Rosmarinic acid attenuates lipopolysaccharide-induced neuroinflammation and cognitive impairment in rats

https://doi.org/10.1016/j.jchemneu.2021.102008Get rights and content

Highlights

  • Rosmarinic acid (RA) improved the LPS-induced memory deficit.

  • RA could prevent hippocampal neural cell loss through non-amyloidogenic pathways.

  • RA attenuated brain inflammation and reversed oxidant-antioxidant balance.

  • RA prevented the overproduction of TNF-α, IL-1β, and IL-6 in rat brain.

  • RA could reverse the hippocampal and cortical levels of MDA, NOx, and SOD.

Abstract

It has been recently demonstrated that rosmarinic acid (RA) through modulation in the amyloidogenic pathway exhibit neuroprotective potential in Alzheimer’s disease. However, its effects on non-amyloidogenic pathways such as neuroinflammation (NI) and oxidative stress have not been elucidated carefully. Hence, this study aimed to investigate the effect of RA on cognitive function, cortical and hippocampal oxidant-antioxidant balance, and proinflammatory cytokines production in lipopolysaccharide (LPS)-induced NI in rats. NI was induced by intracerebroventricular injection of LPS (50 μg/20 μL; 10 μL into each ventricle) in Wistar rats. RA (25 and 50 mg/kg.) was intraperitoneally administrated to the experimental groups 30 min before the LPS injection and continued once per day for seven days. Cognitive function was investigated by the Y-maze test, and the production of proinflammatory cytokines and oxidative stress markers were evaluated in their hippocampi (HIP) and prefrontal cortex (PFC). In addition, neuronal damage was evaluated in the HIP subfields histologically. The RA administration could alleviate cognitive impairments caused by NI in LPS-treated rats as evidenced by improved working memory and attenuated neuronal injury in the HIP subfields. RA treatment in a dose-dependent manner prevented the overproduction of tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), and IL-6 in both the HIP and PFC. RA significantly alleviated the HIP and PFC levels of malondialdehyde (MDA) and nitric oxide (NOx) and enhanced the superoxide dismutase (SOD) activity. These findings demonstrated that RA could also exert its neuroprotective effects by modulating non-amyloidogenic pathways such as inflammation and oxidative stress.

Introduction

Neuroinflammation (NI) is one of the most common denominators in several neurodegenerative diseases. Therefore, in recent years, the role of inflammatory processes in the etiology of many neurological disorders has been studied (Gilhus and Deuschl, 2019). Consequently, a bulk of evidence is rising attention and pointing to the role of NI in the pathophysiology of several neurodegenerative disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (Dorothée, 2018). NI is an inflammatory response of the central nervous system (CNS) mediated by abnormal secretion of proinflammatory cytokines, chemokines, and reactive oxygen/nitrogen species (DiSabato et al., 2016). It can lead to an ongoing pathologic process in the CNS by infiltration of peripheral immune cells, edema, neuronal atrophy, damage, and death overtime. These events affect neuronal structure and function during prolonged NI, resulting in cognitive and functional impairments (Chen et al., 2016). Considering that, modulation of the immune system has emerged as a therapeutic strategy in neurodegenerative diseases (Chen et al., 2016). In this approach, valuable signs of progress are being made as several studies show considerable promise for new substances, mainly natural products. However, more in vivo experiments are still needed to improve our understanding of these substances' mechanisms of action (Rekatsina et al., 2020).

Rosmarinic acid (RA) is naturally found in the Lamiaceae family, such as rosemary, sage, lemon balm, mint, and sweet basil (Nadeem et al., 2019). The accumulating evidence supporting that RA exerts powerful neuroprotective, antimicrobial, anti-inflammatory, anti-aging, and antioxidant effects (Ma et al., 2020; Nadeem et al., 2019). However, its mechanisms of action have not been completely elucidated. For example, a recently published study demonstrated that RA by increasing brain monoamines such as norepinephrine, dopamine, and levodopa could suppress amyloid-beta aggregation in a mouse model of AD (Hase et al., 2019). Since NI, as a key pathological event, triggers and perpetuates several neurodegenerative diseases such as AD, its modulation could influence other pathological mechanisms linked to the neurodegenerative process (Batista et al., 2019). Therefore, we hypothesized that RA probably could affect other pathological mechanisms involved in AD like NI and oxidative stress. Consequently, this study aimed to broaden current knowledge of mechanisms through which RA impacts neurodegenerative diseases' pathogenesis. To test the hypothesis, we established a rat model of NI by intracerebroventricular (ICV) injection of lipopolysaccharide (LPS) and evaluated RA's possible anti-NI efficiency in systemic administration.

Section snippets

Chemical and reagents

RA (molecular formula: C18H16O8; R4033; CAS No: 20283-92-5; purity ≥ 98 %), LPS (Escherichia coli; O127: B8), phosphate-buffered saline (PBS) tablets, and tetrazolium WST-1 solution were purchased from Sigma Aldrich Company (St Louis, MO, USA). The artificial cerebrospinal fluid (aCSF) was used as a vehicle. The aCSF (pH = 7.4) was containing 140 mM NaCl, 3 mM KCl, 25 mM CaCl2, 1 mM MgCl2 and 1.2 mM Na2Hpo4 (Zhao et al., 2017). Enzyme-linked immunosorbent assay (ELISA) kits including tumor

Effects of RA on working memory

The Y-maze test was performed to determine RA's impact on working memory in LPS-treated rats (Fig. 1). LPS treated rats showed significantly lower short-term memory performance than the SO group (p < 0.001). RA treatment, mainly at the higher one (50 mg/kg), significantly improved the percentage of spontaneous alternation behavior in the LPS-treated rats, as compared to the LPS group (p < 0.01). RA (50 mg/kg) improved the short-term working memory in LPS-treated rats close to the SO group

Discussion

A growing body of evidence suggests that the NI could drive the pathogenic process in several neurodegenerative diseases. Therefore, the modulation of NI has been considered a therapeutic goal for drug development. Recent studies have shown that RA, through inhibiting the amyloidogenic pathway increases brain monoamines and, in consequence, suppresses amyloid β aggregation. However, its effects on non-amyloidogenic pathways such as NI and oxidative stress have not been elucidated carefully. In

Conclusion

Taken together, the findings of the present study extend the range of previous results on the neuroprotective mechanisms of RA. These findings demonstrated that RA by modulating non-amyloidogenic pathways such as inflammation and oxidative stress could also exert its beneficial effects on neurodegenerative disease pathogenesis.

Ethical statement

  • 1)

    This material is the authors' own original work, which has not been previously published elsewhere.

  • 2)

    The paper is not currently being considered for publication elsewhere.

  • 3)

    The paper reflects the authors' own research and analysis in a truthful and complete manner.

  • 4)

    The paper properly credits the meaningful contributions of co-authors and co-researchers.

  • 5)

    The results are appropriately placed in the context of prior and existing research.

  • 6)

    All sources used are properly disclosed (correct citation).

Authorship contributions

Mohammadmehdi Hassanzadeh-Taheri: Conceptualization, Supervision, Reviewing and Editing.

Atiyeh Ahmadi-Zohan: Investigation.

Mahtab Mohammadifard: Methodology, Resources, Validation.

Mehran Hosseini: Conceptualization, Methodology, Formal analysis, Writing original draft, Project administration.

Funding

This work was supported by grants from Birjand University of Medical Sciences (456070).

Availability of data and materials

The datasets used in the current study are available from the corresponding author on reasonable request.

Code availability

Not applicable.

Authors’ contribution

M H-T and MH designed the experiments; AA-Z and MH performed the animal experiments; MM and MH participated in histopathological and ELISA assessments; MH discussed the data and wrote the manuscript draft and M H-T revised it. All authors read and approved the findings.

Ethics approval

This study was approved by the Ethics Committee of Birjand University of Medical Sciences (permit code: Ir.bums.REC.1398.316).

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Declaration of Competing Interest

The authors report no declarations of interest.

Acknowledgements

The authors would like to thank Ms. Khadijeh Vazifeshenas (Laboratory technologist) for her technical assistance in ELISA assays.

References (40)

  • D.J. DiSabato et al.

    Neuroinflammation: the devil is in the details

    J. Neurochem.

    (2016)
  • G. Dorothée

    Neuroinflammation in neurodegeneration: role in pathophysiology, therapeutic opportunities and clinical perspectives

    J. Neural Transm. (Vienna)

    (2018)
  • P.M. Fernando et al.

    Rosmarinic acid attenuates cell damage against UVB radiation-induced oxidative stress via enhancing antioxidant effects in human HaCaT cells

    Biomol. Ther. (Seoul)

    (2016)
  • N.E. Gilhus et al.

    Neuroinflammation—a common thread in neurological disorders

    Nat. Rev. Neurol.

    (2019)
  • D.K. Gok et al.

    Effects of rosmarinic acid on cognitive and biochemical alterations in ovariectomized rats treated with D-galactose

    Folia Histochem. Cytobiol.

    (2015)
  • H. Haghir et al.

    Expression of apoptosis-regulatory genes in the hippocampus of rat neonates born to mothers with diabetes

    Metab. Brain Dis.

    (2017)
  • J. Hami et al.

    Effects of L-arginine pre-treatment in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced Parkinson’s diseases in Balb/c mice

    Iran. J. Neurol.

    (2015)
  • T. Hase et al.

    Rosmarinic acid suppresses Alzheimer’s disease development by reducing amyloid β aggregation by increasing monoamine secretion

    Sci. Rep.

    (2019)
  • R. Hoshyar et al.

    The impact of Crocus sativus stigma against methotrexate-induced liver toxicity in rats

    J. Complement. Integr. Med.

    (2019)
  • N. Ito et al.

    Rosmarinic acid from Perillae Herba produces an antidepressant-like effect in mice through cell proliferation in the hippocampus

    Biol. Pharm. Bull.

    (2008)
  • Cited by (19)

    • Phenethyl ester of rosmarinic acid ameliorates experimental autoimmune encephalomyelitis

      2022, Immunology Letters
      Citation Excerpt :

      Importantly, we have recently shown that PERA ameliorates type 1 diabetes in mice [49]. Also, rosmarinic acid was shown to be efficient against neuroinflammation induced by LPS [50], and in hepatic ischemia and reperfusion injury [39] in rats. These data provide further value on PERA as a prominent candidate for therapeutical application in autoimmune and chronic inflammatory disorders.

    • Binding of rosmarinic acid curcumin and capsaicin with PLA2: A comparative study

      2022, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      RA (Fig. 1A) is a naturally occurring ester of caffeic acid and 3,4-dihydroxyphenyllactic acid [13]. Recent studies have shown that the neuroprotective properties of RA is mediated through alteration of the amyloidogenic pathway and attenuates lipopolysaccharide-induced neuroinflammation and cognitive impairment [14]. CUR (Fig. 1B), a plant derived polyphenol compound, isolated from the plant Curcuma longa, commonly known as turmeric, with a variety of pharmacologic properties [15].

    View all citing articles on Scopus
    View full text