Table 1.
Effects of celastrol on CNS cells and diseases | In vitro or in vivo | Models | Mechanisms of action of celastrol | Refs. |
---|---|---|---|---|
Microglia activation | Microglia cell line MG6 | dsRNA [poly(I:C)]-induced activation | preventing F-actin rearrangement, preventing cytoskeletal alteration, attenuating the expression of proinflammatory cytokines and chemokines | 10 |
BV-2 microglia cells | LPS-stimulated activation | inhibiting LPS-induced phosphorylation of MAPK/ERK1/2 and NF-κB activation | 11 | |
Female SD rats | SCI model | inhibiting the activation of microglia and microglia pyroptosis, down-regulating the release of pro-inflammatory cytokines and up-regulating the expression of anti-inflammatory cytokine and reducing the expression of NLRP3 inflammasome by inhibiting the expression of NF-κB/p-p65 | 12 | |
BV-2 microglia cells | LPS/ATP induced microgliosis | |||
Inflammatory responses of astrocytes | CRT-MG human astroglial cells | HIV-1 Tat (trans-activator of transcription)-induced inflammatory responses | inhibiting JNK, AP-1 and NF-κB activation and inducing expression of HO-1 | 16 |
Poly (I:C) activated neuro-inflammation | suppressing ICAM-1/VCAM-1, chemokines expression and activation of JNK-STAT1 and NF-κB signaling pathways | 17 | ||
Neuronal apoptosis and neuro-inflammation | PC12, SH-SY5Y cells and primary neurons | Cells were treated with Cd (10 μM and/or 20 μM) for 24 h | (1) inactivating JNK and Akt/mTOR signaling pathway and elevating PTEN activity; (2) inhibiting CaMKII-dependent Akt/mTOR pathway; (3) suppressing mitochondrial ROS-dependent AMPK-mTOR signaling pathway; (4) targeting NOX2-derived ROS-dependent PP5-JNK signaling pathway. | 22-25 |
BBB dysfunction | Murine brain endothelial bEnd3 cells | OGD model | inducing activation of MAPKs and PI3K/Akt/mTOR pathways | 30 |
AD | Male SD rats | i.p. injection of STZ, and inhale 3% sevoflurane for 2 h | All these dementias like pathology were reversed after celastrol treatment. | 37 |
Male SD rats | Aβ25-35-induced rat model of AD | Celastrol attenuated hippocampal inflammation, improved synaptic function, and maintained hippocampal energy metabolism. | 38 | |
CHO cell line | A CHO cell line overexpressing Aβ | Celastrol inhibited Aβ1-40 and Aβ1-42 production by reducing the β-cleavage of APP, and reduced BACE-1 expression by preventing NF-κB activation. | 18 | |
Transgenic mouse model of AD | A transgenic mouse model of AD overexpressing the human APP695sw mutation and the presenilin-1 mutation M146L (Tg PS1/APPsw) | Celastrol reduced the levels of Aβ, decreased the microgliosis in the cortex, and reduced the levels of both soluble and insoluble Aβ1-38, Aβ1-40 and Aβ1-42. | ||
SH-SY5Y cells | Tau hyperphosphorylation induced by Aβ1-42 | Aβ1-42 induced Tau hyperphosphorylation and HSP90 expression were inhibited by celastrol | 39 | |
SH-SY5Y cells, C57BL/6J and APP23 mice primary hippocampal neurons | None | In addition to increased expression of HSP40, HSP70 and HSP90, celastrol induced activation of HSF1 and promoted the TTR transcription in SH-SY5Y cells. | 40 | |
C57BL/6J, transgenic mouse model of AD | APP23 AD model mice, APP23/Ttr-/-(APP23 mice on Ttr knock-out background) mouse strains | |||
H4 human neuroglioma cells stably transfected to overexpress human full length APP | LPS induced neuroinflammation | Celastrol increased HSP-70 and Bcl-2 expression, decreased NF-κB, COX-2, phosphorylated GSK-3β expression and ROS production. | 43 | |
PD | Drosophila | A Drosophila DJ-1A RNAi model of PD | Celastrol prevented the loss of DN and restored dopamine content to near normal levels. | 45 |
Swiss Webster mice | Dopaminergic neurotoxin MPTP-induced PD model | Celastrol attenuated the loss of dopaminergic neuron in the SNpc and reduced depletion of striatal dopamine levels, increased HSP70 expression to attenuate inflammation by preventing TNF-α and NF-κB activation. | 19 | |
Male C57BL/6 mice and genetically modified mice (Nrf2-KO, NLRP3-KO and Caspase-1-KO) | MPTP-induced PD mouse model and AAV-mediated human α-synuclein overexpression PD model | Celastrol relieved motor deficits and nigrostriatal dopaminergic degeneration through Nrf2-NLRP3-caspase-1 pathway. | 46 | |
SH-SY5Y cells | Rotenone-induced PD model | Celastrol suppressed oxidative stress, provided antiapoptotic effects to maintain the mitochondrial function and induced autophagy to clear damaged mitochondria. | 47 | |
SH-SY5Y cells | SH-SY5Y cells were treated with 1 mM MPP+ for 24 h to induced about 50% neuronal death. | Celastrol treatment suppressed MPP+-induced injuries by activating autophagy through MAPK/p38, MAPK/ERK, MAPK/Akt, or MAPK/JNK signaling pathways. | 26 | |
Male C57BL/6 mice | Mice received i.p. injections of MPTP (10 mg/kg/day for 3 days) 24 h after the last celastrol injection | Celastrol increased Bcl-2 expression in the substantia nigra by enhancing mitophagy to clear impaired mitochondria and further inhibiting dopaminergic neuronal apoptosis | ||
MS | Female C57BL/6 mice | EAE animal model | Celastrol suppressed pathogenic Th17 polarization in the peripheral lymph nodes, downregulated cytokine production in BMDCs and inhibited T cells infiltration into the CNS and Th17 cell responses in the CNS. | 51 |
Male SD rats | EAE animal model | Celastrol attenuated demyelination and inflammatory infiltration in spinal cord. Celastrol also attenuated optic neuritis by inhibiting cytokines and microgliosis production, expression of iNOS and activation of NF-κB in optic nerve, and attenuating ganglion cells apoptosis in the retina of EAE rats. | 52 | |
Male C57BL/6 mice | EAE animal model | Celastrol modulated MAPK (p38, ERK) to regulate the downstream genes encoding SGK1, so as to restore the Th17/Treg balance and enhance BDNF expression in T cells, and lead to protection against EAE. | 53 | |
Female SD rats | EAE animal model | Celastrol transformed cytokines profile from Th1 to Th2 pattern, with decreasing TNF-α and increasing IL-10 correspondingly. Celastrol also decreased NF-κB expression, nitrites levels, and immune-histochemical expression of TLR2 and CD3+ T-lymphocytic count. | 54 | |
ALS | Transgenic ALS mice | G93A transgenic familial ALS mice (high expresser line) | Celastrol inhibited proinflammatory cytokine and glial activation through reducing TNF-α, iNOS, CD40, GFAP and increasing HSP70 immunoreactivity in lumbar spinal cord neurons. | 56 |
Primary motoneuron cultures | Cellular stress, such as staurosporin and H2O2, to induce apoptosis and oxidative stress | Celastrol did not appear any neuroprotective effect and exhibited neurotoxic. | 57, 58 | |
Polyglutamine expansion diseases | HD Male Lewis rats | Succinate dehydrogenase inhibitor 3-NP-induced HD | Celastrol reduced neurotoxicity by decreasing the striatal lesion volumes, inducing HSP70 in the striata, and reducing astrogliosis. | 19 |
Polyglutamine aggregation and toxicity HeLa cells, PC12 cells, HSF1+/+ and HSF1-/- mouse embryo fibroblast (MEF) cells | Polyglutamine aggregation and toxicity is transfection of a Q57-YFP fusion protein into cell lines | Celastrol effectively decreased the aggregation and toxicity of polyglutamine expression in vitro via stimulating HSF1 activity to lead to inducible HSP70 gene expression pathway. | 60, 61 | |
SCA14 | SH-SY5Y, CHO, and COS-7 cells, primary cultured Purkinje cells |
Adenovirus infection | Celastrol induced upregulation of HSP70 and HSP40 to synergistic diminish aggregation formation of mutant PKCγ and cells death. Celastrol activated autophagy also benefited for clearing the PKCγ aggregates. | 63 |
Male C57BL/6N mice | Pharmacological induction of HSPs | Celastrol treatment upregulated HSP70 by penetrating the mouse cerebellum. | ||
Stroke | Male SD rats | pMCAO model | Celastrol downregulated the expression of p-JNK, p-c-Jun and NF-κB. | 67 |
AIS patients | Clinical samples | Celastrol treatment increased IL-33 and IL-10 expression, and decreased IL-1β, IL-6, and TNF-α level in vitro and in vivo. The neuroprotective and anti-inflammatory effects of celastrol for ischemic stroke were derived from promoting growth ST2/IL-33 activation in microglia. | 13 | |
Male SD rats | pMCAO model | |||
Primary rats neurons and microglia | OGD model | |||
Primary microglia-enriched cultures | Microglial polarization: microglia were transfected with a ST2 interference vector before pretreatment with OGD for 3 h, then treated with 50 ng/mL IL-33 | |||
Primary rats neuronal | Neurons underwent OGD for 3 h after which they were treated with different concentrations of IL-33 | |||
Male SD rats | Transient global cerebral ischemia reperfusion | Celastrol inhibited HMGB1/NF-κB signaling pathway. | 69 | |
Primary rats neuronal | OGD model | Celastrol directly bound to HMGB1 to inactivate it, up-regulated HSP70 and down-regulated NF-κB expression to play neuroprotective effect in cerebral ischemia reperfusion injury in vitro and in vivo. | 27 | |
Male SD rats | MCAO model | |||
Male C57BL/6 mice | MCAO model | Celastrol exhibited neuroprotection and anti-apoptosis effects partially by modulating lipid metabolites. | 70 | |
Hippocampal cell line (HT-22) cells | OGD model | Celastrol significantly attenuated I/R-induced hippocampal injury by inhibiting the AK005401/MAP3K12 signaling and activating the PI3K/Akt pathway. | 71 | |
Male C57BL/6 mice | Bilateral common carotid clip reperfusion | |||
Male SD rats | SAH endovascular perforation model | Celastrol attenuated brain swelling and protected BBB integrity after rats SAH by decreasing MMP-9 expression and attenuating pro-inflammatory cytokines expression. | 31 | |
TBI | hsp110-deficient mice, hsp70.1 and hsp70.3 (named hsp70i)-deficient mouse lines with C57BL/6 genetic background | Controlled Cortical Impact (CCI) | By increasing the levels of HSP70/HSP110, celastrol treatment in wild-type mice exhibited lower levels of brain injury, decreased cellular apoptosis, inflammatory cells infiltration and gliosis, and increased Ki-67-positive cells and improved behavior. | 74 |