Skip to main content
. 2022 Oct 19;13:1024955. doi: 10.3389/fphar.2022.1024955

TABLE 1.

Effects of triptolide and celastrol on neurological diseases.

Disease Component In vivo/In vitro Effects References
Alzheimer’s disease Triptolide in vivo; Aβ 1-40-induced AD rats attenuates the degeneration of dendritic spines in hippocampal neurons Wan et al. (2014)
in vivo; APP/PS1 double transgenic AD murine model improves cognitive function, accompanied by reduced neuroinflammation and Aβ deposition Cheng et al. (2014)
in vivo; APP/PS1 double transgenic AD murine model reduces astrocyte proliferation and microglia activation in the hippocampal region Li et al. (2016)
in vivo; APP/PS1 double transgenic AD murine model improves spatial memory deficits by inhibiting of BACE1 Wang et al. (2014)
in vivo; APP/PS1 double transgenic AD murine model improves spatial memory deficits by inhibiting inflammatory responses and MAPKs activity Cui et al. (2016)
in vitro; SH-SY5Y cell lines exerts neuroprotective effects by inhibiting CXCR2 activity and reducing Aβ production Wang et al. (2013b)
in vitro; Aβ 1-42-treated cultured rat microglia exerts protective effects by inhibiting TNF-α and IL-1β expression levels Jiao et al. (2008)
in vitro; glutamate-stimulated PC12 cells inhibits the level of ROS, attenuating apoptosis He et al., 2003; Gu et al., 2004
in vitro; Aβ 25-35-treated differentiated PC12 cells promotes autophagy and inhibits oxidative stress, exerting neuroprotective effects Xu et al., 2015; Xu et al., 2016
in vitro; Aβ 1-42-treated differentiated PC12 cells nanoparticles loaded with triptolide reduces oxidative stress and inhibits cytotoxicity Jia et al. (2021)
in vitro; primary astrocytes from rats increases synthesis and release of nerve growth factor Xue et al. (2007)
in vitro; hippocampal neurons increases the expression of synaptophysin Nie et al. (2012)
in vivo; APP/PS1 double transgenic AD murine model increased hippocampal neuroligin-1 expression through epigenetic mechanisms Lu et al. (2019)
Celastrol in vivo; LPS rat model improves performance in memory and learning activity tests Allison et al. (2001)
in vivo; double transgenic Tg PS1/APPsw AD mice attenuates the accumulation of pathological plaque and microglial activation Paris et al. (2010)
in vivo; diabetes mellitus rat model improves cognitive function and decreases amyloid substance Liao et al. (2018)
in vivo; Aβ 25-35-microinjected rats improves learning and memory deficits by inhibiting NF-kB activity, improving synaptic function and increasing glucose metabolism Xiao et al. (2021)
In vivo and vitro; P301S microtubule associated protein tau mice and 3XTg mice, N2a cells activates transcription factor EB-mediated autophagy and lysosome biogenesis, reduce the accumulation of neurofibrillary tangles, thereby attenuating disease severity Yang et al. (2022b)
in vitro; human monocytes and macrophages, microglia, endothelial cells inhibits the production of TNF-α and IL-1β by human monocytes and macrophages, the expression of MHC II molecules of microglia, and the production of inducible nitric oxide in endothelial cells Allison et al. (2001)
in vitro; HEK293 cells attenuates NF-kB activity Paris et al. (2010)
in vitro; 7 W CHO cells overexpressing wild-type human APP inhibits amyloid-β production by inhibiting BACE-1 Paris et al. (2010)
in vitro; IMR-32 cells exerts neuroprotective effects by inhibiting IKK Veerappan et al. (2017)
in vitro; LPS-treated H4-APP cells celastrol inhibits the production of Aβ, attenuates NF-kB activity and suppresses COX-2 expression. In addition, celastrol increases the expression of Hsp70 and Bcl-2 Zhao et al. (2014)
in vitro; Aβ 1-42-treated SH-SY5Y cells no effect on the expression of Hsp70, while inhibits the expression of Hsp90 Cao et al. (2018)
Parkinson’s disease Triptolide in vivo; LPS rat model exerting neuroprotective effects by protecting dopaminergic neurons and reducing the expression of pro-inflammatory cytokines (TNF-α and IL-1β) Zhou et al. (2005)
in vivo; LPS rat model protecting dopaminergic neurons and inhibiting microglia activation Li et al. (2006b)
in vivo; MPP + -induced rat model improving behavioral performance by protecting dopaminergic neurons and inhibiting microglial activation Gao et al. (2008)
in vitro; LPS-induced primary mesencephalic neuron/glia mixed culture decreases [3H]dopamine uptake and loss of tyrosine hydroxylase-immunoreactive neurons, inhibits microglial activation, and attenuates TNF-α and NO production Li et al. (2004)
in vitro; MPP + -induced primary mesencephalic neurons tripchlorolide; promotes axonal elongation and protects dopaminergic neurons, as well as increases BDNF mRNA expression Li et al. (2003)
in vivo; partially lesioned PD rat model tripchlorolide; protects dopaminergic neurons and inhibits the overproduction of TNF-α and IL-2 Cheng et al. (2002)
in vivo; MPTP-induced PD mouse model tripchlorolide; improves behavioral performance, protects dopaminergic neurons and inhibits astroglial responses Hong et al. (2007)
in vitro; preformed fibrils of human wild-type α-synuclein-induced mouse primary microglia inhibits microglial activation by suppressing NF-κB activity via targeting the miR155–5p/SHIP1 pathway Feng et al. (2019)
in vivo and vitro; LPS-induced PD model inhibits microglial activation by upregulating metabotropic glutamate receptor 5 Huang et al. (2018)
in vitro; MN9D cell line enhances autophagy in neuronal cells, promoting the clearance of α-synuclein Hu et al. (2017)
Celastrol in vivo; Drosophila DJ-1A PD model inhibits the reduction of dopaminergic neurons and increases brain dopamine content Faust et al. (2009)
in vivo; MPTP-induced mouse PD model attenuates the loss of dopaminergic neurons, increases Hsp70 within dopaminergic neurons, and decreases the levels of NF-kB and TNF-α Cleren et al. (2005)
in vivo; MPTP-induced mouse PD model exerts neuroprotective effects by promoting mitophagy Lin et al. (2019)
in vivo; AAV-mediated human α-synuclein overexpression PD model and the MPTP-induced PD mouse model improve motor deficits by modulating the Nrf2-NLRP3-caspase-1 pathway Zhang et al. (2021)
in vivo and vitro; lactacystin-induced Wistar rats, SH-SY5Y cells and mouse primary cortical neurons no neuroprotective effects Konieczny et al. (2014)
in vitro; rotenone-induced SH-SY5Y PD model exerts neuroprotection by inducing autophagy, preserving mitochondrial function and inhibiting p38 MAPK Deng et al., 2013; Choi et al., 2014
in vitro; dendritic cells mediates antigen trafficking in DCs, thus attenuating α-synuclein-specific T cell responses Ng et al. (2022)
Multiple sclerosis Triptolide in vivo; C57 BL/6 mouse EAE model delays the onset of EAE, attenuates the degree of inflammation and demyelination, improves behavirol deficits, and inhibits NF-kB-DNA binding activity Wang et al. (2008)
in vivo; C57 BL/6 mouse EAE model LLDT-8; suppresses the severity of EAE by inhibiting T-cell activation Fu et al. (2006)
in vivo; SJL/J mouse EAE model increases expression levels of Hsp70 and stabilisation of the NF-kB/IkBα complex Kizelsztein et al. (2009)
in vivo; cuprizone-induced toxic model improves behavioral deficits and attenuates neuroinflammation by inhibiting NF-kB activation and promoting intrinsic myelin repair Sanadgol et al. (2018)
Celastrol in vivo; relapsing-remitting EAE rat model inhibits relapses and reduces clinical scores by modulating the Th1/Th2 cytokines profile (increases IL-10 expression but reduces TNF-α expression), inhibiting NF-κB and TLR2 expression, and reducing CD3+ T lymphocytic count O'Brien et al., 2001; Abdin and Hasby, (2014)
in vivo; EAE mouse model exerts neuroprotective effects by inhibiting Th17 cell responses and attenuating cytokine production Wang et al. (2015)
in vivo; EAE mouse model affects T-cell responses through the MAPK pathway, inhibiting SGK1 expression and incresing BDNF expression Venkatesha and Moudgil, (2019)
in vivo; EAE rat model inhibiting the expression of iNOS and NF-kB and attenuating MS and optic neuritis Yang et al. (2017)
Huntington’s diseases Celastrol in vivo; 3-nitropropionic acid-induced HD rat models decreases striatal lesion voulme, increases the expression of Hsp70 in the striata, and attenuates astrogliosis Cleren et al. (2005)
in vitro; cell lines expressing mutant inhibits polyglutamine aggregation by inducing HSF1 and increasing the expression of Hsp70 Zhang and Sarge, (2007)
polyglutamine protein
in vitro; HdhQ111/Q111 knock-in mouse-derived striatal cell line inhibits mutant huntingtin aggregation, and reverses the abnormal cellular localization of full-length mutant huntingtin Wang et al. (2005)
Amyotrophic lateral sclerosis Celastrol in vivo; G93A SOD1 transgenic ALS mouse model delays disease onset, improves motor deficits, increases the number of neurons, promotes Hsp70 expression, and reduces TNF-α and iNOS levels Kiaei et al. (2005)
in vitro; staurosporin or H2O2-induced primary motoneurons activates the heat shock response (i.e. increases Hsp70 expression) Kalmar and Greensmith, (2009)
in vitro; H2O2-treated G93A SOD1 transfected NSC34 cells reduces cell death by activating MEK/ERK and PI3K/AKT signaling pathways Li et al. (2017)
Cerebral ischemia Triptolide in vivo; focal cerebral ischemia reperfusion rat model improves neural function, attenuates neuronal apoptosis, and suppresses infiltration of neutrophils Wei et al. (2004)
in vivo; focal cerebral ischemia reperfusion rat model exerts neuroprotection by inhibiting NF-kB activity Jin et al., 2015; Bai et al., 2016a; Bai et al., 2016b
in vivo; focal cerebral ischemia reperfusion rat model exerts neuroprotective effects by inhibiting NF-kB/PUMA signaling pathway Zhang et al. (2016)
in vivo and vitro; focal cerebral ischemia reperfusion rat model, and OGD and TNF-α-stimulated SH-SY5Y cells inhibits NF-kB and p38 MAPK signaling pathways, exerting neuroprotection Hao et al. (2015)
in vivo; focal cerebral ischemia reperfusion rat model upregulates autophagy and downregulates apoptosis Yang et al. (2015)
in vivo; focal cerebral ischemia reperfusion rat model downregulating apoptosis by activating the PI3K/AKT/mTOR signaling pathway Li et al. (2015)
in vivo; focal cerebral ischemic mouse model improves cerebral ischemia by triggering BDNF-AKT signaling pathway and autophagy Du et al. (2020)
in vivo; focal cerebral ischemia reperfusion rat model improves neurobehavioral scores, reduces brain damage, reduces levels of malondialdehyde and ROS, increases superoxide dismutase level, involving inhibition of Wnt/β-catenin signaling pathway Pan and Xu, (2020)
in vivo and vitro; chronic cerebral hypoperfusion mouse model, and OGD-stimulated primary oligodendrocytes and BV2 cells alleviates white matter injury, protects against oligodendrocyte apoptosis directly, and inhibits microglial inflammation indirectly, involving increase of phosphorylation of the Src/AKT/GSK 3β singnaling pathway Wan et al. (2022)
Celastrol in vivo; permanent middle cerebral artery occlusion mouse model improves neurological function and reduces infarct volume in by attenuating the expression of NF-kB, p-c-Jun, and p-JNK Li et al. (2012)
in vivo; transient global cerebral ischemia reperfusion rat model exerts neuroprotection, inhibits the expression of pro-inflammatory cytokines and MDA and elevates the levels of GSH and SOD, which is mediated by inhibiting HMGB1/NF-kB signaling pathway Zhang et al. (2020)
in vivo and vitro; focal cerebral ischemia reperfusion rat model and OGD-stimulated primary rat cortical neuron directly binds to HMGB1, thus inhibiting the binding of HMGB1 to its downstream inflammatory components; inhibits NF-kB activity Liu M et al. (2021)
in vivo and vitro; permanent focal ischemia rat model and OGD-stimulated rimary neurons and microglia exerts neuroprotective effects through an IL-33/ST2 axis-mediated M2 microglia/macrophage polarization Jiang et al. (2018)
in vivo; focal cerebral ischemia reperfusion mouse model attenuates glycolysis and exerts neuroprotection by inhibiting HIF-1α/PDK1 Chen et al. (2022)
in vivo and vitro; cerebral ischemia reperfusion mouse model and OGD-stimulated HT-22 cells inhibits AK005401/MAP3K12 and activates PI3K/AKT signaling pathway, thus exerting neuroprotective effects Wang et al. (2021)
Traumatic brain injury Triptolide in vivo; TBI rat model improves neurological deficits and attenuates contusion volume, edema, cell apoptosis, decreases expressions of pro-inflammatory cytokines while increases level of anti-inflammatory cytokines Lee et al. (2012)
Celastrol in vivo; TBI mouse model improves neurobehavioral functions and protects neuronal cells by inducing Hsp70/Hsp110 expression Eroglu et al. (2014)
Spinal cord injury Triptolide in vivo and vitro; SCI rat model and LPS-stimulated primary astrocytes promotes spinal cord repair, inhibits inflammation, and attenuates astrogliosis and glial scar by inhibiting the JAK2/STAT3 pathway Su et al. (2010)
in vivo; SCI rat model exerts neuroprotection by targeting the miR-96/IKKβ/NF-κB pathway and thus inhibiting microglial activation Huang et al. (2019)
in vivo; SCI mouse model enhances autophagy and inhibits MAPK/ERK1/2 signaling pathway Zhu et al. (2020)
Celastrol in vitro; SCI spinal cords model reduces motorneuron death by inducing Hsp70 expression, while exerts limited protection on the lumbar motor network Petrović et al. (2019)
in vivo and vitro; SCI rat model and LPS + ATP-induced BV2 cells attenuates microglial activation in the spinal cord, inhibits the expression of NF-kB, thus inhibiting the expression of NLRP3, caspase-1, GSDMD and inflammatory cytokines, while increases the levels of anti-inflammatory cytokines Dai et al. (2019)
Epilepsy Triptolide in vivo; kainic acid-induced epilepsy rat model protects neurons, which is associated with increased expression of neuron kv1.1 in the CA3 region of the hippocampus Pan et al. (2012)
in vitro; kainic acid-stimulated BV2 microglia inhibits microglial activation, decreases MHC II expression in microglia by inhibiting AP-1/class II transactivator, which is related to neuronal death Sun et al. (2018)
Celastrol in vivo; multiple-hit rat model has a therapeutic effect by inhibiting NF-kB Schiavone et al. (2021)
in vivo and vitro; kainic acid-induced rats and hippocampal slices inhibits NOX activation and rapid H2O2 release, thus alleviating epileptic seizure Malkov et al. (2019)
in vivo; mouse amygdala-kindling model increases microglial activation in hippocampal CA1 and CA3 regions and reduces postkindling seizure thresholds von Rüden et al. (2019)