Open Access

Neuroprotective mechanisms of 3‑n‑butylphthalide in neurodegenerative diseases (Review)

  • Authors:
    • Rixin Luo
    • Runqi Wangqin
    • Lihong Zhu
    • Wei Bi
  • View Affiliations

  • Published online on: October 16, 2019     https://doi.org/10.3892/br.2019.1246
  • Pages: 235-240
  • Copyright: © Luo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Since 3‑n‑butylphthalide (NBP) was approved by the China Food and Drug Administration for the treatment of acute ischemia stroke in 2002, a number of studies have investigated NBP worldwide. In recent years, NBP has also demonstrated potential as treatment of several neurodegenerative diseases, which has increased the interest in its mechanisms of protection and action. Clinical studies and studies that used cell or animal models, have directly demonstrated neuroprotective effects of NBP via the following mechanisms: i) Inhibiting the inflammatory reaction; ii) reducing mitochondrial oxidative stress; iii) regulating apoptosis and autophagy; iv) inducing resistance to endoplasmic reticulum stress; and v) decreasing abnormal protein deposition. Therefore, NBP may be a potential drug for neurodegenerative diseases, and it is particularly important to identify the mechanism of NBP as it may assist with the development of new drugs for neurodegeneration. The present review summarizes the neuroprotective mechanisms of NBP and discusses new perspectives and prospects. The aim of the current review is to provide a new summary regarding NBP and its associated mechanisms.

1. Introduction

3-n-butylphthalide (NBP), approved by the China Food and Drug Administration for the treatment of acute ischemic stroke, is a type of compound isolated from the seeds of Chinese celery (1). The molecular structure of NBP is presented in Fig. 1. Therapy using NBP has been recommended by Chinese guidelines for acute ischemic stroke (2). A randomized double-blind trial (clinical trial no. ChiCTR-TRC-09000483) reported that NBP significantly improves clinical outcomes, including the modified Rankin Scale (3) and National institute of Health Stroke Scale scores (4), of patients who experienced ischemic stroke (5). In addition, a study demonstrated that NBP therapy persistently increases the level of endothelial progenitor cells in peripheral blood, ameliorate cerebral blood flow and improve neuronal functions (6). Furthermore, NBP has been reported to be a safe treatment for cerebral ischemia stroke (5-7). A study has indicated that NBP exhibits protective effects in several neurodegenerative diseases (8). However, to the best of our knowledge, the neuroprotective mechanism of NBP remains unclear. Therefore, the present review discusses the potential mechanism of neuroprotective effects of NBP. The aim of the current review is to provide further understanding regarding the advances of NBP.

2. NBP inhibits the inflammatory reaction

Inflammation, a complex biological response to injury, is associated with neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease (PD), multiple sclerosis, amyotrophic lateral sclerosis, traumatic brain injury (TBI) and more (9-11). NBP has exhibited anti-inflammatory effects in various models of these diseases and certain mechanisms have been identified. NBP has been reported to reduce the inflammatory reaction by inhibiting nucleotide binding oligomerization domain like receptor protein 3-inflammasome microglia activation and mitigating the Alzheimer's-like pathology via the nuclear factor erythroid-2-related factor 2-thioredoxin-interacting protein-TXNIP-thioredoxin axis in an APP/PS1 mouse model (12,13). Furthermore, NBP inhibited the inflammatory reaction in lipopolysaccharide (LPS)-induced rats via inhibition of c-Jun N-terminal kinase activation and the NF-κB pathway (14,15). NBP was reported to improve dyskinesia in a LPS-induced PD mouse model via a reduction in the loss of dopaminergic neurons, activation of mouse microglia, an increase in TNF-α levels and α-synuclein deposition in the black substantia of the mouse midbrain (16). Additionally, NBP-treatment reduces NF-κB activation following TBI (17), and NBP also inhibits the inflammatory reaction via the same pathway in spontaneously hypertensive rats (18). Notably, a number of studies have indicated that NBP inhibits the inflammatory reaction in other neuroassociated experimental models, such as an experimental model of autoimmune encephalomyelitis of microglia or autoimmune myositis in guinea pigs (19,20). In addition, NBP-treatment has been demonstrated to significantly ameliorate cerebral ischemia reperfusion-induced brain injury of Sprague-Dawley (SD) rats by inhibiting toll like receptor 4/NF-κB-associated inflammation (21). NBP attenuates advanced glycation end products-induced endothelial dysfunction by ameliorating inflammatory responses (22). In summary, there is some understanding regarding the mechanism of NBP in the inhibition of inflammation.

3. NBP reduces mitochondrial oxidative stress

Mitochondria, the site of oxidative metabolism in eukaryotes, produce energy through the oxidation of carbohydrates, fats and amino acids (23). Therefore, mitochondrial dysfunction in the form of oxidative stress may contribute to the pathogenesis of various neurodegenerative diseases (24). Oxidative stress is considered a condition that is caused by an imbalance between pro- and antioxidant factors, which leads to molecular and cellular damage (25). Oxidative stress serves an essential role in the development of age-related diseases (26). NBP exhibits a cumulative beneficial effect on the process of mitochondrial damage (27). This section will discuss the mechanisms involved in mitochondrial oxidative stress.

Recently, NBP exhibited a powerful effect on antioxidant stress in some different models. NBP inhibited oxidative stress in K141N-induced SH-SY5Y cells and in LPS-induced rats through activation of the Kelch-like ECH-associating protein 1 Nrf2-related factor 2-antioxidant response element signaling pathway (15,28). Similarly, NBP reduced oxidative damage to provide neuroprotection in mice following TBI and in rats following carbon monoxide poisoning (29,30). In addition, NBP protects against cerebral ischemia-reperfusion injury by decreasing antioxidant stress via the ERK signaling pathway (31). NBP also protects against H2O2-induced injury in neural stem cells by activation of the PI3K/Akt and the Mash1 signaling pathways (32). Furthermore, NBP has been reported to increase superoxide dismutase and catalase activity, and reduce malondialdehyde activity in the experimental autoimmune myositis (EAM) model, NBP directly protects muscle mitochondria and muscle cells from oxidative damage (33). However, the protective effect of NBP on mitochondrial function is not only limited to neurodegeneration, but also appears in cardiovascular diseases. A study suggested that NBP exerts a cardioprotective effect on cardiac ischemic injury via the regulation of mitochondrial function both using in vivo and in vitro experiments (34). In summary, the antioxidant effect of NBP has been widely recognized.

4. NBP regulates apoptosis and autophagy

Apoptosis and autophagy are basic biological phenomena of cells, which serve essential roles in removing abnormal cells in multicellular organisms. Disorders in the apoptosis and autophagy processes may cause the occurrence of neuropathy (35). The neuroprotective effect of NBP via the regulation of apoptosis and autophagy has been demonstrated. Treatment with NBP has been reported to reduce apoptotic cell death by increasing the levels of cleaved caspase-3 and caspase-9 following TBI (17). Furthermore, NBP blocks neural apoptosis in areas surrounding cortical contusions on the brain that are induced by TBI (29). The neuroprotective mechanism of NBP involves the mitochondrial apoptotic pathway. NBP inhibits HSPB8 K141N mutation-induced neurotoxicity, attenuates β-amyloid-induced toxicity in SH-SY5Y cells, and protects rat cardiomyocytes from ischemia or reperfusion through regulating mitochondrion-mediated apoptosis (28,36,37). Furthermore, certain studies have demonstrated the inhibition of apoptosis by NBP via the Akt pathway. One study reported that NBP activates Akt/mTOR signaling to inhibit neuronal apoptosis and autophagy in mice with repeated cerebral ischemia reperfusion injury (38). Another study demonstrated that NBP improves cognitive impairment of APP/PS1 mice by inhibiting apoptosis via the PI3K/AKT pathway (39). Additionally, NBP reduces the number of apoptotic cells by regulating Bcl-2 in HUVECs and an EAM model (22,33).

5. NBP resists endoplasmic reticulum stress

ERS is characterized by incorrect folding and aggregation of unfolded proteins in the endoplasmic reticulum lumen and a disturbance of the calcium balance, which can activate the unfolded protein response and lead to disturbance of the cell function and cell death (40). In recent years, certain studies have reported an anti-ERS effect of NBP. One study demonstrated that NBP inhibits doxorubicin-induced ERS in SD rats (41). In addition, NBP alleviates vascular cognitive impairment by regulating ERS and the Sonic hedgehog/Patched homolog 1 signaling pathway in SD rats (42). Both of these studies agreed that NBP attenuates ERS through regulating the expression of 78-kDa glucose-regulated protein (GRP78), CCAAT-enhancer binding protein homologous protein (CHOP) and caspase-12. Furthermore, NBP also inhibits ERS by attenuating activating transcription factory (ATF)-4, ATF-6, X-box binding protein 1, protein disulfide isomerase, GRP78, CHOP and cleaved-caspase-12 in a spinal cord injury (SCI) model, which may improve functional recovery and prevent disruption of the blood-spinal cord barrier (43,44). However, this mechanism has only recently been identified; therefore, there is limited literature about it. Further research on this mechanism may lead to new findings.

6. NBP decreases abnormal protein deposition

Abnormal protein deposition is closely associated with numerous neurodegenerative diseases (45), such as Alzheimer's disease, which is associated with amyloid-β (Aβ) and tau proteins; and PD, which is associated with α-synuclein (46). A study has demonstrated that NBP significantly reduces total cerebral Aβ plaque deposition and lowers Aβ levels in brain homogenates in a triple-transgenic mouse model of Alzheimer's disease via directing amyloid precursor protein processing toward a non-amyloidogenic pathway (47). Furthermore, NBP treatment inhibited tau hyperphosphorylation in AβPP/PS1 mice, which may improve cognitive impairment (48). NBP enhances a 1-methyl-4-phenylpyridiniumion-induced cellular model and a LPS-induced mice model of PD via reducing the accumulation of α-synuclein (16,49). However, the molecular mechanisms of how NBP reduces the accumulation of α-synuclein and inhibits tau hyperphosphorylation remain unclear. Furthermore, to the best of our knowledge, there is no associated study that provides the clinical evidence that NBP is effective in multiple sclerosis or Lewy body dementia via attenuating abnormal protein deposition. Potentially, new findings can be revealed in additional neurodegenerative diseases.

7. Conclusion

In summary, current studies suggest that NBP serves a neuroprotective role through inhibiting inflammation, protecting mitochondrial function, alleviating oxidative stress, regulating apoptosis, resisting ERS and decreasing the abnormal protein deposition (Fig. 2). Details on specific molecular mechanisms are presented in Table I. Taken together, it is suggested that NBP provides a promising therapeutic strategy for neurodegenerative diseases. In further studies, the mechanism of action of NBP may be further clarified, and the understanding regarding its potential uses may be expanded.

Table I

Neuroprotective mechanisms of 3-n-butylphthalide.

Table I

Neuroprotective mechanisms of 3-n-butylphthalide.

A, Inflammation inhibition
Author, yearStudy subjectMethodMolecular mechanismRefs.
Wang et al, 2018APP/PS1 miceTransgenicNLRP3 inflammasome activation inhibition(13)
 A172, SH-SY5YLPS inducedNLRP3 inflammasome activation inhibition 
Yang et al, 2018SD ratsLPS inducedNF-κB pathway inhibition(14)
Zhao et al, 2016C57BL/6 miceLPS inducedDownregulation of JNK activation(15)
Zhao et al, 2017C57BL/6 miceTraumatic brain injuryNF-κB pathway inhibition(17)
Wang et al, 2018EAE Neuroantigen-specific proinflammatory T cells inducedSuppression of PGAM5(19)
Zhang et al, 2016SD ratsCerebral ischemia reperfusion inducedIncreased HGF expression(21)
Liu et al, 2017HUVECsAdvanced glycation end product inducedRAGE/NF-κB pathway inhibition(22)
B, Reduction of mitochondrial oxidative stress
Author, yearStudy subjectMethodMolecular mechanismRefs.
Yang et al, 2017SH-SY5YMissense mutationsIncreased Nrf2 expression(28)
Liu et al, 2017ICR miceTraumatic brain injuryNrf2-ARE pathway activation(29)
Li et al, 2015SD ratsCarbon monoxide poisonedKeap1/Nrf2 pathway activation(30)
Zhu et al, 2018ICR miceCerebral ischemia reperfusion injuryERK signaling inhibition(31)
Wang et al, 2018NSCs from SD ratsHydrogen peroxide inducedPI3K/Akt and Mash1 pathway activation(32)
Chen et al, 2017Guinea pigsExperimental autoimmune myositisEnhanced Na+-K+ and Ca2+-Mg2+ ATPase activities(33)
Tian et al, 2017H9C2Hydrogen peroxide inducedEnhanced Nrf-1 and TFAM expression(34)
C, Regulation of apoptosis and autophagy
Author, yearStudy subjectMethodMolecular mechanismRefs.
Zhao et al, 2017C57BL/6 miceTraumatic brain injuryDownregulated caspase-3 and -9 expression(17)
Liu et al, 2017HUVECsAdvanced glycation end product inducedRegulation of Bcl-2 expression(22)
Lei et al, 2014SH-SY5Yβ-amyloid inducedRegulation of Bcl-2, caspase-3 and -9 expression(37)
Xu et al, 2017C57BL/6 miceRepeated cerebral ischemia reperfusionBcl-2/Bax elevation(38)
Xiang et al, 2014APP/PS1 miceTransgenicBDNF/TrkB/PI3K/Akt pathway regulation(39)
D, Resistance to endoplasmic reticulum stress
Author, yearStudy subjectMethodMolecular mechanismRefs.
Liao et al, 2018SD ratsDoxorubicin inducedGRP78, CHOP and caspase-12 expression regulation(41)
Niu et al, 2018SD ratsBilateral surgical ligation of common carotid arteriesGRP78, CHOP and caspase-12 expression regulation(42)
Zheng et al, 2017SD ratsLaminectomy performed at T9ATF-4, ATF-6, XBP-1, PDI, GRP78, CHOP and cleaved-caspase 12 attenuation(43)
 HBMECsThapsigargin inducedATF-4, ATF-6, XBP-1, PDI, GRP78, CHOP and cleaved-caspase 12 attenuation 
He et al, 2017SD ratsLaminectomy performed at T9ATF-4, ATF-6, XBP-1, PDI, GRP78, CHOP and cleaved-caspase 12 attenuation(44)
 PC12Thapsigargin inducedATF-4, ATF-6, XBP-1, PDI, GRP78, CHOP and cleaved-caspase 12 attenuation 
E, Reduced abnormal protein deposition
Author, yearStudy subjectMethodMolecular mechanismRefs.
Peng et al, 20103xTg-AD miceTransgenicDirection of APP processing towards a non-amyloidogenic pathway(47)
Peng et al, 2012AβPP/PS1 miceTransgenicTau hyperphosphorylation inhibition(48)
Chen et al, 2018C57BL/6 miceLPS inducedReduction of α-synuclein deposition(16)
Huang et al, 2010PC12MPP+ toxicity inducedReduction of α-synuclein deposition(49)

[i] LPS, lipopolysaccharide; SD, Sprague Dawley; JNK, c-Jun N-terminal kinase; HGF, hepatocyte growth factor; PGAM5, PGAM family member 5; RAGE, receptor for advanced glycation end-product; Nrf, nuclear respiratory factor; ARE, antioxidant response element; Keap1, Kelch-like ECH-Associating protein 1; Mash1, mammalian achaete scute homolog-1; TFAM, human mitochondrial transcription factor A; ICR, Institute of Cancer Research; NSC, neural stem cell; BDNF, brain derived neurotrophic factor; TrkB, Tyrosine receptor kinase B; GRP78, glucose regulated protein 78; XBP-1, X-box-binding protein 1; PDI, protein disulfide isomerase; APP, amyloid precursor protein; ATF, activating transcription factory; CHOP, CCAAT-enhancer binding protein homologous protein; MPP+, 1-methyl-4-phenylpyridiniumion.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the Natural Science Foundation of China (grant no. 81371442), the Training program for outstanding young teachers in higher education institutions of Guangdong Province (grant no. YQ2015024) and the Fundamental Research Funds for the Central Universities (grant no. 21617482).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

RL was a major contributor in writing the manuscript. RL, RW, LZ and WB contributed to researching data, discussing content and editing the manuscript. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Xu ZQ, Zhou Y, Shao BZ, Zhang JJ and Liu C: A Systematic Review of Neuroprotective Efficacy and Safety of DL-3-N-Butylphthalide in Ischemic Stroke. Am J Chin Med. 47:507–525. 2019.PubMed/NCBI View Article : Google Scholar

2 

Chinese Society of Cerebral Blood Flow and Metabolism: The Chinese guidelines for the evaluation and management of cerebral collateral circulation in ischemic stroke (2017). Zhonghua Nei Ke Za Zhi 56: 460-471, 2017 (In Chinese).

3 

Banks JL and Marotta CA: Outcomes validity and reliability of the modified Rankin scale: implications for stroke clinical trials: a literature review and synthesis. Stroke. 38:1091–1096. 2007.PubMed/NCBI View Article : Google Scholar

4 

Heldner MR, Zubler C, Mattle HP, Schroth G, Weck A, Mono ML, Gralla J, Jung S, El-Koussy M, Lüdi R, et al: National Institutes of Health stroke scale score and vessel occlusion in 2152 patients with acute ischemic stroke. Stroke. 44:1153–1157. 2013.PubMed/NCBI View Article : Google Scholar

5 

Cui LY, Zhu YC, Gao S, Wang JM, Peng B, Ni J, Zhou LX, He J and Ma XQ: Ninety-day administration of dl-3-n-butylphthalide for acute ischemic stroke: A randomized, double-blind trial. Chin Med J (Engl). 126:3405–3410. 2013.PubMed/NCBI View Article : Google Scholar

6 

Zhao H, Yun W, Zhang Q, Cai X, Li X, Hui G, Zhou X and Ni J: Mobilization of Circulating Endothelial Progenitor Cells by dl-3-n-Butylphthalide in Acute Ischemic Stroke Patients. J Stroke Cerebrovasc Dis. 25:752–760. 2016.PubMed/NCBI View Article : Google Scholar

7 

Zhang C, Zhao S, Zang Y, Gu F, Mao S, Feng S, Hu L and Zhang C: The efficacy and safety of Dl-3n-butylphthalide on progressive cerebral infarction: A randomized controlled STROBE study. Medicine (Baltimore). 96(e7257)2017.PubMed/NCBI View Article : Google Scholar

8 

Huang L, Wang S, Ma F, Zhang Y and Peng Y, Xing C, Feng Y, Wang X and Peng Y: From stroke to neurodegenerative diseases: The multi-target neuroprotective effects of 3-n-butylphthalide and its derivatives. Pharmacol Res. 135:201–211. 2018.PubMed/NCBI View Article : Google Scholar

9 

Skaper SD, Facci L, Zusso M and Giusti P: An Inflammation-Centric View of Neurological Disease: Beyond the Neuron. Front Cell Neurosci. 12(72)2018.PubMed/NCBI View Article : Google Scholar

10 

Liu J and Wang F: Role of Neuroinflammation in Amyotrophic Lateral Sclerosis: Cellular Mechanisms and Therapeutic Implications. Front Immunol. 8(1005)2017.PubMed/NCBI View Article : Google Scholar

11 

Niu F, Sharma A, Feng L, Ozkizilcik A, Muresanu DF, Lafuente JV, Tian ZR, Nozari A and Sharma HS: Nanowired delivery of DL-3-n-butylphthalide induces superior neuroprotection in concussive head injury. Prog Brain Res. 245:89–118. 2019.PubMed/NCBI View Article : Google Scholar

12 

Zhang Y, Huang LJ, Shi S, Xu SF, Wang XL and Peng Y: L-3-n-butylphthalide Rescues Hippocampal Synaptic Failure and Attenuates Neuropathology in Aged APP/PS1 Mouse Model of Alzheimer's Disease. CNS Neurosci Ther. 22:979–987. 2016.PubMed/NCBI View Article : Google Scholar

13 

Wang CY, Xu Y, Wang X, Guo C, Wang T and Wang ZY: Dl-3-n-Butylphthalide Inhibits NLRP3 Inflammasome and Mitigates Alzheimer's-Like Pathology via Nrf2-TXNIP-TrX Axis. Antioxid Redox Signal. 30:1411–1431. 2018.PubMed/NCBI View Article : Google Scholar

14 

Yang M, Dang R, Xu P, Guo Y, Han W, Liao D and Jiang P: Dl-3-n-Butylphthalide improves lipopolysaccharide-induced depressive-like behavior in rats: Involvement of Nrf2 and NF-κB pathways. Psychopharmacology (Berl). 235:2573–2585. 2018.PubMed/NCBI View Article : Google Scholar

15 

Zhao CY, Lei H, Zhang Y, Li L, Xu SF, Cai J, Li PP, Wang L, Wang XL and Peng Y: L-3-n-Butylphthalide attenuates neuroinflammatory responses by downregulating JNK activation and upregulating Heme oxygenase-1 in lipopolysaccharide-treated mice. J Asian Nat Prod Res. 18:289–302. 2016.PubMed/NCBI View Article : Google Scholar

16 

Chen Y, Jiang M, Li L, Ye M, Yu M, Zhang L, Ge B, Xu W and Wei D: DL-3-n-butylphthalide reduces microglial activation in lipopolysaccharide-induced Parkinson's disease model mice. Mol Med Rep. 17:3884–3890. 2018.PubMed/NCBI View Article : Google Scholar

17 

Zhao Y, Lee JH, Chen D, Gu X, Caslin A, Li J, Yu SP and Wei L: DL-3-n-butylphthalide induced neuroprotection, regenerative repair, functional recovery and psychological benefits following traumatic brain injury in mice. Neurochem Int. 111:82–92. 2017.PubMed/NCBI View Article : Google Scholar

18 

Zhu J, Zhang Y and Yang C: Protective effect of 3-n-butylphthalide against hypertensive nephropathy in spontaneously hypertensive rats. Mol Med Rep. 11:1448–1454. 2015.PubMed/NCBI View Article : Google Scholar

19 

Wang Y, Bi Y, Xia Z, Shi W, Li B, Li B, Chen L and Guo L: Butylphthalide ameliorates experimental autoimmune encephalomyelitis by suppressing PGAM5-induced necroptosis and inflammation in microglia. Biochem Biophys Res Commun. 497:80–86. 2018.PubMed/NCBI View Article : Google Scholar

20 

Chen J, Wang J, Zhang J and Pu C: Effect of butylphthalide intervention on experimental autoimmune myositis in guinea pigs. Exp Ther Med. 15:152–158. 2018.PubMed/NCBI View Article : Google Scholar

21 

Zhang P, Guo ZF, Xu YM, Li YS and Song JG: N-Butylphthalide (NBP) ameliorated cerebral ischemia reperfusion-induced brain injury via HGF-regulated TLR4/NF-κB signaling pathway. Biomed Pharmacother. 83:658–666. 2016.PubMed/NCBI View Article : Google Scholar

22 

Liu CY, Zhao ZH, Chen ZT, Che CH, Zou ZY, Wu XM, Chen SG, Li YX, Lin HB, Wei XF, et al: DL-3-n-butylphthalide protects endothelial cells against advanced glycation end product-induced injury by attenuating oxidative stress and inflammation responses. Exp Ther Med. 14:2241–2248. 2017.PubMed/NCBI View Article : Google Scholar

23 

Schapira AHV: Mitochondrial diseases. Lancet. 379:1825–1834. 2012.PubMed/NCBI View Article : Google Scholar

24 

Arun S, Liu L and Donmez G: Mitochondrial Biology and Neurological Diseases. Curr Neuropharmacol. 14:143–154. 2016.PubMed/NCBI View Article : Google Scholar

25 

Hybertson BM, Gao B, Bose SK and McCord JM: Oxidative stress in health and disease: The therapeutic potential of Nrf2 activation. Mol Aspects Med. 32:234–246. 2011.PubMed/NCBI View Article : Google Scholar

26 

Tan BL, Norhaizan ME, Liew WP and Sulaiman Rahman H: Antioxidant and Oxidative Stress: A Mutual Interplay in Age-Related Diseases. Front Pharmacol. 9(1162)2018.PubMed/NCBI View Article : Google Scholar

27 

Abdoulaye IA and Guo YJ: A Review of Recent Advances in Neuroprotective Potential of 3-N-Butylphthalide and Its Derivatives. BioMed Res Int. 2016(5012341)2016.PubMed/NCBI View Article : Google Scholar

28 

Yang XD, Cen ZD, Cheng HP, Shi K, Bai J, Xie F, Wu HW, Li BB and Luo W: L-3-n-Butylphthalide Protects HSPB8 K141N Mutation-Induced Oxidative Stress by Modulating the Mitochondrial Apoptotic and Nrf2 Pathways. Front Neurosci. 11(402)2017.PubMed/NCBI View Article : Google Scholar

29 

Liu Z, Wang H, Shi X, Li L, Zhou M, Ding H, Yang Y, Li X and Ding K: DL-3-n-Butylphthalide (NBP) Provides Neuroprotection in the Mice Models After Traumatic Brain Injury via Nrf2-ARE Signaling Pathway. Neurochem Res. 42:1375–1386. 2017.PubMed/NCBI View Article : Google Scholar

30 

Li Q, Cheng Y, Bi M, Lin H, Chen Y, Zou Y, Liu Y, Kang H and Guo Y: Effects of N-butylphthalide on the activation of Keap1/Nrf-2 signal pathway in rats after carbon monoxide poisoning. Environ Toxicol Pharmacol. 40:22–29. 2015.PubMed/NCBI View Article : Google Scholar

31 

Zhu BL, Xie CL, Hu NN, Zhu XB and Liu CF: Inhibiting of GRASP65 Phosphorylation by DL-3-N-Butylphthalide Protects against Cerebral Ischemia-Reperfusion Injury via ERK Signaling. Behav Neurol. 2018(5701719)2018.PubMed/NCBI View Article : Google Scholar

32 

Wang S, Huang L, Zhang Y and Peng Y, Wang X and Peng Y: Protective Effects of L-3-n-Butylphthalide Against H2O2-Induced Injury in Neural Stem Cells by Activation of PI3K/Akt and Mash1 Pathway. Neuroscience. 393:164–174. 2018.PubMed/NCBI View Article : Google Scholar

33 

Chen J, Wang J, Zhang J and Pu C: 3-n-Butylphthalide reduces the oxidative damage of muscles in an experimental autoimmune myositis animal model. Exp Ther Med. 14:2085–2093. 2017.PubMed/NCBI View Article : Google Scholar

34 

Tian X, He W, Yang R and Liu Y: Dl-3-n-butylphthalide protects the heart against ischemic injury and H9c2 cardiomyoblasts against oxidative stress: Involvement of mitochondrial function and biogenesis. J Biomed Sci. 24(38)2017.PubMed/NCBI View Article : Google Scholar

35 

Booth LA, Tavallai S, Hamed HA, Cruickshanks N and Dent P: The role of cell signalling in the crosstalk between autophagy and apoptosis. Cell Signal. 26:549–555. 2014.PubMed/NCBI View Article : Google Scholar

36 

Wang YG, Li Y, Wang CY, Ai JW, Dong XY, Huang HY, Feng ZY, Pan YM, Lin Y, Wang BX, et al: L-3-n-Butylphthalide protects rats' cardiomyocytes from ischaemia/reperfusion-induced apoptosis by affecting the mitochondrial apoptosis pathway. Acta Physiol (Oxf). 210:524–533. 2014.PubMed/NCBI View Article : Google Scholar

37 

Lei H, Zhao CY, Liu DM, Zhang Y, Li L, Wang XL and Peng Y: l-3-n-Butylphthalide attenuates β-amyloid-induced toxicity in neuroblastoma SH-SY5Y cells through regulating mitochondrion-mediated apoptosis and MAPK signaling. J Asian Nat Prod Res. 16:854–864. 2014.PubMed/NCBI View Article : Google Scholar

38 

Xu J, Huai Y, Meng N, Dong Y, Liu Z, Qi Q, Hu M, Fan M, Jin W and Lv P: L-3-n-Butylphthalide Activates Akt/mTOR Signaling, Inhibits Neuronal Apoptosis and Autophagy and Improves Cognitive Impairment in Mice with Repeated Cerebral Ischemia-Reperfusion Injury. Neurochem Res. 42:2968–2981. 2017.PubMed/NCBI View Article : Google Scholar

39 

Xiang J, Pan J, Chen F, Zheng L, Chen Y, Zhang S and Feng W: L-3-n-butylphthalide improves cognitive impairment of APP/PS1 mice by BDNF/TrkB/PI3K/AKT pathway. Int J Clin Exp Med. 7:1706–1713. 2014.PubMed/NCBI

40 

Iurlaro R and Muñoz-Pinedo C: Cell death induced by endoplasmic reticulum stress. FEBS J. 283:2640–2652. 2016.PubMed/NCBI View Article : Google Scholar

41 

Liao D, Xiang D, Dang R, Xu P, Wang J, Han W, Fu Y, Yao D, Cao L and Jiang P: Neuroprotective Effects of dl-3-n-Butylphthalide against Doxorubicin-Induced Neuroinflammation, Oxidative Stress, Endoplasmic Reticulum Stress, and Behavioral Changes. Oxid Med Cell Longev. 2018(9125601)2018.PubMed/NCBI View Article : Google Scholar

42 

Niu XL, Jiang X, Xu GD, Zheng GM, Tang ZP, Yin N, Li XQ, Yang YY and Lv PY: DL-3-n-butylphthalide alleviates vascular cognitive impairment by regulating endoplasmic reticulum stress and the Shh/Ptch1 signaling-pathway in rats. J Cell Physiol. 234:12604–12614. 2018.PubMed/NCBI View Article : Google Scholar

43 

Zheng B, Zhou Y, Zhang H, Yang G, Hong Z, Han D, Wang Q, He Z, Liu Y, Wu F, et al: Dl-3-n-butylphthalide prevents the disruption of blood-spinal cord barrier via inhibiting endoplasmic reticulum stress following spinal cord injury. Int J Biol Sci. 13:1520–1531. 2017.PubMed/NCBI View Article : Google Scholar

44 

He Z, Zhou Y, Huang Y, Wang Q, Zheng B, Zhang H, Li J, Liu Y, Wu F, Zhang X, et al: Dl-3-n-butylphthalide improves functional recovery in rats with spinal cord injury by inhibiting endoplasmic reticulum stress-induced apoptosis. Am J Transl Res. 9:1075–1087. 2017.PubMed/NCBI

45 

Nonaka T, Masuda-Suzukake M and Hasegawa M: Molecular mechanisms of the co-deposition of multiple pathological proteins in neurodegenerative diseases. Neuropathology. 38:64–71. 2018.PubMed/NCBI View Article : Google Scholar

46 

Goedert M: NEURODEGENERATION. Alzheimer's and Parkinson's diseases: The prion concept in relation to assembled Aβ, tau, and α-synuclein. Science. 349(1255555)2015.PubMed/NCBI View Article : Google Scholar

47 

Peng Y, Sun J, Hon S, Nylander AN, Xia W, Feng Y, Wang X and Lemere CA: L-3-n-butylphthalide improves cognitive impairment and reduces amyloid-beta in a transgenic model of Alzheimer's disease. J Neurosci. 30:8180–8189. 2010.PubMed/NCBI View Article : Google Scholar

48 

Peng Y, Hu Y, Xu S, Li P, Li J, Lu L, Yang H, Feng N, Wang L and Wang X: L-3-n-butylphthalide reduces tau phosphorylation and improves cognitive deficits in AβPP/PS1-Alzheimer's transgenic mice. J Alzheimers Dis. 29:379–391. 2012.PubMed/NCBI View Article : Google Scholar

49 

Huang JZ, Chen YZ, Su M, Zheng HF, Yang YP, Chen J and Liu CF: dl-3-n-Butylphthalide prevents oxidative damage and reduces mitochondrial dysfunction in an MPP(+)-induced cellular model of Parkinson's disease. Neurosci Lett. 475:89–94. 2010.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2019
Volume 11 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luo R, Wangqin R, Zhu L and Bi W: Neuroprotective mechanisms of 3‑n‑butylphthalide in neurodegenerative diseases (Review). Biomed Rep 11: 235-240, 2019
APA
Luo, R., Wangqin, R., Zhu, L., & Bi, W. (2019). Neuroprotective mechanisms of 3‑n‑butylphthalide in neurodegenerative diseases (Review). Biomedical Reports, 11, 235-240. https://doi.org/10.3892/br.2019.1246
MLA
Luo, R., Wangqin, R., Zhu, L., Bi, W."Neuroprotective mechanisms of 3‑n‑butylphthalide in neurodegenerative diseases (Review)". Biomedical Reports 11.6 (2019): 235-240.
Chicago
Luo, R., Wangqin, R., Zhu, L., Bi, W."Neuroprotective mechanisms of 3‑n‑butylphthalide in neurodegenerative diseases (Review)". Biomedical Reports 11, no. 6 (2019): 235-240. https://doi.org/10.3892/br.2019.1246