Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 807
Filter
1.
Eur J Pharmacol ; 974: 176633, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38703975

ABSTRACT

Cardiac arrest (CA) remains a leading cause of death, with suboptimal survival rates despite efforts involving cardiopulmonary resuscitation and advanced life-support technology. Post-resuscitation myocardial dysfunction (PRMD) is an important determinant of patient outcomes. Myocardial ischemia/reperfusion injury underlies this dysfunction. Previous reports have shown that ruthenium red (RR) has a protective effect against cardiac ischemia-reperfusion injury; however, its precise mechanism of action in PRMD remains unclear. This study investigated the effects of RR on PRMD and analyzed its underlying mechanisms. Ventricular fibrillation was induced in rats, which were then subjected to cardiopulmonary resuscitation to establish an experimental CA model. At the onset of return of spontaneous circulation, RR (2.5 mg/kg) was administered intraperitoneally. Our study showed that RR improved myocardial function and reduced the production of oxidative stress markers such as malondialdehyde (MDA), glutathione peroxidase (GSSG), and reactive oxygen species (ROS) production. RR also helped maintain mitochondrial structure and increased ATP and GTP levels. Additionally, RR effectively attenuated myocardial apoptosis. Furthermore, we observed downregulation of proteins closely related to mitophagy, including ubiquitin-specific protease 33 (USP33) and P62, whereas LC3B (microtubule-associated protein light chain 3B) was upregulated. The upregulation of mitophagy may play a critical role in reducing myocardial injury. These results demonstrate that RR may attenuate PRMD by promoting mitophagy through the inhibition of USP33. These effects are likely mediated through diverse mechanisms, including antioxidant activity, apoptosis suppression, and preservation of mitochondrial integrity and energy metabolism. Consequently, RR has emerged as a promising therapeutic approach for addressing post-resuscitation myocardial dysfunction.


Subject(s)
Disease Models, Animal , Heart Arrest , Mitophagy , Rats, Sprague-Dawley , Ruthenium Red , Animals , Mitophagy/drug effects , Heart Arrest/complications , Heart Arrest/drug therapy , Heart Arrest/metabolism , Heart Arrest/physiopathology , Rats , Male , Ruthenium Red/pharmacology , Ruthenium Red/therapeutic use , Oxidative Stress/drug effects , Ubiquitin Thiolesterase/metabolism , Cardiopulmonary Resuscitation , Up-Regulation/drug effects , Myocardium/pathology , Myocardium/metabolism , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology
2.
Mol Med ; 30(1): 58, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720283

ABSTRACT

BACKGROUND: Vascular calcification (VC) is a complication in diabetes mellitus (DM) patients. Osteogenic phenotype switching of vascular smooth muscle cells (VSMCs) plays a critical role in diabetes-related VC. Mitophagy can inhibit phenotype switching in VSMCs. This study aimed to investigate the role of the glucagon-like peptide-1 receptor (GLP-1R) agonist exendin 4 (EX4) in mitophagy-induced phenotype switching. MATERIALS AND METHODS: The status of VC in T2DM mice was monitored using Von Kossa and Alizarin Red S (ARS) staining in mouse aortic tissue. Human aortic smooth muscle cells were cultured in high glucose (HG) and ß-glycerophosphate (ß-GP) conditioned medium. Accumulation of LC3B and p62 was detected in the mitochondrial fraction. The effect of EX4 in vitro and in vivo was investigated by knocking down AMPKα1. RESULTS: In diabetic VC mice, EX4 decreased the percentage of von Kossa/ARS positive area. EX4 inhibited osteogenic differentiation of HG/ß-GP-induced VSMCs. In HG/ß-GP-induced VSMCs, the number of mitophagosomes was increased, whereas the addition of EX4 restored mitochondrial function, increased the number of mitophagosome-lysosome fusions, and reduced p62 in mitochondrial frictions. EX4 increased the phosphorylation of AMPKα (Thr172) and ULK1 (Ser555) in HG/ß-GP-induced VSMCs. After knockdown of AMPKα1, ULK1 could not be activated by EX4. The accumulation of LC3B and p62 could not be reduced after AMPKα1 knockdown. Knockdown of AMPKα1 negated the therapeutic effects of EX4 on VC of diabetic mice. CONCLUSION: EX4 could promote mitophagy by activating the AMPK signaling pathway, attenuate insufficient mitophagy, and thus inhibit the osteogenic phenotype switching of VSMCs.


Subject(s)
AMP-Activated Protein Kinases , Exenatide , Glucagon-Like Peptide-1 Receptor , Mitophagy , Signal Transduction , Vascular Calcification , Animals , Mitophagy/drug effects , Vascular Calcification/etiology , Vascular Calcification/metabolism , Vascular Calcification/drug therapy , Signal Transduction/drug effects , Mice , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Male , AMP-Activated Protein Kinases/metabolism , Humans , Exenatide/pharmacology , Exenatide/therapeutic use , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Mice, Inbred C57BL
3.
Cell Commun Signal ; 22(1): 269, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745240

ABSTRACT

BACKGROUND: The pathway involving PTEN-induced putative kinase 1 (PINK1) and PARKIN plays a crucial role in mitophagy, a process activated by artesunate (ART). We propose that patients with anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis exhibit insufficient mitophagy, and ART enhances mitophagy via the PINK1/PARKIN pathway, thereby providing neuroprotection. METHODS: Adult female mice aged 8-10 weeks were selected to create a passive transfer model of anti-NMDAR encephalitis. We conducted behavioral tests on these mice within a set timeframe. Techniques such as immunohistochemistry, immunofluorescence, and western blotting were employed to assess markers including PINK1, PARKIN, LC3B, p62, caspase3, and cleaved caspase3. The TUNEL assay was utilized to detect neuronal apoptosis, while transmission electron microscopy (TEM) was used to examine mitochondrial autophagosomes. Primary hippocampal neurons were cultured, treated, and then analyzed through immunofluorescence for mtDNA, mtROS, TMRM. RESULTS: In comparison to the control group, mitophagy levels in the experimental group were not significantly altered, yet there was a notable increase in apoptotic neurons. Furthermore, markers indicative of mitochondrial leakage and damage were found to be elevated in the experimental group compared to the control group, but these markers showed improvement following ART treatment. ART was effective in activating the PINK1/PARKIN pathway, enhancing mitophagy, and diminishing neuronal apoptosis. Behavioral assessments revealed that ART ameliorated symptoms in mice with anti-NMDAR encephalitis in the passive transfer model (PTM). The knockdown of PINK1 led to a reduction in mitophagy levels, and subsequent ART intervention did not alleviate symptoms in the anti-NMDAR encephalitis PTM mice, indicating that ART's therapeutic efficacy is mediated through the activation of the PINK1/PARKIN pathway. CONCLUSIONS: At the onset of anti-NMDAR encephalitis, mitochondrial damage is observed; however, this damage is mitigated by the activation of mitophagy via the PINK1/PARKIN pathway. This regulatory feedback mechanism facilitates the removal of damaged mitochondria, prevents neuronal apoptosis, and consequently safeguards neural tissue. ART activates the PINK1/PARKIN pathway to enhance mitophagy, thereby exerting neuroprotective effects and may achieve therapeutic goals in treating anti-NMDAR encephalitis.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis , Artesunate , Disease Models, Animal , Neuroprotective Agents , Protein Kinases , Animals , Artesunate/pharmacology , Artesunate/therapeutic use , Mice , Female , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/pathology , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/drug therapy , Protein Kinases/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Microscopy, Electron, Transmission , Mitophagy/drug effects , Apoptosis/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Hippocampus/pathology , Hippocampus/drug effects , Hippocampus/metabolism
4.
Life Sci Alliance ; 7(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38697845

ABSTRACT

Defective mitophagy in renal tubular epithelial cells is one of the main drivers of renal fibrosis in diabetic kidney disease. Our gene sequencing data showed the expression of PINK1 and BNIP3, two key molecules of mitophagy, was decreased in renal tissues of VDR-knockout mice. Herein, streptozotocin (STZ) was used to induce renal interstitial fibrosis in mice. VDR deficiency exacerbated STZ-induced renal impairment and defective mitophagy. Paricalcitol (pari, a VDR agonist) and the tubular epithelial cell-specific overexpression of VDR restored the expression of PINK1 and BNIP3 in the renal cortex and attenuated STZ-induced kidney fibrosis and mitochondrial dysfunction. In HK-2 cells under high glucose conditions, an increased level of α-SMA, COL1, and FN and a decreased expression of PINK1 and BNIP3 with severe mitochondrial damage were observed, and these alterations could be largely reversed by pari treatment. ChIP-qPCR and luciferase reporter assays showed VDR could positively regulate the transcription of Pink1 and Bnip3 genes. These findings reveal that VDR could restore mitophagy defects and attenuate STZ-induced fibrosis in diabetic mice through regulation of PINK1 and BNIP3.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Ergocalciferols , Membrane Proteins , Mice, Knockout , Mitophagy , Protein Kinases , Receptors, Calcitriol , Streptozocin , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , Mitophagy/genetics , Mitophagy/drug effects , Protein Kinases/metabolism , Protein Kinases/genetics , Humans , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/genetics , Male , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Fibrosis , Kidney Tubules/metabolism , Kidney Tubules/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Mice, Inbred C57BL , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Cell Line , Gene Expression Regulation/drug effects
5.
Discov Med ; 36(184): 992-1001, 2024 May.
Article in English | MEDLINE | ID: mdl-38798258

ABSTRACT

BACKGROUND: Diabetic neuropathic pain (DNP) is a complication of diabetes mellitus (DM). Hyperbaric lidocaine (HL), a local anesthetics drug, has neurotoxicity. The present study aims to study the effect and molecular mechanisms of HL on spinal nerve injury in DNP. METHODS: The DNP rat model was established through a high-fat-glucose diet in combination with Streptozotocin (STZ) administration. SB203580 and PD98059 were utilized to inhibit p38 mitogen-activated protein kinase (p38 MAPK) and extracellular signal-regulated kinase (ERK). The mechanical paw withdrawal threshold (PWT) and the thermal paw withdrawal latency (PWL) were tested to evaluate rats' mechanical allodynia and thermal hyperalgesia. Hematoxylin-eosin (H&E) and terminal deoxynucleotidyltransferase-mediated dUTP nick-end Labeling (TUNEL) staining were performed to evaluate the pathological changes and neuron apoptosis in spinal cord tissues of L4-5. Western blotting analysis and reverse transcription-polymerase chain reaction (RT-qPCR) assay were used to measure the levels of proteins and mRNAs, respectively. RESULTS: PWT and PWL were decreased in DNP rats with serious spinal nerve injury. HL administration downregulated the PWT and PWL and aggravated spinal nerve injury in DNP rats, but isobaric lidocaine had no effects on these changes. Meanwhile, p38 MAPK/ERK signaling and PTEN-induced kinase 1 (PINK1)-mediated mitophagy were activated in DNP, which was enhanced by HL but not isobaric lidocaine. Blocking p38 MAPK/ERK signaling could effectively attenuate HL-induced spinal nerve injury and inhibit mitophagy. CONCLUSION: In summary, HL can aggravate spinal cord tissue damage in DNP rats by inducing PINK1-mediated mitophagy via activating p38 MAPK/ERK signaling. Our data provide a novel insight that supports the potential role of p38 MAPK/ERK signaling in acting as a therapeutic target for HL-induced neurotoxicity.


Subject(s)
Diabetic Neuropathies , Lidocaine , Mitophagy , Protein Kinases , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases , p38 Mitogen-Activated Protein Kinases , Animals , Lidocaine/pharmacology , Rats , Diabetic Neuropathies/pathology , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/etiology , p38 Mitogen-Activated Protein Kinases/metabolism , Mitophagy/drug effects , Male , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , MAP Kinase Signaling System/drug effects , Signal Transduction/drug effects
6.
Bone Res ; 12(1): 32, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789434

ABSTRACT

Extracellular matrix (ECM) stiffening is a typical characteristic of cartilage aging, which is a quintessential feature of knee osteoarthritis (KOA). However, little is known about how ECM stiffening affects chondrocytes and other molecules downstream. This study mimicked the physiological and pathological stiffness of human cartilage using polydimethylsiloxane (PDMS) substrates. It demonstrated that epigenetic Parkin regulation by histone deacetylase 3 (HDAC3) represents a new mechanosensitive mechanism by which the stiffness matrix affected chondrocyte physiology. We found that ECM stiffening accelerated cultured chondrocyte senescence in vitro, while the stiffness ECM downregulated HDAC3, prompting Parkin acetylation to activate excessive mitophagy and accelerating chondrocyte senescence and osteoarthritis (OA) in mice. Contrarily, intra-articular injection with an HDAC3-expressing adeno-associated virus restored the young phenotype of the aged chondrocytes stimulated by ECM stiffening and alleviated OA in mice. The findings indicated that changes in the mechanical ECM properties initiated pathogenic mechanotransduction signals, promoted the Parkin acetylation and hyperactivated mitophagy, and damaged chondrocyte health. These results may provide new insights into chondrocyte regulation by the mechanical properties of ECM, suggesting that the modification of the physical ECM properties may be a potential OA treatment strategy.


Subject(s)
Cellular Senescence , Chondrocytes , Down-Regulation , Extracellular Matrix , Histone Deacetylases , Osteoarthritis , Animals , Chondrocytes/metabolism , Chondrocytes/pathology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Extracellular Matrix/metabolism , Osteoarthritis/pathology , Humans , Mice , Cellular Senescence/drug effects , Mice, Inbred C57BL , Mitophagy/drug effects , Male , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Acetylation , Cells, Cultured
7.
Sci Rep ; 14(1): 11824, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38782946

ABSTRACT

Pathological cardiac hypertrophy is an important cause of heart failure(HF). Recent studies reveal that glucagon-like peptide-1 receptor (GLP1R) agonists can improve mortality and left ventricular ejection fraction in the patients with type 2 diabetes and HF. The present study aims to investigate whether semaglutide, a long-acting GLP1R agonist, can ameliorate cardiac hypertrophy induced by pressure overload, and explore the potential mechanism. The rats were performed transverse aortic constriction (TAC) to mimic pressure overload model. The rats were divided into four groups including Sham, TAC, TAC + semaglutide, and TAC + semaglutide + HCQ (hydroxychloroquine, an inhibitor of mitophagy). The rats in each experimental group received their respective interventions for 4 weeks. The parameters of left ventricular hypertrophy(LVH) were measured by echocardiography, Hematoxylin-eosin (HE) staining, western-blot and immunohistochemistry (IHC), respectively. The changes of mitophagy were reflected by detecting cytochrome c oxidase subunit II (COXII), LC3II/LC3I, mitochondria, and autophagosomes. Meanwhile, NLRP3, Caspase-1, and interleukin-18 were detected to evaluate the activation of NLRP3 inflammasome in each group. The results suggest that LVH, impaired mitophagy, and activation of NLRP3 inflammasome were present in TAC rats. Semaglutide significantly reduced LVH, improve mitophagy, and down-regulated NLRP3 inflammatory signal pathway in TAC rats. However, the reversed effect of semaglutide on cardiac hypertrophy was abolished by HCQ, which restored the activation of NLRP3 inflammasome suppressed by improved mitophagy. In conclusion, semaglutide ameliorates the cardiac hypertrophy by improving cardiac mitophagy to suppress the activation of NLRP3 inflammasome. Semaglutide may be a novel potential option for intervention of cardiac hypertrophy induced by pressure overload.


Subject(s)
Cardiomegaly , Glucagon-Like Peptides , Inflammasomes , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mitophagy/drug effects , Inflammasomes/metabolism , Rats , Male , Glucagon-Like Peptides/pharmacology , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cardiomegaly/etiology , Cardiomegaly/pathology , Disease Models, Animal , Rats, Sprague-Dawley , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hypertrophy, Left Ventricular/drug therapy , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/prevention & control
8.
Cells ; 13(10)2024 May 18.
Article in English | MEDLINE | ID: mdl-38786094

ABSTRACT

Post-stroke cognitive impairment (PSCI) remains the most common consequence of ischemic stroke. In this study, we aimed to investigate the role and mechanisms of melatonin (MT) in improving cognitive dysfunction in stroke mice. We used CoCl2-induced hypoxia-injured SH-SY5Y cells as a cellular model of stroke and photothrombotic-induced ischemic stroke mice as an animal model. We found that the stroke-induced upregulation of mitophagy, apoptosis, and neuronal synaptic plasticity was impaired both in vivo and in vitro. The results of the novel object recognition test and Y-maze showed significant cognitive deficits in the stroke mice, and Nissl staining showed a loss of neurons in the stroke mice. In contrast, MT inhibited excessive mitophagy both in vivo and in vitro and decreased the levels of mitophagy proteins PINK1 and Parkin, and immunofluorescence staining showed reduced co-localization of Tom20 and LC3. A significant inhibition of mitophagy levels could be directly observed under transmission electron microscopy. Furthermore, behavioral experiments and Nissl staining showed that MT ameliorated cognitive deficits and reduced neuronal loss in mice following a stroke. Our results demonstrated that MT inhibits excessive mitophagy and improves PSCI. These findings highlight the potential of MT as a preventive drug for PSCI, offering promising therapeutic implications.


Subject(s)
Cognitive Dysfunction , Melatonin , Mitophagy , Stroke , Animals , Melatonin/pharmacology , Melatonin/therapeutic use , Mitophagy/drug effects , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/pathology , Cognitive Dysfunction/etiology , Mice , Stroke/complications , Stroke/drug therapy , Stroke/pathology , Male , Humans , Disease Models, Animal , Mice, Inbred C57BL , Apoptosis/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuronal Plasticity/drug effects , Cell Line, Tumor , Protein Kinases , Ubiquitin-Protein Ligases
9.
Cancer Lett ; 592: 216927, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38697460

ABSTRACT

Glioblastoma (GBM), one of the most malignant brain tumors in the world, has limited treatment options and a dismal survival rate. Effective and safe disease-modifying drugs for glioblastoma are urgently needed. Here, we identified a small molecule, Molephantin (EM-5), effectively penetrated the blood-brain barrier (BBB) and demonstrated notable antitumor effects against GBM with good safety profiles both in vitro and in vivo. Mechanistically, EM-5 not only inhibits the proliferation and invasion of GBM cells but also induces cell apoptosis through the reactive oxygen species (ROS)-mediated PI3K/Akt/mTOR pathway. Furthermore, EM-5 causes mitochondrial dysfunction and blocks mitophagy flux by impeding the fusion of mitophagosomes with lysosomes. It is noteworthy that EM-5 does not interfere with the initiation of autophagosome formation or lysosomal function. Additionally, the mitophagy flux blockage caused by EM-5 was driven by the accumulation of intracellular ROS. In vivo, EM-5 exhibited significant efficacy in suppressing tumor growth in a xenograft model. Collectively, our findings not only identified EM-5 as a promising, effective, and safe lead compound for treating GBM but also uncovered its underlying mechanisms from the perspective of apoptosis and mitophagy.


Subject(s)
Apoptosis , Brain Neoplasms , Cell Proliferation , Glioblastoma , Mitophagy , Reactive Oxygen Species , Xenograft Model Antitumor Assays , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Reactive Oxygen Species/metabolism , Humans , Mitophagy/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Mice , Cell Proliferation/drug effects , Signal Transduction/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Nude , TOR Serine-Threonine Kinases/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Proto-Oncogene Proteins c-akt/metabolism
10.
Toxicol Appl Pharmacol ; 486: 116952, 2024 May.
Article in English | MEDLINE | ID: mdl-38705399

ABSTRACT

The incidence of contrast-induced acute kidney injury (CI-AKI) has escalated to become the third most prevalent cause of hospital-acquired AKI, with a lack of efficacious interventions. Berberine (BBR) possesses diverse pharmacological effects and exhibits renoprotective properties; however, limited knowledge exists regarding its impact on CI-AKI. Therefore, our study aimed to investigate the protective effects and underlying mechanisms of BBR on CI-AKI in a mice model, focusing on the nucleotide-binding oligomerization domain-like pyrin domain-containing protein 3 (NLRP3) inflammasome and mitophagy. The CI-AKI mice model was established by administering NG-nitro-L-arginine methyl ester (L-NAME) (10 mg/kg), indomethacin (10 mg/kg), and iohexol (11 g/kg) following water deprivation. A pretreatment of 100 mg/kg of BBR was orally administered to the mice for two weeks. Renal injury markers, damage-associated molecular patterns (DAMPs), renal histopathology, mitochondrial morphology, autophagosomes, and potential mechanisms were investigated. BBR effectively reduced levels of renal injury biomarkers such as serum cystatin C, urea nitrogen, and creatinine, downregulated the protein level of kidney injury molecule 1 (KIM1), and mitigated renal histomorphological damage. Moreover, BBR reduced DAMPs, including high mobility group box-1 (HMGB1), heat shock protein 70 (HSP70), and uric acid (UA). It also alleviated oxidative stress and inflammatory factors such as monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1ß). Furthermore, the activation of NLRP3 inflammasome was attenuated in the BBR pretreatment group, as evidenced by both mRNA and protein levels. Electron microscopy and western blotting examination revealed that BBR mitigated mitochondrial damage and enhanced mitophagy. Additionally, BBR increased the P-AMPK/AMPK ratio. These findings indicated that BBR exerted a protective effect against CI-AKI by suppressing NLRP3 inflammasome activation and modulating mitophagy, providing a potential therapeutic strategy for its prevention.


Subject(s)
Acute Kidney Injury , Berberine , Contrast Media , Disease Models, Animal , Inflammasomes , Mice, Inbred C57BL , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mitophagy/drug effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Inflammasomes/metabolism , Inflammasomes/drug effects , Mice , Berberine/pharmacology , Male , Kidney/drug effects , Kidney/pathology , Kidney/metabolism
11.
Mol Med ; 30(1): 72, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822247

ABSTRACT

BACKGROUND: 8-Oxoguanine DNA glycosylase (OGG1), a well-known DNA repair enzyme, has been demonstrated to promote lung fibrosis, while the specific regulatory mechanism of OGG1 during pulmonary fibrosis remains unclarified. METHODS: A bleomycin (BLM)-induced mouse pulmonary fibrosis model was established, and TH5487 (the small molecule OGG1 inhibitor) and Mitochondrial division inhibitor 1 (Mdivi-1) were used for administration. Histopathological injury of the lung tissues was assessed. The profibrotic factors and oxidative stress-related factors were examined using the commercial kits. Western blot was used to examine protein expression and immunofluorescence analysis was conducted to assess macrophages polarization and autophagy. The conditional medium from M2 macrophages was harvested and added to HFL-1 cells for culture to simulate the immune microenvironment around fibroblasts during pulmonary fibrosis. Subsequently, the loss- and gain-of function experiments were conducted to further confirm the molecular mechanism of OGG1/PINK1. RESULTS: In BLM-induced pulmonary fibrosis, OGG1 was upregulated while PINK1/Parkin was downregulated. Macrophages were activated and polarized to M2 phenotype. TH5487 administration effectively mitigated pulmonary fibrosis, M2 macrophage polarization, oxidative stress and mitochondrial dysfunction while promoted PINK1/Parkin-mediated mitophagy in lung tissues of BLM-induced mice, which was partly hindered by Mdivi-1. PINK1 overexpression restricted M2 macrophages-induced oxidative stress, mitochondrial dysfunction and mitophagy inactivation in lung fibroblast cells, and OGG1 knockdown could promote PINK1/Parkin expression and alleviate M2 macrophages-induced mitochondrial dysfunction in HFL-1 cells. CONCLUSION: OGG1 inhibition protects against pulmonary fibrosis, which is partly via activating PINK1/Parkin-mediated mitophagy and retarding M2 macrophage polarization, providing a therapeutic target for pulmonary fibrosis.


Subject(s)
Bleomycin , DNA Glycosylases , Disease Models, Animal , Macrophages , Mitophagy , Protein Kinases , Pulmonary Fibrosis , Animals , Mitophagy/drug effects , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , DNA Glycosylases/metabolism , DNA Glycosylases/genetics , Mice , Macrophages/metabolism , Protein Kinases/metabolism , Bleomycin/adverse effects , Male , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Oxidative Stress/drug effects , Mice, Inbred C57BL , Macrophage Activation , Humans , Quinazolinones
12.
Mol Med Rep ; 30(1)2024 Jul.
Article in English | MEDLINE | ID: mdl-38785143

ABSTRACT

Astragaloside IV (AS­IV) has various pharmacological effects, including antioxidant and immunoregulatory properties, which can improve myasthenia gravis (MG) symptoms. However, the potential mechanism underlying the effects of AS­IV on MG remains to be elucidated. The present study aimed to investigate whether AS­IV has a therapeutic effect on MG and its potential mechanism of action. By subcutaneously immunizing rats with R97­116 peptide, an experimental autoimmune (EA) MG rat model was established. AS­IV (40 or 80 mg/kg/day) treatment was then applied for 28 days after modeling. The results demonstrated that AS­IV significantly ameliorated the weight loss, Lennon score and pathological changes in the gastrocnemius muscle of EAMG rats compared with the model group. Additionally, the levels of acetylcholine receptor antibody (AChR­Ab) were significantly decreased, whereas mitochondrial function [ATPase and cytochrome c (Cyt­C) oxidase activities] and ultrastructure were improved in the AS­IV treated rats. Moreover, the mRNA and protein expression levels of phosphatase and tensin homolog­induced putative kinase 1, Parkin, LC3II and Bcl­2, key signaling molecules for mitophagy and apoptosis, were upregulated, whereas the mRNA and protein expression levels of p62, Cyt­C, Bax, caspase 3 and caspase 9 were downregulated following AS­IV intervention. In conclusion, AS­IV may protect against EAMG in a rat model by modulating mitophagy and apoptosis. These findings indicated the potential mechanism underlying the effects of AS­IV on MG and provided novel insights into treatment strategies for MG.


Subject(s)
Apoptosis , Mitophagy , Myasthenia Gravis, Autoimmune, Experimental , Saponins , Triterpenes , Animals , Saponins/pharmacology , Apoptosis/drug effects , Triterpenes/pharmacology , Mitophagy/drug effects , Rats , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Female , Disease Models, Animal , Mitochondria/drug effects , Mitochondria/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Receptors, Cholinergic/metabolism , Rats, Sprague-Dawley , Protective Agents/pharmacology
13.
Mil Med Res ; 11(1): 32, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38812059

ABSTRACT

Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.


Subject(s)
Mitochondria , Mitophagy , Humans , Mitochondria/metabolism , Mitochondria/physiology , Mitophagy/physiology , Mitophagy/drug effects , Mitochondrial Dynamics/physiology
14.
Acta Biomater ; 181: 425-439, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729544

ABSTRACT

Synovial macrophages play an important role in the progression of osteoarthritis (OA). In this study, we noted that synovial macrophages can activate pyroptosis in a gasdermin d-dependent manner and produce reactive oxygen species (ROS), aberrantly activating the mammalian target of rapamycin complex 1 (mTORC1) pathway and matrix metalloproteinase-9 (MMP9) expression in synovial tissue samples collected from both patients with OA and collagen-induced osteoarthritis (CIOA) mouse model. To overcome this, we constructed rapamycin- (RAPA, a mTORC1 inhibitor) loaded mesoporous Prussian blue nanoparticles (MPB NPs, for catalyzing ROS) and modified the NPs with MMP9-targeted peptides (favor macrophage targeting) to develop RAPA@MPB-MMP9 NPs. The inherent enzyme-like activity and RAPA released from RAPA@MPB-MMP9 NPs synergistically impeded the pyroptosis of macrophages and the activation of the mTORC1 pathway. In particular, the NPs decreased pyroptosis-mediated ROS generation, thereby inhibiting cGAS-STING signaling pathway activation caused by the release of mitochondrial DNA. Moreover, the NPs promoted macrophage mitophagy to restore mitochondrial stability, alleviate pyroptosis-related inflammatory responses, and decrease senescent synoviocytes. After the as-prepared NPs were intra-articularly injected into the CIOA mouse model, they efficiently attenuated synovial macrophage pyroptosis and cartilage degradation. In conclusion, our study findings provide a novel therapeutic strategy for OA that modulates the pyroptosis and mitophagy of synovial macrophage by utilizing functionalized NPs. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) presents a significant global challenge owing to its complex pathogenesis and finite treatment options. Synovial macrophages have emerged as key players in the progression of OA, managing inflammation and tissue destruction. In this study, we discovered a novel therapeutic strategy in which the pyroptosis and mitophagy of synovial macrophages are targeted to mitigate OA pathology. For this, we designed and prepared rapamycin-loaded mesoporous Prussian blue nanoparticles (RAPA@MPB-MMP9 NPs) to specifically target synovial macrophages and modulate their inflammatory responses. These NPs could efficiently suppress macrophage pyroptosis, diminish reactive oxygen species production, and promote mitophagy, thereby alleviating inflammation and protecting cartilage integrity. Our study findings not only clarify the intricate mechanisms underlying OA pathogenesis but also present a promising therapeutic approach for effectively managing OA by targeting dysregulation in synovial macrophages.


Subject(s)
Macrophages , Mitophagy , Nanoparticles , Osteoarthritis , Pyroptosis , Reactive Oxygen Species , Osteoarthritis/pathology , Osteoarthritis/drug therapy , Animals , Pyroptosis/drug effects , Nanoparticles/chemistry , Macrophages/metabolism , Macrophages/drug effects , Macrophages/pathology , Mitophagy/drug effects , Mice , Humans , Reactive Oxygen Species/metabolism , Male , Sirolimus/pharmacology , Matrix Metalloproteinase 9/metabolism , Disease Progression , Mechanistic Target of Rapamycin Complex 1/metabolism , Synovial Membrane/pathology , Synovial Membrane/drug effects , Mice, Inbred C57BL , Ferrocyanides
15.
Cell Death Dis ; 15(5): 382, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821960

ABSTRACT

Impairment of autophagy leads to an accumulation of misfolded proteins and damaged organelles and has been implicated in plethora of human diseases. Loss of autophagy in actively respiring cells has also been shown to trigger metabolic collapse mediated by the depletion of nicotinamide adenine dinucleotide (NAD) pools, resulting in cell death. Here we found that the deficit in the autophagy-NAD axis underpins the loss of viability in cell models of a neurodegenerative lysosomal storage disorder, Niemann-Pick type C1 (NPC1) disease. Defective autophagic flux in NPC1 cells resulted in mitochondrial dysfunction due to impairment of mitophagy, leading to the depletion of both the reduced and oxidised forms of NAD as identified via metabolic profiling. Consequently, exhaustion of the NAD pools triggered mitochondrial depolarisation and apoptotic cell death. Our chemical screening identified two FDA-approved drugs, celecoxib and memantine, as autophagy activators which effectively restored autophagic flux, NAD levels, and cell viability of NPC1 cells. Of biomedical relevance, either pharmacological rescue of the autophagy deficiency or NAD precursor supplementation restored NAD levels and improved the viability of NPC1 patient fibroblasts and induced pluripotent stem cell (iPSC)-derived cortical neurons. Together, our findings identify the autophagy-NAD axis as a mechanism of cell death and a target for therapeutic interventions in NPC1 disease, with a potential relevance to other neurodegenerative disorders.


Subject(s)
Autophagy , Induced Pluripotent Stem Cells , NAD , Niemann-Pick Disease, Type C , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/genetics , Humans , Autophagy/drug effects , NAD/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Mitochondria/metabolism , Mitochondria/drug effects , Memantine/pharmacology , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Cell Death/drug effects , Cell Survival/drug effects , Mitophagy/drug effects , Apoptosis/drug effects
16.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732105

ABSTRACT

Multiple myeloma is an incurable plasma cell malignancy. Most patients end up relapsing and developing resistance to antineoplastic drugs, like bortezomib. Antibiotic tigecycline has activity against myeloma. This study analyzed tigecycline and bortezomib combination on cell lines and plasma cells from myeloma patients. Apoptosis, autophagic vesicles, mitochondrial mass, mitochondrial superoxide, cell cycle, and hydrogen peroxide were studied by flow cytometry. In addition, mitochondrial antioxidants and electron transport chain complexes were quantified by reverse transcription real-time PCR (RT-qPCR) or western blot. Cell metabolism and mitochondrial activity were characterized by Seahorse and RT-qPCR. We found that the addition of tigecycline to bortezomib reduces apoptosis in proportion to tigecycline concentration. Supporting this, the combination of both drugs counteracts bortezomib in vitro individual effects on the cell cycle, reduces autophagy and mitophagy markers, and reverts bortezomib-induced increase in mitochondrial superoxide. Changes in mitochondrial homeostasis and MYC upregulation may account for some of these findings. These data not only advise to avoid considering tigecycline and bortezomib combination for treating myeloma, but caution on the potential adverse impact of treating infections with this antibiotic in myeloma patients under bortezomib treatment.


Subject(s)
Apoptosis , Bortezomib , Mitochondria , Multiple Myeloma , Reactive Oxygen Species , Tigecycline , Bortezomib/pharmacology , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tigecycline/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Mitophagy/drug effects , Cell Cycle/drug effects
17.
J Transl Med ; 22(1): 436, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720350

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH) represents a form of cerebrovascular event characterized by a notable mortality and morbidity rate. Fibroblast growth factor 21 (FGF21), a versatile hormone predominantly synthesized by the hepatic tissue, has emerged as a promising neuroprotective agent. Nevertheless, the precise impacts and underlying mechanisms of FGF21 in the context of SAH remain enigmatic. METHODS: To elucidate the role of FGF21 in inhibiting the microglial cGAS-STING pathway and providing protection against SAH-induced cerebral injury, a series of cellular and molecular techniques, including western blot analysis, real-time polymerase chain reaction, immunohistochemistry, RNA sequencing, and behavioral assays, were employed. RESULTS: Administration of recombinant fibroblast growth factor 21 (rFGF21) effectively mitigated neural apoptosis, improved cerebral edema, and attenuated neurological impairments post-SAH. Transcriptomic analysis revealed that SAH triggered the upregulation of numerous genes linked to innate immunity, particularly those involved in the type I interferon (IFN-I) pathway and microglial function, which were notably suppressed upon adjunctive rFGF21 treatment. Mechanistically, rFGF21 intervention facilitated mitophagy in an AMP-activated protein kinase (AMPK)-dependent manner, thereby preventing mitochondrial DNA (mtDNA) release into the cytoplasm and dampening the activation of the DNA-sensing cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Conditional knockout of STING in microglia markedly ameliorated the inflammatory response and mitigated secondary brain injuries post-SAH. CONCLUSION: Our results present the initial evidence that FGF21 confers a protective effect against neuroinflammation-associated brain damage subsequent to SAH. Mechanistically, we have elucidated a novel pathway by which FGF21 exerts this neuroprotection through inhibition of the cGAS-STING signaling cascade.


Subject(s)
Fibroblast Growth Factors , Membrane Proteins , Mice, Inbred C57BL , Mitophagy , Neuroinflammatory Diseases , Nucleotidyltransferases , Signal Transduction , Subarachnoid Hemorrhage , Animals , Membrane Proteins/metabolism , Fibroblast Growth Factors/metabolism , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Mitophagy/drug effects , Signal Transduction/drug effects , Nucleotidyltransferases/metabolism , Male , Mice , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Microglia/metabolism , Microglia/pathology , Microglia/drug effects , Apoptosis/drug effects
18.
Eur J Med Chem ; 272: 116478, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38718624

ABSTRACT

Metallodrugs exhibiting distinct mechanisms of action compared with cisplatin hold promise for overcoming cisplatin resistance and improving the efficacy of anticancer drugs. In this study, a new series of rhodium (Rh)(III) complexes containing tris(triphenylphosphine)rhodium(I) chloride [(TPP)3RhCl] (TPP = triphenylphosphine, TPP=O = triphenylphosphine oxide) and 8-hydroxyquinoline derivatives (H-XR1-H-XR4), namely [Rh(XR1)2(TPP)Cl]·(TPP=O) (Yulin Normal University-1a [YNU-1a]), [Rh(XR2)2(TPP)Cl] (YNU-1b), [Rh(XR3)2(TPP)Cl] (YNU-1c), and [Rh(XR4)2(TPP)Cl] (YNU-1d), was synthesized and characterized via X-ray diffraction, mass spectrometry and IR. The cytotoxicity of the compounds YNU-1a-YNU-1d in Hep-G2 and HCC1806 human cancer cell lines and normal HL-7702 cell line was evaluated. YNU-1c exhibited cytotoxicity and selectivity in HCC1806 cells (IC50 = 0.13 ± 0.06 µM, selectivity factor (SF) = 384.6). The compounds YNU-1b and YNU-1c, which were selected for mechanistic studies, induced the activation of apoptotic pathways and mitophagy. In addition, these compounds released cytochrome c, cleaved caspase-3/pro-caspase-3 and downregulated the levels of mitochondrial respiratory chain complexes I/IV (M1 and M4) and ATP. The compound YNU-1c, which was selected for in vivo experiments, exhibited tumor growth inhibition (58.9 %). Importantly, hematoxylin and eosin staining and TUNEL revealed that HCC1806 tumor tissues exhibited significant apoptotic characteristics. YNU-1a-YNU-1d compounds are promising drug candidates that can be used to overcome cisplatin resistance.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Coordination Complexes , Drug Screening Assays, Antitumor , Mitophagy , Oxyquinoline , Rhodium , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Rhodium/chemistry , Rhodium/pharmacology , Oxyquinoline/chemistry , Oxyquinoline/pharmacology , Oxyquinoline/chemical synthesis , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Animals , Structure-Activity Relationship , Cell Proliferation/drug effects , Mitophagy/drug effects , Molecular Structure , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/chemical synthesis , Dose-Response Relationship, Drug , Apoptosis/drug effects , Mice , Cell Line, Tumor
19.
Sci Rep ; 14(1): 11721, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38777823

ABSTRACT

It has recently been shown that KAT8, a genome-wide association study candidate risk gene for Parkinson's Disease, is involved in PINK1/Parkin-dependant mitophagy. The KAT8 gene encodes a lysine acetyltransferase and represents the catalytically active subunit of the non-specific lethal epigenetic remodelling complex. In the current study, we show that contrary to KAT5 inhibition, dual inhibition of KAT5 and KAT8 via the MG149 compound inhibits the initial steps of the PINK1-dependant mitophagy process. More specifically, our study shows that following mitochondrial depolarisation induced by mitochondrial toxins, MG149 treatment inhibits PINK1-dependant mitophagy initiation by impairing PINK1 activation, and subsequent phosphorylation of Parkin and ubiquitin. While this inhibitory effect of MG149 on PINK1-activation is potent, MG149 treatment in the absence of mitochondrial toxins is sufficient to depolarise the mitochondrial membrane, recruit PINK1 and promote partial downstream recruitment of the autophagy receptor p62, leading to an increase in mitochondrial delivery to the lysosomes. Altogether, our study provides additional support for KAT8 as a regulator of mitophagy and autophagy processes.


Subject(s)
Mitochondria , Mitophagy , Protein Kinases , Ubiquitin-Protein Ligases , Mitophagy/drug effects , Humans , Protein Kinases/metabolism , Protein Kinases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Phosphorylation/drug effects , Membrane Potential, Mitochondrial/drug effects , HeLa Cells
20.
Eur J Pharmacol ; 972: 176586, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38615891

ABSTRACT

Coronary atherosclerotic heart disease (CHD) is a primary cardiovascular disease caused by atherosclerosis (AS), which is characterized by chronic inflammation and lipid oxidative deposition. Molecular hydrogen (H2) is an effective anti-inflammatory agent and has potential to ameliorate glycolipid metabolism disorders, which is believed to exert beneficial effects on the prevention and treatment of CHD. It is suggested that H2 reduces inflammation in CHD by regulating multiple pathways, including NF-κB inflammatory pathway, pyroptosis, mitophagy, endoplasmic reticulum (ER) stress, and Nrf2 antioxidant pathway. Additionally, H2 may improve glycolipid metabolism by mediation of PI3K and AMPK signalling pathways, contributing to inhibition of the occurrence and development of CHD. This review elaborates pathogenesis of CHD and evaluates the role of H2 in CHD. Moreover, possible molecular mechanisms have been discussed and speculated, aiming to provide more strategies and directions for subsequent studies of H2 in CHD.


Subject(s)
Coronary Artery Disease , Hydrogen , Humans , Hydrogen/therapeutic use , Hydrogen/pharmacology , Animals , Coronary Artery Disease/prevention & control , Coronary Artery Disease/drug therapy , Coronary Artery Disease/metabolism , Signal Transduction/drug effects , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Endoplasmic Reticulum Stress/drug effects , Mitophagy/drug effects , Oxidative Stress/drug effects , Glycolipids/metabolism , Glycolipids/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , NF-kappa B/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...