Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Cell Rep ; 38(1): 110197, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986346

ABSTRACT

AMP-activated protein kinase (AMPK) regulates the balance between cellular anabolism and catabolism dependent on energy resources to maintain proliferation and survival. Small-compound AMPK activators show anti-cancer activity in preclinical models. Using the direct AMPK activator GSK621, we show that the unfolded protein response (UPR) is activated by AMPK in acute myeloid leukemia (AML) cells. Mechanistically, the UPR effector protein kinase RNA-like ER kinase (PERK) represses oxidative phosphorylation, tricarboxylic acid (TCA) cycle, and pyrimidine biosynthesis and primes the mitochondrial membrane to apoptotic signals in an AMPK-dependent manner. Accordingly, in vitro and in vivo studies reveal synergy between the direct AMPK activator GSK621 and the Bcl-2 inhibitor venetoclax. Thus, selective AMPK-activating compounds kill AML cells by rewiring mitochondrial metabolism that primes mitochondria to apoptosis by BH3 mimetics, holding therapeutic promise in AML.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , Unfolded Protein Response/physiology , eIF-2 Kinase/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Line, Tumor , Citric Acid Cycle/drug effects , Drug Evaluation, Preclinical , Female , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Middle Aged , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , THP-1 Cells , U937 Cells , Young Adult
2.
Molecules ; 27(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35011413

ABSTRACT

Proliferating cancer cells have high energy demands, which is mainly obtained through glycolysis. The transmembrane trafficking of lactate, a major metabolite produced by glycolytic cancer cells, relies on monocarboxylate transporters (MCTs). MCT1 optimally imports lactate, although it can work bidirectionally, and its activity has been linked to cancer aggressiveness and poor outcomes. AZD3965, a specific MCT1 inhibitor, was tested both in vitro and in vivo, with encouraging results; a phase I clinical trial has already been undertaken. Thus, analysis of the experimental evidence using AZD3965 in different cancer types could give valuable information for its clinical use. This systematic review aimed to assess the in vivo anticancer activity of AZD3965 either alone (monotherapy) or with other interventions (combination therapy). Study search was performed in nine different databases using the keywords "AZD3965 in vivo" as search terms. The results show that AZD3965 successfully decreased tumor growth and promoted intracellular lactate accumulation, which confirmed its effectiveness, especially in combined therapy. These results support the setup of clinical trials, but other important findings, namely AZD3965 enhanced activity when given in combination with other therapies, or MCT4-induced treatment resistance, should be further considered in the clinical trial design to improve therapy response.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Monocarboxylic Acid Transporters/antagonists & inhibitors , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Symporters/antagonists & inhibitors , Thiophenes/pharmacology , Thiophenes/therapeutic use , Animals , Cell Line, Tumor , Disease Management , Disease Progression , Drug Evaluation, Preclinical , Energy Metabolism/drug effects , Glycolysis , Humans , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Symporters/metabolism , Tumor Microenvironment/drug effects , Warburg Effect, Oncologic/drug effects , Xenograft Model Antitumor Assays
3.
Comb Chem High Throughput Screen ; 24(3): 441-454, 2021.
Article in English | MEDLINE | ID: mdl-32748740

ABSTRACT

BACKGROUND: Coronavirus Disease 2019 (COVID-19) pandemic continues to threaten patients, societies and healthcare systems around the world. There is an urgent need to search for possible medications. OBJECTIVE: This article intends to use virtual screening and molecular docking methods to find potential inhibitors from existing drugs that can respond to COVID-19. METHODS: To take part in the current research investigation and to define a potential target drug that may protect the world from the pandemic of corona disease, a virtual screening study of 129 approved drugs was carried out which showed that their metabolic characteristics, dosages used, potential efficacy and side effects are clear as they have been approved for treating existing infections. Especially 12 drugs against chronic hepatitis B virus, 37 against chronic hepatitis C virus, 37 against human immunodeficiency virus, 14 anti-herpesvirus, 11 anti-influenza, and 18 other drugs currently on the market were considered for this study. These drugs were then evaluated using virtual screening and molecular docking studies on the active site of the (SARS-CoV-2) main protease (6lu7). Once the efficacy of the drug is determined, it can be approved for its in vitro and in vivo activity against the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), which can be beneficial for the rapid clinical treatment of patients. These drugs were considered potentially effective against SARS-CoV-2 and those with high molecular docking scores were proposed as novel candidates for repurposing. The N3 inhibitor cocrystallized with protease (6lu7) and the anti-HIV protease inhibitor Lopinavir were used as standards for comparison. RESULTS: The results suggest the effectiveness of Beclabuvir, Nilotinib, Tirilazad, Trametinib and Glecaprevir as potent drugs against SARS-CoV-2 since they tightly bind to its main protease. CONCLUSION: These promising drugs can inhibit the replication of the virus; hence, the repurposing of these compounds is suggested for the treatment of COVID-19. No toxicity measurements are required for these drugs since they were previously tested prior to their approval by the FDA. However, the assessment of these potential inhibitors as clinical drugs requires further in vivo tests of these drugs.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Coronavirus 3C Proteases/metabolism , Drug Evaluation, Preclinical/methods , SARS-CoV-2/drug effects , Antiviral Agents/metabolism , Binding Sites , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/chemistry , Drug Repositioning , Hepacivirus/drug effects , Influenza A virus/drug effects , Lopinavir/chemistry , Lopinavir/pharmacology , Molecular Docking Simulation , Pyridones/chemistry , Pyridones/pharmacology , Pyrimidinones/chemistry , Pyrimidinones/pharmacology
4.
Microbiologyopen ; 10(1): e1143, 2021 01.
Article in English | MEDLINE | ID: mdl-33269542

ABSTRACT

Carotenoids are widely used in functional foods, cosmetics, and health supplements, and their importance and scope of use are continuously expanding. Here, we characterized carotenoid biosynthetic genes of the plant-pathogenic bacterium Pantoea ananatis, which carries a carotenoid biosynthetic gene cluster (including crtE, X, Y, I, B, and Z) on a plasmid. Reverse transcription-polymerase chain reaction (RT-PCR) analysis revealed that the crtEXYIB gene cluster is transcribed as a single transcript and crtZ is independently transcribed in the opposite direction. Using splicing by overlap extension with polymerase chain reaction (SOE by PCR) based on asymmetric amplification, we reassembled crtE-B, crtE-B-I, and crtE-B-I-Y. High-performance liquid chromatography confirmed that Escherichia coli expressing the reassembled crtE-B, crtE-B-I, and crtE-B-I-Y operons produced phytoene, lycopene, and ß-carotene, respectively. We found that the carotenoids conferred tolerance to UV radiation and toxoflavin. Pantoea ananatis shares rice environments with the toxoflavin producer Burkholderia glumae and is considered to be the first reported example of producing and using carotenoids to withstand toxoflavin. We confirmed that carotenoid production by P. ananatis depends on RpoS, which is positively regulated by Hfq/ArcZ and negatively regulated by ClpP, similar to an important regulatory network of E. coli (HfqArcZ →RpoS Í° ClpXP). We also demonstrated that Hfq-controlled quorum signaling de-represses EanR to activate RpoS, thereby initiating carotenoid production. Survival genes such as those responsible for the production of carotenoids of the plant-pathogenic P. ananatis must be expressed promptly to overcome stressful environments and compete with other microorganisms. This mechanism is likely maintained by a brake with excellent performance, such as EanR.


Subject(s)
Carotenoids/metabolism , Host Factor 1 Protein/metabolism , Pantoea/drug effects , Pantoea/metabolism , Pyrimidinones/pharmacology , Quorum Sensing/physiology , Triazines/pharmacology , Bacterial Proteins/metabolism , Endopeptidase Clp/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Multigene Family/genetics , Plasmids/genetics , Sigma Factor/metabolism , Ultraviolet Rays
5.
Proc Natl Acad Sci U S A ; 117(48): 30670-30678, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33199632

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is typically diagnosed at an advanced stage, which limits surgical options and portends a dismal prognosis. Current oncologic PDAC therapies confer marginal benefit and, thus, a significant unmet clinical need exists for new therapeutic strategies. To identify effective PDAC therapies, we leveraged a syngeneic orthotopic PDAC transplant mouse model to perform a large-scale, in vivo screen of 16 single-agent and 41 two-drug targeted therapy combinations in mice. Among 57 drug conditions screened, combined inhibition of heat shock protein (Hsp)-90 and MEK was found to produce robust suppression of tumor growth, leading to an 80% increase in the survival of PDAC-bearing mice with no significant toxicity. Mechanistically, we observed that single-agent MEK inhibition led to compensatory activation of resistance pathways, including components of the PI3K/AKT/mTOR signaling axis, which was overcome with the addition of HSP90 inhibition. The combination of HSP90(i) + MEK(i) was also active in vitro in established human PDAC cell lines and in vivo in patient-derived organoid PDAC transplant models. These findings encourage the clinical development of HSP90(i) + MEK(i) combination therapy and highlight the power of clinically relevant in vivo model systems for identifying cancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/therapeutic use , Benzodioxoles/pharmacology , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Drug Screening Assays, Antitumor/methods , Drug Synergism , Gene Expression , Humans , Immunohistochemistry , MAP Kinase Signaling System/drug effects , Mice , Molecular Targeted Therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Signal Transduction/drug effects , Survival Rate , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Mol Pain ; 16: 1744806920955103, 2020.
Article in English | MEDLINE | ID: mdl-32880221

ABSTRACT

Neuropathic pain is a chronic disease state resulting from injury to the nervous system. This type of pain often responds poorly to standard treatments and occasionally may get worse instead of better over time. Patients who experience neuropathic pain report sensitivity to cold and mechanical stimuli. Since the nociceptive system of African naked mole-rats contains unique adaptations that result in insensitivity to some pain types, we investigated whether naked mole-rats may be resilient to sensitivity following nerve injury. Using the spared nerve injury model of neuropathic pain, we showed that sensitivity to mechanical stimuli developed similarly in mice and naked mole-rats. However, naked mole-rats lacked sensitivity to mild cold stimulation after nerve injury, while mice developed robust cold sensitivity. We pursued this response deficit by testing behavior to activators of transient receptor potential (TRP) receptors involved in detecting cold in naïve animals. Following mustard oil, a TRPA1 activator, naked mole-rats responded similarly to mice. Conversely, icilin, a TRPM8 agonist, did not evoke pain behavior in naked mole-rats when compared with mice. Finally, we used RNAscope to probe for TRPA1 and TRPM8 messenger RNA expression in dorsal root ganglia of both species. We found increased TRPA1 messenger RNA, but decreased TRPM8 punctae in naked mole-rats when compared with mice. Our findings likely reflect species differences due to evolutionary environmental responses that are not easily explained by differences in receptor expression between the species.


Subject(s)
Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , Neuralgia/metabolism , TRPA1 Cation Channel/metabolism , TRPM Cation Channels/metabolism , Animals , Cold Temperature , Disease Models, Animal , Female , Ganglia, Spinal/injuries , Male , Mice , Mole Rats , Mustard Plant , Neurons/metabolism , Neurons/physiology , Nociception , Pain Measurement , Plant Oils/pharmacology , Pyrimidinones/pharmacology , TRPA1 Cation Channel/genetics , TRPM Cation Channels/agonists , TRPM Cation Channels/genetics
7.
Leukemia ; 34(11): 2951-2963, 2020 11.
Article in English | MEDLINE | ID: mdl-32576961

ABSTRACT

To establish novel and effective treatment combinations for chronic myelomonocytic leukemia (CMML) preclinically, we hypothesized that supplementation of CMML cells with the human oncogene Meningioma 1 (MN1) promotes expansion and serial transplantability in mice, while maintaining the functional dependencies of these cells on their original genetic profile. Using lentiviral expression of MN1 for oncogenic supplementation and transplanting transduced primary mononuclear CMML cells into immunocompromised mice, we established three serially transplantable CMML-PDX models with disease-related gene mutations that recapitulate the disease in vivo. Ectopic MN1 expression was confirmed to enhance the proliferation of CMML cells, which otherwise did not engraft upon secondary transplantation. Furthermore, MN1-supplemented CMML cells were serially transplantable into recipient mice up to 5 generations. This robust engraftment enabled an in vivo RNA interference screening targeting CMML-related mutated genes including NRAS, confirming that their functional relevance is preserved in the presence of MN1. The novel combination treatment with azacitidine and the MEK-inhibitor trametinib additively inhibited ERK-phosphorylation and thus depleted the signal from mutated NRAS. The combination treatment significantly prolonged survival of CMML mice compared to single-agent treatment. Thus, we identified the combination of azacitidine and trametinib as an effective treatment in NRAS-mutated CMML and propose its clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical , Leukemia, Myelomonocytic, Chronic/drug therapy , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/pharmacology , Clonal Evolution , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Drug Synergism , Female , GTP Phosphohydrolases/genetics , Humans , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Membrane Proteins/genetics , Mice , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , Xenograft Model Antitumor Assays/methods
8.
Drug Des Devel Ther ; 14: 1779-1798, 2020.
Article in English | MEDLINE | ID: mdl-32440103

ABSTRACT

BACKGROUND: Radiotherapy has an ameliorative effect on a wide variety of tumors, but hepatocellular carcinoma (HCC) is insensitive to this treatment. Overactivated mammalian target of rapamycin (mTOR) plays an important part in the resistance of HCC to radiotherapy; thus, mTOR inhibitors have potential as novel radiosensitizers to enhance the efficacy of radiotherapy for HCC. METHODS: A lead compound was found based on pharmacophore modeling and molecular docking, and optimized according to the differences between the ATP-binding pockets of mTOR and PI3K. The radiosensitizing effect of the optimized compound (2a) was confirmed by colony formation assays and DNA double-strand break assays in vitro. The discovery and preclinical characteristics of this compound are described. RESULTS: The key amino acid residues in mTOR were identified, and a precise virtual screening model was constructed. Compound 2a, with a 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidine scaffold, exhibited promising potency against mTOR (mTOR IC50=7.1 nmol/L (nM)) with 126-fold selectivity over PI3Kα. Moreover, 2a significantly enhanced the sensitivity of HCC to radiotherapy in vitro in a dose-dependent manner. CONCLUSION: A new class of selective mTOR inhibitors was developed and their radiosensitization effects were confirmed. This study also provides a basis for developing mTOR-specific inhibitors for use as radiosensitizers for HCC radiotherapy.


Subject(s)
Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Pyrimidinones/pharmacology , Radiation-Sensitizing Agents/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Models, Molecular , Molecular Structure , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/chemistry , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism
9.
Curr Med Sci ; 40(2): 354-362, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32337697

ABSTRACT

Imatinib mesylate (IM) is the first-line treatment for Philadelphia (Ph) chromosomal positive leukemia by inhibiting phosphorylation of substrates via binding to the ABL kinase domain. Because of the drug resistance, side effects and the high cost of IM, it is necessary to find anti-cancer drugs with relatively low toxicity and cost, and enhanced efficacy, such as traditional Chinese medicines (TCMs). As one of TCMs, Huai Qi Huang (HQH) was chosen to treat BV173 and K562 cells. Various concentrations of HQH were added to cells for 24-72 h. Co-treatment of HQH and trametinib, an MEK inhibitor, was used to verify the synergistic effects on cell viability and apoptosis. Knockdown and overexpression of mitogen-activated protein kinase kinase 4 (MEK4) were implemented to demonstrate the role of MEK in cell apoptosis. Cell viability and apoptosis were measured by cell counting kit-8 assay (CCK8) and flow cytometry, respectively. Western blotting and real-time quantitative PCR (RT-qPCR) were used to assess protein and mRNA expression levels, respectively. The results showed that HQH inhibited survival and promoted apoptosis of BV173 and K562 cells in a dose-dependent manner, accompanied with down-regulation of PRKCH mRNA as well as CRAF, MEK4, phospho-ERK (pERK) and BCL2 proteins, and up-regulation of cleaved caspase3 protein. Co-treatment of HQH and trametinib had a synergistic effect on inhibiting survival and promoting apoptosis. MEK4 knockdown increased apoptosis, and had a synergistic effect with HQH. In contrast, MEK4 overexpression decreased apoptosis, and had the opposite effect with HQH. Collectively, the results of this study may identify a therapeutic mechanism of HQH on promoting apoptosis, and provide a potential option for treatment of Ph+ leukemia.


Subject(s)
Down-Regulation , Drugs, Chinese Herbal/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Protein Kinase C/metabolism , Pyridones/pharmacology , Pyrimidinones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MAP Kinase Signaling System/drug effects , Protein Kinase C/genetics
10.
Theranostics ; 10(2): 782-796, 2020.
Article in English | MEDLINE | ID: mdl-31903150

ABSTRACT

Nanobubble (NB), which simultaneously enhances ultrasound (US) images and access therapeutic platforms, is required for future cancer treatment. Methods: We designed a theranostic agent for novel cancer treatment by using an NB-encapsulated hybrid nanosystem that can be monitored by US and fluorescent imaging and activated by near-infrared (NIR) light. The nanosystem was transported to the tumor through the enhanced permeability and retention effect. The hybrid nanosystem comprised upconversion nanoparticle (UCNP) and mesoporous silica-coated gold nanorod (AuNR@mS) with the photosensitizer merocyanine 540 to realize dual phototherapy. Results: With the NIR light-triggered, the luminous intensity of the UCNP was enhanced by doping holmium ion and emitted visible green and red lights at 540 and 660 nm. The high optical density state between the UCNP and AuNR@mS can induce plasmonic enhancement to improve the photothermal and photodynamic effects, resulting in cell death by apoptosis. The nanosystem showed excellent stability to avoid the aggregation of nanoparticles during the treatment. JC-1 dye was used as an indicator of mitochondrial membrane potential to identify the mechanism of cell death. The results of in vitro and in vivo analyses confirmed the curative effect of improved dual phototherapy. Conclusion: We developed and showed the therapeutic functions of a novel nanosystem with the combination of multiple theranostic nanoplatforms that can be triggered and activated by 808 nm NIR laser and US.


Subject(s)
Gold/chemistry , Lung Neoplasms/therapy , Nanoparticles/administration & dosage , Phototherapy/methods , Pyrimidinones/pharmacology , Theranostic Nanomedicine/methods , Animals , Cell Death , Cell Line, Tumor , Diagnostic Imaging/methods , Humans , Hyperthermia, Induced/methods , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Mice , Models, Animal , Nanoparticles/chemistry , Photosensitizing Agents/pharmacology
11.
Kidney Int ; 96(4): 927-941, 2019 10.
Article in English | MEDLINE | ID: mdl-31377057

ABSTRACT

Transforming growth factor-ß (TGF-ß) plays crucial roles in the development of focal segmental glomerulosclerosis, but key molecular pathways remain unknown. Here, we identified the regulation of mammalian target of rapamycin complex1 (mTORC1) by TGF-ß via ERK1/2 in the Adriamycin-induced murine model of focal segmental glomerulosclerosis. Adriamycin administration elicited early activation of TGF-ß-ERK1/2-mTORC1 in podocytes, which persisted at later stages of albuminuria and glomerulosclerosis. Phosphorylation of the TGF-ß receptor-I (TGF-ßRI), Smad3, ERK1/2 and ribosomal protein S6 were evident in the glomeruli of adriamycin-treated mice. Targeting TGFß-RI and mTORC1 with pharmacological inhibitors suppressed TGF-ß signaling in glomeruli and significantly reduced albuminuria, glomerulosclerosis, protein levels of collagen 4α3, plasminogen activator inhibitor-1, and vimentin and restored mRNA levels of podocyte markers. Low dose US Food and Drug Administration (FDA)-approved MEK/ERK inhibitor trametinib/GSK1120212 blunted TGF-ß1-induced mTORC1 activation in podocytes, ameliorated up-regulation of TGF-ß, plasminogen activator inhibitor-1, monocyte chemoattractant protein-1, fibronectin and α-smooth muscle actin and prevented albuminuria and glomerulosclerosis with improved serum albumin. In cultured podocytes, this pathway was found to be associated with translation of fibrogenic collagen 4α3 and plasminogen activator inhibitor-1, without influencing their transcription. Notably, rapamycin suppressed upstream p-TGF-ßRI, p-Smad3 and p-ERK1/2, and trametinib down-regulated upstream p-Smad3 in ex vivo and in vivo studies, indicating that harmful paracrine signaling among glomerular cells amplified the TGF-ß-ERK1/2-mTORC1 axis by forming a positive feedback loop. Thus, an accentuated TGF-ß-ERK1/2-mTORC1 pathway is suggested as a central upstream mediator to develop proteinuria and glomerulosclerosis. Hence, preventing activation of this vicious loop by trametinib may offer a new therapeutic strategy for glomerular disease treatment.


Subject(s)
Glomerulosclerosis, Focal Segmental/drug therapy , MAP Kinase Signaling System/drug effects , Proteinuria/drug therapy , Pyridones/pharmacology , Pyrimidinones/pharmacology , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Disease Models, Animal , Doxorubicin/toxicity , Drug Evaluation, Preclinical , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Phosphorylation/drug effects , Proteinuria/chemically induced , Proteinuria/pathology , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Rats
12.
Exp Lung Res ; 45(7): 188-199, 2019 09.
Article in English | MEDLINE | ID: mdl-31298961

ABSTRACT

Purpose/Aim of the Study: Wnt/ß-catenin signaling was reported to be activated in pulmonary fibrosis, and was focused on as a target for antifibrotic therapy. However, the mechanism how the inhibition of Wnt/ß-catenin signaling ameliorate pulmonary fibrosis has not been fully elucidated. The purpose of this study is to explore the target cells of Wnt/ß-catenin inhibition in pulmonary fibrosis and to examine the antifibrotic effect of the novel inhibitor PRI-724 specifically disrupting the interaction of ß-catenin and CBP. Materials and Methods: The effect of C-82, an active metabolite of PRI-724, on the expression of TGF-ß1 and α-smooth muscle actin (SMA) was examined on fibroblasts and macrophages. We also examined the effects of PRI-724 in mouse model of bleomycin-induced pulmonary fibrosis. Results: The activation and increased accumulation of ß-catenin in the canonical pathway were detected in lung fibroblasts as well as macrophages stimulated by Wnt3a using Western blotting. Treatment with C-82 reduced CBP protein and increased p300 protein binding to ß-catenin in the nucleus of lung fibroblasts. In addition, C-82 inhibited the expression of SMA in lung fibroblasts treated with TGF-ß, indicating the inhibition of myofibroblast differentiation. In the fibrotic lungs induced by bleomycin, ß-catenin was stained strongly in macrophages, but the staining of ß-catenin in alveolar epithelial cells and fibroblasts was weak. The administration of PRI-724 ameliorated pulmonary fibrosis induced by bleomycin in mice when administered with a late, but not an early, treatment schedule. Analysis of bronchoalveolar fluid (BALF) showed a decreased number of alveolar macrophages. In addition, the level of TGF-ß1 in BALF was decreased in mice treated with PRI-724. C-82 also inhibited the production of TGF-ß1 by alveolar macrophages. Conclusions: These results suggest that the ß-catenin/CBP inhibitor PRI-724 is a potent antifibrotic agent that acts by modulating the activity of macrophages in the lungs.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Pulmonary Fibrosis/drug therapy , Pyrimidinones/therapeutic use , beta Catenin/antagonists & inhibitors , Animals , Bleomycin , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pyrimidinones/pharmacology , Transforming Growth Factor beta1/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
13.
J Comp Physiol B ; 189(3-4): 413-424, 2019 08.
Article in English | MEDLINE | ID: mdl-31123821

ABSTRACT

Saturated fatty acids are implicated in the development of metabolic diseases, including obesity and type 2 diabetes. There is evidence, however, that polyunsaturated fatty acids can counteract the pathogenic effects of saturated fatty acids. To gain insight into the early molecular mechanisms by which fatty acids influence hypothalamic inflammation and insulin signalling, we performed time-course experiments in a hypothalamic cell line, using different durations of treatment with the saturated fatty acid palmitate, and the omega-3 polyunsaturated fatty acid, docosahexaenoic acid (DHA). Western blot analysis revealed that palmitate elevated the protein levels of phospho(p)AKT in a time-dependent manner. This effect is involved in the pathogenicity of palmitate, as temporary inhibition of the PI3K/AKT pathway by selective PI3K inhibitors prevented the palmitate-induced attenuation of insulin signalling. Similar to palmitate, DHA also increased levels of pAKT, but to a weaker extent. Co-administration of DHA with palmitate decreased pAKT close to the basal level after 8 h, and prevented the palmitate-induced reduction of insulin signalling after 12 h. The monounsaturated fatty acid oleate had a similar effect on the palmitate-induced attenuation of insulin signalling, the polyunsaturated fatty acid linoleate had no effect. Measurement of the inflammatory markers pJNK and pNFκB-p65 revealed tonic elevation of both markers in the presence of palmitate alone. DHA alone transiently induced elevation of pJNK, returning to basal levels by 12 h treatment. Co-administration of DHA with palmitate prevented palmitate-induced inflammation after 12 h, but not at earlier timepoints.


Subject(s)
Gene Expression Regulation/drug effects , Hypothalamus/cytology , Neurons/drug effects , Palmitic Acid/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Docosahexaenoic Acids/pharmacology , Hydrazones/pharmacology , Insulin/metabolism , Mice , Morpholines/pharmacology , Oleic Acid/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pyrimidinones/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology
14.
Appl Environ Microbiol ; 85(9)2019 05 01.
Article in English | MEDLINE | ID: mdl-30824447

ABSTRACT

Fungal infections not only cause extensive agricultural damage but also result in serious diseases in the immunodeficient populations of human beings. Moreover, the increasing emergence of drug resistance has led to a decrease in the efficacy of current antifungals. Thus, screening of new antifungal agents is imperative in the fight against antifungal drug resistance. In this study, we show that an endophytic bacterium, Burkholderia gladioli HDXY-02, isolated from the medicinal plant Lycoris aurea, showed broad-spectrum antifungal activity against plant and human fungal pathogens. An antifungal ability assay indicated that the bioactive component was produced from strain HDXY-02 having an extracellular secreted component with a molecular weight lower than 1,000 Da. In addition, we found that this new antifungal could be produced effectively by liquid fermentation of HDXY-02. Furthermore, the purified component contributing to the antifungal activity was identified to be toxoflavin, a yellow compound possessing a pyrimido[5,4-e][1,2,4]triazine ring. In vitro bioactivity studies demonstrated that purified toxoflavin from B. gladioli HDXY-02 cultures had a significant antifungal activity against the human fungal pathogen Aspergillus fumigatus, resulting in abolished germination of conidia. More importantly, the growth inhibition by toxoflavin was observed in both wild-type and drug-resistant mutants (cyp51A and non-cyp51A) of A. fumigatus Finally, an optimized protocol for the large-scale production of toxoflavin (1,533 mg/liter) has been developed. Taken together, our findings provide a promising biosynthetic resource for producing a new antifungal reagent, toxoflavin, from isolates of the endophytic bacterium B. gladioliIMPORTANCE Human fungal infections are a growing problem associated with increased morbidity and mortality. Moreover, a growing number of antifungal-resistant fungal isolates have been reported over the past decade. Thus, the need for novel antifungal agents is imperative. In this study, we show that an endophytic bacterium, Burkholderia gladioli, isolated from the medicinal plant Lycoris aurea, is able to abundantly secrete a compound, toxoflavin, which has a strong fungicidal activity not only against plant fungal pathogens but also against human fungal pathogens Aspergillus fumigatus and Candida albicans, Cryptococcus neoformans, and the model filamentous fungus Aspergillus nidulans More importantly, toxoflavin also displays an efficacious inhibitory effect against azole antifungal-resistant mutants of A. fumigatus Consequently, our findings provide a promising approach to abundantly produce toxoflavin, which has novel broad-spectrum antifungal activity, especially against those currently problematic drug-resistant isolates.


Subject(s)
Antifungal Agents/pharmacology , Burkholderia gladioli/chemistry , Fungicides, Industrial/pharmacology , Pyrimidinones/pharmacology , Triazines/pharmacology , Lycoris/microbiology
15.
Anal Bioanal Chem ; 410(28): 7441-7448, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30218128

ABSTRACT

Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI TOF MS) is a well-established method with a unique set of qualities including sensitivity, minute sample consumption, and label-free detection, all of which are highly desired in enzyme assays. On the other hand, the application of MALDI TOF MS is usually limited by high concentrations of MS-incompatible compounds in the reaction mixture such as salts or organic solvents. Here, we introduce kinetic and inhibition studies of ß-secretase (BACE1), a key enzyme of the progression of Alzheimer's disease. Compatibility of the enzyme assay with MALDI TOF MS was achieved, providing both a complex protocol including a desalting step designed for rigorous kinetic studies and a simple mix-and-measure protocol designed for high-throughput inhibitor screening. In comparison with fluorescent or colorimetric assays, MALDI TOF MS represents a sensitive, fast, and label-free technique with minimal sample preparation. In contrast to other MS-based methodological approaches typically used in drug discovery processes, such as a direct injection MS or MS-coupled liquid chromatography or capillary electrophoresis, MALDI TOF MS enables direct analysis and is a highly suitable approach for high-throughput screening. The method's applicability is strongly supported by the high correlation of the acquired kinetic and inhibition parameters with data from the literature as well as from our previous research. Graphical abstract ᅟ.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amino Acids/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Drug Evaluation, Preclinical/methods , HEK293 Cells , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Kinetics , Picolinic Acids/pharmacology , Pyrimidinones/pharmacology
16.
J Med Chem ; 61(17): 7671-7686, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30106292

ABSTRACT

We previously reported Chalcone-4 (1) that binds the chemokine CXCL12, not its cognate receptors CXCR4 or CXCR7, and neutralizes its biological activity. However, this neutraligand suffers from limitations such as poor chemical stability, solubility, and oral activity. Herein, we report on the discovery of pyrimidinone 57 (LIT-927), a novel neutraligand of CXCL12 which displays a higher solubility than 1 and is no longer a Michael acceptor. While both 1 and 57 reduce eosinophil recruitment in a murine model of allergic airway hypereosinophilia, 57 is the only one to display inhibitory activity following oral administration. Thereby, we here describe 57 as the first orally active CXCL12 neutraligand with anti-inflammatory properties. Combined with a high binding selectivity for CXCL12 over other chemokines, 57 represents a powerful pharmacological tool to investigate CXCL12 physiology in vivo and to explore the activity of chemokine neutralization in inflammatory and related diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chemokine CXCL12/metabolism , Hypereosinophilic Syndrome/drug therapy , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Chemokine CXCL12/chemistry , Disease Models, Animal , Drug Evaluation, Preclinical , Fluorescence Resonance Energy Transfer , Humans , Hypersensitivity/drug therapy , Hypersensitivity/etiology , Male , Mice, Inbred BALB C , Models, Molecular , Pyrimidinones/administration & dosage , Pyrimidinones/metabolism , Pyrimidinones/pharmacokinetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 28(10): 1887-1891, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29655979

ABSTRACT

During the course of our research efforts to develop potent and selective AKT inhibitors, we discovered enatiomerically pure substituted dihydropyridopyrimidinones (DHP) as potent inhibitors of protein kinase B/AKT with excellent selectivity against ROCK2. A key challenge in this program was the poor physicochemical properties of the initial lead compound 5. Integration of structure-based drug design and physical properties-based design resulted in replacement of a highly hydrophobic poly fluorinated aryl ring by a simple trifluoromethyl that led to identification of compound 6 with much improved physicochemical properties. Subsequent SAR studies led to the synthesis of new pyran analog 7 with improved cell potency. Further optimization of pharmacokintetics properties by increasing permeability with appropriate fluorinated alkyl led to compound 8 as a potent, selective AKT inhibitors that blocks the phosphorylation of GSK3ß in vivo and had robust, dose and concentration dependent efficacy in the U87MG tumor xenograft model.


Subject(s)
Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidinones/chemistry , Animals , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Drug Evaluation, Preclinical , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice , Molecular Dynamics Simulation , Neoplasms/drug therapy , Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous
18.
Cell Death Dis ; 9(3): 269, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449645

ABSTRACT

The epidermal growth factor receptor (EGFR) pathway and Hippo signaling play an important role in the carcinogenesis of hepatocellular carcinoma (HCC). However, the crosstalk between these two pathways and its implications in targeted therapy remains unclear. We found that the activated EGFR signaling could bypass RhoA to promote the expression of YAP(Yes-associated protein), the core effector of the Hippo signaling, and its downstream target Cyr61. Further studies indicated that EGFR signaling mainly acted through the PI3K-PDK1 (Phosphoinositide 3-kinase-Phosphoinositide-dependent kinase-1) pathway to activate YAP, but not the AKT and MAPK pathways. While YAP knockdown hardly affected the EGFR signaling. In addition, EGF could promote the proliferation of HCC cells in a YAP-independent manner. Combined targeting of YAP and EGFR signaling by simvastatin and the EGFR signaling inhibitors, including the EGFR tyrosine kinase inhibitor (TKI) gefitinib, the RAF inhibitor sorafenib and the MEK inhibitor trametinib, presented strong synergistic cytotoxicities in HCC cells. Therefore, the EGFR-PI3K-PDK1 pathway could activate the YAP signaling, and the activated EGFR signaling could promote the HCC cell growth in a YAP-independent manner. Combined use of FDA-approved inhibitors to simultaneously target YAP and EGFR signaling presented several promising therapeutic approaches for HCC treatment.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/pharmacology , Simvastatin/pharmacology , Transcription Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gefitinib/pharmacology , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Molecular Targeted Therapy , Pyridones/pharmacology , Pyrimidinones/pharmacology , Signal Transduction/drug effects , Sorafenib/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
19.
Sci Rep ; 8(1): 3627, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483620

ABSTRACT

Cells grown in three-dimensional (3D) cultures are more likely to have native cell-cell and cell-matrix interactions than in 2D cultures that impose mechanical constraints to cells. However, most 3D cultures utilise gel matrix which, while serving as a scaffold, limits application due to its solid and opaque nature and inconsistency in cell exposure to exogenous signals. In 3D culture without gel matrix, cells tend to adhere to each other and form clumps with necrotic zone at the centre, making them unsuitable for analyses. Here we report that addition of low-molecular-weight agar named LA717 to culture media allows cells to grow as dispersed clonal spheroids in 3D. LA717 maintains cells dispersed and settled to the bottom of the medium while keeping the medium clear with little additional viscosity, making it suitable for microscopic observation. Importantly, cancer spheroids formed in LA717-containing medium show higher sensitivity to anti-cancer drugs such as Trametinib and MK-2206 that are not as effective in 2D. Because of the small and consistent size of spheroids, cell viability and drug toxicity are readily detectable in automated imaging analysis. These results demonstrate that LA717 offers a novel 3D culture system with great in vivo reflection and practicality.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , A549 Cells , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Drug Evaluation, Preclinical , HCT116 Cells , HeLa Cells , Hep G2 Cells , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Pyridones/pharmacology , Pyrimidinones/pharmacology , Real-Time Polymerase Chain Reaction , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects
20.
Sci Rep ; 8(1): 1155, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348452

ABSTRACT

The threat of antibiotic resistant bacteria has called for alternative antimicrobial strategies that would mitigate the increase of classical resistance mechanism. Many bacteria employ quorum sensing (QS) to govern the production of virulence factors and formation of drug-resistant biofilms. Targeting the mechanism of QS has proven to be a functional alternative to conventional antibiotic control of infections. However, the presence of multiple QS systems in individual bacterial species poses a challenge to this approach. Quorum sensing inhibitors (QSI) and quorum quenching enzymes (QQE) have been both investigated for their QS interfering capabilities. Here, we first simulated the combination effect of QQE and QSI in blocking bacterial QS. The effect was next validated by experiments using AiiA as QQE and G1 as QSI on Pseudomonas aeruginosa LasR/I and RhlR/I QS circuits. Combination of QQE and QSI almost completely blocked the P. aeruginosa las and rhl QS systems. Our findings provide a potential chemical biology application strategy for bacterial QS disruption.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/pharmacology , Biofilms/drug effects , Gene Expression Regulation, Bacterial/drug effects , Metalloendopeptidases/pharmacology , Pseudomonas aeruginosa/drug effects , Quorum Sensing/drug effects , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , Drug Combinations , Drug Synergism , Ligases/antagonists & inhibitors , Ligases/genetics , Ligases/metabolism , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Microbial Sensitivity Tests , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Pyrimidinones/pharmacology , Quorum Sensing/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL