrossRef]
- Mazuryk, O.; Gurgul, I.; Oszajca, M.; Polaczek, J.; Kieca, K.; Bieszczad-Zak, E.; Martyka, T.; Stochel, G. Nitric Oxide Signaling and Sensing in Age-Related Diseases. Antioxidants 2024, 13, 1213. [Google Scholar] [CrossRef]
- Blanco, S.; Molina, F.J.; Castro, L.; Del Moral, M.L.; Hernandez, R.; Jimenez, A.; Rus, A.; Martinez-Lara, E.; Siles, E.; Peinado, M.A. Study of the nitric oxide system in the rat cerebellum during aging. BMC Neurosci. 2010, 11, 78. [Google Scholar] [CrossRef]
- Andrew, P.J.; Mayer, B. Enzymatic function of nitric oxide synthases. Cardiovasc. Res. 1999, 43, 521–531. [Google Scholar] [CrossRef]
- Lowenstein, C.J.; Dinerman, J.L.; Snyder, S.H. Nitric oxide: A physiologic messenger. Ann. Intern. Med. 1994, 120, 227–237. [Google Scholar] [CrossRef]
- Acuna-Castroviejo, D.; Escames, G.; Lopez, L.C.; Hitos, A.B.; Leon, J. Melatonin and nitric oxide: Two required antagonists for mitochondrial homeostasis. Endocrine 2005, 27, 159–168. [Google Scholar] [CrossRef]
- Chen, Y. Recent developments of fluorescent probes for detection and bioimaging of nitric oxide. Nitric Oxide Biol. Chem./Off. J. Nitric Oxide Soc. 2020, 98, 1–19. [Google Scholar] [CrossRef]
- Shukla, P.; Sun, C.; O'Rourke, S.T. Melatonin inhibits nitric oxide signaling by increasing PDE5 phosphorylation in coronary arteries. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H1418–H1425. [Google Scholar] [CrossRef]
- Hardeland, R. Melatonin and the electron transport chain. Cell. Mol. Life Sci. 2017, 74, 3883–3896. [Google Scholar] [CrossRef]
- Zhang, R.; Song, B.; Hong, X.; Shen, Z.; Sui, L.; Wang, S. microRNA-9 Inhibits Vulnerable Plaque Formation and Vascular Remodeling via Suppression of the SDC2-Dependent FAK/ERK Signaling Pathway in Mice With Atherosclerosis. Front. Physiol. 2020, 11, 804. [Google Scholar] [CrossRef]
- Santos, A.I.; Martinez-Ruiz, A.; Araujo, I.M. S-nitrosation and neuronal plasticity. Br. J. Pharmacol. 2015, 172, 1468–1478. [Google Scholar] [CrossRef] [PubMed]
- Hardingham, N.; Dachtler, J.; Fox, K. The role of nitric oxide in pre-synaptic plasticity and homeostasis. Front. Cell Neurosci. 2013, 7, 190. [Google Scholar] [CrossRef]
- Szabo, C. Physiological and pathophysiological roles of nitric oxide in the central nervous system. Brain Res. Bull. 1996, 41, 131–141. [Google Scholar] [CrossRef]
- Potes, Y.; Cachan-Vega, C.; Antuna, E.; Garcia-Gonzalez, C.; Menendez-Coto, N.; Boga, J.A.; Gutierrez-Rodriguez, J.; Bermudez, M.; Sierra, V.; Vega-Naredo, I.; et al. Benefits of the Neurogenic Potential of Melatonin for Treating Neurological and Neuropsychiatric Disorders. Int. J. Mol. Sci. 2023, 24, 4803. [Google Scholar] [CrossRef] [PubMed]
- Deng, W.G.; Tang, S.T.; Tseng, H.P.; Wu, K.K. Melatonin suppresses macrophage cyclooxygenase-2 and inducible nitric oxide synthase expression by inhibiting p52 acetylation and binding. Blood 2006, 108, 518–524. [Google Scholar] [CrossRef]
- Vilar, A.; de Lemos, L.; Patraca, I.; Martinez, N.; Folch, J.; Junyent, F.; Verdaguer, E.; Pallas, M.; Auladell, C.; Camins, A. Melatonin suppresses nitric oxide production in glial cultures by pro-inflammatory cytokines through p38 MAPK inhibition. Free Radic. Res. 2014, 48, 119–128. [Google Scholar] [CrossRef]
- Yin, C.; Zhang, M.; Cheng, L.; Ding, L.; Lv, Q.; Huang, Z.; Zhou, J.; Chen, J.; Wang, P.; Zhang, S.; et al. Melatonin modulates TLR4/MyD88/NF-kappaB signaling pathway to ameliorate cognitive impairment in sleep-deprived rats. Front. Pharmacol. 2024, 15, 1430599. [Google Scholar] [CrossRef]
- Leon, J.; Vives, F.; Crespo, E.; Camacho, E.; Espinosa, A.; Gallo, M.A.; Escames, G.; Acuna-Castroviejo, D. Modification of nitric oxide synthase activity and neuronal response in rat striatum by melatonin and kynurenine derivatives. J. Neuroendocrinol. 1998, 10, 297–302. [Google Scholar] [CrossRef] [PubMed]
- Xie, S.S.; Fan, W.G.; Liu, Q.; Li, J.Z.; Zheng, M.M.; He, H.W.; Huang, F. Involvement of nNOS in the antinociceptive activity of melatonin in inflammatory pain at the level of sensory neurons. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7399–7411. [Google Scholar] [CrossRef]
- Yang, H.J.; Kim, M.J.; Kim, S.S.; Cho, Y.W. Melatonin modulates nitric oxide-regulated WNK-SPAK/OSR1-NKCC1 signaling in dorsal raphe nucleus of rats. Korean J. Physiol. Pharmacol. 2021, 25, 449–457. [Google Scholar] [CrossRef] [PubMed]
- Pei, Z.; Cheung, R.T. Pretreatment with melatonin exerts anti-inflammatory effects against ischemia/reperfusion injury in a rat middle cerebral artery occlusion stroke model. J. Pineal Res. 2004, 37, 85–91. [Google Scholar] [CrossRef]
- Koh, P.O. Melatonin regulates nitric oxide synthase expression in ischemic brain injury. J. Vet. Med. Sci./Jpn. Soc. Vet. Sci. 2008, 70, 747–750. [Google Scholar] [CrossRef]
- Smith Petri, E.; Ajao, M.S.; Olaleye, O.; Ihunwo, A.O. Effect of Melatonin on Neuronal Nitric Oxide Synthase Expressing Cells in the Brain Following Global Cerebral Ischemia. J. Anim. Vet. Adv. 2011, 10, 395–400. [Google Scholar] [CrossRef]
- Beker, M.C.; Caglayan, A.B.; Kelestemur, T.; Caglayan, B.; Yalcin, E.; Yulug, B.; Kilic, U.; Hermann, D.M.; Kilic, E. Effects of normobaric oxygen and melatonin on reperfusion injury: Role of cerebral microcirculation. Oncotarget 2015, 6, 30604–30614. [Google Scholar] [CrossRef] [PubMed]
- Sarti, P.; Magnifico, M.C.; Altieri, F.; Mastronicola, D.; Arese, M. New evidence for cross talk between melatonin and mitochondria mediated by a circadian-compatible interaction with nitric oxide. Int. J. Mol. Sci. 2013, 14, 11259–11276. [Google Scholar] [CrossRef] [PubMed]
- Pechanova, O.; Paulis, L.; Simko, F. Impact of melatonin on central blood pressure regulation. Act. Nerv. Super. Rediviva 2016, 58, 99–104. [Google Scholar]
- Klimentova, J.; Cebova, M.; Barta, A.; Matuskova, Z.; Vrankova, S.; Rehakova, R.; Kovacsova, M.; Pechanova, O. Effect of melatonin on blood pressure and nitric oxide generation in rats with metabolic syndrome. Physiol. Res./Acad. Sci. Bohemoslov. 2016, 65, S373–S380. [Google Scholar] [CrossRef] [PubMed]
- Mendes, L.; Queiroz, M.; Sena, C.M. Melatonin and Vascular Function. Antioxidants 2024, 13, 747. [Google Scholar] [CrossRef]
- Li, D.; Pan, J.H.; Huang, X.F.; Liao, Y.Q.; Ling, Y.J.; Luo, J.Y. Effect of melatonin on oxidative stress indicators in animal models of fibrosis: A systematic review and meta-analysis. Free Radic. Biol. Med. 2023, 195, 158–177. [Google Scholar] [CrossRef]
- Monteiro, K.; Shiroma, M.E.; Damous, L.L.; Simoes, M.J.; Simoes, R.D.S.; Cipolla-Neto, J.; Baracat, E.C.; Soares-Jr, J.M. Antioxidant Actions of Melatonin: A Systematic Review of Animal Studies. Antioxidants 2024, 13, 439. [Google Scholar] [CrossRef]
- Siu, A.W.; Ortiz, G.G.; Benitez-King, G.; To, C.H.; Reiter, R.J. Effects of melatonin on the nitric oxide treated retina. Br. J. Ophthalmol. 2004, 88, 1078–1081. [Google Scholar] [CrossRef]
- Yilmaz, U.; Tanbek, K.; Gul, S.; Gul, M.; Koc, A.; Sandal, S. Melatonin Attenuates Cerebral Ischemia/Reperfusion Injury through Inducing Autophagy. Neuroendocrinology 2023, 113, 1035–1050. [Google Scholar] [CrossRef]
- Carretero, V.J.; Ramos, E.; Segura-Chama, P.; Hernandez, A.; Baraibar, A.M.; Alvarez-Merz, I.; Munoz, F.L.; Egea, J.; Solis, J.M.; Romero, A.; et al. Non-Excitatory Amino Acids, Melatonin, and Free Radicals: Examining the Role in Stroke and Aging. Antioxidants 2023, 12, 1844. [Google Scholar] [CrossRef]
- Tozihi, M.; Shademan, B.; Yousefi, H.; Avci, C.B.; Nourazarian, A.; Dehghan, G. Melatonin: A promising neuroprotective agent for cerebral ischemia-reperfusion injury. Front. Aging Neurosci. 2023, 15, 1227513. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.; Wang, S.; Wang, X.; Li, Y.; Dai, Z. Melatonin improves stroke by inhibiting autophagy-dependent ferroptosis mediated by NCOA4 binding to FTH1. Exp. Neurol. 2024, 379, 114868. [Google Scholar] [CrossRef] [PubMed]
- Reiter, R.J.; Manchester, L.C.; Tan, D.X. Neurotoxins: Free radical mechanisms and melatonin protection. Curr. Neuropharmacol. 2010, 8, 194–210. [Google Scholar] [CrossRef]
- Tejero, J.; Shiva, S.; Gladwin, M.T. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol. Rev. 2019, 99, 311–379. [Google Scholar] [CrossRef]
- Pei, Z.; Fung, P.C.; Cheung, R.T. Melatonin reduces nitric oxide level during ischemia but not blood-brain barrier breakdown during reperfusion in a rat middle cerebral artery occlusion stroke model. J. Pineal Res. 2003, 34, 110–118. [Google Scholar] [CrossRef]
- Liu, L.; Cao, Q.; Gao, W.; Li, B.; Xia, Z.; Zhao, B. Melatonin protects against focal cerebral ischemia-reperfusion injury in diabetic mice by ameliorating mitochondrial impairments: Involvement of the Akt-SIRT3-SOD2 signaling pathway. Aging 2021, 13, 16105–16123. [Google Scholar] [CrossRef]
- Li, L.; Yuan, Y.; Zhang, C.; Li, Y.; Xu, R.; Zhang, X.; Shang, W. Melatonin Promotes Cerebral Angiogenesis in Ischemic Mice via BMP6/Smad1/5/9 Pathway. Mol. Neurobiol. 2025. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.C.; Lai, C.J.; Tsai, M.H.; Wu, Y.C.; Chen, K.T.; Jou, M.J.; Fu, P.I.; Wu, C.H.; Wei, I.H. Effects of melatonin on the nitric oxide system and protein nitration in the hypobaric hypoxic rat hippocampus. BMC Neurosci. 2015, 16, 61. [Google Scholar] [CrossRef]
- Ran, Y.; Ye, L.; Ding, Z.; Gao, F.; Yang, S.; Fang, B.; Liu, Z.; Xi, J. Melatonin Protects Against Ischemic Brain Injury by Modulating PI3K/AKT Signaling Pathway via Suppression of PTEN Activity. ASN Neuro 2021, 13, 17590914211022888. [Google Scholar] [CrossRef]
- Xie, S.; Fan, W.; He, H.; Huang, F. Role of Melatonin in the Regulation of Pain. J. Pain. Res. 2020, 13, 331–343. [Google Scholar] [CrossRef]
- Yang, B.; Zang, L.E.; Cui, J.W.; Zhang, M.Y.; Ma, X.; Wei, L.L. Melatonin Plays a Protective Role by Regulating miR-26a-5p-NRSF and JAK2-STAT3 Pathway to Improve Autophagy, Inflammation and Oxidative Stress of Cerebral Ischemia-Reperfusion Injury. Drug Des. Devel Ther. 2020, 14, 3177–3188. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.J.; Ran, Y.Y.; Qie, S.Y.; Gong, W.J.; Gao, F.H.; Ding, Z.T.; Xi, J.N. Melatonin protects against ischemic stroke by modulating microglia/macrophage polarization toward anti-inflammatory phenotype through STAT3 pathway. CNS Neurosci. Ther. 2019, 25, 1353–1362. [Google Scholar] [CrossRef]
- Chen, D.; Zhang, T.; Lee, T.H. Cellular Mechanisms of Melatonin: Insight from Neurodegenerative Diseases. Biomolecules 2020, 10, 1158. [Google Scholar] [CrossRef]
- Qin, T.; Feng, D.; Zhou, B.; Bai, L.; Yin, Y. Melatonin Suppresses LPS-Induced Oxidative Stress in Dendritic Cells for Inflammatory Regulation via the Nrf2/HO-1 Axis. Antioxidants 2022, 11, 2012. [Google Scholar] [CrossRef] [PubMed]
- Nacarkucuk, E.; Bernis, M.E.; Bremer, A.S.; Grzelak, K.; Zweyer, M.; Maes, E.; Burkard, H.; Sabir, H. Neuroprotective Effect of Melatonin in a Neonatal Hypoxia-Ischemia Rat Model Is Regulated by the AMPK/mTOR Pathway. J. Am. Heart Assoc. 2024, 13, e036054. [Google Scholar] [CrossRef] [PubMed]
- Kilic, E.; Caglayan, B.; Caglar Beker, M. Physiological and pharmacological roles of melatonin in the pathophysiological components of cellular injury after ischemic stroke. Turk. J. Med. Sci. 2020, 50, 1655–1664. [Google Scholar] [CrossRef]
- Young, R.; Bush, S.J.; Lefevre, L.; McCulloch, M.E.B.; Lisowski, Z.M.; Muriuki, C.; Waddell, L.A.; Sauter, K.A.; Pridans, C.; Clark, E.L.; et al. Species-Specific Transcriptional Regulation of Genes Involved in Nitric Oxide Production and Arginine Metabolism in Macrophages. Immunohorizons 2018, 2, 27–37. [Google Scholar] [CrossRef]
- Kennaway, D.J. Melatonin research in mice: A review. Chronobiol. Int. 2019, 36, 1167–1183. [Google Scholar] [CrossRef]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Blanco, S.; Muñoz-Gallardo, M.d.M.; Hernández, R.; Peinado, M.Á. The Interplay Between Melatonin and Nitric Oxide: Mechanisms and Implications in Stroke Pathophysiology. Antioxidants 2025, 14, 724. https://doi.org/10.3390/antiox14060724
Blanco S, Muñoz-Gallardo MdM, Hernández R, Peinado MÁ. The Interplay Between Melatonin and Nitric Oxide: Mechanisms and Implications in Stroke Pathophysiology. Antioxidants. 2025; 14(6):724. https://doi.org/10.3390/antiox14060724
Chicago/Turabian StyleBlanco, Santos, María del Mar Muñoz-Gallardo, Raquel Hernández, and María Ángeles Peinado. 2025. "The Interplay Between Melatonin and Nitric Oxide: Mechanisms and Implications in Stroke Pathophysiology" Antioxidants 14, no. 6: 724. https://doi.org/10.3390/antiox14060724
APA StyleBlanco, S., Muñoz-Gallardo, M. d. M., Hernández, R., & Peinado, M. Á. (2025). The Interplay Between Melatonin and Nitric Oxide: Mechanisms and Implications in Stroke Pathophysiology. Antioxidants, 14(6), 724. https://doi.org/10.3390/antiox14060724