Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29.786
Filter
1.
Immun Inflamm Dis ; 12(5): e1077, 2024 May.
Article in English | MEDLINE | ID: mdl-38722267

ABSTRACT

BACKGROUND: Considering the antihepatitis effects of Tectorigenin (TEC), and the same adenosine mitogen-activated protein kinase (MAPK) pathway in both hepatitis and inflammatory bowel disease (IBD) models, exploring the role of TEC in IBD is contributive to develop a new treatment strategy against IBD. METHODS: The IBD mouse model was constructed by feeding with dextran sodium sulfate (DSS) and injection of TEC. Afterward, the mouse body weight, colon length, and disease activity index (DAI) were tested to assess the enteritis level. Mouse intestine lesions were detected by hematoxylin and eosin staining. Murine macrophages underwent lipopolysaccharide (LPS) induction to establish an inflammation model. Cell viability was determined by cell counting kit-8 assay. Enzyme-linked immunosorbent assay was performed to measure interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α) levels. Cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expressions were quantified via quantitative reverse transcription polymerase chain reaction. Levels of MAPK pathway-related proteins (p-P38, P38, p-Jun N-terminal kinase (JNK), JNK, signal-regulated kinase (ERK), p-ERK), COX-2 and iNOS were quantitated by Western blot. RESULTS: TEC improved the inflammatory response through ameliorating weight loss, shortening colon, and increasing DAI score in IBD mouse. Expressions of intestinal inflammatory factors (IL-6, TNF-α, iNOS and COX-2) and MAPK pathway-related proteins (p-P38, p-JNK, and p-ERK) were increased both in DSS-induced mouse intestinal tissue, but TEC inhibited expressions of inflammatory factors. The same increased trend was identified in LPS-induced macrophages, but TEC improved macrophage inflammation, as evidenced by downregulation of inflammatory factors. CONCLUSION: TEC mitigates IBD and LPS-induced macrophage inflammation in mice via inhibiting MAPK signaling pathway.


Subject(s)
Inflammatory Bowel Diseases , Isoflavones , Lipopolysaccharides , MAP Kinase Signaling System , Macrophages , Animals , Mice , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , MAP Kinase Signaling System/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , Isoflavones/pharmacology , Isoflavones/therapeutic use , Disease Models, Animal , Dextran Sulfate/toxicity , Inflammation/drug therapy , Inflammation/immunology , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism
2.
PLoS One ; 19(5): e0303060, 2024.
Article in English | MEDLINE | ID: mdl-38723008

ABSTRACT

In the current study we investigated the impact of combination of rutin and vitamin A on glycated products, the glyoxalase system, oxidative markers, and inflammation in animals fed a high-fat high-fructose (HFFD) diet. Thirty rats were randomly divided into six groups (n = 5). The treatments, metformin (120 mg/kg), rutin (100 mg/kg), vitamin A (43 IU/kg), and a combination of rutin (100 mg/kg) and vitamin A (43 IU/kg) were given to relevant groups of rats along with high-fructose high-fat diet for 42 days. HbA1c, D-lactate, Glyoxylase-1, Hexokinase 2, malondialdehyde (MDA), glutathione peroxidase (GPx), catalase (CAT), nuclear transcription factor-B (NF-κB), interleukin-6 (IL-6), interleukin-8 (IL-8) and histological examinations were performed after 42 days. The docking simulations were conducted using Auto Dock package. The combined effects of rutin and vitamin A in treated rats significantly (p < 0.001) reduced HbA1c, hexokinase 2, and D-lactate levels while preventing cellular damage. The combination dramatically (p < 0.001) decreased MDA, CAT, and GPx in treated rats and decreased the expression of inflammatory cytokines such as IL-6 andIL-8, as well as the transcription factor NF-κB. The molecular docking investigations revealed that rutin had a strong affinity for several important biomolecules, including as NF-κB, Catalase, MDA, IL-6, hexokinase 2, and GPx. The results propose beneficial impact of rutin and vitamin A as a convincing treatment strategy to treat AGE-related disorders, such as diabetes, autism, alzheimer's, atherosclerosis.


Subject(s)
Diet, High-Fat , Fructose , Hyperglycemia , Inflammation , Oxidative Stress , Rutin , Vitamin A , Animals , Rutin/pharmacology , Oxidative Stress/drug effects , Fructose/adverse effects , Rats , Diet, High-Fat/adverse effects , Vitamin A/pharmacology , Vitamin A/metabolism , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Male , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hyperglycemia/chemically induced , Molecular Docking Simulation , Rats, Wistar , Disease Models, Animal , Glycosylation/drug effects , Metformin/pharmacology , Glycated Hemoglobin/metabolism , NF-kappa B/metabolism , Hexokinase/metabolism , Catalase/metabolism
3.
J Nanobiotechnology ; 22(1): 233, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38725011

ABSTRACT

BACKGROUND: Dry Eye Disease (DED) is a prevalent multifactorial ocular disease characterized by a vicious cycle of inflammation, oxidative stress, and mitochondrial dysfunction on the ocular surface, all of which lead to DED deterioration and impair the patients' quality of life and social functioning. Currently, anti-inflammatory drugs have shown promising efficacy in treating DED; however, such drugs are associated with side effects. The bioavailability of ocular drugs is less than 5% owing to factors such as rapid tear turnover and the presence of the corneal barrier. This calls for investigations to overcome these challenges associated with ocular drug administration. RESULTS: A novel hierarchical action liposome nanosystem (PHP-DPS@INS) was developed in this study. In terms of delivery, PHP-DPS@INS nanoparticles (NPs) overcame the ocular surface transport barrier by adopting the strategy of "ocular surface electrostatic adhesion-lysosomal site-directed escape". In terms of therapy, PHP-DPS@INS achieved mitochondrial targeting and antioxidant effects through SS-31 peptide, and exerted an anti-inflammatory effect by loading insulin to reduce mitochondrial inflammatory metabolites. Ultimately, the synergistic action of "anti-inflammation-antioxidation-mitochondrial function restoration" breaks the vicious cycle associated with DED. The PHP-DPS@INS demonstrated remarkable cellular uptake, lysosomal escape, and mitochondrial targeting in vitro. Targeted metabolomics analysis revealed that PHP-DPS@INS effectively normalized the elevated level of mitochondrial proinflammatory metabolite fumarate in an in vitro hypertonic model of DED, thereby reducing the levels of key inflammatory factors (IL-1ß, IL-6, and TNF-α). Additionally, PHP-DPS@INS strongly inhibited reactive oxygen species (ROS) production and facilitated mitochondrial structural repair. In vivo, the PHP-DPS@INS treatment significantly enhanced the adhesion duration and corneal permeability of the ocular surface in DED mice, thereby improving insulin bioavailability. It also restored tear secretion, suppressed ocular surface damage, and reduced inflammation in DED mice. Moreover, it demonstrated favorable safety profiles both in vitro and in vivo. CONCLUSION: In summary, this study successfully developed a comprehensive DED management nanosystem that overcame the ocular surface transmission barrier and disrupted the vicious cycle that lead to dry eye pathogenesis. Additionally, it pioneered the regulation of mitochondrial metabolites as an anti-inflammatory treatment for ocular conditions, presenting a safe, efficient, and innovative therapeutic strategy for DED and other inflammatory diseases.


Subject(s)
Dry Eye Syndromes , Inflammation , Liposomes , Mitochondria , Oxidative Stress , Dry Eye Syndromes/drug therapy , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Mice , Oxidative Stress/drug effects , Liposomes/chemistry , Inflammation/drug therapy , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/chemistry , Nanoparticles/chemistry , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cornea/metabolism , Cornea/drug effects , Drug Delivery Systems , Oligopeptides
4.
Cells ; 13(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38727269

ABSTRACT

The histone deacetylase inhibitor (HDACi) valproic acid (VPA) has neuroprotective and anti-inflammatory effects in experimental traumatic brain injury (TBI), which have been partially attributed to the epigenetic disinhibition of the transcription repressor RE1-Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). Additionally, VPA changes post-traumatic brain injury (TBI) brain metabolism to create a neuroprotective environment. To address the interconnection of neuroprotection, metabolism, inflammation and REST/NRSF after TBI, we subjected C57BL/6N mice to experimental TBI and intraperitoneal VPA administration or vehicle solution at 15 min, 1, 2, and 3 days post-injury (dpi). At 7 dpi, TBI-induced an up-regulation of REST/NRSF gene expression and HDACi function of VPA on histone H3 acetylation were confirmed. Neurological deficits, brain lesion size, blood-brain barrier permeability, or astrogliosis were not affected, and REST/NRSF target genes were only marginally influenced by VPA. However, VPA attenuated structural damage in the hippocampus, microgliosis and expression of the pro-inflammatory marker genes. Analyses of plasma lipidomic and polar metabolomic patterns revealed that VPA treatment increased lysophosphatidylcholines (LPCs), which were inversely associated with interleukin 1 beta (Il1b) and tumor necrosis factor (Tnf) gene expression in the brain. The results show that VPA has mild neuroprotective and anti-inflammatory effects likely originating from favorable systemic metabolic changes resulting in increased plasma LPCs that are known to be actively taken up by the brain and function as carriers for neuroprotective polyunsaturated fatty acids.


Subject(s)
Brain Injuries, Traumatic , Inflammation , Lysophosphatidylcholines , Mice, Inbred C57BL , Neurons , Valproic Acid , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/complications , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Mice , Male , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Inflammation/pathology , Inflammation/drug therapy , Lysophosphatidylcholines/blood , Cell Death/drug effects , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Repressor Proteins/metabolism , Repressor Proteins/genetics
5.
PLoS One ; 19(5): e0302015, 2024.
Article in English | MEDLINE | ID: mdl-38728332

ABSTRACT

Nature has proven to be a treasure resource of bioactive metabolites. In this regard, Tamarix aphylla (F. Tamaricaceae) leaves crude extract was investigated for its gastroprotective effect against indomethacin-induced damage to the gastric mucosa. Additionally, phytochemical investigation of the methanolic extract afforded eight flavonoids' derivatives (1-8). On pharmacology networking study, the isolated compounds identified 123 unique targets where only 45 targets were related to peptic ulcer conditions, these 45 targets include 11 targets specifically correlate to gastric ulcer. The protein-protein interaction defined the PTGS2 gene as one of the highly interacted genes and the complete pharmacology network defined the PTGS2 gene as the most represented gene. The top KEGG signaling pathways according to fold enrichment analysis was the EGFR tyrosine kinase inhibitor resistance pathway. As a result, these findings highlighted the significance of using T. aphylla leaves crude extract as an anti-gastric ulcer candidate, which provides a safer option to chemical antisecretory medicines, which are infamous for their negative side effects. Our findings have illuminated the potent anti-inflammatory and antioxidant effects of T. aphylla, which are likely mediated by suppressing IL-1ß, IL-6, TNF-α, and MAPK signaling pathways, without compromising gastric acidity.


Subject(s)
Indomethacin , MAP Kinase Signaling System , Oxidative Stress , Plant Extracts , Stomach Ulcer , Tamaricaceae , Stomach Ulcer/drug therapy , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Animals , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Indomethacin/adverse effects , Indomethacin/toxicity , Rats , Tamaricaceae/chemistry , MAP Kinase Signaling System/drug effects , Male , Plant Leaves/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/chemically induced , Rats, Sprague-Dawley , Network Pharmacology , Gastric Mucosa/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Anti-Ulcer Agents/chemistry , Flavonoids/pharmacology , Flavonoids/chemistry
6.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731964

ABSTRACT

Cannabidiol (CBD) is a non-psychoactive compound derived from Cannabis sativa. It has demonstrated promising effects in combating inflammation and holds potential as a treatment for the progression of chronic inflammation. However, the clinical application of CBD is limited due to its poor solubility and bioavailability. This study introduces an effective method for preparing CBD-loaded solid lipid nanoparticles (CBD-SLNs) using a combination of low-energy hot homogenization and ultrasonication. We enhanced this process by employing statistical optimization with response surface methodology (RSM). The optimized CBD-SLN formulation utilizes glyceryl monostearate as the primary lipid component of the nanocarrier. The CBD-SLN formulation is screened as a potential tool for managing chronic inflammation. Stable, uniformly dispersed spherical nanoparticles with a size of 123 nm, a surface charge of -32.1 mV, an encapsulation efficiency of 95.16%, and a drug loading of 2.36% were obtained. The CBD-SLNs exhibited sustained release properties, ensuring prolonged and controlled CBD delivery, which could potentially amplify its therapeutic effects. Additionally, we observed that CBD-SLNs significantly reduced both reactive oxygen and nitrogen species and proinflammatory cytokines in chondrocyte and macrophage cell lines, with these inhibitory effects being more pronounced than those of free CBD. In conclusion, CBD-SLNs demonstrated superiority over free CBD, highlighting its potential as an effective delivery system for CBD.


Subject(s)
Cannabidiol , Cytokines , Inflammation , Nanoparticles , Cannabidiol/chemistry , Cannabidiol/pharmacology , Nanoparticles/chemistry , Cytokines/metabolism , Inflammation/drug therapy , Humans , Animals , Free Radicals , Mice , Drug Carriers/chemistry , Lipids/chemistry , Cell Line , Reactive Oxygen Species/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/administration & dosage , Liposomes
7.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732062

ABSTRACT

Prunella vulgaris (PV) is one of the most commonly used nutraceuticals as it has been proven to have anti-inflammatory and antioxidant properties. The aim of this study was to evaluate the phytochemical composition of PV and its in vivo antioxidant properties. A phytochemical analysis measuring the total phenolic content (TPC), the identification of phenolic compounds by HPLC-DAD-ESI, and the evaluation of the in vitro antioxidant activity by the DPPH assay of the extract were performed. The antioxidant effects on inflammation induced by turpentine oil were experimentally tested in rats. Seven groups with six animals each were used: a control group, the experimental inflammation treatment group, the experimental inflammation and diclofenac sodium (DS) treatment group, and four groups with their inflammation treated using different dilutions of the extract. Serum redox balance was assessed based on total oxidative status (TOS), nitric oxide (NO), malondialdehyde (MDA), total antioxidant capacity (TAC), total thiols, and an oxidative stress index (OSI) contents. The TPC was 0.28 mg gallic acid equivalents (GAE)/mL extract, while specific representatives were represented by caffeic acid, p-coumaric acid, dihydroxybenzoic acid, gentisic acid, protocatechuic acid, rosmarinic acid, vanillic acid, apigenin-glucuronide, hesperidin, kaempferol-glucuronide. The highest amount (370.45 µg/mL) was reported for hesperidin, which is a phenolic compound belonging to the flavanone subclass. The antioxidant activity of the extracts, determined using the DPPH assay, was 27.52 mmol Trolox/mL extract. The PV treatment reduced the oxidative stress by lowering the TOS, OSI, NO, and MDA and by increasing the TAC and thiols. In acute inflammation, treatment with the PV extract reduced oxidative stress, with lower concentrations being more efficient and having a better effect than DS.


Subject(s)
Antioxidants , Inflammation , Oxidative Stress , Phytochemicals , Plant Extracts , Prunella , Animals , Antioxidants/pharmacology , Antioxidants/chemistry , Rats , Prunella/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Phytochemicals/pharmacology , Phytochemicals/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Oxidative Stress/drug effects , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Phenols/pharmacology , Phenols/analysis , Rats, Wistar
8.
J Transl Med ; 22(1): 457, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745204

ABSTRACT

BACKGROUND AND PURPOSE: Interstitial lung disease (ILD) represents a significant complication of rheumatoid arthritis (RA) that lacks effective treatment options. This study aimed to investigate the intrinsic mechanism by which resveratrol attenuates rheumatoid arthritis complicated with interstitial lung disease through the AKT/TMEM175 pathway. METHODS: We established an arthritis model by combining chicken type II collagen and complete Freund's adjuvant. Resveratrol treatment was administered via tube feeding for 10 days. Pathological changes in both the joints and lungs were evaluated using HE and Masson staining techniques. Protein expression of TGF-ß1, AKT, and TMEM175 was examined in lung tissue. MRC-5 cells were stimulated using IL-1ß in combination with TGF-ß1 as an in vitro model of RA-ILD, and agonists of AKT, metabolic inhibitors, and SiRNA of TMEM175 were used to explore the regulation and mechanism of action of resveratrol RA-ILD. RESULTS: Resveratrol mitigates fibrosis in rheumatoid arthritis-associated interstitial lung disease and reduces oxidative stress and inflammation in RA-ILD. Furthermore, resveratrol restored cellular autophagy. When combined with the in vitro model, it was further demonstrated that resveratrol could suppress TGF-ß1 expression, and reduce AKT metamorphic activation, consequently inhibiting the opening of AKT/MEM175 ion channels. This, in turn, lowers lysosomal pH and enhances the fusion of autophagosomes with lysosomes, ultimately ameliorating the progression of RA-ILD. CONCLUSION: In this study, we demonstrated that resveratrol restores autophagic flux through the AKT/MEM175 pathway to attenuate inflammation as well as fibrosis in RA-ILD by combining in vivo and in vitro experiments. It further provides a theoretical basis for the selection of therapeutic targets for RA-ILD.


Subject(s)
Arthritis, Rheumatoid , Fibrosis , Inflammation , Lung Diseases, Interstitial , Proto-Oncogene Proteins c-akt , Resveratrol , Signal Transduction , Resveratrol/pharmacology , Resveratrol/therapeutic use , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/metabolism , Humans , Inflammation/pathology , Inflammation/drug therapy , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Membrane Proteins/metabolism , Autophagy/drug effects , Oxidative Stress/drug effects , Cell Line , Lung/pathology , Lung/drug effects , Male
9.
Eur J Med Res ; 29(1): 285, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745325

ABSTRACT

INTRODUCTION: Hydrogen (H2) is regarded as a novel therapeutic agent against several diseases owing to its inherent biosafety. Bronchopulmonary dysplasia (BPD) has been widely considered among adverse pregnancy outcomes, without effective treatment. Placenta plays a role in defense, synthesis, and immunity, which provides a new perspective for the treatment of BPD. This study aimed to investigate if H2 reduced the placental inflammation to protect the neonatal rat against BPD damage and potential mechanisms. METHODS: We induced neonatal BPD model by injecting lipopolysaccharide (LPS, 1 µg) into the amniotic fluid at embryonic day 16.5 as LPS group. LPS + H2 group inhaled 42% H2 gas (4 h/day) until the samples were collected. We primarily analyzed the neonatal outcomes and then compared inflammatory levels from the control group (CON), LPS group and LPS + H2 group. HE staining was performed to evaluate inflammatory levels. RNA sequencing revealed dominant differentially expressed genes. Bioinformatics analysis (GO and KEGG) of RNA-seq was applied to mine the signaling pathways involved in protective effect of H2 on the development of LPS-induced BPD. We further used qRT-PCR, Western blot and ELISA methods to verify differential expression of mRNA and proteins. Moreover, we verified the correlation between the upstream signaling pathways and the downstream targets in LPS-induced BPD model. RESULTS: Upon administration of H2, the inflammatory infiltration degree of the LPS-induced placenta was reduced, and infiltration significantly narrowed. Hydrogen normalized LPS-induced perturbed lung development and reduced the death ratio of the fetus and neonate. RNA-seq results revealed the importance of inflammatory response biological processes and Toll-like receptor signaling pathway in protective effect of hydrogen on BPD. The over-activated upstream signals [Toll-like receptor 4 (TLR4), nuclear factor kappa-B p65 (NF-κB p65), Caspase1 (Casp1) and NLR family pyrin domain containing 3 (NLRP3) inflammasome] in LPS placenta were attenuated by H2 inhalation. The downstream targets, inflammatory cytokines/chemokines [interleukin (IL)-6, IL-18, IL-1ß, C-C motif chemokine ligand 2 (CCL2) and C-X-C motif chemokine ligand 1 (CXCL1)], were decreased both in mRNA and protein levels by H2 inhalation in LPS-induced placentas to rescue them from BPD. Correlation analysis displayed a positive association of TLR4-mediated signaling pathway both proinflammatory cytokines and chemokines in placenta. CONCLUSION: H2 inhalation ameliorates LPS-induced BPD by inhibiting excessive inflammatory cytokines and chemokines via the TLR4-NFκB-IL6/NLRP3 signaling pathway in placenta and may be a potential therapeutic strategy for BPD.


Subject(s)
Bronchopulmonary Dysplasia , Hydrogen , Inflammation , Lipopolysaccharides , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Placenta , Signal Transduction , Toll-Like Receptor 4 , Female , Pregnancy , Lipopolysaccharides/toxicity , Hydrogen/pharmacology , Hydrogen/therapeutic use , Animals , Placenta/metabolism , Placenta/drug effects , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Signal Transduction/drug effects , Rats , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NF-kappa B/metabolism , Inflammation/metabolism , Inflammation/drug therapy , Administration, Inhalation , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/drug therapy , Bronchopulmonary Dysplasia/prevention & control , Interleukin-6/metabolism , Interleukin-6/genetics , Rats, Sprague-Dawley , Disease Models, Animal
10.
Front Immunol ; 15: 1394925, 2024.
Article in English | MEDLINE | ID: mdl-38690282

ABSTRACT

Sepsis is a life-threatening organ dysfunction caused by the host's dysfunctional response to infection. Abnormal activation of the immune system and disturbance of energy metabolism play a key role in the development of sepsis. In recent years, the Sirtuins (SIRTs) family has been found to play an important role in the pathogenesis of sepsis. SIRTs, as a class of histone deacetylases (HDACs), are widely involved in cellular inflammation regulation, energy metabolism and oxidative stress. The effects of SIRTs on immune cells are mainly reflected in the regulation of inflammatory pathways. This regulation helps balance the inflammatory response and may lessen cell damage and organ dysfunction in sepsis. In terms of energy metabolism, SIRTs can play a role in immunophenotypic transformation by regulating cell metabolism, improve mitochondrial function, increase energy production, and maintain cell energy balance. SIRTs also regulate the production of reactive oxygen species (ROS), protecting cells from oxidative stress damage by activating antioxidant defense pathways and maintaining a balance between oxidants and reducing agents. Current studies have shown that several potential drugs, such as Resveratrol and melatonin, can enhance the activity of SIRT. It can help to reduce inflammatory response, improve energy metabolism and reduce oxidative stress, showing potential clinical application prospects for the treatment of sepsis. This review focuses on the regulation of SIRT on inflammatory response, energy metabolism and oxidative stress of immune cells, as well as its important influence on multiple organ dysfunction in sepsis, and discusses and summarizes the effects of related drugs and compounds on reducing multiple organ damage in sepsis through the pathway involving SIRTs. SIRTs may become a new target for the treatment of sepsis and its resulting organ dysfunction, providing new ideas and possibilities for the treatment of this life-threatening disease.


Subject(s)
Energy Metabolism , Oxidative Stress , Sepsis , Sirtuins , Humans , Sepsis/drug therapy , Sepsis/immunology , Sepsis/metabolism , Animals , Sirtuins/metabolism , Energy Metabolism/drug effects , Reactive Oxygen Species/metabolism , Inflammation/drug therapy , Inflammation/immunology
11.
Sci Rep ; 14(1): 11047, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38744989

ABSTRACT

Callicarpa kwangtungensis Chun (CK) is a common remedy exhibits anti-inflammatory properties and has been used in Chinese herbal formulations, such as KangGongYan tablets. It is the main component of KangGongYan tablets, which has been used to treat chronic cervicitis caused by damp heat, red and white bands, cervical erosion, and bleeding. However, the anti-inflammatory effects of CK water extract remains unknown. This study assessed the anti-inflammatory effects of CK in vivo and in vitro, characterized its main components in the serum of rats and verified the anti-inflammatory effects of serum containing CK. Nitric oxide (NO), tumour necrosis factor α (TNF-α) and interleukin-6 (IL-6) release by RAW264.7 cells was examined by ELISA and Griess reagents. Inflammation-related protein expression in LPS-stimulated RAW264.7 cells was measured by western blotting. Furthermore, rat model of foot swelling induced by λ-carrageenan and a collagen-induced arthritis (CIA) rat model were used to explore the anti-inflammatory effects of CK. The components of CK were characterized by LC-MS, and the effects of CK-containing serum on proinflammatory factors levels and the expression of inflammation-related proteins were examined by ELISA, Griess reagents and Western blotting. CK suppressed IL-6, TNF-α, and NO production, and iNOS protein expression in LPS-stimulated RAW264.7 cells. Mechanistic studies showed that CK inhibited the phosphorylation of ERK, P38 and JNK in the MAPK signaling pathway, promoted the expression of IκBα in the NF-κB signaling pathway, and subsequently inhibited the expression of iNOS, thereby exerting anti-inflammatory effects. Moreover, CK reduced the swelling rates with λ-carrageenan induced foot swelling, and reduced the arthritis score and incidence in the collagen-induced arthritis (CIA) rat model. A total of 68 compounds in CK water extract and 31 components in rat serum after intragastric administration of CK were characterized. Serum pharmacological analysis showed that CK-containing serum suppressed iNOS protein expression and NO, TNF-α, and IL-6 release. CK may be an anti-inflammatory agent with therapeutic potential for acute and chronic inflammatory diseases, especially inflammatory diseases associated with MAPK activation.


Subject(s)
Anti-Inflammatory Agents , Arthritis, Experimental , Nitric Oxide , Plant Extracts , Animals , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Rats , RAW 264.7 Cells , Plant Extracts/pharmacology , Plant Extracts/chemistry , Nitric Oxide/metabolism , Arthritis, Experimental/drug therapy , Water/chemistry , Carrageenan , Disease Models, Animal , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/blood , Male , Interleukin-6/metabolism , Interleukin-6/blood , Edema/drug therapy , Inflammation/drug therapy
12.
Sci Rep ; 14(1): 11079, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38745047

ABSTRACT

N-acetyl glucosamine (NAG) is a natural amino sugar found in various human tissues with previously described anti-inflammatory effects. Various chemical modifications of NAG have been made to promote its biomedical applications. In this study, we synthesized two bi-deoxygenated NAG, BNAG1 and BNAG2 and investigated their anti-inflammatory properties, using an in vivo and in vitro inflammation mouse model induced by lipopolysaccharide (LPS). Among the parent molecule NAG, BNAG1 and BNAG2, BNAG1 showed the highest inhibition against serum levels of IL-6 and TNF α and the leukocyte migration to lungs and peritoneal cavity in LPS challenged mice, as well as IL-6 and TNF α production in LPS-stimulated primary peritoneal macrophages. BNAG2 displayed an anti-inflammatory effect which was comparable to NAG. These findings implied potential application of these novel NAG derivatives, especially BNAG1, in treatment of certain inflammation-related diseases.


Subject(s)
Acetylglucosamine , Anti-Inflammatory Agents , Lipopolysaccharides , Macrophages, Peritoneal , Tumor Necrosis Factor-alpha , Animals , Acetylglucosamine/pharmacology , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemical synthesis , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Interleukin-6/blood , Inflammation/drug therapy , Male , Disease Models, Animal
13.
Cell Biochem Funct ; 42(4): e4030, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38720663

ABSTRACT

Diabetes mellitus (DM) is a collection of metabolic disorder that is characterized by chronic hyperglycemia. Recent studies have demonstrated the crucial involvement of oxidative stress (OS) and inflammatory reactions in the development of DM. Curcumin (CUR), a natural compound derived from turmeric, exerts beneficial effects on diabetes mellitus through its interaction with the nuclear factor kappa B (NF-κB) pathway. Research indicates that CUR targets inflammatory mediators in diabetes, including tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6), by modulating the NF-κB signaling pathway. By reducing the expression of these inflammatory factors, CUR demonstrates protective effects in DM by improving pancreatic ß-cells function, normalizing inflammatory cytokines, reducing OS and enhancing insulin sensitivity. The findings reveal that CUR administration effectively lowered blood glucose elevation, reinstated diminished serum insulin levels, and enhanced body weight in Streptozotocin -induced diabetic rats. CUR exerts its beneficial effects in management of diabetic complications through regulation of signaling pathways, such as calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII), peroxisome proliferator-activated receptor gamma (PPAR-γ), NF-κB, and transforming growth factor ß1 (TGFB1). Moreover, CUR reversed the heightened expression of inflammatory cytokines (TNF-α, Interleukin-1 beta (IL-1ß), IL-6) and chemokines like MCP-1 in diabetic specimens, vindicating its anti-inflammatory potency in counteracting hyperglycemia-induced alterations. CUR diminishes OS, avert structural kidney damage linked to diabetic nephropathy, and suppress NF-κB activity. Furthermore, CUR exhibited a protective effect against diabetic cardiomyopathy, lung injury, and diabetic gastroparesis. Conclusively, the study posits that CUR could potentially offer therapeutic benefits in relieving diabetic complications through its influence on the NF-κB pathway.


Subject(s)
Curcumin , Inflammation , NF-kappa B , Oxidative Stress , Signal Transduction , Curcumin/pharmacology , Curcumin/therapeutic use , Oxidative Stress/drug effects , NF-kappa B/metabolism , Animals , Inflammation/drug therapy , Inflammation/metabolism , Signal Transduction/drug effects , Humans , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Rats
14.
Autoimmunity ; 57(1): 2350202, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38721694

ABSTRACT

Yinchenhao Decoction (YCHD) is a classic prescription in traditional Chinese medicine (TCM). It appears to play an important role in anti-inflammation and autoimmunity protection. As one of the key active ingredients in YCHD, quercetin is a novel anti-inflammatory metabolite that exerts protective effects in many autoimmune diseases. However, its role in autoimmune hepatitis (AIH)-related hepatic injury has not been studied. The aim of this study was to reveal the hepatocyte protective mechanism of quercetin. In this study, we used Concanavalin A (Con A) to establish an in vitro hepatocyte injury-associated AIH model. Brl3a hepatocyte injury was induced by the supernatant of J774A.1 cells treated with Con A. We found that quercetin mitigated Con A-induced via macrophage-mediated Brl3a hepatocyte injury. Quercetin administration reduced the levels of alanine transaminase (ALT) and aspartate transaminase (AST) in the supernatant of Con A-treated Brl3a cells and attenuated the infiltration of J774A.1 macrophages induced by Con A. Moreover, quercetin effectively inhibited the expression of proinflammatory cytokines including interleukin-1ß (IL-1ß) by Con A. Furthermore, quercetin decreased hepatocyte apoptosis and ferroptosis levels in the macrophage-induced hepatocyte injury model. In conclusion, our study indicates that quercetin alleviates macrophage-induced hepatocyte damage by reducing the inflammatory response, apoptosis and ferroptosis. Our work suggests that quercetin might be a potential therapeutic strategy for AIH.


Subject(s)
Anti-Inflammatory Agents , Apoptosis , Ferroptosis , Hepatocytes , Macrophages , Quercetin , Quercetin/pharmacology , Quercetin/therapeutic use , Animals , Hepatocytes/drug effects , Hepatocytes/metabolism , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Ferroptosis/drug effects , Apoptosis/drug effects , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Line , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Hepatitis, Autoimmune/metabolism , Hepatitis, Autoimmune/etiology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/immunology , Concanavalin A , Cytokines/metabolism
15.
Carbohydr Polym ; 337: 122139, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38710550

ABSTRACT

A novel RG-I pectin-like polysaccharide, YJ3A1, was purified from the flowers of Rosa chinensis and its structure and hepatoprotective effect in vivo and in vitro were investigated. The backbone of this polysaccharide is mainly composed of 1, 4-galactan, 1, 4-linked α-GalpA and 1, 2-linked α-Rhap disaccharide repeating unit attached by 1, 6-linked ß-Galp or 1, 5-linked α-Araf on C-4 of the Rhap. Interestingly, oral administration of YJ3A1 significantly ameliorates NASH-associated inflammation, oxidative stress and fibrosis and does not affect the liver morphology of normal mice at a dose of 50 mg/kg. The mechanism study suggests that the biological activity may associate to inactivating of high-mobility group box 1 protein (HMGB1)/TLR4/NF-κB and Akt signaling pathways by restraining the expression and release of HMGB1, thereby impeding the effect of NASH. The current findings outline a novel leading polysaccharide for new drug candidate development against NASH.


Subject(s)
HMGB1 Protein , NF-kappa B , Non-alcoholic Fatty Liver Disease , Pectins , Rosa , Signal Transduction , Toll-Like Receptor 4 , Animals , Rosa/chemistry , Toll-Like Receptor 4/metabolism , HMGB1 Protein/metabolism , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction/drug effects , Mice , Pectins/pharmacology , Pectins/chemistry , Pectins/isolation & purification , Male , Humans , Inflammation/drug therapy , Inflammation/metabolism , Mice, Inbred C57BL , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Oxidative Stress/drug effects
16.
Balkan Med J ; 41(3): 193-205, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38700358

ABSTRACT

Background: Paclitaxel (PAX) is a widely used chemotherapy drug for various cancer types but often induces significant toxicity in multiple organ systems. Silymarin (SIL), a natural flavonoid, has shown therapeutic potential due to its multiple benefits. Aims: To evaluate the therapeutic efficacy of SIL in mitigating liver and kidney damage induced by PAX in rats, focusing on oxidative stress, inflammation, and apoptosis pathways. Study Design: Experimental animal model. Methods: The study included 28 male Wistar rats aged 12-14 weeks weighing 270-300 g. The rats were divided into four groups: control, SIL, PAX, and PAX + SIL, with seven in each group. The rats received intraperitoneal (i.p.) injections at a dose of 2 mg per kilogram of body weight of PAX for 5 successive days, followed by oral gavage with 200 mg/kg body mass of SIL for 10 uninterrupted days. We examined the effect of SIL on specific serum biochemical parameters using an autoanalyzer and rat-specific kits. The spectrophotometric methods was used to investigate oxidative stress indicators in kidney and liver tissues. Aquaporin-2 (AQP-2), B-cell lymphoma-2 (Bcl-2), cysteine aspartate-specific protease-3 (caspase-3), interleukin-6 (IL-6), nuclear factor kappa B (NF-κB), and streptavidin-biotin staining were used to assess immunoreactivity in PAX-induced liver and kidney injury models. Results: SIL treatment significantly reduced serum levels of alanine aminotransferase, aspartate aminotransferase, creatinine, urea, and C-reactive protein, indicating its effectiveness in treating PAX-induced liver and kidney injury. SIL treatment significantly reduced oxidative stress by increasing essential antioxidant parameters, such as superoxide dismutase, catalase, glutathione peroxidase, and glutathione. It also reduced malondialdehyde levels in liver and kidney tissues of SIL-PAX groups (p < 0.05). SIL administration reduced NF-κB, caspase-3, and IL-6 expression while increasing Bcl-2 and AQP2 levels in liver and kidney tissues of rats treated with SIL and PAX (p < 0.05). Conclusion: Our findings indicate the potential of SIL to alleviate PAX-induced liver and kidney damage in rats by reducing oxidative stress, inflammation, and apoptotic processes.


Subject(s)
Apoptosis , Inflammation , Oxidative Stress , Paclitaxel , Rats, Wistar , Silymarin , Animals , Oxidative Stress/drug effects , Rats , Male , Apoptosis/drug effects , Inflammation/drug therapy , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Silymarin/pharmacology , Silymarin/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/etiology , Antioxidants/pharmacology , Antioxidants/therapeutic use , Liver/drug effects , Kidney/drug effects , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 644-651, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708496

ABSTRACT

OBJECTIVE: To observe the effect of Shenqi Chongcao (SQCC) Formula on the ASS1/src/STAT3 signaling pathway in a rat model of lung fibrosis and explore its therapeutic mechanism. METHODS: A total of 120 male SD rats were divided equally into 5 groups, including a blank control group with saline treatment and 4 groups of rat models of idiopathic pulmonary fibrosis induced by intratracheal instillation of bleomycin. One day after modeling, the rat models were treated with daily gavage of 10 mL/kg saline, SQCC decoction (0.423 g/kg), pirfenidone (10 mL/kg), or intraperitoneal injection of arginine deiminase (ADI; 2.25 mg/kg, every 3 days) for 28 days. After the treatments, the lung tissues of the rats were collected for calculating the lung/body weight ratio, observing histopathology using HE and Masson staining, and analyzing the inflammatory cells in BALF using Giemsa staining. Serum chemokine ligand 2 (CCL2) and transforming growth factor-ß1 (TGF-ß1) levels were measured with ELISA. The protein expressions of src, p-srcTry529, STAT3, and p-STAT3Try705 and the mRNA expressions of ASS1, src and STAT3 in the lung tissues were detected using Western blotting and RT-qPCR. RESULTS: The neutrophil, macrophage and lymphocyte counts and serum levels of CCL2 and TGF-ß1 were significantly lower in SQCC, pirfenidone and ADI treatment groups than in the model group at each time point of measurement (P < 0.05). P-srcTry529 and p-STAT3Try705 protein expression levels and ASS1, src, and STAT3 mRNA in the lung tissues were also significantly lower in the 3 treatment groups than in the model group (P < 0.05). CONCLUSION: SQCC Formula can alleviate lung fibrosis in rats possibly by activating the ASS1/src/STAT3 signaling pathway in the lung tissues.


Subject(s)
Drugs, Chinese Herbal , Pulmonary Fibrosis , Rats, Sprague-Dawley , STAT3 Transcription Factor , Signal Transduction , Animals , STAT3 Transcription Factor/metabolism , Rats , Male , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Signal Transduction/drug effects , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Lung/metabolism , Lung/pathology , Lung/drug effects , Transforming Growth Factor beta1/metabolism , Inflammation/metabolism , Inflammation/drug therapy , Disease Models, Animal , Bleomycin , Chemokine CCL2/metabolism , src-Family Kinases/metabolism
18.
Clin Perinatol ; 51(2): 497-510, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705654

ABSTRACT

This review examines the complexities of preterm birth (PTB), emphasizes the pivotal role of inflammation in the pathogenesis of preterm labor, and assesses current available interventions. Antibiotics, progesterone analogs, mechanical approaches, nonsteroidal anti-inflammatory drugs, and nutritional supplementation demonstrate a limited efficacy. Tocolytic agents, targeting uterine activity and contractility, inadequately prevent PTB by neglecting to act on uteroplacental inflammation. Emerging therapies targeting toll-like receptors, chemokines, and interleukin receptors exhibit promise in mitigating inflammation and preventing PTB.


Subject(s)
Premature Birth , Tocolytic Agents , Humans , Pregnancy , Female , Premature Birth/prevention & control , Tocolytic Agents/therapeutic use , Infant, Newborn , Inflammation/drug therapy , Inflammation/prevention & control , Obstetric Labor, Premature/prevention & control
19.
Front Immunol ; 15: 1344098, 2024.
Article in English | MEDLINE | ID: mdl-38711511

ABSTRACT

Inflammatory responses, especially chronic inflammation, are closely associated with many systemic diseases. There are many ways to treat and alleviate inflammation, but how to solve this problem at the molecular level has always been a hot topic in research. The use of nanoparticles (NPs) as anti-inflammatory agents is a potential treatment method. We synthesized new hollow cerium oxide nanomaterials (hCeO2 NPs) doped with different concentrations of Cu5.4O NPs [the molar ratio of Cu/(Ce + Cu) was 50%, 67%, and 83%, respectively], characterized their surface morphology and physicochemical properties, and screened the safe concentration of hCeO2@Cu5.4O using the CCK8 method. Macrophages were cultured, and P.g-lipopolysaccharide-stimulated was used as a model of inflammation and co-cultured with hCeO2@Cu5.4O NPs. We then observe the effect of the transcription levels of CTSB, NLRP3, caspase-1, ASC, IL-18, and IL-1ß by PCR and detect its effect on the expression level of CTSB protein by Western blot. The levels of IL-18 and IL-1ß in the cell supernatant were measured by enzyme-linked immunosorbent assay. Our results indicated that hCeO2@Cu5.4O NPs could reduce the production of reactive oxygen species and inhibit CTSB and NLRP3 to alleviate the damage caused by the inflammatory response to cells. More importantly, hCeO2@Cu5.4O NPs showed stronger anti-inflammatory effects as Cu5.4O NP doping increased. Therefore, the development of the novel nanomaterial hCeO2@Cu5.4O NPs provides a possible new approach for the treatment of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents , Cerium , Copper , Inflammation , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Cerium/pharmacology , Cerium/chemistry , Signal Transduction/drug effects , Animals , Mice , Inflammation/drug therapy , Inflammation/metabolism , Anti-Inflammatory Agents/pharmacology , Nanoparticles , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
20.
Brain Behav ; 14(5): e3515, 2024 May.
Article in English | MEDLINE | ID: mdl-38702895

ABSTRACT

INTRODUCTION: Maternal sleep deprivation (MSD), which induces inflammation and synaptic dysfunction in the hippocampus, has been associated with learning and memory impairment in offspring. Melatonin (Mel) has been shown to have anti-inflammatory, antioxidant, and neuroprotective function. However, the beneficial effect of Mel on MSD-induced cognitive impairment and its mechanisms are unknown. METHODS: In the present study, adult offspring suffered from MSD were injected with Mel (20 mg/kg) once a day during postnatal days 61-88. The cognitive function was evaluated by the Morris water maze test. Levels of proinflammatory cytokines were examined by enzyme-linked immunosorbent assay. The mRNA and protein levels of synaptic plasticity associated proteins were examined using reverse transcription-polymerase chain reaction and western blotting. RESULTS: The results showed that MSD impaired learning and memory in the offspring mice. MSD increased the levels of interleukin (IL)-1creIL-6, and tumor necrosis factor-α and decreased the expression levels of brain-derived neurotrophic factor, tyrosine kinase receptor B, postsynaptic density protein-95, and synaptophysin in the hippocampus. Furthermore, Mel attenuated cognitive impairment and restored markers of inflammation and synaptic plasticity to control levels. CONCLUSIONS: These findings indicated that Mel could ameliorate learning and memory impairment induced by MSD, and these beneficial effects were related to improvement in inflammation and synaptic dysfunction.


Subject(s)
Hippocampus , Melatonin , Memory Disorders , Neuronal Plasticity , Sleep Deprivation , Animals , Melatonin/pharmacology , Melatonin/administration & dosage , Sleep Deprivation/complications , Sleep Deprivation/drug therapy , Sleep Deprivation/physiopathology , Mice , Male , Hippocampus/metabolism , Hippocampus/drug effects , Female , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/physiopathology , Neuronal Plasticity/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Pregnancy , Maternal Deprivation , Cognitive Dysfunction/etiology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Neuroinflammatory Diseases/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...