Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Steroids ; 196: 109245, 2023 08.
Article in English | MEDLINE | ID: mdl-37141980

ABSTRACT

Cassia occidentalis L., from Fabaceae family phytochemical screening, revealed several biologically active principles mainly flavonoids and anthraquinones. GLC analysis of the lipoidal matter afforded 12 hydrocarbons: 9-dodecyl-tetradecahydro-anthracene (48.97 %), 9-dodecyl-tetradecahydro-phenanthrene (14.43 %), and 6 sterols/triterpenes: isojaspisterol (11.99%) and fatty acids were palmitic acid (50 %), and Linoleic acid (16.06%). Column chromatography led to the isolation of fifteen compounds (1-15), elucidated using spectroscopic evidence. First report of undecanoic acid (4) from the family Fabaceae, while p-dimethyl amino-benzaldehyde (15) was first time isolated from a natural origin. Eight compounds isolated for the first time from C. occidentalis L.; ß-amyrin (1), ß-sitosterol (2), stigmasterol (3), camphor (5), lupeol (6), chrysin (7), pectolinargenin (8), and 1, 2, 5-trihydroxy anthraquinone (14) besides five known compounds previously isolated; apigenin (9), kaempferol (10), chrysophanol (11), physcion (12), and aloe-emodin (13). In-vivo evaluation of anti-inflammatory and analgesic effects of C. occidentalis L. extracts where the n-butanol and total extracts showed the highest activities. The percentage of the inhibitory effect of the n-butanol extract was 29.7 at a dose of 400 mg/Kg. Furthermore, identified phytoconstituents were docked into the active sites of enzymes nAChRs, COX-1, and COX-2 to evaluate binding affinity. Phyto-compounds Physcion, aloe-emodin, and chrysophanol were found to have a good affinity for targeted receptors compared to co-crystalized inhibitors, validating the analgesic and anti-inflammatory effects of the phytochemicals.


Subject(s)
Emodin , Senna Plant , Plant Extracts/pharmacology , Plant Extracts/chemistry , Senna Plant/chemistry , 1-Butanol , Egypt , Anthraquinones/pharmacology , Anti-Inflammatory Agents/pharmacology , Analgesics/pharmacology , Phytochemicals/pharmacology
2.
Molecules ; 27(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35164118

ABSTRACT

Diabetes mellitus is a chronic disease affecting the globe and its incidence is increasing pandemically. The use of plant-derived natural products for diabetes management is of great interest. Polar fraction of Artemisia annua L. leaves has shown antidiabetic activity in vivo. In the present study, three major compounds were isolated from this polar fraction; namely, 3,5-dicaffeoylquinic acid (1); 4,5-dicaffeoylquinic acid (2), and 3,4- dicaffeoylquinic acid methyl ester (3), using VLC-RP-18 and HPLC techniques. The potential protective effects of these compounds against diabetes and its complications were investigated by employing various in vitro enzyme inhibition assays. Furthermore, their antioxidant and wound healing effectiveness were evaluated. Results declared that these dicaffeoylquinic acids greatly inhibited DPPIV enzyme while moderately inhibited α-glucosidase enzyme, where compounds 1 and 3 displayed the most prominent effects. In addition, compound 3 showed pronounced inhibition of α-amylase enzyme. Moreover, these compounds markedly inhibited aldose reductase enzyme and exerted powerful antioxidant effects, among which compound 3 exhibited the highest activity implying a notable potentiality in impeding diabetes complications. Interestingly, compounds 2 and 3 moderately accelerated scratch wound healing. Our findings suggest that these dicaffeoylquinic acids can be promising therapeutic agents for managing diabetes and its complications.


Subject(s)
Artemisia annua/chemistry , Diabetes Complications/prevention & control , Glycoside Hydrolase Inhibitors , Hypoglycemic Agents , Plant Leaves/chemistry , Quinic Acid/analogs & derivatives , Cell Line , Diabetes Complications/metabolism , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/isolation & purification , Glycoside Hydrolase Inhibitors/pharmacology , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/isolation & purification , Hypoglycemic Agents/pharmacology , Quinic Acid/chemistry , Quinic Acid/isolation & purification , Quinic Acid/pharmacology
3.
Arch Biochem Biophys ; 717: 109121, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35065059

ABSTRACT

Acute liver failure (ALF) is considered a fatal clinical disorder and novel therapeutic interventions are mandatory. Naringenin is a flavonoid with anti-inflammatory, antioxidant and antiapoptotic effects that have displayed beneficial effects in different animal models of ALF. The current study aimed at investigating the hepatoprotective effect and the possible underlying molecular mechanisms of naringenin in lipopolysaccharide (LPS)/D-galactosamine (D-Gal) mouse model of ALF. Interestingly, naringenin pretreatment substantially alleviated LPS/D-Gal-induced liver injury, enhanced survival, improved liver function and ameliorated histopathological liver changes. Importantly, naringenin potently activated autophagy as evidenced by the increased Beclin-1 expression and LC3 II/LC3 I ratio. Furthermore, results demonstrated that naringenin alleviated oxidative stress by inducing nuclear factor-erythroid 2-related factor 2 (Nrf2) and increasing hepatic SOD activity and GSH level as well as ameliorated endoplasmic reticulum (ER) stress. Likewise, naringenin mitigated LPS/D-Gal-triggered inflammation by suppressing NF-κB and NLRP3 pathways. Accordingly, apoptotic cell death provoked by LPS/D-Gal challenge was markedly attenuated as depicted by the decrease in caspase-3 and p53 in naringenin-treated mice. To investigate the contribution of autophagy to naringenin-conferred hepatoprotection, autophagy was inhibited using 3-methyladenine (3 MA). Strikingly, 3 MA co-treatment abolished the hepatoprotective effect of naringenin, a finding that strongly suggests that naringenin-afforded protection is, at least in part, attributed to autophagy. Taken together, the present study revealed that naringenin exerted a prominent hepatoprotective effect by promoting autophagy with consequent attenuation of inflammatory responses, oxidative stress, ER stress and apoptosis. Our results provide evidence that naringenin use holds a promise as a potential therapeutic agent for ALF management.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Autophagy/drug effects , Flavanones/pharmacology , Galactosamine/metabolism , Lipopolysaccharides/metabolism , Liver Failure, Acute/chemically induced , Animals , Apoptosis/drug effects , Beclin-1/genetics , Beclin-1/metabolism , Caspase 3/metabolism , Disease Models, Animal , Humans , Liver , Liver Failure, Acute/prevention & control , Male , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , NF-E2-Related Factor 2 , NF-kappa B , Oxidative Stress , Signal Transduction , Superoxide Dismutase-1/metabolism , Tumor Suppressor Protein p53/metabolism
4.
Molecules ; 25(24)2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33322431

ABSTRACT

Diabetes is a major health problem that is associated with high risk of various complications. Medicinal plants hold great promise against diabetes. The traditional use of Cleome droserifolia as an antidiabetic agent was correlated to its flavonol glycosides content. In the current study, five major flavonol glycosides appeared on the RP-HPLC chromatogram of the aqueous extract namely; quercetin-3-O-ß-d-glucosyl-7-O-α-rhamnoside (1), isorhamnetin-7-O-ß-neohesperidoside (2), isorhamnetin-3-O-ß-d-glucoside (3) kaempferol-4'-methoxy-3,7-O-α-dirhamnoside (4), and isorhamnetin-3-O-α-(4″-acetylrhamnoside)-7-O-α-rhamnoside (5). The inhibitory activities of these compounds were tested in vitro against several enzymes involved in diabetes management. Only the relatively less polar methoxylated flavonol glycosides (4, 5) showed mild to moderate α-amylase and α-glucosidase inhibitory activities. Compounds 1-4 displayed remarkable inhibition of dipeptidyl peptidase IV (DPPIV) enzyme (IC50 0.194 ± 0.06, 0.573 ± 0.03, 0.345 ± 0.02 and 0.281 ± 0.05 µg/mL, respectively) comparable to vildagliptin (IC50 0.154 ± 0.02 µg/mL). Moreover, these compounds showed high potential in preventing diabetes complications through inhibiting aldose reductase enzyme and combating oxidative stress. Both isorhamnetin glycoside derivatives (2, 3) exhibited the highest activities in aldose reductase inhibition and compound 2 (IC50 5.45 ± 0.26 µg/mL) was even more potent than standard quercetin (IC50 7.77 ± 0.43 µg/mL). Additionally, these flavonols exerted excellent antioxidant capacities through 2, 2-diphenyl-1-picrylhydrazil (DPPH) and ferric reducing antioxidant (FRAP) assays.


Subject(s)
Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Glycosides/pharmacology , Aldehyde Reductase/chemistry , Aldehyde Reductase/metabolism , Antioxidants/chemistry , Biphenyl Compounds/chemistry , Chemistry, Pharmaceutical/methods , Chromatography, High Pressure Liquid , Cleome , Drug Design , Free Radical Scavengers , Humans , Hypoglycemic Agents , In Vitro Techniques , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Models, Chemical , Oxidative Stress , Picrates/chemistry , Vildagliptin/pharmacology , alpha-Amylases/chemistry , alpha-Glucosidases/metabolism
5.
Chem Biol Interact ; 331: 109276, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-33002459

ABSTRACT

Ulcerative colitis (UC) is a chronic disease driven primarily by uncontrolled pervasive inflammatory responses affecting the colon and rectum. Currently available medications carry multiple detrimental adverse effects, which have emphasized the mandatory need for safer and more efficient novel therapeutic alternatives. Melittin is the main constituent of bee venom and exhibits potent anti-inflammatory properties. The antiulcerogenic effect of oral melittin (40 µg/kg) was explored in the current study using the acetic acid-induced colitis model. Increase in body weight and decrease in colon mass index were observed in the melittin group. Microscopically, melittin ameliorated acetic acid-induced histological damage. Melittin administration has efficiently amended the elevated levels of the cytokines, tumor necrosis factor (TNF-α) and interleukin 6 (IL-6) seen in the colitis group. This was accompanied by inhibition of the upstream signaling molecules, Toll-like receptor 4 (TLR4), tumor necrosis factor receptor (TNF-R)-associated factor (TRAF6), mitogen-activated protein kinase 38 (p38 MAPK), and nuclear factor kappaB (NF-κB) in the melittin group. Moreover, treatment with melittin resulted in marked decrease in colonic level of prostaglandin E2 (PGE2) together with the enzymes involved in its synthesis, secretory phospholipase A2 (sPLA2) and cyclooxygenase 2 (COX-2). Additionally, melittin has attenuated acetic acid-induced oxidative stress as manifested by the significant diminishment in malondialdehyde (MDA) as well as the increase in superoxide dismutase (SOD) and reduced glutathione (GSH) levels. Therefore, melittin mitigated UC pathogenesis and could be considered as a potent and promising therapeutic alternative for UC treatment.


Subject(s)
Anti-Ulcer Agents/pharmacology , Melitten/pharmacology , Signal Transduction/drug effects , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 4/metabolism , Acetic Acid/toxicity , Administration, Oral , Animals , Anti-Ulcer Agents/therapeutic use , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Colon/metabolism , Colon/pathology , Cyclooxygenase 2/metabolism , Interleukin-6/metabolism , Malondialdehyde/metabolism , Melitten/therapeutic use , Mice , NF-kappa B/metabolism , Oxidative Stress/drug effects , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
J Endocrinol ; 244(2): 323-337, 2020 02.
Article in English | MEDLINE | ID: mdl-31682591

ABSTRACT

Maturity-onset diabetes of the young (MODY) is a group of monogenetic forms of diabetes mellitus caused by mutations in genes regulating ß-cell development and function. MODY represents a heterogeneous group of non-insulin-dependent diabetes arising in childhood or adult life. Interestingly, clinical heterogeneity in MODY patients like variable disease onset and severity is observed even among individual family members sharing the same mutation, an issue that is not well understood. As high blood glucose levels are a well-known factor promoting ß-cell stress and ultimately leading to cell death, we asked whether additional ß-cell stress might account for the occurrence of disease heterogeneity in mice carrying a MODY4 mutation. In order to challenge ß-cells, we established a MODY4 animal model based on Pdx1 (pancreatic and duodenal homeobox 1) haploinsufficiency, which allows conditional modulation of cell stress by genetic inhibition of the stress-responsive IKK/NF-κB signalling pathway. While Pdx1+/- mice were found glucose intolerant without progressing to diabetes, additional challenge of ß-cell function by IKK/NF-κB inhibition promoted rapid diabetes development showing hyperglycaemia, hypoinsulinemia and loss of ß-cell mass. Disease pathogenesis was characterized by deregulation of genes controlling ß-cell homeostasis and function. Importantly, restoration of normal IKK/NF-κB signalling reverted the diabetic phenotype including normalization of glycaemia and ß-cell mass. Our findings implicate that the avoidance of additional ß-cell stress can delay a detrimental disease progression in MODY4 diabetes. Remarkably, an already present diabetic phenotype can be reversed when ß-cell stress is normalized.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Blood Glucose/metabolism , Cell Death , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Stress, Physiological , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Molecules ; 24(3)2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30717217

ABSTRACT

A novel series of thiazole-based heterocycles was synthesized using 1,3-dipolar cycloaddition reactions in the presence of chitosan-grafted-poly(vinylpyridine) as an eco-friendly biopolymeric basic catalyst. The molecular structure of the synthesized compounds was illustrated by spectroscopic and elemental analysis. Various in vitro biological assays were performed to explore the potential antitumor, antimicrobial and hepatoprotective activities of the newly synthesized compounds. The cytotoxic activities were assessed against human hepatocellular carcinoma (HepG-2), colorectal carcinoma (HCT-116) and breast cancer (MCF-7) cell lines and results revealed that all compounds displayed antitumor activities with the chlorine-containing derivatives, 11c and 6g, being the most potent. The majority of the tested thiazole derivatives exhibited satisfactory antibacterial activity towards the used gram positive and gram-negative bacterial species. Moreover, many derivatives showed weak hepatoprotective activity against CCl4-induced hepatotoxicity.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Antineoplastic Agents/chemical synthesis , Carbon Tetrachloride/antagonists & inhibitors , Green Chemistry Technology , Heterocyclic Compounds/chemical synthesis , Thiazoles/chemical synthesis , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/growth & development , Candida albicans/drug effects , Candida albicans/growth & development , Carbon Tetrachloride/toxicity , Catalysis , Chitosan/chemistry , Cycloaddition Reaction , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , HCT116 Cells , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Heterocyclic Compounds/pharmacology , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Microbial Sensitivity Tests , Polyvinyls/chemistry , Primary Cell Culture , Pyridines/chemistry , Structure-Activity Relationship , Thiazoles/pharmacology
8.
Diabetes ; 63(3): 960-75, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24296718

ABSTRACT

Type 1 diabetes is a multifactorial inflammatory disease in genetically susceptible individuals characterized by progressive autoimmune destruction of pancreatic ß-cells initiated by yet unknown factors. Although animal models of type 1 diabetes have substantially increased our understanding of disease pathogenesis, heterogeneity seen in human patients cannot be reflected by a single model and calls for additional models covering different aspects of human pathophysiology. Inhibitor of κB kinase (IKK)/nuclear factor-κB (NF-κB) signaling is a master regulator of inflammation; however, its role in diabetes pathogenesis is controversially discussed by studies using different inhibition approaches. To investigate the potential diabetogenic effects of NF-κB in ß-cells, we generated a gain-of-function model allowing conditional IKK2/NF-κB activation in ß-cells. A transgenic mouse model that expresses a constitutively active mutant of human IKK2 dependent on Pdx-1 promoter activity (IKK2-CA(Pdx-1)) spontaneously develops full-blown immune-mediated diabetes with insulitis, hyperglycemia, and hypoinsulinemia. Disease development involves a gene expression program mimicking virus-induced diabetes and allergic inflammatory responses as well as increased major histocompatibility complex class I/II expression by ß-cells that could collectively promote diabetes development. Potential novel diabetes candidate genes were also identified. Interestingly, animals successfully recovered from diabetes upon transgene inactivation. Our data give the first direct evidence that ß-cell-specific IKK2/NF-κB activation is a potential trigger of immune-mediated diabetes. Moreover, IKK2-CA(Pdx-1) mice provide a novel tool for studying critical checkpoints in diabetes pathogenesis and mechanisms governing ß-cell degeneration/regeneration.


Subject(s)
Diabetes Mellitus, Type 1/etiology , I-kappa B Kinase/physiology , Insulin-Secreting Cells/physiology , NF-kappa B/physiology , Signal Transduction/physiology , Animals , Apoptosis , Chemokine CCL17/physiology , Chemokine CCL22/physiology , Endoplasmic Reticulum Stress , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Trans-Activators/genetics , Transcriptome
9.
Curr Diab Rep ; 12(5): 499-509, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22791179

ABSTRACT

Type 1 diabetes (T1D) is a multi-factorial, organ-specific autoimmune disease in genetically susceptible individuals, which is characterized by a selective and progressive loss of insulin-producing ß-cells. Cells mediating innate as well as adaptive immunity infiltrate pancreatic islets, thereby generating an aberrant inflammatory process called insulitis that can be mirrored by a pathologic autoantibody production and autoreactive T-cells. In tight cooperation with infiltrating innate immune cells, which secrete high levels of pro-inflammatory cytokines like IL-1ß, TNFα, and INFγ effector T-cells trigger the fatal destruction process of ß-cells. There is ongoing discussion on the contribution of inflammation in T1D pathogenesis, ranging from a bystander reaction of autoimmunity to a dysregulation of immune responses that initiate inflammatory processes and thereby actively promoting ß-cell death. Here, we review recent advances in anti-inflammatory interventions in T1D animal models and preclinical studies and discuss their mode of action as well as their capacity to interfere with T1D development.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Autoimmunity/drug effects , Clinical Trials as Topic , Cytokines/metabolism , Cytokines/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Models, Biological , NF-kappa B/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...