Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell Death Dis ; 15(1): 53, 2024 01 15.
Article in English | MEDLINE | ID: mdl-38225221

ABSTRACT

Chronic metabolic stress paradoxically elicits pro-tumorigenic signals that facilitate cancer stem cell (CSC) development. Therefore, elucidating the metabolic sensing and signaling mechanisms governing cancer cell stemness can provide insights into ameliorating cancer relapse and therapeutic resistance. Here, we provide convincing evidence that chronic metabolic stress triggered by hyaluronan production augments CSC-like traits and chemoresistance by partially impairing nucleotide sugar metabolism, dolichol lipid-linked oligosaccharide (LLO) biosynthesis and N-glycan assembly. Notably, preconditioning with either low-dose tunicamycin or 2-deoxy-D-glucose, which partially interferes with LLO biosynthesis, reproduced the promoting effects of hyaluronan production on CSCs. Multi-omics revealed characteristic changes in N-glycan profiles and Notch signaling activation in cancer cells exposed to mild glycometabolic stress. Restoration of N-glycan assembly with glucosamine and mannose supplementation and Notch signaling blockade attenuated CSC-like properties and further enhanced the therapeutic efficacy of cisplatin. Therefore, our findings uncover a novel mechanism by which tolerable glycometabolic stress boosts cancer cell resilience through altered N-glycosylation and Notch signaling activation.


Subject(s)
Hyaluronic Acid , Resilience, Psychological , Humans , Glycosylation , Hyaluronic Acid/metabolism , Neoplasm Recurrence, Local/metabolism , Polysaccharides/metabolism , Dietary Supplements , Neoplastic Stem Cells/metabolism
2.
PLoS One ; 18(7): e0274479, 2023.
Article in English | MEDLINE | ID: mdl-37418356

ABSTRACT

Cordyceps cicadae (Miq.) is an edible fungus with unique and valuable medicinal properties that is commonly used in traditional Chinese medicine, but its anti-aging effects on the skin fibroblast are not well studied. The aim of the present study was to analyze the active components of aqueous C. cicadae extract (CCE), determine the effects of CCE on hyaluronan synthesis in human skin fibroblasts, and explore the underlying mechanisms. The results of this study indicate that CCE was rich in polysaccharides, five alditols (mainly mannitol), eight nucleosides, protein, and polyphenols, which were present at concentrations of 62.7, 110, 8.26, 35.7, and 3.8 mg/g, respectively. The concentration of extract required to inhibit 50% of 2,2-azino-bis (3-ethylbenzothiazo-line-6-sulphonic acid) (ABTS) and 2,2-diphenyl-1-picrylhydrazil (DPPH) radical scavenging capacities were 0.36 ± 0.03 and 4.54 ± 0.10 mg/mL, respectively, indicating that CCE exhibits excellent antioxidant activities. CCE showed no cytotoxicity to skin fibroblasts at concentrations ≤ 100 µg/mL, and promoted HA synthesis in fibroblasts. Treatment of fibroblast cells with 100 µg/mL CCE enhances the HA content to 1293 ± 142 ng/mL, which is significantly more than that in the non-treatment (NT) group (p = 0.0067). Further, RNA sequencing detected 1,192 differentially expressed genes (DEGs) in CCE-treated fibroblasts, among which 417 were upregulated and 775 were downregulated. Kyoto Encyclopedia of Genes (KEGG) and Genomes pathway (GO) analysis based on RNA sequencing revealed that CCE mainly affected cytokine-cytokine receptor interaction regulated by HA synthesis-related genes. CCE upregulated HA synthase 2 (HAS2), epidermal growth factor (EGF)-related genes, heparin-binding EGF-like growth factor, C-C motif chemokine ligand 2, interleukin 1 receptor-associated kinase 2, and other genes related to fibroblast differentiation and proliferation. CCE downregulated the gene of matrix metallopeptidase 12 (MMP12), which leads to cell matrix loss. RT-qPCR further verified CCE significantly upregulated HAS2 expression and significantly downregulated MMP12 expression, thus promoting hyaluronan synthesis. CCE shows potential as a moisturizer and anti-aging agent in functional foods and cosmetics.


Subject(s)
Cordyceps , Hyaluronic Acid , Humans , Hyaluronic Acid/pharmacology , Hyaluronic Acid/metabolism , Matrix Metalloproteinase 12/metabolism , Hyaluronan Synthases , Cordyceps/metabolism , Aging , Fibroblasts/metabolism
3.
Int J Mol Sci ; 23(21)2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36361854

ABSTRACT

We investigated the effects of bactericidal/permeability-increasing protein (BPI) alone or in combination with hyaluronic acid (HA) in two animal models: collagen-induced arthritis (CIA) and crystal-induced inflammation. In CIA, mice were intraperitoneally injected with PBS, HA, or BPI plus or minus HA, twice a week for 2 months, and then euthanized to collect paw and blood. Arthritis was assessed in ankle joints by clinical and histological evaluation. Pathogenic crystals were intraperitoneally injected in mice plus or minus BPI, or with a composition of BPI and HA. After sacrifice, total and differential leukocyte counts were determined. Cytokine levels were measured in serum and peritoneal fluids. In CIA mice, BPI improved clinical and histological outcomes (histological scores ≥2-fold), and downregulated inflammatory mediators (47-93%). In crystal-induced inflammation, BPI reduced leukocyte infiltration (total count: ≥60%; polymorphonuclear cells: ≥36%) and inhibited cytokine production (35-74%). In both models, when mice were co-treated with BPI and HA, the improvement of all parameters was greater than that observed after administration of the two substances alone. Results show that BPI attenuates CIA and inflammation in mice, and this effect is enhanced by HA co-administration. Combined use of BPI and HA represents an interesting perspective for new potential treatments in arthritis.


Subject(s)
Arthritis, Experimental , Mice , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Inflammation Mediators/metabolism , Cytokines/metabolism , Inflammation/drug therapy , Inflammation/pathology , Hyaluronic Acid/metabolism , Permeability
4.
J Ethnopharmacol ; 296: 115523, 2022 Oct 05.
Article in English | MEDLINE | ID: mdl-35809756

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The meadowsweet family (genus Filipendula) includes about 30 species, which have been traditionally used in folk medicine to treat various inflammatory diseases. Particularily, F. palmata (Pall.) Maxim. (Siberian meadowsweet) were traditionally and widely used as an ethnic herb in the Oroqen application. AIM OF THE STUDY: Limited studies have been documented on most species, except for two main species, F. ulmaria (L.) Maxim. and F. vulgaris Moench. Thus, this study aimed to investigate the anti-inflammatory and skin-moisturizing effects of 70% ethanolic extract (FPE) of F. palmata on human epidermal keratinocytes. MATERIALS AND METHODS: HaCaT keratinocytes were treated with FPE under different conditions. Quantitative real time-PCR, enzyme-linked immunosorbent assay, western blotting methods were used to evaluate the effect and molecular mechanism of the cells treated with FPE. The bioactive compounds in FPE, which are responsible for biological activities, was explored using mass spectrometric analysis. RESULTS: FPE did not show a cytotoxic effect on the cells at concentrations below 200 µg/mL. FPE significantly suppressed the intracellular reactive oxygen species and mitochondrial superoxide of inflamed HaCaT cells induced by tumor necrosis factor-α and interferon-γ (T + I) and inflammatory chemokine genes and proteins, such as CC chemokine ligands (CCL5, CCL17, and CCL27) and CXC chemokine ligand (CXCL8). These anti-inflammatory activities of FPE were mediated by the downregulation of mitogen-activated protein kinase (MAPK) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways. In normal HaCaT cells, FPE significantly promoted the production of hyaluronic acid (HA) via the downregulation of hyaluronidase (HYAL1 and HYAL2) and upregulation of hyaluronic acid synthase (HAS1, HAS2, and HAS3) genes, and these effects seemed to be associated with the PI3K/Akt/NF-κB signaling. Ultraperformance liquid chromatography-tandem mass spectrometry indicated that FPE contains four flavonoids, including (+)-catechin, miquelianin, scutellarin, and quercitrin, as its major phytochemicals. Finally, we demonstrated that miquelianin and quercitrin contribute partially to the anti-inflammatory and HA-producing activity of FPE without cytotoxic effects on HaCaT cells. CONCLUSIONS: Our findings suggest that topical applications of FPE can be utilized as an alternative therapy for treating skin inflammation. Additionally, our findings serve as a reference in applying FPE as a functional ingredient to treat inflammatory skin diseases and promote skin health.


Subject(s)
Filipendula , Anti-Inflammatory Agents/therapeutic use , Filipendula/chemistry , Humans , Hyaluronic Acid/metabolism , Hyaluronic Acid/pharmacology , Keratinocytes , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phytochemicals/metabolism , Phytochemicals/pharmacology , Tumor Necrosis Factor-alpha/metabolism
5.
Biomed Pharmacother ; 145: 112461, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34839253

ABSTRACT

Skin aging is accompanied by an increase in the number of senescent cells, resulting in various pathological outcomes. These include inflammation, impaired barrier function, and susceptibility to skin disorders such as cancer. Kaempferia parviflora (Thai black ginger), a medicinal plant native to Thailand, has been shown to counteract inflammation, cancer, and senescence. This study demonstrates that polymethoxyflavones (5,7-dimethoxyflavone, 5,7,4'-trimethoxyflavone, and 3,5,7,3',4'-pentamethoxyflavone) purified from K. parviflora rhizomes suppressed cellular senescence, reactive oxygen species, and the senescence-associated secretory phenotype in primary human dermal fibroblasts. In addition, they increased tropocollagen synthesis and alleviated free radical-induced cellular and mitochondrial damage. Moreover, the compounds mitigated chronological aging in a human ex vivo skin model by attenuating senescence and restoring expression of essential components of the extracellular matrix, including collagen type I, fibrillin-1, and hyaluronic acid. Finally, we report that polymethoxyflavones enhanced epidermal thickness and epidermal-dermal stability, while blocking age-related inflammation in skin explants. Our findings support the use of polymethoxyflavones from K. parviflora as natural anti-aging agents, highlighting their potential as active ingredients in cosmeceutical and nutraceutical products.


Subject(s)
Collagen Type I/metabolism , Extracellular Matrix , Flavonoids/pharmacology , Hyaluronic Acid/metabolism , Skin Aging , Skin , Zingiberaceae , Cell Line , Extracellular Matrix/drug effects , Extracellular Matrix/physiology , Fibrillin-1/metabolism , Fibroblasts/metabolism , Flavones/pharmacology , Geroscience , Humans , Rhizome , Skin/drug effects , Skin/metabolism , Skin Aging/drug effects , Skin Aging/physiology , Thailand
6.
Biomed Res Int ; 2021: 5598110, 2021.
Article in English | MEDLINE | ID: mdl-34754881

ABSTRACT

Dermal papilla cells (DPCs) are a source of nutrients and growth factors, which support the proliferation and growth of keratinocytes as well as promoting the induction of new hair follicles and maintenance of hair growth. The protection from reactive oxygen species (ROS) and the promotion of angiogenesis are considered two of the basal mechanisms to preserve the growth of the hair follicle. In this study, a noncrosslinked hyaluronic acid (HA) filler (HYDRO DELUXE BIO, Matex Lab S.p.A.) containing several amino acids was tested with in vitro assays on human follicle dermal papilla cells (HFDPCs). The experiments were carried out to investigate the possible protection against oxidative stress and the ability to increase the vascular endothelial growth factor (VEGF) release. The results demonstrated the restoration of cell viability against UVB-induced cytotoxicity and an increase in the VEGF secretion. These data demonstrate the capability of the product to modulate human dermal papilla cells, suggesting a future use in mesotherapy, a minimally invasive local intradermal therapy (LIT), after further clinical investigations.


Subject(s)
Dermis/metabolism , Hair Follicle/metabolism , Hyaluronic Acid/pharmacology , Cells, Cultured , Dermis/drug effects , Dermis/growth & development , Hair/growth & development , Hair Follicle/drug effects , Humans , Hyaluronic Acid/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
7.
Bioorg Chem ; 117: 105441, 2021 12.
Article in English | MEDLINE | ID: mdl-34695731

ABSTRACT

Under the guidance of bioassay against HSC-LX2, the EtOH extract and the EtOAc fraction of Artemisia capillaris (Yin-Chen) exhibited cytotoxic activity against HSC-LX2 with inhibitory ratios of 39.7% and 68.7% at the concentration of 400.0 µg/mL. Bioassay-guided investigation of Fr. D (the active fraction) yielded 14 new coumaric acid analogues, artemicapillasins A-N (1-14). The structures of the isolates were elucidated by spectroscopic analyses involving UV, IR, MS, 1D and 2D NMR spectra and ECD calculations. Cytotoxic activity against HSC-LX2 cells of these isolates was performed to reveal that 12 compounds demonstrated cytotoxicity with inhibitory ratios more than 50% at 400 µM. The most active artemicapillasin B (2) gave an IC50 value of 24.5 µM, which was about 7 times more toxic than the positive drug silybin (IC50, 162.3 µM). Importantly, artemicapillasin B (2) showed significant inhibition on the deposition of human collagen type I (Col I), human laminin (HL) and human hyaluronic acid (HA) with IC50 values of 11.0, 14.4 and 13.8 µM, which was about 7, 11 and 5 times more active than silybin. Artemicapillasin B (2) as an interesting antihepatic fibrosis candidate is worth in-depth study.


Subject(s)
Artemisia/chemistry , Hepatic Stellate Cells/drug effects , Cell Survival/drug effects , Collagen Type I/antagonists & inhibitors , Collagen Type I/metabolism , Dose-Response Relationship, Drug , Humans , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/metabolism , Laminin/antagonists & inhibitors , Laminin/metabolism , Molecular Structure , Structure-Activity Relationship
8.
J Plast Reconstr Aesthet Surg ; 74(7): 1610-1614, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34024740

ABSTRACT

Hyaluronidase is a family of enzymes that degrade hyaluronic acid (HA). It is found to increase vascular permeability and temporarily disrupt the extracellular matrix, promoting diffusion of substances through tissues. Alongside its applications in ophthalmology, obstetrics and gynaecology, musculoskeletal medicine, radiology and drug and fluid administration, hyaluronidase has a number of roles in the field of plastic surgery. The popularity of HA fillers in recent years has led to an increase in the usage of hyaluronidase in the treatment of filler-related complications. The purpose of this article is to review the current and future uses of hyaluronidase within the field of plastic surgery. Hyaluronidase is used as an adjunct to local anaesthetics in skin infiltration, skin graft harvesting, tumescent analgesia, managing complications of dermal fillers, treatment of extravasation injury, prevention and management of oedema, treatment of ganglion and management of scars. However, it has some limitations. Hyaluronidase is known to interact with a number of common medications. Several case reports also highlight the risk of allergic reaction to the substance. Although rare and usually mild, hyaluronidase has the potential to cause anaphylaxis. Other adverse effects include bruising and swelling. Overall, hyaluronidase appears to be a very safe, cheap and effective medication for a variety of uses in the field of plastic surgery and beyond.


Subject(s)
Adjuvants, Anesthesia , Anesthesia, Local/methods , Cicatrix/drug therapy , Dermal Fillers/adverse effects , Hyaluronic Acid/adverse effects , Hyaluronoglucosaminidase/therapeutic use , Plastic Surgery Procedures , Dermal Fillers/metabolism , Drug Hypersensitivity/etiology , Humans , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/adverse effects , Hyaluronoglucosaminidase/metabolism
9.
Clin Exp Dermatol ; 46(5): 825-833, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33522006

ABSTRACT

Curcuma and its derivatives are associated with anti-inflammatory and antioxidant activities in the skin. They exhibit beneficial effects in wound healing and prevention of chronic ultraviolet B damage and may prevent facial redness such as rosacea and flushing. This review aims to provide an up-to-date and rigorous synthesis of studies that demonstrated the clinical efficacy of curcuminoids in the skin. We evaluated studies published in the MEDLINE-PubMed/PMC (National Library of Medicine) databases, and followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines for this review. This search included papers published in the past 10 years in controlled clinical trials, double-blind and randomized controlled studies, and case studies. The search resulted in 12 studies published in the past 10 years. Curcuma species (Curcuma longa and Curcuma aeruginosa) and curcumin were found to produce various dermatological effects, including influencing antioxidant and anti-inflammatory processes in the production of hyaluronan, increasing skin moisture, and reducing axillary hair growth. Curcuma was also found to reduce thickness, erythema, pruritus, burning and pain in psoriasis lesions and to improve radiodermatitis lesions. Our review results show that Curcuma species may play a role in skin health management and may exhibit various dermatological effects, thus it could be a new therapeutic arsenal for dermatology professionals. Nevertheless, more clinical trials should be conducted with humans to establish the optimum delivery method and dosages for different dermatological conditions.


Subject(s)
Curcuma/chemistry , Curcumin/pharmacology , Plant Extracts/pharmacology , Skin/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Curcuma/adverse effects , Curcumin/therapeutic use , Double-Blind Method , Female , Humans , Hyaluronic Acid/metabolism , Male , Psoriasis/drug therapy , Radiodermatitis/drug therapy , Radiodermatitis/prevention & control , Randomized Controlled Trials as Topic , Rosacea/prevention & control , Skin/pathology , Treatment Outcome , Wound Healing/drug effects
10.
J Orthop Surg Res ; 16(1): 8, 2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33407721

ABSTRACT

BACKGROUND: Deer antler is considered as a precious traditional Chinese medicinal material and has been widely used to reinforce kidney's yang, nourish essence, and strengthen bone function. The most prominent bioactive components in deer antler are water-soluble proteins that play potential roles in bone formation and repair. The aim of this study was to explore the molecular control and therapeutic targets of deer antler extract (DAE) on articular cartilage. METHODS: DAE was prepared as previously described. All rats were randomly divided into Blank group and DAE group (10 rats per group) after 7-day adaptive feeding. The rats in DAE group were orally administrated with DAE at a dose of 0.2 g/kg per day for 3 weeks, and the rats in Blank group were fed with drinking water. Total RNA was isolated from the articular cartilage of knee joints. RNA sequencing (RNA-seq) experiment combined with quantitative real-time polymerase chain reaction (qRT-PCR) verification assay was carried out to explore the molecular control and therapeutic targets of DAE on articular cartilage. RESULTS: We demonstrated that DAE significantly increased the expression levels of functional genes involved in cartilage formation, growth, and repair and decreased the expression levels of susceptibility genes involved in the pathophysiology of osteoarthritis. CONCLUSIONS: DAE might serve as a candidate supplement for maintaining cartilage homeostasis and preventing cartilage degeneration and inflammation. These effects were possibly achieved by accelerating the expression of functional genes involved in chondrocyte commitment, survival, proliferation, and differentiation and suppressing the expression of susceptibility genes involved in the pathophysiology of osteoarthritis. Thus, our findings will contribute towards deepening the knowledge about the molecular control and therapeutic targets of DAE on the treatment of cartilage-related diseases.


Subject(s)
Antlers/chemistry , Cartilage, Articular/metabolism , Cartilage, Articular/physiology , Deer , Osteogenesis/drug effects , Osteogenesis/genetics , Tissue Extracts/administration & dosage , Tissue Extracts/pharmacology , Administration, Oral , Animals , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Genetic Predisposition to Disease/genetics , Hyaluronic Acid/genetics , Hyaluronic Acid/metabolism , Male , Medicine, Chinese Traditional , Molecular Targeted Therapy , Osteoarthritis/genetics , Proteoglycans/genetics , Proteoglycans/metabolism , RNA/genetics , RNA/isolation & purification , Rats, Sprague-Dawley , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism
11.
Int J Mol Sci ; 23(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35008494

ABSTRACT

Skin barrier damage is present in the patients with hereditary disorders of the magnesium channel, but the molecular mechanism has not been fully understood. We found that the expressions of hyaluronan synthase (HAS), HAS2 and HAS3 are influenced by MgCl2 concentration in human keratinocyte-derived HaCaT cells. The exposure of cells to a high concentration (5.8 mM) of MgCl2 induced the elevation of HAS2/3 expression, which was inhibited by mRNA knockdown of nonimprinted in Prader-Willi/Angelman syndrome-like domain containing 4 (NIPAL4). Similarly, the content of hyaluronic acid (HA) was changed according to MgCl2 concentration and the expression of NIPAL4. The MgCl2 supplementation increased the reporter activities of HAS2/3, which were inhibited by NIPAL4 knockdown, indicating that the expressions of HAS2/3 are up-regulated at the transcriptional level. The reporter activities and mRNA levels of HAS2/3, and the production of HA were inhibited by CHIR-99021, a glycogen synthase kinase-3 (GSK3) inhibitor, and naphthol AS-E, a cyclic AMP-response element binding protein (CREB) inhibitor. Furthermore, the mutation in putative CREB-binding sites of promoter region in HAS2/3 genes inhibited the MgCl2 supplementation-induced elevation of promoter activity. Our results indicate that the expressions of HAS2/3 are up-regulated by MgCl2 supplementation in HaCaT cells mediated through the activation of GSK3 and CREB. Magnesium may play a pivotal role in maintaining the skin barrier function and magnesium supplementation may be useful to enhance moisturization and wound repair in the skin.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , Hyaluronan Synthases/metabolism , Keratinocytes/drug effects , Magnesium/pharmacology , Cell Line , Dietary Supplements , HaCaT Cells , Humans , Hyaluronic Acid/metabolism , Keratinocytes/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism , Up-Regulation/drug effects
12.
ACS Appl Mater Interfaces ; 12(36): 40085-40093, 2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32791825

ABSTRACT

Immune checkpoint blockade of the programmed cell death-ligand 1/programmed cell death-1 (PD-L1/PD-1) pathway via an antibody is a potent strategy for T cell remodeling. Nevertheless, the potency of the antibody is partly compromised by its high price, instability, risk of autoimmune disease, and so forth. Small-molecule inhibitors are interesting alternatives to antibodies. However, tumor-specific delivery of small-molecule inhibitors to the target site for boosting the interruption of the PD-L1/PD-1 pathway is rarely reported. Herein, we designed a tumor-specific delivery nanoplatform that could efficiently deliver the small-molecule inhibitor to the precise target site, greatly enhancing the blocking effect of the PD-L1/PD-1 pathway. Hyaluronic acid (HA) was conjugated with chlorin e6 (Ce6), resulting in a HA-Ce6 conjugate (HC). The nanoplatform was constructed by the HC micelles with the encapsulation of small-molecule inhibitor, BMS 202 (BMS), to form BMS/HC micelles. The target property of HA, combined with the hyaluronidase-induced degradation of HA in the tumor site, enables the as-prepared micelles with tumor-specific delivery of BMS for blocking the PD-L1/PD-1 pathway. With cooperative treatment with the photosensitizer Ce6, the present therapeutic nanoplatform demonstrated excellent photoimmunotherapy for tumor regression in distant tumors and lung metastasis. This strategy of tumor-specific delivery of small-molecule inhibitors provides an effective pathway to strengthen the blocking efficacy of PD-L1/PD-1 on effective photoimmunotherapy.


Subject(s)
Acetamides/pharmacology , Immunotherapy , Melanoma, Experimental/drug therapy , Nanoparticles/chemistry , Phototherapy , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Acetamides/chemical synthesis , Acetamides/chemistry , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Chlorophyllides , Female , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Micelles , Particle Size , Porphyrins/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Reactive Oxygen Species/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Surface Properties , Tumor Cells, Cultured
13.
Anim Sci J ; 91(1): e13434, 2020.
Article in English | MEDLINE | ID: mdl-32696560

ABSTRACT

Liver fibrosis is a major health concern, which might progress to cirrhosis. To date, treatment trials rely mainly on the removal of the causative factor. The current study investigated the potential ameliorative role of sidr honey on thioacetamide (TAA)-induced liver fibrosis in rats. Forty-eight Wistar albino rats were equally allocated into four groups: control; sidr honey (5g/kg body weight (BW), orally); TAA (200 mg/kg BW, IP three times weekly/15 weeks); and sidr honey plus TAA at the same dose and administration rout. Rats co-treated with sidr honey plus TAA revealed significant reduction in hepatic malondialdehyde, hyaluronic acid (HA), alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, gamma glutamyl transferase, direct bilirubin, and hepatic mRNA expression of transforming growth factor (TGF)-ß1 and collagen type I alpha 1 chain (COL1a1) compared to TAA-exposed rats. In addition, the hepatoprotective potential of sidr honey was indicated via improvement of histopathologic picture of hepatocytes and upregulation of total antioxidant capacity, reduced glutathione, catalase, glutathione peroxidase, superoxide dismutase, total protein, and albumin compared to TAA-treated rats. In conclusion, daily administration of sidr honey (5 g/kg BW) is a promising natural antioxidant and fibrosuppressive agent that could ameliorate liver fibrosis via downregulation of fibrosis genes including TGF-ß1 and COL1a1 and HA and via enhancement of antioxidant system.


Subject(s)
Collagen Type I/genetics , Collagen Type I/metabolism , Down-Regulation/genetics , Gene Expression , Honey , Hyaluronic Acid/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/therapy , Oxidative Stress/genetics , Phytotherapy , Thioacetamide/adverse effects , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ziziphus , Animals , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Liver Cirrhosis/chemically induced , Male , Rats, Wistar
14.
J Histochem Cytochem ; 68(12): 907-927, 2020 12.
Article in English | MEDLINE | ID: mdl-32639183

ABSTRACT

Inter-α-trypsin inhibitor (IαI) family members are ancient and unique molecules that have evolved over several hundred million years of vertebrate evolution. IαI is a complex containing the proteoglycan bikunin to which heavy chain proteins are covalently attached to the chondroitin sulfate chain. Besides its matrix protective activity through protease inhibitory action, IαI family members interact with extracellular matrix molecules and most notably hyaluronan, inhibit complement, and provide cell regulatory functions. Recent evidence for the diverse roles of the IαI family in both biology and pathology is reviewed and gives insight into their pivotal roles in tissue homeostasis. In addition, the clinical uses of these molecules are explored, such as in the treatment of inflammatory conditions including sepsis and Kawasaki disease, which has recently been associated with severe acute respiratory syndrome coronavirus 2 infection in children.


Subject(s)
Alpha-Globulins/metabolism , Alpha-Globulins/analysis , Animals , Arthritis/metabolism , Arthritis/pathology , Asthma/metabolism , Asthma/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Humans , Hyaluronic Acid/metabolism , Inflammation/metabolism , Inflammation/pathology , Sepsis/metabolism , Sepsis/pathology
15.
Mol Pharm ; 17(7): 2411-2425, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32437163

ABSTRACT

Tumor-targeted drug delivery via chemotherapy is very effective on cancer treatment. For potential anticancer agent such as Camptothecin (CPT), high chemotherapeutic efficacy and accurate tumor targeting are equally crucial. Inspired by special CD44 binding capability from hyaluronic acid (HA), in this study, novel HA-coated CPT nanocrystals were successfully prepared by an antisolvent precipitation method for tumor-targeted delivery of hydrophobic drug CPT. These HA-coated CPT nanocrystals demonstrated high drug loading efficiency, improved aqueous dispersion, prolonged circulation, and enhanced stability resulting from their nanoscaled sizes and hydrophilic HA layer. Moreover, as compared to crude CPT and naked CPT nanocrystals, HA-coated CPT nanocrystals displayed dramatically enhanced in vitro anticancer activity, apoptosis-inducing potency against CD44 overexpressed cancer cells, and lower toxic effect toward normal cells due to pH-responsive drug release behavior and specific HA-CD44 mediated endocytosis. Additionally, HA-coated CPT nanocrystals performed fairly better antimigration activity and biocompatibility. The possible molecular mechanism regarding this novel drug formulation might be linked to intrinsic mitochondria-mediated apoptosis by an increase of Bax to Bcl-2 ratio and upregulation of P53. Consequently, HA-coated CPT nanocrystals are expected to be an effective nanoplatform in drug delivery for cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Camptotheca/chemistry , Camptothecin/administration & dosage , Drug Delivery Systems/methods , Hyaluronic Acid/chemistry , Nanoparticles/chemistry , Neoplasms/metabolism , Phytotherapy/methods , Plant Extracts/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Camptothecin/chemistry , Cell Movement/drug effects , Cell Survival/drug effects , Drug Liberation , Drug Stability , Hep G2 Cells , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Hydrogen-Ion Concentration , MCF-7 Cells , Neoplasms/pathology , Particle Size , Plant Extracts/chemistry , Protein Binding
16.
Mater Sci Eng C Mater Biol Appl ; 112: 110885, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32409042

ABSTRACT

A fixed combination of bimatoprost/timolol eye drop solution is used to manage the elevated intra-ocular pressure in glaucoma patients, including individuals whose condition is poorly controlled by monotherapy. Eye drop solutions are generally given in high dose, due to poor ocular bioavailability. The high ocular dose of bimatoprost and timolol lead to hyperaemia and systemic cardiac side effects respectively. Here, we introduce multiple implant-laden contact lenses (IM) to passively deliver timolol, bimatoprost and hyaluronic acid at therapeutically relevant doses without high burst release. The drug-loaded implants were individually implanted in the outer periphery of the silicone contact lenses. Atomic force microscopy showed the smooth surface of the implant contact lens, as the implants were inside the contact lens matrix. The implant lens (IM) showed major loss of drugs [timolol = 60.60%, bimatoprost = 61.75% and HA = 46.03%] during the monomer extraction and wet sterilization, while the option of dry radiation sterilization (IM-R lens) and hydration for 24 h prior to use showed relatively lower loss of drugs [timolol = 16.87%, bimatoprost = 47.95% and HA = 24.41%]. The in-vitro drugs release data of IM-R lens, showed sustained release for 72 h, with low burst release in comparison to the soaked (SM) and direct drug-laden contact lenses (DL). The in vivo drug release data in the rabbit tear fluid showed sustained release using IM-R lens in comparison to the SM lens and eye drop therapy. The burst release with the IM-R lens was many folds reduced, which could bypass the side effects associated with multiple eye drop therapy. The in vivo pharmacodynamic study in the rabbit model showed peak and valley profile with multiple eye drop therapy, while IM-R lens showed prolong reduction in intra ocular pressure (IOP) for 120 h. The study demonstrates the application of implantation technology to deliver multiple drug through contact lenses to treat glaucoma.


Subject(s)
Bimatoprost/metabolism , Contact Lenses , Drug Carriers/chemistry , Silicones/chemistry , Timolol/metabolism , Animals , Bimatoprost/administration & dosage , Bimatoprost/chemistry , Drug Implants/chemistry , Drug Liberation , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Intraocular Pressure , Rabbits , Surface Properties , Timolol/administration & dosage , Timolol/chemistry
17.
Nanoscale ; 12(17): 9541-9556, 2020 May 07.
Article in English | MEDLINE | ID: mdl-32314997

ABSTRACT

Atherosclerosis is associated with inflammation in the arteries, which is a major cause of heart attacks and strokes. Reducing the extent of local inflammation at atherosclerotic plaques can be an attractive strategy to combat atherosclerosis. While statins can exhibit direct anti-inflammatory activities, the high dose required for such a therapy renders it unrealistic due to their low systemic bioavailabilities and potential side effects. To overcome this, a new hyaluronan (HA)-atorvastatin (ATV) conjugate was designed with the hydrophobic statin ATV forming the core of the nanoparticle (HA-ATV-NP). The HA on the NPs can selectively bind with CD44, a cell surface receptor overexpressed on cells residing in atherosclerotic plaques and known to play important roles in plaque development. HA-ATV-NPs exhibited significantly higher anti-inflammatory effects on macrophages compared to ATV alone in vitro. Furthermore, when administered in an apolipoprotein E (ApoE)-knockout mouse model of atherosclerosis following a 1-week treatment regimen, HA-ATV-NPs markedly decreased inflammation in advanced atherosclerotic plaques, which were monitored through contrast agent aided magnetic resonance imaging. These results suggest CD44 targeting with HA-ATV-NPs is an attractive strategy to reduce harmful inflammation in atherosclerotic plaques.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Atorvastatin/administration & dosage , Hyaluronic Acid/chemistry , Nanoparticles/administration & dosage , Plaque, Atherosclerotic/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atorvastatin/chemistry , Atorvastatin/pharmacology , Drug Delivery Systems , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Inflammation , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Nanoparticles/chemistry , Nanoparticles/metabolism , Plaque, Atherosclerotic/pathology , RAW 264.7 Cells
18.
Nanoscale ; 12(15): 8139-8146, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32236229

ABSTRACT

Recent studies have suggested that the anticancer activity of disulfiram (DSF, an FDA-approved alcohol-abuse drug) is Cu-dependent. Low system toxicity and explicit pharmacokinetic characteristics of DSF necessitate safe and effective Cu supplementation in local lesion for further applications. Herein, we presented a new conceptual 'nanosized coordination transport' strategy of Cu(ii) that was realized in porphyrin-based metal-organic frameworks, Sm-TCPP, with strong binding ability to Cu(ii) due to their coordination interactions. Sm-TCPP(Cu) was coated by hyaluronic acid (HA) that termed by Sm-TCPP(Cu)@HA, acting as 'beneficial horse' to target the tumor-localized HA receptor (CD44), thus liberating Cu(ii) ions in cellular overexpressed reductants. The CD44-mediated Cu(ii) accumulation efficiency of Sm-TCPP(Cu)@HA was benchmarked in vitro and vivo against the free TCPP (Cu) via ICP-MS analysis. More importantly, the sensitization effects of Sm-TCPP(Cu)@HA on the anticancer activity of DSF were demonstrated in vivo and in vitro. This study offered a new class of targeted Cu supplements to sensitize DSF for the effective treatment of cancer and established a versatile methodology for constructing a safe and specific delivery of metal ions within living organisms.


Subject(s)
Copper/administration & dosage , Disulfiram/administration & dosage , Drug Delivery Systems , Hyaluronan Receptors/metabolism , Nanostructures/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Animals , Cell Line, Tumor , Copper/chemistry , Drug Carriers , Female , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Mice , Nanostructures/chemistry , Porphyrins/chemistry , Samarium/chemistry , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
Invest Ophthalmol Vis Sci ; 61(3): 39, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32196098

ABSTRACT

Purpose: Inflammation, hyaluronan production, and adipogenesis are the main pathological events leading to Graves' orbitopathy (GO). Guggulsterone (GS), a phytosterol found in the resin of the guggul plant, is a well-known treatment for several inflammatory disorders, such as arthritis, obesity, and hyperlipidemia. Here we investigated the effects of GS treatment on GO pathology. Methods: Using primary cultures of orbital fibroblasts from GO patients and non-GO controls, we examined the effects of GS on hyaluronan production and the production of proinflammatory cytokines induced by interleukin (IL)-1ß, using real-time reverse transcription-polymerase chain reaction analysis, western blots, and enzyme-linked immunosorbent assays. Further, adipogenic differentiation was evaluated by quantification of Oil Red O staining and assessment of protein levels of peroxisome proliferator activator gamma (PPARγ), CCAAT-enhancer-binding proteins (C/EBP) α and ß, and sterol regulatory element-binding protein-1 (SREBP-1). Results: Treatment with noncytotoxic concentrations of GS resulted in the dose-dependent inhibition of IL-1ß-induced inflammatory cytokines, including IL-6, IL-8, MCP-1, and COX-2, at both mRNA and protein levels. The hyaluronan level was also significantly suppressed by GS. Moreover, GS significantly decreased the formation of lipid droplets and expression of PPARγ, C/EBP α/ß, and SREBP-1 in a dose-dependent manner. GS pretreatment attenuated the phosphorylation of nuclear factor-kappa B induced by IL-1ß. Conclusions: Our data show significant inhibitory effects of GS on inflammation, production of hyaluronan, and adipogenesis in orbital fibroblasts. To our knowledge, this is the first in vitro preclinical evidence of the therapeutic effect of GS in GO.


Subject(s)
Fibroblasts/drug effects , Graves Ophthalmopathy/drug therapy , Orbit/drug effects , Pregnenediones/therapeutic use , Adipogenesis/drug effects , Adult , Aged , Blotting, Western , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation , Cells, Cultured , Commiphora/chemistry , Cytokines/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/metabolism , Graves Ophthalmopathy/metabolism , Humans , Hyaluronic Acid/metabolism , Male , Middle Aged , Orbit/metabolism , PPAR gamma/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/metabolism , Young Adult
20.
Mol Vis ; 26: 818-829, 2020.
Article in English | MEDLINE | ID: mdl-33456301

ABSTRACT

Purpose: A growing number of studies on animal models have demonstrated that some ocular diseases are the result of the interaction between hyalocytes and the ocular inflammatory setting. Endogenous and exogenous substances might alter the structure and behavior of hyalocytes that can contribute to the pathogenesis of some ocular diseases. Obtaining primary cultures of human hyalocytes could help understand the role of these cells in response to different treatments. Methods: Hyalocytes were isolated from eyes of 54 patient volunteers subjected to vitrectomy for different clinical reasons. By testing different matrices and growth media, we reproducibly generated primary cultures of hyalocytes that we characterized morphologically and biologically, basally and upon treatment with several agents (basic fibroblast growth factor (bFGF), transforming growth factor beta 1 (TGF-ß), platelet-derived growth factor subunit-BB (PDGF-BB), ascorbic acid, dexamethasone, and hydrogen peroxide). Results: We were able to generate primary cultures from vitreous human samples, growing the cells on collagen-coated plates in Iscove's modified Dulbecco's medium supplemented with 10% fetal bovine serum; primary cells expressed the hyalocyte markers. Specific cytoskeletal modifications were observed upon treatment with bFGF, TGF-ß, PDGF-BB, ascorbic acid, dexamethasone, and hydrogen peroxide. Only bFGF was able to promote cell growth and hyaluronic acid production. Conclusions: We describe for the first time the generation and the characterization of primary cultures of human hyalocytes from living donors. Although human hyalocytes share some characteristics with animal hyalocytes, human hyalocytes have their own features typical of the species, confirming how important human experimental models are for investigating human pathologies and their treatments.


Subject(s)
Cell Culture Techniques/methods , Vitreous Body/cytology , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Proliferation , Cells, Cultured , Female , Humans , Hyaluronic Acid/metabolism , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL