Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.849
Filter
1.
Plant Cell Rep ; 43(6): 160, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38825616

ABSTRACT

KEY MESSAGE: LeBAHD56 is preferentially expressed in tissues where shikonin and its derivatives are biosynthesized, and it confers shikonin acylation in vivo. Two WRKY transcriptional factors might regulate LeBAHD56's expression. Shikonin and its derivatives, found in the roots of Lithospermum erythrorhizon, have extensive application in the field of medicine, cosmetics, and other industries. Prior research has demonstrated that LeBAHD1(LeSAT1) is responsible for the biochemical process of shikonin acylation both in vitro and in vivo. However, with the exception of its documented in vitro biochemical function, there is no in vivo genetic evidence supporting the acylation function of the highly homologous gene of LeSAT1, LeBAHD56(LeSAT2), apart from its reported role. Here, we validated the critical acylation function of LeBAHD56 for shikonin using overexpression (OE) and CRISPR/Cas9-based knockout (KO) strategies. The results showed that the OE lines had a significantly higher ratio of acetylshikonin, isobutyrylshikonin or isovalerylshikonin to shikonin than the control. In contrast, the KO lines had a significantly lower ratio of acetylshikonin, isobutyrylshikonin or isovalerylshikonin to shikonin than controls. As for its detailed expression patterns, we found that LeBAHD56 is preferentially expressed in roots and callus cells, which are the biosynthesis sites for shikonin and its derivatives. In addition, we anticipated that a wide range of putative transcription factors might control its transcription and verified the direct binding of two crucial WRKY members to the LeBAHD56 promoter's W-box. Our results not only confirmed the in vivo function of LeBAHD56 in shikonin acylation, but also shed light on its transcriptional regulation.


Subject(s)
Gene Expression Regulation, Plant , Lithospermum , Naphthoquinones , Plant Proteins , Plants, Genetically Modified , Naphthoquinones/metabolism , Lithospermum/genetics , Lithospermum/metabolism , Acylation , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Roots/genetics , Plant Roots/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , CRISPR-Cas Systems , Anthraquinones
2.
Oncoimmunology ; 13(1): 2363000, 2024.
Article in English | MEDLINE | ID: mdl-38846085

ABSTRACT

NAD(P)H:quinone oxidoreductase 1 (NQO1) is overexpressed in most solid cancers, emerging as a promising target for tumor-selective killing. ß-Lapachone (ß-Lap), an NQO1 bioactivatable drug, exhibits significant antitumor effects on NQO1-positive cancer cells by inducing immunogenic cell death (ICD) and enhancing tumor immunogenicity. However, the interaction between ß-Lap-mediated antitumor immune responses and neutrophils, novel antigen-presenting cells (APCs), remains unknown. This study demonstrates that ß-Lap selectively kills NQO1-positive murine tumor cells by significantly increasing intracellular ROS formation and inducing DNA double strand breaks (DSBs), resulting in DNA damage. Treatment with ß-Lap efficiently eradicates immunocompetent murine tumors and significantly increases the infiltration of tumor-associated neutrophils (TANs) into the tumor microenvironment (TME), which plays a crucial role in the drug's therapeutic efficacy. Further, the presence of ß-Lap-induced antigen medium leads bone marrow-derived neutrophils (BMNs) to directly kill murine tumor cells, aiding in dendritic cells (DCs) recruitment and significantly enhancing CD8+ T cell proliferation. ß-Lap treatment also drives the polarization of TANs toward an antitumor N1 phenotype, characterized by elevated IFN-ß expression and reduced TGF-ß cytokine expression, along with increased CD95 and CD54 surface markers. ß-Lap treatment also induces N1 TAN-mediated T cell cross-priming. The HMGB1/TLR4/MyD88 signaling cascade influences neutrophil infiltration into ß-Lap-treated tumors. Blocking this cascade or depleting neutrophil infiltration abolishes the antigen-specific T cell response induced by ß-Lap treatment. Overall, this study provides comprehensive insights into the role of tumor-infiltrating neutrophils in the ß-Lap-induced antitumor activity against NQO1-positive murine tumors.


Subject(s)
NAD(P)H Dehydrogenase (Quinone) , Naphthoquinones , Neutrophils , Tumor Microenvironment , Animals , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/immunology , Mice , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Cell Line, Tumor , Neutrophil Infiltration/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Female , Phenotype
4.
J Exp Clin Cancer Res ; 43(1): 159, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38840237

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) was historically considered to be less responsive to radiation therapy (RT) compared to other cancer indications. However, advancements in precision high-dose radiation delivery through single-fraction and multi-fraction stereotactic ablative radiotherapy (SABR) have led to better outcomes and reduced treatment-related toxicities, sparking renewed interest in using RT to treat RCC. Moreover, numerous studies have revealed that certain therapeutic agents including chemotherapies can increase the sensitivity of tumors to RT, leading to a growing interest in combining these treatments. Here, we developed a rational combination of two radiosensitizers in a tumor-targeted liposomal formulation for augmenting RT in RCC. The objective of this study is to assess the efficacy of a tumor-targeted liposomal formulation combining the mTOR inhibitor everolimus (E) with the survivin inhibitor YM155 (Y) in enhancing the sensitivity of RCC tumors to radiation. EXPERIMENTAL DESIGN: We slightly modified our previously published tumor-targeted liposomal formulation to develop a rational combination of E and Y in a single liposomal formulation (EY-L) and assessed its efficacy in RCC cell lines in vitro and in RCC tumors in vivo. We further investigated how well EY-L sensitizes RCC cell lines and tumors toward radiation and explored the underlying mechanism of radiosensitization. RESULTS: EY-L outperformed the corresponding single drug-loaded formulations E-L and Y-L in terms of containing primary tumor growth and improving survival in an immunocompetent syngeneic mouse model of RCC. EY-L also exhibited significantly higher sensitization of RCC cells towards radiation in vitro than E-L and Y-L. Additionally, EY-L sensitized RCC tumors towards radiation therapy in xenograft and murine RCC models. EY-L mediated induction of mitotic catastrophe via downregulation of multiple cell cycle checkpoints and DNA damage repair pathways could be responsible for the augmentation of radiation therapy. CONCLUSION: Taken together, our study demonstrated the efficacy of a strategic combination therapy in sensitizing RCC to radiation therapy via inhibition of DNA damage repair and a substantial increase in mitotic catastrophe. This combination therapy may find its use in the augmentation of radiation therapy during the treatment of RCC patients.


Subject(s)
Carcinoma, Renal Cell , DNA Repair , Kidney Neoplasms , Survivin , TOR Serine-Threonine Kinases , Xenograft Model Antitumor Assays , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/radiotherapy , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Animals , Survivin/metabolism , Humans , Mice , Cell Line, Tumor , Kidney Neoplasms/pathology , Kidney Neoplasms/radiotherapy , Kidney Neoplasms/drug therapy , DNA Repair/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Mitosis/drug effects , Mitosis/radiation effects , Imidazoles/pharmacology , DNA Damage , Everolimus/pharmacology , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Liposomes/pharmacology , MTOR Inhibitors/pharmacology , MTOR Inhibitors/therapeutic use
5.
J Ovarian Res ; 17(1): 101, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745186

ABSTRACT

BACKGROUND: Shikonin (SK), a naphthoquinone with anti-tumor effects, has been found to decrease production of tumor-associated exosomes (exo). This study aims to verify the treatment effect of SK on ovarian cancer (OC) cells, especially on the production of exo and their subsequent effect on macrophage polarization. METHODS: OC cells SKOV3 and A2780 were treated with SK. The exo were isolated from OC cells with or without SK treatment, termed OC exo and SK OC exo, respectively. These exo were used to treat PMA-induced THP-1 cells (M0 macrophages). M2 polarization of macrophages was determined by measuring the M2 specific cell surface markers CD163 and CD206 as well as the secretion of M2 cytokine IL-10. The functions of galectin 3 (LGALS3/GAL3) and ß-catenin in macrophage polarization were determined by gain- or loss-of-function assays. CB-17 SCID mice were subcutaneously injected with SKOV3 cells to generate xenograft tumors, followed by OC exo or SK OC exo treatment for in vivo experiments. RESULTS: SK suppressed viability, migration and invasion, and apoptosis resistance of OC cells in vitro. Compared to OC exo, SK OC exo reduced the M2 polarization of macrophages. Regarding the mechanism, SK reduced exo production in cancer cells, and it decreased the protein level of GAL3 in exo and recipient macrophages, leading to decreased ß-catenin activation. M2 polarization of macrophages was restored by LGALS3 overexpression but decreased again by the ß-catenin inhibitor FH535. Compared to OC exo, the SK OC exo treatment reduced the xenograft tumor growth in mice, and it decreased the M2 macrophage infiltration within tumor tissues. CONCLUSION: This study suggests that SK reduces M2 macrophage population in OC by repressing exo production and blocking exosomal GAL3-mediated ß-catenin activation.


Subject(s)
Exosomes , Galectin 3 , Macrophages , Naphthoquinones , Ovarian Neoplasms , beta Catenin , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Female , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Humans , Exosomes/metabolism , Animals , Macrophages/metabolism , Macrophages/drug effects , beta Catenin/metabolism , Galectin 3/metabolism , Mice , Cell Line, Tumor , Xenograft Model Antitumor Assays , Cell Movement/drug effects , Apoptosis/drug effects , Mice, SCID
6.
An Acad Bras Cienc ; 96(2): e20230375, 2024.
Article in English | MEDLINE | ID: mdl-38747836

ABSTRACT

In pursuit of potential agents to treat Chagas disease and leishmaniasis, we report the design, synthesis, and identification novel naphthoquinone hydrazide-based molecular hybrids. The compounds were subjected to in vitro trypanocide and leishmanicidal activities. N'-(1,4-Dioxo-1,4-dihydronaphthalen-2-yl)-3,5-dimethoxybenzohydrazide (13) showed the best performance against Trypanosoma cruzi (IC50 1.83 µM) and Leishmania amazonensis (IC50 9.65 µM). 4-Bromo-N'-(1,4-dioxo-1,4-dihydronaphthalen-2-yl)benzohydrazide (16) exhibited leishmanicidal activity (IC50 12.16 µM). Regarding trypanocide activity, compound 13 was low cytotoxic to LLC-MK2 cells (SI = 95.28). Furthermore, through molecular modeling studies, the cysteine proteases cruzain, rhodesain and CPB2.8 were identified as the potential biological targets.


Subject(s)
Drug Design , Hydrazines , Leishmania , Naphthoquinones , Trypanocidal Agents , Trypanosoma cruzi , Naphthoquinones/pharmacology , Naphthoquinones/chemistry , Naphthoquinones/chemical synthesis , Trypanosoma cruzi/drug effects , Trypanocidal Agents/pharmacology , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry , Leishmania/drug effects , Hydrazines/chemistry , Hydrazines/pharmacology , Animals , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Parasitic Sensitivity Tests , Inhibitory Concentration 50 , Structure-Activity Relationship , Cysteine Endopeptidases
7.
Cell Mol Biol Lett ; 29(1): 63, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698330

ABSTRACT

BACKGROUND: Endometrial cancer (EC) is one of the most common gynecological cancers. Herein, we aimed to define the role of specific myosin family members in EC because this protein family is involved in the progression of various cancers. METHODS: Bioinformatics analyses were performed to reveal EC patients' prognosis-associated genes in patients with EC. Furthermore, colony formation, immunofluorescence, cell counting kit 8, wound healing, and transwell assays as well as coimmunoprecipitation, cycloheximide chase, luciferase reporter, and cellular thermal shift assays were performed to functionally and mechanistically analyze human EC samples, cell lines, and a mouse model, respectively. RESULTS: Machine learning techniques identified MYH14, a member of the myosin family, as the prognosis-associated gene in patients with EC. Furthermore, bioinformatics analyses based on public databases showed that MYH14 was associated with EC chemoresistance. Moreover, immunohistochemistry validated MYH14 upregulation in EC cases compared with that in normal controls and confirmed that MYH14 was an independent and unfavorable prognostic indicator of EC. MYH14 impaired cell sensitivity to carboplatin, paclitaxel, and progesterone, and increased cell proliferation and metastasis in EC. The mechanistic study showed that MYH14 interacted with MYH9 and impaired GSK3ß-mediated ß-catenin ubiquitination and degradation, thus facilitating the Wnt/ß-catenin signaling pathway and epithelial-mesenchymal transition. Sesamolin, a natural compound extracted from Sesamum indicum (L.), directly targeted MYH14 and attenuated EC progression. Additionally, the compound disrupted the interplay between MYH14 and MYH9 and repressed MYH9-regulated Wnt/ß-catenin signaling. The in vivo study further verified sesamolin as a therapeutic drug without side effects. CONCLUSIONS: Herein, we identified that EC prognosis-associated MYH14 was independently responsible for poor overall survival time of patients, and it augmented EC progression by activating Wnt/ß-catenin signaling. Targeting MYH14 by sesamolin, a cytotoxicity-based approach, can be applied synergistically with chemotherapy and endocrine therapy to eventually mitigate EC development. This study emphasizes MYH14 as a potential target and sesamolin as a valuable natural drug for EC therapy.


Subject(s)
Endometrial Neoplasms , Glycogen Synthase Kinase 3 beta , Myosin Heavy Chains , beta Catenin , Humans , Female , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Cell Line, Tumor , beta Catenin/metabolism , beta Catenin/genetics , Mice , Cell Proliferation/drug effects , Mice, Nude , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects , Prognosis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Middle Aged , Naphthoquinones/pharmacology
8.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732208

ABSTRACT

The current study investigated the impact of cold stress on the morphological, physiological, and phytochemical properties of Juglans regia L. (J. regia) using in vitro microclone cultures. The study revealed significant stress-induced changes in the production of secondary antioxidant metabolites. According to gas chromatography-mass spectrometry (GC-MS) analyses, the stress conditions profoundly altered the metabolism of J. regia microclones. Although the overall spectrum of metabolites was reduced, the production of key secondary antioxidant metabolites significantly increased. Notably, there was a sevenfold (7×) increase in juglone concentration. These findings are crucial for advancing walnut metabolomics and enhancing our understanding of plant responses to abiotic stress factors. Additionally, study results aid in identifying the role of individual metabolites in these processes, which is essential for developing strategies to improve plant resilience and tolerance to adverse conditions.


Subject(s)
Antioxidants , Cold-Shock Response , Juglans , Phytochemicals , Juglans/metabolism , Juglans/chemistry , Phytochemicals/metabolism , Antioxidants/metabolism , Secondary Metabolism , Metabolomics/methods , Gas Chromatography-Mass Spectrometry , Metabolome , Naphthoquinones
9.
Luminescence ; 39(5): e4761, 2024 May.
Article in English | MEDLINE | ID: mdl-38807512

ABSTRACT

The fear of an increase in blood sugar can be very traumatic. Being diabetic either type I or type II leads to a disorder called diabetes distress having traits of stress, depression, and anxiety. Among risk factors of diabetes mellitus heavy and trace metal toxicity emerges as new risk factors reported in many studies. In this study we target toxic metals, viz., Ni2+, Zn2+, and Cu2+, involved in the pathogenesis of diabetes and diabetic stress with naphthazarin esters. The compounds C1-C3 isolated from the leaves and roots of Arnebia guttata were tested for their metal-binding ability in an aqueous medium in UV-Visible and nuclear magnetic resonance (NMR) studies. These probes are well-known naphthoquinones present in the Arnebia species. In the UV-Visible titrations of compounds C1-C3 with Na2+, K2+, Zn2+, Ca2+, Cu2+, Mg2+, Co2+, and Ni2+ ions, significant binding was observed with Ni2+, Cu2+, and Zn2+ ions in MeOH/H2O. There occurs a beautiful formation of red-shifted bands between the 520 to 620 nm range with a synergistic increase in absorbance. Also, the disappearance of proton peaks in the 1H NMR spectrum on addition of metal ions confirmed binding. Compounds C1-C3 isolated from A. guttata came out as potent Ni2+, Zn2+, and Cu2+ sensors that are reportedly involved in islet function and induction of diabetes.


Subject(s)
Esters , Naphthoquinones , Esters/chemistry , Naphthoquinones/chemistry , Diabetes Mellitus/metabolism , Neurotoxins/chemistry , Neurotoxins/metabolism , Water/chemistry , Molecular Structure , Plant Leaves/chemistry
10.
Zhongguo Zhong Yao Za Zhi ; 49(9): 2345-2354, 2024 May.
Article in Chinese | MEDLINE | ID: mdl-38812135

ABSTRACT

To investigate the effects of plumbagin on the proliferation and apoptosis of human hepatoma Huh-7 cells and its mechanism based on the creatine kinase B(CKB)/p53 signaling pathway. Huh-7 cells were treated with plumbagin from 1 to 12 µmol·L~(-1) for cell counting kit-8(CCK-8) assay, and 1, 3, and 6 µmol·L~(-1) were determined as low, medium, and high concentrations of plumbagin for subsequent experiments. CKB gene was knocked out in Huh-7 cells by clustered regularly interspaced short palindromic repeats(CRISPR)/CRISPR-associated proteins(Cas)-9 gene editing technology. CKB overexpression lentivirus was transfected into Huh-7 cells to up-regulate the expression of CKB. Cell proliferation and apoptosis were detected by plate cloning assay and flow cytometry. The mRNA expression of CKB was detected by quantitative real-time PCR(qRT-PCR). CKB, p53, mouse double minute 2 homolog(MDM2), B-cell lymphoma 2(Bcl-2), Bcl-2 associated X protein(Bax), and caspase-3 protein were detected by Western blot(WB). The results showed that plumbagin significantly inhibited the proliferation of Huh-7 cells and induced cell apoptosis. Compared with the control group, the apoptosis level was significantly increased in the plumbagin group, while the apoptosis level was significantly decreased in the plumbagin combined with the apoptosis inhibitor group. Plumbagin significantly down-regulated the protein expression levels of CKB, Bcl-2, and MDM2 and up-regulated the protein expression levels of p53, Bax, and caspase-3. Knockdown of the CKB gene decreased the proliferative ability of Huh-7 cells, increased the apoptotic rate, decreased the expression levels of Bcl-2 and MDM2 proteins, and increased the expression levels of p53, Bax, and caspase-3 proteins. After up-regulation of CKB expression, the proliferation ability of Huh-7 cells was enhanced, and the protein expression levels of Bcl-2 and MDM2 were elevated. The protein expression levels of p53, Bax, and caspase-3 were decreased. In addition, plumbagin reversed the effect of overexpression of CKB on the proliferation and apoptosis of Huh-7 cells. In conclusion, plumbagin significantly inhibited the proliferative ability of Huh-7 cells, and the mechanism may be related to the inhibition of CKB expression, activation of the p53 signaling pathway, and regulation of the expression of mitochondrial-associated apoptotic proteins, ultimately inducing cell apoptosis.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular , Cell Proliferation , Liver Neoplasms , Naphthoquinones , Signal Transduction , Tumor Suppressor Protein p53 , Humans , Naphthoquinones/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Signal Transduction/drug effects , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/drug therapy , Cell Line, Tumor , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism
11.
Biomaterials ; 309: 122608, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38744189

ABSTRACT

Necroptotic immunogenic cell death (ICD) can activate the human immune system to treat the metastasis and recurrence of triple-negative breast cancer (TNBC). However, developing the necroptotic inducer and precisely delivering it to the tumor site is the key issue. Herein, we reported that the combination of shikonin (SHK) and chitosan silver nanoparticles (Chi-Ag NPs) effectively induced ICD by triggering necroptosis in 4T1 cells. Moreover, to address the lack of selectivity of drugs for in vivo application, we developed an MUC1 aptamer-targeted nanocomplex (MUC1@Chi-Ag@CPB@SHK, abbreviated as MUC1@ACS) for co-delivering SHK and Chi-Ag NPs. The accumulation of MUC1@ACS NPs at the tumor site showed a 6.02-fold increase compared to the free drug. Subsequently, upon reaching the tumor site, the acid-responsive release of SHK and Chi-Ag NPs from MUC1@ACS NPs cooperatively induced necroptosis in tumor cells by upregulating the expression of RIPK3, p-RIPK3, and tetrameric MLKL, thereby effectively triggering ICD. The sequential maturation of dendritic cells (DCs) subsequently enhanced the infiltration of CD8+ and CD4+ T cells in tumors, while inhibiting regulatory T cells (Treg cells), resulting in the effective treatment of primary and distal tumor growth and the inhibition of TNBC metastasis. This work highlights the importance of nanoparticles in mediating drug interactions during necroptotic ICD.


Subject(s)
Chitosan , Metal Nanoparticles , Naphthoquinones , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Silver , Triple Negative Breast Neoplasms , Naphthoquinones/pharmacology , Naphthoquinones/chemistry , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Chitosan/chemistry , Silver/chemistry , Silver/pharmacology , Animals , Metal Nanoparticles/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Female , Necroptosis/drug effects , Humans , Mice , Immunogenic Cell Death/drug effects , Mice, Inbred BALB C , Mucin-1/metabolism , Drug Synergism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry
12.
J Enzyme Inhib Med Chem ; 39(1): 2357174, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38814149

ABSTRACT

Tyrosinase, a pivotal enzyme in melanin synthesis, is a primary target for the development of depigmenting agents. In this work, in vitro and in silico techniques were employed to identify novel tyrosinase inhibitors from a set of 12 anilino-1,4-naphthoquinone derivatives. Results from the mushroom tyrosinase activity assay indicated that, among the 12 derivatives, three compounds (1, 5, and 10) demonstrated the most significant inhibitory activity against mushroom tyrosinase, surpassing the effectiveness of the kojic acid. Molecular docking revealed that all studied derivatives interacted with copper ions and amino acid residues at the enzyme active site. Molecular dynamics simulations provided insights into the stability of enzyme-inhibitor complexes, in which compounds 1, 5, and particularly 10 displayed greater stability, atomic contacts, and structural compactness than kojic acid. Drug likeness prediction further strengthens the potential of anilino-1,4-naphthoquinones as promising candidates for the development of novel tyrosinase inhibitors for the treatment of hyperpigmentation disorders.


Subject(s)
Agaricales , Dose-Response Relationship, Drug , Enzyme Inhibitors , Monophenol Monooxygenase , Naphthoquinones , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Naphthoquinones/pharmacology , Naphthoquinones/chemistry , Naphthoquinones/chemical synthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Agaricales/enzymology , Structure-Activity Relationship , Molecular Structure , Molecular Docking Simulation , Molecular Dynamics Simulation
13.
Arch Dermatol Res ; 316(6): 235, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38795154

ABSTRACT

The aim of this study is to delineate the expression patterns of prolyl cis-trans isomerase NIMA-interacting protein 1 (Pin1), Glial cell-derived neurotrophic factor (GDNF), and Angiotensin II (ANG II) during the process of wound repair, and to ascertain the effects of Pin1, GDNF, and ANG II on the healing of wounds in a rat model. A total of 18 rats were allocated into three groups-sham (control), DMSO (vehicle control), and Pin1 inhibitor (treatment with juglone)-with six animals in each group. An animal model of wound healing was established, followed by the intraperitoneal administration of juglone. Tissue samples from the wounds were subsequently collected for histopathological evaluation. Expression levels of Pin1, GDNF, and Ang II were quantified. In addition, an in vitro model of wound healing was created using human umbilical vein endothelial cells (HUVEC), to assess cell proliferation, migration, and tube formation under conditions of juglone pre-treatment. The expression levels of Pin1, GDNF, and ANG II were notably elevated on 7-, and 10- days post-wound compared to those measured on 3-day. Contrastingly, pre-treatment with juglone significantly inhibited the expression of these molecules. Histological analyses, including HE (Hematoxylin and Eosin), Masson's trichrome, and EVG (Elastic van Gieson) staining, demonstrated that vascular angiogenesis, as well as collagen and elastin deposition, were substantially reduced in the juglone pre-treated group when compared to the normal group. Further, immunohistochemical analysis revealed a considerable decrease in CD31 expression in the juglone pre-treatment group relative to the normal control group. Pin1 serves as a pivotal facilitator of wound repair. The findings indicate that the modulation of Pin1, GDNF, and ANG II expression impacts the wound healing process in rats, suggesting potential targets for therapeutic intervention in human wound repair.


Subject(s)
Angiotensin II , Cell Proliferation , Glial Cell Line-Derived Neurotrophic Factor , Human Umbilical Vein Endothelial Cells , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones , Wound Healing , Animals , Wound Healing/drug effects , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Humans , Rats , Naphthoquinones/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Male , Cell Proliferation/drug effects , Angiotensin II/metabolism , Cell Movement/drug effects , Disease Models, Animal , Rats, Sprague-Dawley , Skin/pathology , Skin/metabolism , Skin/injuries , Skin/drug effects , Adaptor Proteins, Signal Transducing
14.
Int J Biol Macromol ; 270(Pt 2): 132413, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761911

ABSTRACT

Herein, 5-fluorouracil and shikonin (extracted from Fusarium tricinctum) were loaded in chitosan/pectin nanoparticle (CS/PEC-NPs), prepared by blending (B-CS/PEC-NPs) and coating (C-CS/PEC-NPs) methods. The nanoparticles characterized by Fourier Transform Infrared (FTIR), X-ray diffraction (XRD), Energy-dispersive X-ray (EDX), Scanning Electron Microscope (SEM) and Differential Light Scattering (DLS). Then, some properties of the nanoparticles such as drug release rate and the nanoparticles cytotoxicity were studied. The FTIR, XRD, EDX, SEM and DLS results showed that the nanoparticles synthesized properly with an almost spherical morphology, an average size of 82-93 nm for B-CS/PEC-NPs, an average diameter of below 100 nm (mostly 66-89 nm) for C-CS/PEC-NPs, and hydrodynamic diameter of 310-817 nm. The drug release results indicated the lower release rate of drugs for B-CS/PEC-NPs relative to C-CS/PEC-NPs at different pHs, high release rate of drugs for the nanoparticles in the simulated large intestinal fluids containing pectinase, and Korsmeyer-Peppas model for release of the drugs. The results showed more cytotoxicity of B-CS/PEC-NPs containing drugs, especially B-CS/PEC-NPs containing both drugs (B-CS/PEC/5-FU/SHK-NPs) after treating with pectinase (IC50 of 18.6 µg/mL). In conclusion, despite the limitation of C-CS/PEC-NPs for simultaneous loading of hydrophilic and hydrophobic drugs, B-CS/PEC-NPs showed suitable potency for loading and targeted delivery of the drugs.


Subject(s)
Chitosan , Colonic Neoplasms , Drug Carriers , Drug Liberation , Fluorouracil , Nanoparticles , Naphthoquinones , Pectins , Fluorouracil/chemistry , Fluorouracil/pharmacology , Fluorouracil/administration & dosage , Chitosan/chemistry , Pectins/chemistry , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Naphthoquinones/administration & dosage , Nanoparticles/chemistry , Drug Carriers/chemistry , Humans , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Drug Delivery Systems , Cell Line, Tumor , Particle Size
15.
PLoS One ; 19(5): e0299921, 2024.
Article in English | MEDLINE | ID: mdl-38814975

ABSTRACT

Non-small cell lung cancer (NSCLC) is one of the most common malignancies worldwide, and oxidative stress plays a crucial role in its development. Juglone, a naturally occurring naphthoquinone in J. mandshurica, exhibits significant cytotoxic activity against various cancer cell lines. However, whether the anticancer activity of juglone is associated with oxidative stress remains unexplored. In this study, mouse Lewis lung cancer (LLC) and human non-small cell lung cancer A549 cells were used to explore the anticancer mechanisms of juglone. Juglone inhibited LLC and A549 cells viability, with IC50 values of 10.78 µM and 9.47 µM, respectively, for 24 h, and substantially suppressed the migration and invasion of these two lung cancer cells. Additionally, juglone arrested the cell cycle, induced apoptosis, increased the cleavage of caspase 3 and the protein expression of Bax and Cyt c, and decreased the protein expression of Bcl-2 and caspase-3. Furthermore, juglone treatment considerably increased intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) levels, but suppressed glutathione peroxidase 4 (GPX4) and superoxide dismutase (SOD) activities. It also inhibited the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, which was attenuated by 1,3-diCQA (an activator of PI3K/Akt). Moreover, N-acetylcysteine (a ROS scavenger) partially reversed the positive effects of juglone in terms of migration, invasion, ROS production, apoptosis, and PI3K/Akt pathway-associated protein expression. Finally, in tumor-bearing nude mouse models, juglone inhibited tumor growth without any apparent toxicity and significantly induced apoptosis in NSCLC cells. Collectively, our findings suggest that juglone triggers apoptosis via the ROS-mediated PI3K/Akt pathway. Therefore, juglone may serve as a potential therapeutic agent for the treatment of NSCLC.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Naphthoquinones , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species , Signal Transduction , Naphthoquinones/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Reactive Oxygen Species/metabolism , Humans , Animals , Apoptosis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Signal Transduction/drug effects , A549 Cells , Cell Movement/drug effects , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor
16.
J Nat Prod ; 87(4): 1217-1221, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38630559

ABSTRACT

Two unusual naphthoquinones, named here as pleonotoquinones A (1) and B (2), were isolated along with two known anthraquinones (3 and 4) via chromatographic separations of an ethyl acetate extract of the roots of Pleonotoma jasminifolia. Compounds 1 and 2 are the first examples of quinones bearing a 2-methyloxepine moiety. The compounds were isolated with the aid of mass spectrometry and molecular networking, and their structures were resolved using 1D and 2D NMR and HRESIMS data. The isolated compounds were evaluated for their antiproliferative activity against human cancer cell lines, and compounds 1 and 2 displayed cytotoxicity against human colon cancer HCT116 cells (IC50 = 2.6 µM for compound 1 and IC50 = 4.3 µM for compound 2) and human liver cancer HepG2 cells (IC50 = 1.9 µM for compound 1 and IC50 = 6.4 µM for compound 2).


Subject(s)
Antineoplastic Agents, Phytogenic , Drug Screening Assays, Antitumor , Naphthoquinones , Plant Roots , Humans , Naphthoquinones/pharmacology , Naphthoquinones/chemistry , Naphthoquinones/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Molecular Structure , Plant Roots/chemistry , Hep G2 Cells , HCT116 Cells , Boraginaceae/chemistry
17.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(2): 416-421, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38660845

ABSTRACT

OBJECTIVE: To explore the effect of shikonin on autophagy and apoptosis of human promyelocytic leukemia cells and its possible mechanism. METHODS: Human promyelocytic leukemia cells NB4 in the logarithmic growth phase were divided into control group (untreated NB4 cells), shikonin group (0.3 µmol/L shikonin treatment), 740Y-P group (15 µmol/L PI3K/Akt/mTOR pathway activator 740Y-P treatment), shikonin+740Y-P group (0.3 µmol/L shikonin and 15 µmol/L 740Y-P co-treatment), after 24 hours of treatment, the cells were used for subsequent experiments. CCK-8 method was used to detect cell viability, monodansylcadaverine (MDC) staining to detect the aggregation of autophagic vesicles, flow cytometry to detect cell apoptosis, and Western blot to detect the expression of Beclin1, LC3, p62, Bax, cleaved caspase-3, Bcl-2 and PI3K/Akt/mTOR pathway related proteins. RESULTS: Compared with the control group, the purple punctate fluorescence intensity, apoptosis rate, Beclin1, LC3-Ⅱ/LC3-Ⅰ, cleaved caspase-3, and Bax protein expression in NB4 cells were increased in the shikonin group, while OD450 value (24, 48 h) and the expressions of Bcl-2 and p62 proteins were decreased (all P < 0.05). Compared with the control group, the purple punctate fluorescence intensity, apoptosis rate, Beclin1, LC3-Ⅱ/LC3-Ⅰ, cleaved caspase-3, and Bax protein expression in NB4 cells were decreased, while OD450 value (24, 48 h) and the expressions of Bcl-2 and p62 proteins were increased in the 740Y-P group (all P < 0.05). Compared with the shikonin group, the purple punctate fluorescence intensity, apoptosis rate, Beclin1, LC3-Ⅱ/LC3-Ⅰ, cleaved caspase-3, and Bax protein expression in NB4 cells were decreased, while OD450 value (24, 48 h) and the expressions of Bcl-2 and p62 proteins were increased in the shikonin+740Y-P group (all P < 0.05). Compared with the control group, the expression of PI3K/Akt/mTOR pathway related proteins p-PI3K, p-Akt, and p-mTOR in NB4 cells were significantly decreased in the shikonin group, while those in the 740Y-P group were increased (all P < 0.05). Compared with the shikonin group, the expressions of p-PI3K, p-Akt, and p-mTOR proteins in NB4 cells were significantly increased in the shikonin+740Y-P group (all P < 0.05). CONCLUSION: Shikonin may promote autophagy and apoptosis of NB4 cells by inhibiting PI3K/Akt/mTOR pathway.


Subject(s)
Apoptosis , Autophagy , Leukemia, Promyelocytic, Acute , Naphthoquinones , Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases , Humans , Autophagy/drug effects , Apoptosis/drug effects , Naphthoquinones/pharmacology , Cell Line, Tumor , Leukemia, Promyelocytic, Acute/pathology , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Cell Survival/drug effects , Caspase 3/metabolism , bcl-2-Associated X Protein/metabolism , Beclin-1/metabolism
18.
Eur J Med Chem ; 270: 116387, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38593589

ABSTRACT

Activating apoptosis has long been viewed as an anti-cancer process, but recently increasing evidence has accumulated that induction of ferroptosis has emerged as a promising strategy for cancer therapeutics. Glutathione peroxidase 4 (GPX4) is one of the pivotal factors regulating ferroptosis that targeted inhibition or degradation of GPX4 could effectively trigger ferroptosis. In this study, a series of ML162-quinone conjugates were constructed by using pharmacophore hybridization and bioisosterism strategies, with the aim of obtaining more active anticancer agents via the ferroptosis and apoptosis dual cell death processes. Of these compounds, GIC-20 was identified as the most active one that exhibited promising anticancer activity both in vitro and in vivo via ferroptosis and apoptosis dual-targeting processes, without obvious toxicity compared with ML162. On one hand, GIC-20 could trigger ferroptosis in cells by inducing intracellular lipid peroxide and ROS accumulation, and destroying mitochondrial structure. In addition to GPX4 inhibition, GIC-20 can also trigger ferroptosis via proteasomal-mediated degradation of GPX4, suggesting GIC-20 may function as a molecule glue degrader. On the other hand, GIC-20 can also induce apoptosis via upregulating the level of apoptotic protein Bax and downregulating the level of anti-apoptotic protein Bcl-2 in HT1080 cells. Furthermore, GIC-20 also enhanced the sensitivity of resistant MIA-PaCa-2-AMG510R cells to AMG510, suggesting the great potential of GIC-20 in overcoming the acquired resistance of KRASG12C inhibitors. Overall, GIC-20 represents a novel dual ferroptosis/apoptosis inducer warranting further development for cancer therapeutics and overcoming drug resistance.


Subject(s)
Aniline Compounds , Ferroptosis , Naphthoquinones , Neoplasms , Thiophenes , Humans , Naphthoquinones/pharmacology , Apoptosis
19.
ACS Appl Mater Interfaces ; 16(15): 18411-18421, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38584383

ABSTRACT

Cell necroptosis has presented great potential, acting as an effective approach against tumor apoptotic resistance, and it could be further enhanced via accompanying reactive oxygen species (ROS) overexpression. However, whether overproduced ROS assists the necroptotic pathway remains unclear. Thus, iron-palladium nanozyme (FePd NZ)- and shikonin (SKN)-encapsulated functional lipid nanoparticles (FPS-LNPs) were designed to investigate the ROS overexpression-enhanced SKN-induced necroptosis. In this system, SKN acts as an effective necroptosis inducer for cancer cells, and FePd NZ, a sensitive Fenton reaction catalyst, produces extra-intracellular ROS to reinforce the necroptotic pathway. Both in vitro and in vivo antitumor evaluation revealed that FPS-LNPs presented the best tumor growth inhibition efficacy compared with FP-LNPs or SKN-LNPs alone. Meanwhile, induced necroptosis by FPS-LNPs can further trigger the release of damage-associated molecular patterns (DAMPs) and antigens from dying tumor cells to activate the innate immune response. Taking biosafety into consideration, this study has provided a potential nanoplatform for cancer nanotherapy via inducing necroptosis to avoid apoptosis resistance and activate CD8+ T cell immune response.


Subject(s)
Liposomes , Nanoparticles , Naphthoquinones , Necroptosis , Neoplasms , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Apoptosis
20.
Mar Drugs ; 22(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38667780

ABSTRACT

Approximately 75,000 tons of different sea urchin species are globally harvested for their edible gonads. Applying a circular economy approach, we have recently demonstrated that non-edible parts of the Mediterranean Sea urchin Paracentrotus lividus can be fully valorized into high-value products: antioxidant pigments (polyhydroxynaphthoquinones-PHNQs) and fibrillar collagen can be extracted to produce innovative biomaterials for biomedical applications. Can waste from other edible sea urchin species (e.g., Sphaerechinus granularis) be similarly valorised? A comparative study on PHNQs and collagen extraction was conducted. PHNQ extraction yields were compared, pigments were quantified and identified, and antioxidant activities were assessed (by ABTS assay) and correlated to specific PHNQ presence (i.e., spinochrome E). Similarly, collagen extraction yields were evaluated, and the resulting collagen-based biomaterials were compared in terms of their ultrastructure, degradation kinetics, and resistance to compression. Results showed a partially similar PHNQ profile in both species, with significantly higher yield in P. lividus, while S. granularis exhibited better antioxidant activity. P. lividus samples showed higher collagen extraction yield, but S. granularis scaffolds showed higher stability. In conclusion, waste from different species can be successfully valorised through PHNQ and collagen extraction, offering diverse applications in the biomedical field, according to specific technical requirements.


Subject(s)
Antioxidants , Collagen , Paracentrotus , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Antioxidants/isolation & purification , Collagen/chemistry , Paracentrotus/chemistry , Naphthoquinones/chemistry , Naphthoquinones/isolation & purification , Sea Urchins/chemistry , Waste Products , Biocompatible Materials/chemistry , Food Loss and Waste
SELECTION OF CITATIONS
SEARCH DETAIL
...