Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
J Control Release ; 369: 604-616, 2024 May.
Article in English | MEDLINE | ID: mdl-38582337

ABSTRACT

Corneal stromal fibrosis is a common cause of visual impairment resulting from corneal injury, inflammation and surgery. Therefore, there is an unmet need for inhibiting corneal stromal fibrosis. However, bioavailability of topical eye drops is very low due to the tear and corneal barriers. In situ delivery offers a unique alternative to improve efficacy and minimize systemic toxicity. Herein, a drug delivery platform based on thermoresponsive injectable hydrogel/nano-micelles composite with in situ drug-controlled release and long-acting features is developed to prevent corneal scarring and reduce corneal stromal fibrosis in lamellar keratoplasty. The in-situ gelation hydrogels enabled direct delivery of celastrol to the corneal stroma. In vivo evaluation with a rabbit anterior lamellar keratoplasty model showed that hydrogel/micelles platform could effectively inhibit corneal stromal fibrosis. This strategy achieves controlled and prolonged release of celastrol in the corneal stroma of rabbit. Following a single corneal interlamellar injection, celastrol effectively alleviated fibrosis via mTORC1 signal promoting autophagy and inhibiting TGF-ß1/Smad2/3 signaling pathway. Overall, this strategy demonstrates promise for the clinical application of celastrol in preventing corneal scarring and reducing corneal stromal fibrosis post-lamellar keratoplasty, highlighting the potential benefits of targeted drug delivery systems in ocular therapeutics.


Subject(s)
Corneal Transplantation , Hydrogels , Pentacyclic Triterpenes , Animals , Rabbits , Pentacyclic Triterpenes/administration & dosage , Hydrogels/administration & dosage , Corneal Transplantation/methods , Cicatrix/prevention & control , Cicatrix/drug therapy , Delayed-Action Preparations , Fibrosis , Drug Delivery Systems , Cornea/drug effects , Cornea/metabolism , Triterpenes/administration & dosage , Drug Liberation , Corneal Stroma/drug effects , Humans
2.
Avian Dis ; 68(1): 25-32, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38687104

ABSTRACT

Acute myocardial injury (AMI) induced by lipopolysaccharide (LPS) can cause cardiovascular dysfunction and lead to death in poultry. Traditional antibiotic therapy has been found to have many limitations and negative effects. Asiatic acid (AA) is a naturally occurring pentacyclic triterpenoid that is extracted from Centella asiatica and has anti-inflammatory, antioxidant, and anticancer pharmacological properties. Previously, we studied the effect of AA on LPS-induced liver and kidney injury; however, the impact of AA on LPS-induced AMI remained unclear. Sixty 1-day-old broilers were randomly divided into control group, LPS group, LPS + AA 15 mg/kg group, LPS + AA 30 mg/kg group, LPS + AA 60 mg/kg group, and control + AA 60 mg/kg group. The histopathology of cardiac tissues was detected by hematoxylin and eosin (H&E) staining. The mRNA and protein expressions related to mitochondrial dynamics and mitophagy were detected by quantitative real-time PCR, western blot, immunofluorescence, and immunohistochemistry. Disorganized myocardial cells and fractured myocardial fibers were found in the LPS group, and obvious red-blood-cell filling can be seen in the gaps between the myocardial fibers in the low-dose AA group. Nevertheless, the medium and high dose of AA obviously attenuated these changes. Our results showed that AA significantly restored the mRNA and protein expressions related to mitochondrial dynamic through further promoting mitophagy. This study revealed the effect of AA on LPS-induced AMI in broilers. Mechanically, AA regulated mitochondrial dynamic homeostasis and further promoted mitophagy. These novel findings indicate that AA may be a potential drug for LPS-induced AMI in broilers.


El ácido asiático como mitigante de las lesiones miocárdicas agudas inducidas por lipopolisacáridos al promover la mitofagia y regular la dinámica mitocondrial en pollos de engorde. La lesión miocárdica aguda (con siglas en inglés IAM) inducida por lipopolisacáridos (LPS) puede causar disfunción cardiovascular y provocar la muerte en las aves comerciales. Se ha descubierto que la terapia tradicional con antibióticos tiene muchas limitaciones y efectos negativos. El ácido asiático (AA) es un triterpenoide pentacíclico natural que se extrae de la planta Centella asiática y que tiene propiedades farmacológicas antiinflamatorias, antioxidantes y anticancerígenas. Anteriormente, se estudió el efecto del ácido asiático sobre la lesión hepática y renal inducida por lipopolisacáridos; sin embargo, el impacto del ácido asiático en las lesiones miocárdicas agudas inducidas por lipopolisacáridos continua sin estar completamente determinada. Sesenta pollos de engorde de un día de edad se dividieron aleatoriamente en los siguientes grupos experimentales: grupo control, grupo que recibió LPS solamente, grupo LPS + ácido asiático 15 mg/kg, grupo LPS + ácido asiático 30 mg/kg, grupo LPS + ácido asiático 60 mg/kg y control + ácido asiático 60 mg./kg grupo. La histopatología de los tejidos cardíacos se detectó mediante tinción con hematoxilina y eosina (H&E). Las expresiones de ARN mensajero y proteínas relacionadas con la dinámica mitocondrial y la mitofagia se detectaron mediante PCR cuantitativa en tiempo real, inmunotransferencia Western, inmunofluorescencia e inmunohistoquímica. Se encontraron células miocárdicas desorganizadas y fibras miocárdicas fracturadas en el grupo que recibió lipopolisacáridos, y se puede observar un evidente acúmulo de glóbulos rojos en los espacios entre las fibras miocárdicas en el grupo de dosis bajas de ácido asiático. Sin embargo, las dosis medias y altas de ácido asiático obviamente atenuaron estos cambios. Nuestros resultados mostraron que el ácido asiático restableció significativamente las expresiones de ARN mensajero y proteínas relacionadas con la dinámica mitocondrial mediante la promoción adicional de la mitofagia. Este estudio reveló el efecto del ácido asiático sobre las lesiones miocárdicas agudas inducidas por lipopolisacáridos en pollos de engorde. Basicamente, el ácido asiático reguló la homeostasis dinámica mitocondrial y promovió aún más la mitofagia. Estos nuevos hallazgos indican que el ácido asiático puede ser un fármaco potencial para mitigar lesiones miocárdicas agudas inducidas por lipopolisacáridos en pollos de engorde.


Subject(s)
Chickens , Lipopolysaccharides , Mitophagy , Pentacyclic Triterpenes , Poultry Diseases , Animals , Pentacyclic Triterpenes/pharmacology , Pentacyclic Triterpenes/administration & dosage , Poultry Diseases/chemically induced , Mitophagy/drug effects , Mitochondrial Dynamics/drug effects , Random Allocation
3.
Metab Brain Dis ; 37(6): 1773-1785, 2022 08.
Article in English | MEDLINE | ID: mdl-35554794

ABSTRACT

Aluminium (Al) is a potent neurotoxic metal known to cause neurodegeneration. Al exposure causes oxidative stress by accumulation of reactive oxygen species, followed by the activation of neuronal cell death in the brain. Asiatic acid (AA), the major bioactive compound of Centella asiatica (a medicinal plant), act as multifunctional drug as well as an antioxidant. Thus, the present study aimed to investigate the potential neuroprotective effect of AA against Al neurotoxicity. Rats were orally administered aluminium chloride (AlCl3; 100 mg/kg b. wt.) dissolved in distilled water for 8 weeks or AA (75 mg/kg b. wt.) in combination with AlCl3. The results showed that AlCl3-intoxication causes significant impairment of memory, enhances anxiety-like behavior, acetyl cholinesterase (AChE) activity, malondialdehydes (MDA) level, and concomitant decrease in the activities of superoxide dismutase (SOD) and catalase (CAT) in the cortex and hippocampus regions of rat brain. In addition, AlCl3-intoxication enhanced neuronal loss and reactive astrogliosis in both regions. However, co-administration of AA with AlCl3 significantly attenuated the behavioral alterations, restored SOD and CAT activities, while reduced AChE activity and MDA content. Further, the study demonstrated that AA attenuates neuronal loss and reactive astrogliosis in rat brain. In conclusion, the study suggests that AA protects rat brain from Al neurotoxicity by inhibiting oxidative stress, neuronal loss and reactive astrogliosis.


Subject(s)
Aluminum Chloride , Antioxidants , Mental Disorders , Neuroprotective Agents , Neurotoxicity Syndromes , Pentacyclic Triterpenes , Aluminum Chloride/antagonists & inhibitors , Aluminum Chloride/toxicity , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Astrocytes/metabolism , Gliosis , Mental Disorders/chemically induced , Mental Disorders/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/prevention & control , Oxidative Stress , Pentacyclic Triterpenes/administration & dosage , Pentacyclic Triterpenes/therapeutic use , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
4.
Life Sci ; 296: 120435, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35247437

ABSTRACT

AIMS: Glioblastoma (GBM) is the most common and deadliest type of brain cancer, and the current therapeutic options are not curative, imposing the need for novel strategies. Asiatic acid (AA) is a natural compound and has been explored due to its anti-glioma activity and lower toxicity to healthy tissues compared with conventional chemotherapeutic agents. However, its poor water-solubility is an obstacle for clinical application. Poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) were proposed in this work for Asiatic acid (AA) delivery. MAIN METHODS: A central composite design was implemented to optimize the NPs, and their surface was further modified with transferrin (Tf), for targeted delivery to GBM cells. The anti-glioma activity of the NPs was studied in vitro using human GBM cells and immortalized human astrocytes. KEY FINDINGS: The NPs exhibited a mean size smaller than 200 nm, with low polydispersity and negative zeta potential, indicating their suitability for brain tumor delivery. The NPs also exhibited high encapsulation efficiency and maintained a slow and controlled release of AA for 20 days. In vitro cell studies showed that NPs were able to maintain the anti-glioma activity of the natural compound and that the surface modification with Tf molecules was able to increase the cellular uptake in GBM cells, enhancing their selectivity and decreasing toxicity in healthy cells. SIGNIFICANCE: Overall, this work provided guidance for designing brain-targeting delivery systems of natural compounds.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Nanoparticles/administration & dosage , Pentacyclic Triterpenes/administration & dosage , Transferrin/chemistry , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Astrocytes/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems/methods , Drug Liberation , Glioblastoma/pathology , Humans , Nanoparticles/chemistry , Pentacyclic Triterpenes/pharmacokinetics , Pentacyclic Triterpenes/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Receptors, Transferrin/metabolism
5.
Drug Deliv ; 29(1): 454-465, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35119317

ABSTRACT

Rheumatoid arthritis (RA) is an ordinarily occurring autoimmune disease with systemic inflammatory. Targeted drug delivery systems have many successful applications in the treatment of rheumatoid arthritis. In order to develop nanoparticles for targeted delivery of Celastrol (Cel) to rheumatoid arthritis and specific drug release, the dextran sulfate (DS) was modified as the targeting molecular by binding to the scavenger receptor of macrophage. The dextran-sulfate-PVGLIG-celastrol (DS-PVGLIG-Cel), named DPC, amphiphilic polymeric prodrug was synthesized and characterized. The resulting DPC@Cel micelles had the average size of 189.9 nm. Moreover, the micelles had ultrahigh entrapment efficiency (about 44.04%) and zeta potential of -11.91 mV. In the in vitro release study, due to the excessive production of matrix metalloproteinase-2 (MMP-2) at the inflammatory joint, the MMP-2 reactive peptide was used to crack in the inflammatory microenvironment to accelerate the release of Cel. The results have shown that the nanoparticles can effectively deliver Cel to activated macrophages and significantly improve the bioavailability. In vivo experiments showed that DPC@Cel have better anti-rheumatoid arthritis effects and lower systemic toxicity than free Cel. This study provided a new therapeutic strategy for the treatment of RA.


Subject(s)
Arthritis, Rheumatoid/pathology , Dextran Sulfate/chemistry , Matrix Metalloproteinase 2/chemistry , Nanoparticles/chemistry , Pentacyclic Triterpenes/pharmacology , Animals , Cell Survival/drug effects , Chemistry, Pharmaceutical , Disease Models, Animal , Drug Carriers/chemistry , Drug Liberation , Mice , Micelles , Particle Size , Pentacyclic Triterpenes/administration & dosage , RAW 264.7 Cells , Random Allocation , Rats , Rats, Sprague-Dawley , Surface Properties
6.
Drug Deliv ; 29(1): 89-98, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34964425

ABSTRACT

Celastrol is a promising antitumor drug candidate, but the poor water solubility and cytotoxicity limit its clinical application. Herein, we synthesized a Celastrol (Cel)-chitosan oligosaccharide (CSO) conjugate (Cel-CSO) for drug delivery. Celastrol was conjugated to a CSO backbone via amide bond formation, which was verified by infrared spectrum (IR) analyses. The Cel-CSO contained ∼10 wt% of Celastrol showed excellent aqueous solubility (18.6 mg/mL) in comparation with the parent Celastrol. Cel-CSO significantly inhibited tumor growth, induced apoptosis, and effectively suppressed tumor metastasis in human pancreatic cancer cells (BxPC-3). While the cytotoxicity of Cel-CSO in hepatic cells (HL7702) was lower than that of the free Celastrol. Cel-CSO enhanced the anticancer efficacy, promoted the circulation time of Celastrol, and reduced the subacute toxicity, which indicated that CSO can be a promising Celastrol delivery system for pancreatic cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Chitosan/chemistry , Pancreatic Neoplasms/pathology , Pentacyclic Triterpenes/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chemistry, Pharmaceutical , Drug Liberation , Humans , Mice , Mice, Inbred BALB C , Oligosaccharides/chemistry , Particle Size , Pentacyclic Triterpenes/administration & dosage , Solubility , Tumor Burden/drug effects
7.
Drug Deliv ; 28(1): 2534-2547, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34854788

ABSTRACT

Liver fibrosis is a major global health concern. Management of chronic liver disease is severely restricted in clinics due to ineffective treatment approaches. However, a lack of targeted therapy may aggravate this condition. Asiatic acid (AA), a pentacyclic triterpenoid acid, can effectively protect the liver from hepatic disorders. However, the pharmaceutical application of AA is limited by low oral bioavailability and poor targeting efficiency. This study synthesized a novel liver-targeting material from PEG-SA, chemically linked to ursodeoxycholic acid (UA), and utilized it to modify AA nanostructured lipid carriers (UP-AA-NLC) with enhanced targeting and improved efficacy. The formulation of UP-AA-NLC was optimized via the Box-Behnken Experimental Design (BBD) and characterized by size, zeta potential, TEM, DSC, and XRD. Furthermore, in vitro antifibrotic activity and proliferation of AA and NLCs were assessed in LX-2 cells. The addition of UP-AA-NLC significantly stimulated the TGF-beta1-induced expression of α-SMA, FN1, and Col I α1. In vivo near-infrared fluorescence imaging and distribution trials in rats demonstrated that UP-AA-NLC could significantly improve oral absorption and liver-targeting efficiency. Oral UP-AA-NLC greatly alleviated carbon tetrachloride-induced liver injury and fibrosis in rats in a dosage-dependent manner, as reflected by serum biochemical parameters (AST, ALT, and ALB), histopathological features (H&E and Masson staining), and antioxidant activity parameters (SOD and MDA). Also, treatment with UP-AA-NLC lowered liver hydroxyproline levels, demonstrating a reduction of collagen accumulation in the fibrotic liver. Collectively, optimized UP-AA-NLC has potential application prospects in liver-targeted therapy and holds great promise as a drug delivery system for treating liver diseases.


Subject(s)
Liver Cirrhosis/drug therapy , Nanostructures/chemistry , Pentacyclic Triterpenes/pharmacology , Animals , Carbon Tetrachloride/pharmacology , Cell Line , Cell Proliferation/drug effects , Chemistry, Pharmaceutical , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Liberation , Lipids/chemistry , Liver/drug effects , Liver Function Tests , Male , Mice , Mice, Inbred ICR , Particle Size , Pentacyclic Triterpenes/administration & dosage , Pentacyclic Triterpenes/pharmacokinetics , Polyethylene Glycols/chemistry , Rats , Rats, Sprague-Dawley , Surface Properties , Ursodeoxycholic Acid/chemistry
8.
J Nanobiotechnology ; 19(1): 341, 2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34702273

ABSTRACT

Effectively promoting corneal allograft survival remains a challenge in corneal transplantation. The emerging therapeutic agents with high pharmacological activities and their appropriate administration routes provide attractive solutions. In the present study, a celastrol-loaded positive nanomedicine (CPNM) was developed to enhance corneal penetration and to promote corneal allograft survival. The in vitro, in vivo and ex vivo results demonstrated the good performance of CPNM prolonging the retention time on ocular surface and opening the tight junction in cornea, which resulted in enhanced corneal permeability of celastrol. Both in vitro and in vivo results demonstrated that celastrol inhibited the recruitment of M1 macrophage and the expression of TLR4 in corneal allografts through the TLR4/MyD88/NF-κB pathway, thereby significantly decreasing secretion of multiple pro-inflammatory cytokines to promote corneal allograft survival. This is the first celastrol-based topical instillation against corneal allograft rejection to provide treatment more potent than conventional eye drops for ocular anterior segment diseases.


Subject(s)
Corneal Transplantation , Graft Survival/drug effects , Nanomedicine/methods , Pentacyclic Triterpenes/pharmacology , Animals , Cytokines/metabolism , Female , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Ophthalmic Solutions/administration & dosage , Ophthalmic Solutions/pharmacology , Pentacyclic Triterpenes/administration & dosage , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Rats, Wistar
9.
Drug Deliv ; 28(1): 1962-1971, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34565273

ABSTRACT

Betulinic acid (3ß-Hydroxy-20(29)-lupaene-28-oic acid, BA) has excellent anti-cancer activity but poor solubility and low bioavailability. To improve the antitumor activity of BA, a polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol (PVCL-PVA-PEG) graft copolymer (Soluplus) encapsulated BA micelle (Soluplus-BA) was fabricated. The Soluplus-BA micelles presented a mean size of 54.77 ± 1.26 nm and a polydispersity index (PDI) of 0.083. The MTT assay results showed that Soluplus-BA micelles increased the inhibitory effect of BA on MDA-MB-231 cells, mainly due to the enhanced accumulation of reactive oxygen species (ROS) and the destruction of mitochondrial membrane potential (MMP). Soluplus-BA micelles induced the DNA double-strand breaks (DSBs) as the γH2AX foci increased. Moreover, Soluplus-BA also inhibited the tube formation and migration of human umbilical vein endothelial cells (HUVECs), and inhibited the neovascularization of the chicken chorioallantoic membrane (CAM). This angiogenesis inhibitory effect may be accomplished by regulating the HIF-1/VEGF-FAK signaling pathway. The in vivo study confirmed the improved anti-tumor effect of Soluplus-BA and its inhibitory effect on angiogenesis, demonstrating the possibility of Soluplus-BA as an effective anti-breast cancer drug delivery system.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/pathology , Micelles , Pentacyclic Triterpenes/administration & dosage , Polyethylene Glycols/chemistry , Polyvinyls/chemistry , Animals , Animals, Outbred Strains , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Chickens , Drug Carriers/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Neovascularization, Pathologic/metabolism , Pentacyclic Triterpenes/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Surface Properties , Vascular Endothelial Growth Factor A/drug effects , Betulinic Acid
10.
Pharm Biol ; 59(1): 1351-1358, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34590530

ABSTRACT

CONTEXT: Sepsis is a systemic inflammatory disease; pristimerin exhibits strong antibacterial, anti-inflammatory and antioxidant properties. OBJECTIVES: We explored whether pristimerin protected against cognitive dysfunction and neuroinflammation in C57BL/6 J mice with sepsis-induced brain injuries. MATERIALS AND METHODS: Sepsis was induced by intraperitoneal administration of 2 mg/kg lipopolysaccharide (LPS). C57BL/6 J mice were separated into four groups (n = 10 per group): positive control, negative control, pristimerin 10 mg/kg and pristimerin 100 mg/kg. Pristimerin was administered orally for 28 days prior to LPS administration and for six days thereafter. Behavioural changes were assessed one day after LPS administration using the Morris water maze and via neurological dysfunction scoring. Molecular pathogenesis was explored by measurement of malondialdehyde, superoxide dismutase, reactive oxygen species and inflammatory cytokine levels in mouse brains. Neuronal apoptosis was evaluated using the TUNEL assay. The levels of p-Akt/Akt, p-PI3K/PI3K, mTOR, Bax, Bcl-2 and caspase-3 proteins were determined via Western blotting. RESULTS: Pristimerin improved cognitive function and reduces the neurological score to 1.15 ± 0.03. Pristimerin significantly reduced all cytokine levels: TNF-α by 18 ± 0.6 pg/mg, IL-1ß by 43 ± 1.3 pg/mg and IL-6 by 34 ± 1.12 pg/mg. There was significant (p < 0.01) improvement in PI3K/Akt signalling and histopathological changes in the brain tissue of sepsis induced brain injured rats. CONCLUSIONS: Pristimerin ameliorated neuronal injury by regulating PI3K/Akt signalling in mice with sepsis-induced brain injuries. Pristimerin may merit further development for clinical applications.


Subject(s)
Brain Injuries/prevention & control , Cognitive Dysfunction/prevention & control , Pentacyclic Triterpenes/pharmacology , Sepsis/drug therapy , Animals , Apoptosis/drug effects , Brain Injuries/etiology , Cognition/drug effects , Cognitive Dysfunction/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/prevention & control , Pentacyclic Triterpenes/administration & dosage , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sepsis/complications , Signal Transduction/drug effects
11.
Biomed Pharmacother ; 143: 112145, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34507119

ABSTRACT

Lupeol is a pentacyclic triterpene with known anti-inflammatory effects. However, its role in the treatment of noninfectious uveitis has not been explored. This work investigated anti-inflammatory activity of lupeol in ocular tissues with in vitro and in vivo models. First, we evaluated the effect of lupeol (100 µM) on inflammatory response induced by lipopolysaccharide (LPS) in retinal pigment epithelium cells (ARPE-19) by measuring levels of released interleukins (IL-6 and IL-8). Then, we investigated the anti-inflammatory action of intravitreal lupeol in a rodent model of panuveitis induced by Mycobacterium bovis Calmette-Guérin Bacillus (BCG). Rats were submitted to electroretinography and clinical analyses on days 3, 7, and 15 after uveitis induction. In addition, histopathological analysis, and indirect quantification of myeloperoxidase (MPO) and N-acetylglucosaminidase (NAG) in the posterior segment were performed. Treatment with lupeol (100 µM) significantly decreased IL-6 and IL-8 levels in comparison to untreated LPS-activated ARPE-19 cells. This reduction was similar to that detected in ARPE-19 cells treated with dexamethasone. The results of the in vivo assay demonstrated that intravitreal lupeol is able to modulate inflammation in the anterior and posterior segment of the rat eyes, indicating that it should be further investigated as a novel potential candidate for management of uveitis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Eye/drug effects , Pentacyclic Triterpenes/administration & dosage , Uveitis/drug therapy , Acetylglucosaminidase/metabolism , Animals , BCG Vaccine , Cell Line , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Eye/metabolism , Eye/pathology , Humans , Inflammation Mediators/metabolism , Intravitreal Injections , Lipopolysaccharides/toxicity , Male , Peroxidase/metabolism , Rats, Wistar , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Uveitis/chemically induced , Uveitis/metabolism , Uveitis/pathology
12.
Molecules ; 26(14)2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34299450

ABSTRACT

The skin integrity is essential due to its pivotal role as a biological barrier against external noxious factors. Pentacyclic triterpenes stand as valuable plant-derived natural compounds in the treatment of skin injuries due to their anti-inflammatory, antioxidant, antimicrobial, and healing properties. Consequently, the primary aim of the current investigation was the development as well as the physicochemical and pharmaco-toxicological characterization of betulin- and lupeol-based oleogels (Bet OG and Lup OG) for topical application in skin injuries. The results revealed suitable pH as well as organoleptic, rheological, and textural properties. The penetration and permeation of Bet and Lup oleogels through porcine ear skin as well as the retention of both oleogels in the skin were demonstrated through ex vivo studies. In vitro, Bet OG and Lup OG showed good biocompatibility on HaCaT human immortalized cells. Moreover, Bet OG exerted a potent wound-healing property by stimulating the migration of the HaCaT cells. The in ovo results demonstrated the non-irritative potential of the developed formulations. Additionally, the undertaken in vivo investigation indicated a positive effect of oleogels treatment on skin parameters by increasing skin hydration and decreasing erythema. In conclusion, oleogel formulations are ideal for the local delivery of betulin and lupeol in skin disorders.


Subject(s)
Pentacyclic Triterpenes/administration & dosage , Skin/injuries , Triterpenes/administration & dosage , Administration, Cutaneous , Animals , Anti-Inflammatory Agents/pharmacology , Drug Compounding , Female , Mice , Organic Chemicals/chemistry , Organic Chemicals/pharmacology , Pentacyclic Triterpenes/pharmacology , Skin/metabolism , Swine , Triterpenes/pharmacology , Wound Healing/drug effects
13.
J Toxicol Environ Health A ; 84(22): 922-931, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34304725

ABSTRACT

Atopic dermatitis is a chronic inflammatory skin disease, of which incidence is closely related to exposure to environmental pollutants and allergens. Thymic stromal lymphopoietin (TSLP) plays an important role in the early stages of atopic dermatitis development by inducing Th2 immune responses. In addition, TSLP regulates activation of group 2 innate lymphoid cells (ILC2), promoting the pathogenesis of atopic dermatitis. The aim of this study was to investigate whether celastrol alleviated atopic dermatitis symptoms by regulating TSLP expression and ILC2 stimulation. Celastrol suppressed TSLP production in mouse keratinocyte cells by inhibiting NF-ĸB activation. Topical application of celastrol significantly improved atopic dermatitis symptoms induced by house dust mite (HDM) in NC/Nga mice as determined by dermatitis score and histological assessment. Celastrol decreased the levels of TSLP in atopic dermatitis skin lesions of HDM-stimulated NC/Nga mice. Celastrol reduced levels of Th2 cytokines including IL-4, IL-5, and IL-13 in atopic dermatitis skin lesions of NC/Nga mice. Further, celastrol significantly reduced ILC2 population in atopic dermatitis skin lesions of NC/Nga mice. These results indicate that topical application of celastrol improved atopic dermatitis symptoms by lowering TSLP levels and concomitant immune responses. Data demonstrated that reduced TSLP levels and associated lower number of ILC2 cells alleviate atopic dermatitis symptoms induced by house dust mite.


Subject(s)
Cytokines/immunology , Dermatitis, Atopic/drug therapy , Lymphocytes/drug effects , Pentacyclic Triterpenes/administration & dosage , Allergens/adverse effects , Allergens/immunology , Animals , Cell Line, Tumor , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Disease Models, Animal , Immunity, Innate/drug effects , Inflammation , Keratinocytes/drug effects , Keratinocytes/immunology , Lymphocytes/immunology , Mice , NF-kappa B/immunology , Pentacyclic Triterpenes/pharmacology , Pyroglyphidae/immunology , Skin/drug effects , Skin/immunology , Skin/pathology , Thymic Stromal Lymphopoietin
14.
Cancer Sci ; 112(8): 3218-3232, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34080260

ABSTRACT

Skp2 is overexpressed in multiple cancers and plays a critical role in tumor development through ubiquitin/proteasome-dependent degradation of its substrate proteins. Drugs targeting Skp2 have exhibited promising anticancer activity. Here, we identified a plant-derived Skp2 inhibitor, betulinic acid (BA), via high-throughput structure-based virtual screening of a phytochemical library. BA significantly inhibited the proliferation and migration of non-small cell lung cancer (NSCLC) through targeting Skp2-SCF E3 ligase both in vitro and in vivo. Mechanistically, BA binding to Skp2, especially forming H-bonds with residue Lys145, decreases its stability by disrupting Skp1-Skp2 interactions, thereby inhibiting the Skp2-SCF E3 ligase and promoting the accumulation of its substrates; that is, E-cadherin and p27. In both subcutaneous and orthotopic xenografts, BA significantly inhibited the proliferation and metastasis of NSCLC through targeting Skp2-SCF E3 ligase and upregulating p27 and E-cadherin protein levels. Taken together, BA can be considered a valuable therapeutic candidate to inhibit metastasis of NSCLC.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Enzyme Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Pentacyclic Triterpenes/administration & dosage , S-Phase Kinase-Associated Proteins/metabolism , A549 Cells , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Binding Sites , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Early Detection of Cancer , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Lung Neoplasms/metabolism , Male , Mice , Pentacyclic Triterpenes/pharmacology , Protein Binding/drug effects , S-Phase Kinase-Associated Proteins/chemistry , Xenograft Model Antitumor Assays , Betulinic Acid
15.
Phytomedicine ; 86: 153563, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33951569

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-associated mortality worldwide. Sorafenib (SORA), as a first-line therapeutic drug, has been used to treat HCC, but resistance poses a major limitation on the efficacy of SORA chemotherapy. Pristimerin (PRIS), a natural bioactive component isolated from various plant species in the Celastraceae and Hippocrateaceae families, has been reported to exhibit outstanding antitumor effects in several types of cells in vitro. PURPOSE: The aim of this study was to investigate whether PRIS can exert synergistic anti-tumor effects with the combination of SORA, and if so, through what mechanism. METHODS: Conditionally reprogrammed patient derived-primary hepatocellular carcinoma cells (CRHCs) were isolated from human liver cancer tissues and treated with SORA and PRIS. Cell proliferation, apoptosis, migration and tube formation ability were detected by DNA content quantification, flow cytometry, transwell assay and Matrigel-based angiogenesis assay. Gene and protein expression were assessed by qRT-PCR and Western blot respectively. RESULTS: Initially, we observed that the combination of the two drugs had a much stronger inhibitory effect on CRHCs growth than either drug alone. Moreover, the combination of 2 µM SORA and 1 µM PRIS exhibited a significant anti­migrative and anti-invaded effect on CRHCs, and remarkably inhibited capillary structure formation of Human Umbilical Vein Endothelial Cells (HUVECs). Furthermore, the combined treatment with SORA and PRIS synergistically induced intrinsic apoptosis in CRHCs, involving a caspase-4-dependent mechanism paralleled by an increased Bax/Bcl-xL ratio. These activities were mediated through ROS generation and the induction of endoplasmic reticulum (ER) stress and mitochondrial dysfunction. GRP78 silencing or ER stress inhibitor 4-phenylbutyric acid administration was revealed to abolish the anticancer effects of PRIS, indicating the critical role of GRP78 in mediating the bioactivity of PRIS. The present study also provides mechanistic evidence that PRIS modulated the Akt/FoxO1/p27kip1 signaling pathway, which is required for mitochondrial-mediated intrinsic apoptosis, activation of ER stress, and stimulation of caspase-4 induced by PRIS, and, consequently resulting in suppressed cell viability, migration and angiogenesis co-treated with SORA in CRHCs. CONCLUSION: Our results suggest the use of PRIS as sensitizers of chemotherapy paving the way for innovative and promising targeted chemotherapy-based therapeutic strategies in human HCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms/drug therapy , Adult , Aged , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endoplasmic Reticulum Chaperone BiP , Forkhead Box Protein O1/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Middle Aged , Pentacyclic Triterpenes/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sorafenib/administration & dosage , Tumor Cells, Cultured
16.
J Vet Pharmacol Ther ; 44(1): 47-57, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32845519

ABSTRACT

The naturally occurring betulinic acid (BA) and its derivative NVX-207 show anticancer effects against equine malignant melanoma (EMM) cells and a potent permeation in isolated equine skin in vitro. The aim of the study was to determine the in vivo concentration profiles of BA and NVX-207 in equine skin and assess the compounds' local and systemic tolerability with the intent of developing a topical therapy against EMM. Eight horses were treated percutaneously in a crossover design with 1% BA, 1% NVX-207 or a placebo in a respective vehicle twice a day for seven consecutive days with a seven-day washout period between each formulation. Horses were treated at the neck and underneath the tail. Concentration profiles of the compounds were assessed by high-performance liquid chromatography in the cervical skin. Clinical and histopathological examinations and blood analyses were performed. Higher concentrations of NVX-207 were found in the skin compared to BA. Good systemic tolerability and only mild local adverse effects were observed in all three groups. This study substantiates the topical application of BA and NVX-207 in further clinical trials with horses suffering from EMM; however, penetration and permeation of the compounds may be altered in skin affected by tumors.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Horses/metabolism , Pentacyclic Triterpenes/pharmacokinetics , Propanolamines/pharmacokinetics , Triterpenes/pharmacokinetics , Administration, Topical , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cross-Over Studies , Female , Male , Pentacyclic Triterpenes/administration & dosage , Pentacyclic Triterpenes/adverse effects , Permeability , Pilot Projects , Propanolamines/administration & dosage , Propanolamines/adverse effects , Triterpenes/administration & dosage , Triterpenes/adverse effects , Betulinic Acid
17.
Exp Clin Endocrinol Diabetes ; 129(5): 339-348, 2021 May.
Article in English | MEDLINE | ID: mdl-32176932

ABSTRACT

OBJECTIVE: High-fat diet (HFD) increases the risk of inflammatory reaction and acute arterial thrombosis. Celastrol has been confirmed to regulate inflammatory cytokine levels in atherosclerotic animal models. However, the anti-thrombotic effects of celastrol have remained to be fully demonstrated. The present study was performed to investigate the beneficial effect of celastrol in HFD-induced inflammatory reaction and thrombosis in apolipoprotein (apo)E-/- mice. MATERIALS AND METHODS: Thrombogenic mice model was established using HFD-fed apoE-/- mice. The levels of mRNA and protein were assayed by RT-qPCR and western blotting, respectively. Immunohistochemistry (IHC) staining was performed to measure the protein expression of matrix metalloproteinase-2 and matrix metalloproteinase-9 in the aortic endothelium of HFD-fed apoE-/- mice. RESULTS: The results demonstrated that the effect of HFD on inflammatory cytokines in mice with apoE-/- background was reversed by celastrol administration, and celastrol treatment inhibited the NOD-like receptor family, pyrin domain containing 3 (NLRP3)/caspase-1/interleukin-1ß signaling cascades in peripheral blood mononuclear cells from HFD-fed apoE-/- mice. In addition, HFD enhanced adenosine diphosphate-induced platelet aggregation in normal C57BL/6 and apoE-/- mice, while celastrol administration reversed this. Furthermore, celastrol inhibited the pro-thrombotic effects of HFD in apoE-/- mice, and the underlying mechanism was mediated, at least partially, through the suppression of matrix metalloproteinase-2 and -9 expression. CONCLUSIONS: Celastrol administration significantly attenuated HFD-induced inflammatory reaction, platelet aggregation and thrombosis in apoE-/- mice, and celastrol may be used as a drug for the prevention of HFD-induced inflammatory reaction and thrombus.


Subject(s)
Cytokines/drug effects , Diet, High-Fat/adverse effects , Inflammation/drug therapy , Pentacyclic Triterpenes/pharmacology , Thrombosis/drug therapy , Animals , Apolipoproteins E , Disease Models, Animal , Inflammation/blood , Inflammation/etiology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Pentacyclic Triterpenes/administration & dosage , Thrombosis/blood , Thrombosis/etiology , Thrombosis/immunology
18.
Food Chem Toxicol ; 147: 111919, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33301843

ABSTRACT

It has been shown that checkpoint kinase inhibitors can enhance chemosensitivity to gemcitabine by disrupting the replication stress response (RSR). In the present study, we aimed to describe the chemical synthetic lethal effects of the combination of gemcitabine and quinone-methide triterpenoid pristimerin in pancreatic cancer (PC) cells. The drug interaction assay indicated effective synergy between gemcitabine and pristimerin at sub-IC50 concentrations. Interestingly, pristimerin induced lysosomal degradation of checkpoint kinase 1 (Chk1), decreased the percentage of cells at the G1/S boundary and triggered significant double-stranded DNA breaks compared to gemcitabine treatment alone. Moreover, gemcitabine activated the phosphorylation of Chk1 and induced the formation of poly (ADP-ribose) polymers (PARs) as well as the accumulation of 53BP1, which was either partially or completely impaired by pristimerin. Meanwhile, pristimerin augmented the expression of γH2AX upon gemcitabine treatment. Finally, the combination of gemcitabine with pristimerin increased the apoptotic potential of PC cells. These results show that pristimerin acts as a naturally occurring inhibitor of RSR, and a novel therapeutic strategy of combining pristimerin and gemcitabine deserves further detailed investigation in PC models in vivo.


Subject(s)
Checkpoint Kinase 1/metabolism , DNA Repair/drug effects , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Pentacyclic Triterpenes/pharmacokinetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Survival , Checkpoint Kinase 1/genetics , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Drug Synergism , Humans , Pentacyclic Triterpenes/administration & dosage , Pentacyclic Triterpenes/pharmacology , Tumor Suppressor p53-Binding Protein 1/genetics , Gemcitabine
19.
Drug Des Devel Ther ; 14: 4659-4670, 2020.
Article in English | MEDLINE | ID: mdl-33173276

ABSTRACT

BACKGROUND: Ectopic pregnancy (EP) is an ectopic embryo implantation occurred outside the uterine cavity. Nowadays, more attention have garnered in fast and effective treatment with less side effects. Pristimerin is known as the clinical application for anti-cancer, and the effect on EP therapy is still unclear. MATERIALS AND METHODS: Trophoblast cell line HTR-8/SVneo was used; then, we performed cell counting kit-8 assay, wound healing assay, flow cytometry and real-time polymerase chain reaction analysis (RT-PCR) to detect the cell viability, migration ability, apoptosis and epithelial-mesenchymal transition (EMT) under pristimerin treatment. In addition, public bioinformatic database was used to discover the connection between molecular and genes. Finally, we used miRNA transfection and RT-PCR techniques to determine the underlying molecular mechanism. RESULTS: We revealed that pristimerin inhibited trophoblast cells proliferation, migration and EMT, while induced trophoblast cell apoptosis. Furthermore, expression of miR-542-5p, AGO2 and EGFR was suppressed in HTR-8/SVneo cells post pristimerin treatment, and miR-542-5p silence showed the same effect. Combing pristimerin treatment and miR-542-5p silence showed a synergistic action. CONCLUSION: Pristimerin could be an effective treatment to block embryo implantation by miR-542-5p and EGFR down-regulation.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/antagonists & inhibitors , MicroRNAs/antagonists & inhibitors , Pentacyclic Triterpenes/pharmacology , Trophoblasts/drug effects , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Structure , Pentacyclic Triterpenes/administration & dosage , Signal Transduction/drug effects , Structure-Activity Relationship , Trophoblasts/metabolism
20.
Int J Mol Sci ; 21(21)2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33114200

ABSTRACT

Plant-extracted triterpenoids belong to a class of bioactive compounds with pleotropic functions, including antioxidant, anti-cancer, and anti-inflammatory effects. In this work, we investigated the anti-inflammatory and anti-oxidative activities of a semisynthetic derivative of 18ßH-glycyrrhetinic acid (18ßH-GA), soloxolone methyl (methyl 2-cyano-3,12-dioxo-18ßH-olean-9(11),1(2)-dien-30-oate, or SM) in vitro on lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages and in vivo in models of acute inflammation: LPS-induced endotoxemia and carrageenan-induced peritonitis. SM used at non-cytotoxic concentrations was found to attenuate the production of reactive oxygen species and nitric oxide (II) and increase the level of reduced glutathione production by LPS-stimulated RAW264.7 cells. Moreover, SM strongly suppressed the phagocytic and migration activity of activated macrophages. These effects were found to be associated with the stimulation of heme oxigenase-1 (HO-1) expression, as well as with the inhibition of nuclear factor-κB (NF-κB) and Akt phosphorylation. Surprisingly, it was found that SM significantly enhanced LPS-induced expression of the pro-inflammatory cytokines interleukin-6 (IL-6), tumour necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß) in RAW264.7 cells via activation of the c-Jun/Toll-like receptor 4 (TLR4) signaling axis. In vivo pre-exposure treatment with SM effectively inhibited the development of carrageenan-induced acute inflammation in the peritoneal cavity, but it did not improve LPS-induced inflammation in the endotoxemia model.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Endotoxemia/drug therapy , Lipopolysaccharides/adverse effects , Macrophages/cytology , Pentacyclic Triterpenes/administration & dosage , Peritonitis/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/genetics , Endotoxemia/metabolism , Glutathione/metabolism , Glycyrrhetinic Acid/chemistry , Heme Oxygenase-1/genetics , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Membrane Proteins/genetics , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/pharmacology , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...