Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Pediatr Pharmacol Ther ; 29(2): 130-134, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38596416

ABSTRACT

OBJECTIVES: Enoxaparin for the prevention of venous thromboembolism (VTE) in pediatric patients is -typically dosed twice a day. The use of once-daily dosing like that used in adult patients is limited because of a lack of safety and efficacy data. The aim of this study was to evaluate the safety and efficacy of -once-daily versus twice-daily dosing of enoxaparin for pediatric VTE prophylaxis based on incidence of thrombotic and bleeding events. METHODS: This was a 3-year retrospective chart review of enoxaparin received for VTE prophylaxis at -Cohen Children's Medical Center, New Hyde Park, NY. Exclusion criteria were age 18 years or older, and renal dysfunction. RESULTS: A total of 177 enoxaparin courses (81 in the once-daily and 96 in the twice-daily group) were included. The median dose in the once-daily group was 0.68 mg/kg/dose with dose capping at 40 mg/dose in 70% of patients. One patient in the once-daily group had a VTE, whereas no patients in the twice-daily group experienced a VTE. One major bleeding event occurred in the once-daily group (p = 0.46); however, minor bleeding events were comparable between the 2 groups (p = 0.69). CONCLUSIONS: Once-daily enoxaparin prophylaxis appears to be safe and effective based on minimal -differences in incidence of thrombotic and bleeding events when compared to twice-daily dosing. Based on this study, it may be reasonable to consider once-daily enoxaparin dosing for prophylaxis, especially in older children. A larger multicenter cohort study evaluating once-daily dosing for prophylaxis is warranted to validate the safety and efficacy specifically for risk-based dosing strategies.

2.
ACS Appl Mater Interfaces ; 16(10): 12217-12231, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38480984

ABSTRACT

Psoriasis, a prevalent chronic inflammatory skin ailment affecting approximately 2-3% of the global population, is characterized by persistent symptoms. Dexamethasone, a primary corticosteroid for treating psoriasis, demonstrates notable efficacy; however, its limited skin permeation results in documented adverse effects. To address this, the presented study employed a novel strategy to conjugate gold nanorod and dexamethasone and evaluate their potential for mitigating psoriatic inflammation using an imiquimod-induced mouse model and human skin cells. Our findings revealed enhanced cutaneous penetration of gold nanorod and dexamethasone conjugates compared with that of dexamethasone, owing to superior skin penetration. Gold nanorod and dexamethasone conjugates demonstrated an optimal pharmacological impact at minimal dosages without toxicity during extended use. To further enhance the effectiveness of gold nanorod and dexamethasone conjugates, 808 nm near-infrared laser irradiation, which reacts to gold, was additionally applied to achieve thermal elevation to expedite drug skin penetration. Supplementary laser irradiation at 808 nm significantly ameliorated psoriatic symptoms following deep gold nanorod and dexamethasone conjugates penetration. This corresponded with restored peroxisome proliferator-activated receptor-γ levels and accelerated dexamethasone release from the gold nanorod and dexamethasone conjugates complex. These findings highlight the potential of gold nanorod and dexamethasone conjugates to enhance drug penetration through dermal layers, thereby aiding psoriasis treatment. Moreover, its compatibility with photothermal therapy offers prospects for novel therapeutic interventions across various inflammatory skin disorders.


Subject(s)
Nanotubes , Psoriasis , Animals , Mice , Humans , Photothermal Therapy , Gold/pharmacology , Gold/therapeutic use , Psoriasis/drug therapy , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Inflammation/drug therapy
3.
Br J Cancer ; 128(4): 626-637, 2023 02.
Article in English | MEDLINE | ID: mdl-36522480

ABSTRACT

BACKGROUND: Malignant phyllodes tumour (MPT) is a rare breast malignancy with epithelial and mesenchymal features. Currently, there are no appropriate research models or effective targeted therapeutic approaches for MPT. METHODS: We collected fresh frozen tissues from nine patients with MPT and performed whole-exome and RNA sequencing. Additionally, we established patient-derived xenograft (PDX) models from patients with MPT and tested the efficacy of targeting dysregulated pathways in MPT using the PDX model from one MPT. RESULTS: MPT has unique molecular characteristics when compared to breast cancers of epithelial origin and can be classified into two groups. The PDX model derived from one patient with MPT showed that the mouse epithelial component increased during tumour growth. Moreover, targeted inhibition of platelet-derived growth factor receptor (PDGFR) and phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) by imatinib mesylate and PKI-587 showed in vivo tumour suppression effects. CONCLUSIONS: This study revealed the molecular profiles of MPT that can lead to molecular classification and potential targeted therapy, and suggested that the MPT PDX model can be a useful tool for studying the pathogenesis of fibroepithelial neoplasms and for preclinical drug screening to find new therapeutic strategies for MPT.


Subject(s)
Breast Neoplasms , Neoplasms, Fibroepithelial , Phyllodes Tumor , Humans , Animals , Mice , Female , Phosphatidylinositol 3-Kinases , Cell Line, Tumor , Imatinib Mesylate , Breast Neoplasms/pathology , Phyllodes Tumor/pathology , Xenograft Model Antitumor Assays , Mammals
4.
Int J Nanomedicine ; 17: 4599-4617, 2022.
Article in English | MEDLINE | ID: mdl-36199478

ABSTRACT

Purpose: The protein corona surrounding nanoparticles has attracted considerable attention as it induces subsequent inflammatory responses. Although mesoporous silica nanoparticles (MSN) are commonly used in medicines, cosmetics, and packaging, the inflammatory effects of the MSN protein corona on the cutaneous system have not been investigated till date. Methods: We examined the greater plasma protein adsorption on MSN leads to serious inflammatory reactions in Dermatophagoides farinae extract (DFE)-induced mouse atopic dermatitis (AD)-like skin inflammation because of increased uptake by keratinocytes. Results: We compare the AD lesions induced by MSN and colloidal (non-porous) silica nanoparticles (CSN), which exhibit different pore architectures but similar dimensions and surface chemistry. MSN-corona treatment of severe skin inflammation in a DFE-induced in vivo AD model greatly increases mouse ear epidermal thickness and infiltration of immune cells compared with the CSN-corona treatment. Moreover, MSN-corona significantly increase AD-specific immunoglobulins, serum histamine, and Th1/Th2/Th17 cytokines in the ear and lymph nodes. MSN-corona induce more severe cutaneous inflammation than CSN by significantly decreasing claudin-1 expression. Conclusion: This study demonstrates the novel impact of the MSN protein corona in inducing inflammatory responses through claudin-1 downregulation and suggests useful clinical guidelines for MSN application in cosmetics and drug delivery systems.


Subject(s)
Dermatitis, Atopic , Nanoparticles , Protein Corona , Adsorption , Animals , Claudin-1/therapeutic use , Cytokines/metabolism , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Histamine , Immunoglobulin E , Inflammation/drug therapy , Mice , Mice, Inbred BALB C , Plant Extracts/pharmacology , Silicon Dioxide/therapeutic use
5.
Adv Sci (Weinh) ; 9(34): e2202800, 2022 12.
Article in English | MEDLINE | ID: mdl-36180414

ABSTRACT

Although plasma is a promising technology in various fields, its clinical application is restricted by several limitations. A cold atmospheric plasma (CAP) patch is fabricated to help overcome hurdles, especially when treating skin diseases. This patch has surface dielectric barrier discharge, which generates reactive oxygen species (ROS) and reactive nitrogen species (RNS) on a flexible polymer film surface on which the embedded electrode induces a locally strong electric field. The effect of the CAP patch on psoriasis is also evaluated. The distinct characteristics of psoriasis between the lesion and non-lesion area allow the CAP patch to be suitable for only lesion area for its treatment. The CAP patch induces the opening of calcium channels in keratinocytes, thereby restoring abnormal keratinocyte differentiation and the collapse of the tight junction; thus, alleviating psoriatic symptoms. In addition, the favorable effect is due to the induction of ROS/RNS by the CAP patch, not the electric field generated during plasma generation. The findings indicate that the proposed portable CAP patch can help treat inflammatory skin disorders, especially psoriasis. As this can be used easily as a combination therapy with existing drugs, it may help reduce side effects caused by existing drugs.


Subject(s)
Plasma Gases , Plasma Gases/therapeutic use , Anti-Inflammatory Agents
6.
Mol Med Rep ; 24(6)2021 12.
Article in English | MEDLINE | ID: mdl-34664681

ABSTRACT

Psoriasis, a chronic inflammatory skin disease, is characterized by the excessive proliferation and impaired differentiation of epidermal keratinocytes and is accompanied by the increased infiltration of inflammatory cells. The condition requires long­term treatment and has no definitive cure. Hence, supplements and therapeutic agents have been intensely investigated. Gomisin M2 (GM2), a lignan extracted from Schisandra chinensis (Turcz). Baill. (Schisandraceae; S. chinensis), has demonstrated diverse pharmacological properties, including anticancer, anti­inflammatory and antiallergic effects. Based on these findings, the present study examined the effects of GM2 on an imiquimod (IMQ)­induced psoriasis mouse model and on keratinocytes stimulated by tumor necrosis factor (TNF)­α and interferon­Î³. IMQ was topically applied to the back skin of mice for 7 consecutive days, and the mice were orally administered CD. These results showed that the oral administration of GM2 suppressed the symptoms of psoriasis, as evidenced by reductions in skin thickness, psoriasis area severity index scores for psoriasis lesions, transepidermal water loss and myeloperoxidase (MPO)­associated cell infiltration. Furthermore, GM2 reduced the pathologically increased levels of immunoglobulin G2a, MPO and TNF­α in the serum and T helper (Th)1 and Th17 cell populations in the spleen. GM2 decreased the gene expression of inflammatory­related cytokines and chemokines and inhibited the expression of signal transducer and activator of transcription 1 and nuclear factor­κB in the activated keratinocytes. These results suggested that GM2 from S. chinensis is a potential therapeutic candidate to alleviate psoriasis­like skin inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Lignans/pharmacology , Psoriasis/drug therapy , Psoriasis/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Line , Cytokines/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Female , Humans , Imiquimod/toxicity , Inflammation/chemically induced , Inflammation/genetics , Interferon-gamma/toxicity , Keratinocytes/drug effects , Keratinocytes/metabolism , Lignans/therapeutic use , Mice, Inbred C57BL , NF-kappa B/metabolism , Psoriasis/chemically induced , Psoriasis/pathology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Th1 Cells/drug effects , Th17 Cells/drug effects , Tumor Necrosis Factor-alpha/toxicity
7.
Molecules ; 26(19)2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34641629

ABSTRACT

Psoriasis is a chronic inflammatory skin disease accompanied by excessive keratinocyte proliferation. Corticosteroids, vitamin D3 analogs, and calcineurin inhibitors, which are used to treat psoriasis, have diverse adverse effects, whereas natural products are popular due to their high efficiency and relatively low toxicity. The roots of the Cudrania tricuspidata (C. tricuspidata) are known to have diverse pharmacological effects, among which the anti-inflammatory effect is reported as a potential therapeutic agent in skin cells. Nevertheless, its effectiveness against skin diseases, especially psoriasis, is not fully elucidated. Here, we investigated the effect of cudraxanthone D (CD), extracted from the roots the C. tricuspidata Bureau, on psoriasis using an imiquimod (IMQ)-induced mouse model and the tumor necrosis factor (TNF)-α/interferon (IFN)-γ-activated keratinocytes. IMQ was topically applied to the back skin of C57BL/6 mice for seven consecutive days, and the mice were orally administered with CD. This resulted in reduced psoriatic characteristics, such as the skin thickness and Psoriasis Area Severity Index score, and the infiltration of neutrophils in IMQ-induced skin. CD inhibited the serum levels of TNF-α, immunoglobulin G2a, and myeloperoxidase, and the expression of Th1/Th17 cells in splenocytes. In TNF-α/IFN-γ-activated keratinocytes, CD reduced the expressions of CCL17, IL-1ß, IL-6, and IL-8 by inhibiting the phosphorylation of STAT1 and the nuclear translocation of NF-kB. Taken together, these results suggest that CD could be a potential drug candidate for the treatment of psoriasis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Imiquimod/adverse effects , Keratinocytes/cytology , Moraceae/chemistry , Psoriasis/drug therapy , Xanthones/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Disease Models, Animal , Female , Humans , Interferon-gamma/adverse effects , Keratinocytes/drug effects , Keratinocytes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/pharmacology , Plant Extracts/chemistry , Plant Roots/chemistry , Psoriasis/chemically induced , Psoriasis/immunology , Treatment Outcome , Tumor Necrosis Factor-alpha/pharmacology , Xanthones/pharmacology
8.
Nat Commun ; 12(1): 4840, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376661

ABSTRACT

Gastric cancer (GC) is commonly treated by chemotherapy using 5-fluorouracil (5-FU) derivatives and platinum combination, but predictive biomarker remains lacking. We develop patient-derived xenografts (PDXs) from 31 GC patients and treat with a combination of 5-FU and oxaliplatin, to determine biomarkers associated with responsiveness. When the PDXs are defined as either responders or non-responders according to tumor volume change after treatment, the responsiveness of PDXs is significantly consistent with the respective clinical outcomes of the patients. An integrative genomic and transcriptomic analysis of PDXs reveals that pathways associated with cell-to-cell and cell-to-extracellular matrix interactions enriched among the non-responders in both cancer cells and the tumor microenvironment (TME). We develop a 30-gene prediction model to determine the responsiveness to 5-FU and oxaliplatin-based chemotherapy and confirm the significant poor survival outcomes among cases classified as non-responder-like in three independent GC cohorts. Our study may inform clinical decision-making when designing treatment strategies.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Stomach Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Adenocarcinoma/genetics , Animals , Female , Fluorouracil/administration & dosage , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Oxaliplatin/administration & dosage , Stomach Neoplasms/genetics , Survival Analysis , Tumor Burden/drug effects , Tumor Burden/genetics
9.
Molecules ; 26(15)2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34361560

ABSTRACT

The extracts of Schisandra chinensis (Turcz.) Baill. (Schisandraceae) have various therapeutic effects, including inflammation and allergy. In this study, gomisin M2 (GM2) was isolated from S. chinensis and its beneficial effects were assessed against atopic dermatitis (AD). We evaluated the therapeutic effects of GM2 on 2,4-dinitrochlorobenzene (DNCB) and Dermatophagoides farinae extract (DFE)-induced AD-like skin lesions with BALB/c mice ears and within the tumor necrosis factor (TNF)-α and interferon (IFN)-γ-stimulated keratinocytes. The oral administration of GM2 resulted in reduced epidermal and dermal thickness, infiltration of tissue eosinophils, mast cells, and helper T cells in AD-like lesions. GM2 suppressed the expression of IL-1ß, IL-4, IL-5, IL-6, IL-12a, and TSLP in ear tissue and the expression of IFN-γ, IL-4, and IL-17A in auricular lymph nodes. GM2 also inhibited STAT1 and NF-κB phosphorylation in DNCB/DFE-induced AD-like lesions. The oral administration of GM2 reduced levels of IgE (DFE-specific and total) and IgG2a in the mice sera, as well as protein levels of IL-4, IL-6, and TSLP in ear tissues. In TNF-α/IFN-γ-stimulated keratinocytes, GM2 significantly inhibited IL-1ß, IL-6, CXCL8, and CCL22 through the suppression of STAT1 phosphorylation and the nuclear translocation of NF-κB. Taken together, these results indicate that GM2 is a biologically active compound that exhibits inhibitory effects on skin inflammation and suggests that GM2 might serve as a remedy in inflammatory skin diseases, specifically on AD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cyclooctanes/pharmacology , Dermatitis, Atopic , Dermatophagoides farinae/immunology , Dermis/immunology , Dinitrochlorobenzene/toxicity , Epidermis/immunology , NF-kappa B/immunology , STAT1 Transcription Factor/immunology , Animals , Anti-Inflammatory Agents/chemistry , Cyclooctanes/chemistry , Cytokines/immunology , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Mice , Mice, Inbred BALB C
10.
Int Immunopharmacol ; 98: 107883, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34153674

ABSTRACT

Specialized proresolving mediators are enzymatically oxygenated natural molecules derived from polyunsaturated fatty acids and are considered novel. These novel mediators include lipoxins from arachidonic acid, resolvins and protectins from omega-3 essential fatty acids, and new maresins. These mediators harbor potent dual proresolving and anti-inflammatory properties. Resolvins and protectins are known to be potent when administered to various inflammation-associated animal models of human diseases. Although psoriasis' etiology remains unknown, there is accumulating evidence indicating that cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-23, and IL-17, play pivotal roles in its development. Experimentally, resolvins, maresins, and lipoxins downregulate the cytokine expression of the IL-23/IL-17 axis and inhibition of mitogen-activated protein kinases and nuclear factor kappa-light-chain-enhancer of activated B (NF-κB) cell signaling transduction pathways. Here, we assessed the effects of protectin D1 (PD1) on imiquimod (IMQ)-induced psoriasiform skin inflammation and keratinocytes. PD1 showed clinical improvement in skin thickness, redness, and scaling in psoriasis mouse models. Moreover, PD1 decreased IL-1ß, IL-6, IL-17, and CXCL1 mRNA expressions and reduced STAT1 and NF-κB signaling pathway activation in lesions. Serum myeloperoxidase, IgG2a, IL-1ß, IL-6, IL-17, and TNF-α and spleen CD4+IFN-γ+IL-17+ T lymphocytes were reduced after PD1 treatment in IMQ-induced psoriasiform mouse models. In addition, IL-1ß, IL-6, IL-8, and IL-18BP gene expressions were decreased in PD1-treated keratinocytes. Moreover, a decrease in the expression levels of CCL17 and IL-6 and an inhibition of the STAT1 and NF-κB signaling transduction pathways was observed in keratinocytes. These PD1 anti-inflammatory effects suggest that it is a good therapeutic candidate for psoriasis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Docosahexaenoic Acids/pharmacology , Psoriasis/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Cytokines/metabolism , Disease Models, Animal , Docosahexaenoic Acids/therapeutic use , Female , HaCaT Cells , Humans , Imiquimod/administration & dosage , Imiquimod/immunology , Injections, Subcutaneous , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/metabolism , Mice , Phosphorylation/drug effects , Phosphorylation/immunology , Psoriasis/immunology , Psoriasis/pathology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/drug effects , Skin/immunology , Skin/pathology , Spleen/cytology , Spleen/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
11.
Oncogene ; 40(18): 3287-3302, 2021 05.
Article in English | MEDLINE | ID: mdl-33846570

ABSTRACT

Metastatic or recurrent colorectal cancer (CRC) patients require systemic chemotherapy, but the therapeutic options of targeted agents remain limited. CRC patients with KRAS or BRAF gene mutations exhibit a worse prognosis and are resistant to anti-EGFR treatment. Previous studies have shown that the expression of anti-apoptotic protein BCL-XL is increased in CRC patients with KRAS/BRAF mutations, suggesting BCL-XL as a therapeutic target for this subgroup. Here, we performed genome-wide CRISPR/Cas9 screens of cell lines with KRAS mutations to investigate the factors required for sensitivity to BCL-XL inhibitor ABT-263 using single-guide RNAs (sgRNAs) that induce loss-of-function mutations. In the presence of ABT-263, sgRNAs targeting negative regulators of WNT signaling (resulting in WNT activation) were enriched, whereas sgRNAs targeting positive regulators of WNT signaling (resulting in WNT inhibition) were depleted in ABT-263-resistant cells. The activation of WNT signaling was highly associated with an increased expression ratio of anti- to pro-apoptotic BCL-2 family genes in CRC samples. Genetic and pharmacologic inhibition of WNT signaling using ß-catenin short hairpin RNA or TNIK inhibitor NCB-0846, respectively, augmented ABT-263-induced cell death in KRAS/BRAF-mutated cells. Inhibition of WNT signaling resulted in transcriptional repression of the anti-apoptotic BCL-2 family member, MCL1, via the functional inhibition of the ß-catenin-containing complex at the MCL1 promoter. In addition, the combination of ABT-263 and NCB-0846 exhibited synergistic effects in in vivo patient-derived xenograft (PDX) models with KRAS mutations. Our data provide a novel targeted combination treatment strategy for the CRC patient subgroup with KRAS or BRAF mutations.


Subject(s)
Proto-Oncogene Proteins B-raf , Colorectal Neoplasms , Humans , Proto-Oncogene Proteins p21(ras) , Wnt Signaling Pathway
12.
Biomed Pharmacother ; 137: 111359, 2021 May.
Article in English | MEDLINE | ID: mdl-33761595

ABSTRACT

Atopic dermatitis (AD) is a chronic inflammatory skin disorder that affects 10-20% of the world's population. Therefore, the discovery of drugs for the treatment of AD is important for human health. Hispidulin (HPD; also known as scutellarein 6-methyl ether or dinatin) is a natural flavone that exerts anti-inflammatory effects. In the present study, the effectiveness of HPD on AD-like skin inflammation was investigated. We used a mouse AD model through repeated exposure of mice to 2,4-dinitrochlorobenzene and house dust mite extract (Dermatophagoides farinae extract, DFE) to the ears. In addition, tumor necrosis factor-α and interferon-γ-activated keratinocytes (HaCaT cells) were used to investigate the underlying mechanism of HPD action. Oral administration of HPD alleviated AD-like skin inflammations: it reduced ear thickness; serum immunoglobulin (Ig)E, DFE-specific IgE, and IgG2a levels; and inflammatory cell infiltration. HPD reduced the expression of pro-inflammatory cytokines and chemokines through inhibition of signal transducer and activator of transcription 1 nuclear factor-κB in HaCaT cells. Taken together, these results suggest that HPD could be a potential drug candidate for the treatment of AD.


Subject(s)
Anti-Allergic Agents/therapeutic use , Dermatitis, Atopic/drug therapy , Dinitrochlorobenzene , Flavones/therapeutic use , Pyroglyphidae/immunology , Skin/pathology , Animals , Antigens, Dermatophagoides , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Eosinophils/drug effects , Female , Immunoglobulins/metabolism , Keratinocytes/drug effects , Mast Cells/drug effects , Mice , Mice, Inbred BALB C
13.
Cancer Lett ; 497: 123-136, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33068701

ABSTRACT

Cancer chemotherapeutic drugs exert cytotoxic effects by modulating intracellular reactive oxygen species (ROS) levels. However, whether ROS modulates the efficacy of targeted therapeutics remains poorly understood. Previously, we reported that upregulation of the anti-apoptotic protein, BCL-XL, by KRAS activating mutations was a potential target for KRAS-mutant colorectal cancer (CRC) treatment. Here, we demonstrated that the BCL-XL targeting agent, ABT-263, increased intracellular ROS levels and targeting antioxidant pathways augmented the therapeutic efficacy of this BH3 mimetic. ABT-263 induced expression of genes associated with ROS response and increased intracellular ROS levels by enhancing mitochondrial superoxide generation. The superoxide dismutase inhibitor, 2-methoxyestradiol (2-ME), exhibited synergism with ABT-263 in KRAS-mutant CRC cell lines. This synergistic effect was attributed to the inhibition of mTOR-dependent translation of the anti-apoptotic MCL-1 protein via caspase 3-mediated cleavage of AKT and S6K. In addition, combination treatment of ABT-263 and 2-ME demonstrated a synergistic effect in in vivo patient-derived xenografts harboring KRAS mutations. Our data suggest a novel role for ROS in BH3 mimetic-based targeted therapy and provide a novel strategy for treatment of CRC patients with KRAS mutations.


Subject(s)
Aniline Compounds/pharmacology , Antioxidants/metabolism , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , 2-Methoxyestradiol/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Superoxide Dismutase/antagonists & inhibitors , Thioredoxins/antagonists & inhibitors , Transcriptome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Int Immunopharmacol ; 87: 106767, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32679548

ABSTRACT

Psoriasis is a chronic inflammatory skin disease characterized by hyperproliferation and abnormal differentiation of epidermal keratinocytes accompanied by increased infiltration of immune cells. Previous studies have demonstrated that hispidulin (4',5,7-trihydroxy-6-methoxyflavone, HPD) has various pharmacological benefits such as anti-fungal, anti-inflammation, and anti-allergic effects. This study investigated the effectiveness of HPD to treat psoriasis using an imiquimod (IMQ)-induced mouse model and activated keratinocytes. IMQ was topically applied to the back skin of mice for six consecutive days, and the mice were orally administered HPD. Based on the histological observation and immunological analysis, oral administration of HPD suppressed psoriatic characteristics including skin thickness, psoriasis area severity index, transepidermal water loss, and neutrophil infiltration. HPD alleviated pathologically increased levels of immunoglobulin G2a, myeloperoxidase, and tumor necrosis factor-α. Splenic Th1 and Th17 cell populations were also reduced by HPD in the murine model. In addition, in activated keratinocytes, HPD inhibited gene expression of Th1- and Th17-associated cytokines and chemokines, and phosphorylation of mitogen-activated protein kinases and nuclear factor-κB. In summary, HPD alleviates psoriasis skin inflammation in vivo and in vitro. Therefore, we suggest that HPD would be a potent therapeutic candidate for the treatment of psoriasis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Flavones/therapeutic use , Psoriasis/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Cytokines/immunology , Female , Flavones/pharmacology , Humans , Imiquimod , Keratinocytes/drug effects , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/immunology , Neutrophils/drug effects , Neutrophils/immunology , Psoriasis/chemically induced , Psoriasis/immunology , Spleen/drug effects , Spleen/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
15.
Dis Model Mech ; 13(5)2020 05 27.
Article in English | MEDLINE | ID: mdl-32179549

ABSTRACT

High-salt intake and high-fructose intake are risk factors for hypertension via oxidative stress and inflammation. T helper (Th)17 lymphocytes play an important role in the development of hypertension. Here, we tested the hypothesis that activation of pathogenic Th17 lymphocytes induces hypertension after high-fructose intake in Dahl salt-sensitive (SS) but not Dahl salt-resistant (SR) rats. Eight-week-old male SS and SR rats were offered 20% fructose solution or tap water only for 4 weeks. Systolic blood pressure was measured by the tail-cuff method. T lymphocyte [Th17 and T regulatory (Treg)] profiling was determined via flow cytometry. The expression of Th17-related (IL-17A, IL-17RA, IL-23R and RORγt) and Treg-related (IL-10, CD25, FOXP3 and TGFß) factors were measured via ELISA or qRT-PCR. Th17 lymphocytes isolated from high-fructose-fed SS rats were intraperitoneally injected into recipient SS and SR rats, and recombinant IL-23 protein was subcutaneously injected into SS and SR rats to induce hypertension.High-fructose intake induced hypertension via the activation of pathogenic Th17 lymphocytes in SS but not SR rats. Injection of activated Th17 lymphocytes isolated from fructose-fed SS rats induced hypertension via increase of serum IL-17A only in recipient SS rats. In addition, injection of IL-23 induced hypertension via activation of pathogenic Th17 lymphocytes only in SS rats.Thus, activation of pathogenic Th17 lymphocytes induces hypertension after high-fructose intake in SS but not SR rats. These results indicate that immunologic tolerance plays an important role in protection against hypertension in SR rats.


Subject(s)
Hypertension/immunology , Lymphocyte Activation/immunology , Th17 Cells/immunology , Animals , Blood Pressure , Body Weight , Cytokines/blood , Cytokines/metabolism , Forkhead Transcription Factors/metabolism , Fructose , Glucose Intolerance/blood , Glucose Intolerance/complications , Glucose Intolerance/immunology , Hypertension/blood , Hypertension/complications , Immediate-Early Proteins/metabolism , Interleukin-23/metabolism , Male , Models, Biological , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats, Inbred Dahl , Signal Transduction , Systole , T-Lymphocytes, Regulatory/immunology
16.
Front Pharmacol ; 10: 869, 2019.
Article in English | MEDLINE | ID: mdl-31427975

ABSTRACT

Mast cells are effector cells that induce allergic inflammation by secreting inflammatory mediators. Gomisin M2 (G.M2) is a lignan isolated from Schisandra chinensis (Turcz). Baill. exhibiting anti-cancer activities. We aimed to investigate the anti-allergic effects and the underlying mechanism of G.M2 in mast cell-mediated allergic inflammation. For the in vitro study, we used mouse bone marrow-derived mast cells, RBL-2H3, and rat peritoneal mast cells. G.M2 inhibited mast cell degranulation upon immunoglobulin E (IgE) stimulation by suppressing the intracellular calcium. In addition, G.M2 inhibited the secretion of pro-inflammatory cytokines. These inhibitory effects were dependent on the suppression of FcεRI-mediated activation of signaling molecules. To confirm the anti-allergic effects of G.M2 in vivo, IgE-mediated passive cutaneous anaphylaxis (PCA) and ovalbumin-induced active systemic anaphylaxis (ASA) models were utilized. Oral administration of G.M2 suppressed the PCA reactions in a dose-dependent manner. In addition, G.M2 reduced the ASA reactions, including hypothermia, histamine, interleukin-4, and IgE production. In conclusion, G.M2 exhibits anti-allergic effects through suppression of the Lyn and Fyn pathways in mast cells. According to these findings, we suggest that G.M2 has potential as a therapeutic agent for the treatment of allergic inflammatory diseases via suppression of mast cell activation.

17.
Clin Cancer Res ; 25(9): 2821-2834, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30670495

ABSTRACT

PURPOSE: Genomic and transcriptomic alterations during metastasis are considered to affect clinical outcome of colorectal cancers, but detailed clinical implications of metastatic alterations are not fully uncovered. We aimed to investigate the effect of metastatic evolution on in vivo treatment outcome, and identify genomic and transcriptomic alterations associated with drug responsiveness. EXPERIMENTAL DESIGN: We developed and analyzed patient-derived xenograft (PDX) models from 35 patients with colorectal cancer including 5 patients with multiple organ metastases (MOMs). We performed whole-exome, DNA methylation, and RNA sequencing for patient and PDX tumors. With samples from patients with MOMs, we conducted phylogenetic and subclonal analysis and in vivo drug efficacy test on the corresponding PDX models. RESULTS: Phylogenetic analysis using mutation, expression, and DNA methylation data in patients with MOMs showed that mutational alterations were closely connected with transcriptomic and epigenomic changes during the tumor evolution. Subclonal analysis revealed that initial primary tumors with larger number of subclones exhibited more dynamic changes in subclonal architecture according to metastasis, and loco-regional and distant metastases occurred in a parallel or independent fashion. The PDX models from MOMs demonstrated therapeutic heterogeneity for targeted treatment, due to subclonal acquisition of additional mutations or transcriptomic activation of bypass signaling pathway during tumor evolution. CONCLUSIONS: This study demonstrated in vivo therapeutic heterogeneity of colorectal cancers using PDX models, and suggests that acquired subclonal alterations in mutations or gene expression profiles during tumor metastatic processes can be associated with the development of drug resistance and therapeutic heterogeneity of colorectal cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Genetic Heterogeneity , Genome, Human , Mutation , Transcriptome , Animals , Apoptosis , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Phylogeny , Prognosis , Tumor Cells, Cultured , Exome Sequencing , Xenograft Model Antitumor Assays
18.
Blood ; 131(17): 1931-1941, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29475961

ABSTRACT

Epstein-Barr virus (EBV)-positive diffuse large B-cell lymphomas (EBV+-DLBLs) tend to occur in immunocompromised patients, such as the elderly or those undergoing solid organ transplantation. The pathogenesis and genomic characteristics of EBV+-DLBLs are largely unknown because of the limited availability of human samples and lack of experimental animal models. We observed the development of 25 human EBV+-DLBLs during the engraftment of gastric adenocarcinomas into immunodeficient mice. An integrated genomic analysis of the human-derived EBV+-DLBLs revealed enrichment of mutations in Rho pathway genes, including RHPN2, and Rho pathway transcriptomic activation. Targeting the Rho pathway using a Rho-associated protein kinase (ROCK) inhibitor, fasudil, markedly decreased tumor growth in EBV+-DLBL patient-derived xenograft (PDX) models. Thus, alterations in the Rho pathway appear to contribute to EBV-induced lymphomagenesis in immunosuppressed environments.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Viral , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , rho GTP-Binding Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/virology , Animals , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/virology , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/virology , rho GTP-Binding Proteins/genetics
19.
Mol Cancer Ther ; 16(10): 2178-2190, 2017 10.
Article in English | MEDLINE | ID: mdl-28611106

ABSTRACT

Colorectal cancer is the third most commonly diagnosed cancer in the world, and exhibits heterogeneous characteristics in terms of genomic alterations, expression signature, and drug responsiveness. Although there have been considerable efforts to classify this disease based on high-throughput sequencing techniques, targeted treatments for specific subgroups have been limited. KRAS and BRAF mutations are prevalent genetic alterations in colorectal cancers, and patients with mutations in either of these genes have a worse prognosis and are resistant to anti-EGFR treatments. In this study, we have found that a subgroup of colorectal cancers, defined by having either KRAS or BRAF (KRAS/BRAF) mutations and BCL2L1 (encoding BCL-XL) amplification, can be effectively targeted by simultaneous inhibition of BCL-XL (with ABT-263) and MCL1 (with YM-155). This combination treatment of ABT-263 and YM-155 was shown to have a synergistic effect in vitro as well as in in vivo patient-derived xenograft models. Our data suggest that combined inhibition of BCL-XL and MCL1 provides a promising treatment strategy for this genomically defined colorectal cancer subgroup. Mol Cancer Ther; 16(10); 2178-90. ©2017 AACR.


Subject(s)
Colorectal Neoplasms/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , bcl-X Protein/genetics , Aged , Aniline Compounds/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Humans , Imidazoles/administration & dosage , Mice , Mutation , Naphthoquinones/administration & dosage , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays
20.
Mol Cells ; 39(2): 77-86, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26831452

ABSTRACT

Cancer is a heterogeneous disease caused by diverse genomic alterations in oncogenes and tumor suppressor genes. Despite recent advances in high-throughput sequencing technologies and development of targeted therapies, novel cancer drug development is limited due to the high attrition rate from clinical studies. Patient-derived xenografts (PDX), which are established by the transfer of patient tumors into immunodeficient mice, serve as a platform for co-clinical trials by enabling the integration of clinical data, genomic profiles, and drug responsiveness data to determine precisely targeted therapies. PDX models retain many of the key characteristics of patients' tumors including histology, genomic signature, cellular heterogeneity, and drug responsiveness. These models can also be applied to the development of biomarkers for drug responsiveness and personalized drug selection. This review summarizes our current knowledge of this field, including methodologic aspects, applications in drug development, challenges and limitations, and utilization for precision cancer medicine.


Subject(s)
Graft Survival , Molecular Targeted Therapy/methods , Stomach Neoplasms/immunology , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Pharmacological/metabolism , Biphenyl Compounds/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Drug Synergism , Humans , Irinotecan , Mice , Mice, Nude , Mice, SCID , Nitrophenols/pharmacology , Piperazines/pharmacology , Precision Medicine , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Sulfonamides/pharmacology , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL