Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 349
Filter
1.
Biochem Biophys Res Commun ; 595: 1-6, 2022 03 05.
Article in English | MEDLINE | ID: mdl-35091108

ABSTRACT

Farnesoid X receptor (FXR) is a bile acid-related nuclear receptor and is considered a promising target to treat several liver disorders. Cilofexor is a selective FXR agonist and has already entered phase III trials in primary sclerosing cholangitis (PSC) patients. Pruritis caused by cilofexor treatment is dose dependent. The binding characteristics of cilofexor with FXR and its pruritogenic mechanism remain unclear. In our research, the affinity of cilofexor bound to FXR was detected using an isothermal titration calorimetry (ITC) assay. The binding mechanism between cilofexor and FXR-LBD is explained by the cocrystal structure of the FXR/cilofexor complex. Structural models indicate the possibility that cilofexor activates Mas-related G protein-coupled receptor X4 (MRGPRX4) or G protein-coupled bile acid receptor 1 (GPBAR1), leading to pruritus. In summary, our analyses provide a molecular mechanism of cilofexor binding to FXR and provide a possible explanation for the dose-dependent pruritis of cilofexor.


Subject(s)
Azetidines/chemistry , Isonicotinic Acids/chemistry , Molecular Docking Simulation , Protein Domains , Receptors, Cytoplasmic and Nuclear/chemistry , Azetidines/metabolism , Azetidines/pharmacology , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Binding Sites , Binding, Competitive , Calorimetry/methods , Crystallization , Humans , Hydrogen Bonding , Isonicotinic Acids/metabolism , Isonicotinic Acids/pharmacology , Isoxazoles/chemistry , Isoxazoles/metabolism , Isoxazoles/pharmacology , Ligands , Molecular Structure , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism
2.
Chem Biol Drug Des ; 98(5): 787-827, 2021 11.
Article in English | MEDLINE | ID: mdl-34397161

ABSTRACT

Tuberculosis (TB) is an infection caused by Mycobacterium tuberculosis (Mtb) and one of the deadliest infectious diseases in the world. Mtb has the ability to become dormant within the host and to develop resistance. Hence, new antitubercular agents are required to overcome problems in the treatment of multi-drug-resistant Tb (MDR-Tb) and extensively drug-resistant Tb (XDR-Tb) along with shortening the treatment time. Several efforts are being made to develop very effective new drugs for Tb, within the pharmaceutical industry, the academia and through public-private partnerships. This review will address the antitubercular activities, biological target, mode of action, synthetic approaches and thoughtful concept for the development of several new drugs currently in the clinical trial pipeline (up to October 2019) for tuberculosis. The aim of this review may be very useful in scheming new chemical entities (NCEs) for Mtb.


Subject(s)
Antitubercular Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Adamantane/analogs & derivatives , Adamantane/chemical synthesis , Adamantane/pharmacology , Animals , Antitubercular Agents/pharmacology , DNA Gyrase/metabolism , Drug Development , Enzyme Inhibitors/pharmacology , Humans , Isonicotinic Acids/chemical synthesis , Isonicotinic Acids/pharmacology , Oxazolidinones/chemical synthesis , Oxazolidinones/pharmacology , Piperidines/chemical synthesis , Piperidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Structure-Activity Relationship , Tuberculosis, Multidrug-Resistant , Uridine/analogs & derivatives , Uridine/chemical synthesis , Uridine/pharmacology
3.
Cancer Chemother Pharmacol ; 88(5): 795-804, 2021 11.
Article in English | MEDLINE | ID: mdl-34309733

ABSTRACT

PURPOSE: CHMFL-KIT-110, a selective c-KIT kinase inhibitor for gastrointestinal stromal tumors (GISTs), possesses a poorly water-soluble, limiting the further development of the drug. This study was to investigate the antitumor efficacy of CHMFL-KIT-110 and CHMFL-KIT-110 solid dispersion (laboratory code: HYGT-110 SD) in GIST tumor xenograft models and to explore the PK/PD relationship of HYGT-110 SD. METHODS: Plasma concentrations of HYGT-110 and HYGT-110 SD were determined by LC-MS/MS in KM mice. Antitumor activity was evaluated by measuring tumor volume and weight in c-KIT-dependent GIST xenograft models. PK/PD relationship was assessed by LC-MS/MS and Western Blot in the GIST-T1 xenografted mice. RESULTS: HYGT-110 exhibited a low oral bioavailability (10.91%) in KM mice. Compared with HYGT-110 treatment, the Cmax and AUC0-t of HYGT-110 SD in mice plasma were substantially increased by 18.81 and 6.76-fold, respectively. HYGT-110 SD (10, 30, and 100 mg/kg/day) also could dose-dependently decrease the tumor volume and weight in the GIST-882 cell-inoculated xenograft mouse models and show 86.35% tumor growth inhibition (TGI) at 28 days at a 25 mg/kg bid dosage in the GIST-T1 cell-inoculated xenograft mouse model. The free concentration of HYGT-110 in plasma was closely correlated with the inhibition of c-KIT phosphorylation levels in tumor tissues. CONCLUSIONS: In comparison with the HPMC formulation, both improved PK and PD characteristics of the solid dispersion formulation of CHMFL-KIT-110 were observed in in vivo animal experiments.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Stromal Tumors/drug therapy , Isonicotinic Acids/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Benzamides/administration & dosage , Benzamides/pharmacokinetics , Cell Line, Tumor , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/pathology , Humans , Injections, Intravenous , Isonicotinic Acids/administration & dosage , Isonicotinic Acids/pharmacokinetics , Male , Mice , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/blood , Xenograft Model Antitumor Assays
4.
Plant J ; 107(1): 149-165, 2021 07.
Article in English | MEDLINE | ID: mdl-33866633

ABSTRACT

Recent studies have shown that global metabolic reprogramming is a common event in plant innate immunity; however, the relevant molecular mechanisms remain largely unknown. Here, we identified a pathogen-induced glycosyltransferase, UGT73C7, that plays a critical role in Arabidopsis disease resistance through mediating redirection of the phenylpropanoid pathway. Loss of UGT73C7 function resulted in significantly decreased resistance to Pseudomonas syringae pv. tomato DC3000, whereas constitutive overexpression of UGT73C7 led to an enhanced defense response. UGT73C7-activated immunity was demonstrated to be dependent on the upregulated expression of SNC1, a Toll/interleukin 1 receptor-type NLR gene. Furthermore, in vitro and in vivo assays indicated that UGT73C7 could glycosylate p-coumaric acid and ferulic acid, the upstream metabolites in the phenylpropanoid pathway. Mutations that lead to the loss of UGT73C7 enzyme activities resulted in the failure to induce SNC1 expression. Moreover, glycosylation activity of UGT73C7 resulted in the redirection of phenylpropanoid metabolic flux to biosynthesis of hydroxycinnamic acids and coumarins. The disruption of the phenylpropanoid pathway suppressed UGT73C7-promoted SNC1 expression and the immune response. This study not only identified UGT73C7 as an important regulator that adjusts phenylpropanoid metabolism upon pathogen challenge, but also provided a link between phenylpropanoid metabolism and an NLR gene.


Subject(s)
Arabidopsis Proteins/immunology , Arabidopsis/physiology , Glycosyltransferases/metabolism , Plant Immunity/physiology , Arabidopsis/drug effects , Arabidopsis Proteins/genetics , Coumaric Acids/metabolism , Disease Resistance/immunology , Gene Expression Regulation, Plant , Glycosylation , Glycosyltransferases/genetics , Glycosyltransferases/immunology , Host-Pathogen Interactions/physiology , Isonicotinic Acids/pharmacology , Plant Diseases/immunology , Plants, Genetically Modified , Pseudomonas syringae/pathogenicity
5.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33830087

ABSTRACT

The Janus kinase/signal transducers and activators of transcription (JAK/STAT) are key intracellular mediators in the signal transduction of many cytokines and growth factors. Common γ chain cytokines and interferon-γ that use the JAK/STAT pathway to induce biological responses have been implicated in the pathogenesis of alopecia areata (AA), a T cell-mediated autoimmune disease of the hair follicle. We previously showed that therapeutic targeting of JAK/STAT pathways using the first-generation JAK1/2 inhibitor, ruxolitinib, and the pan-JAK inhibitor, tofacitinib, was highly effective in the treatment of human AA, as well as prevention and reversal of AA in the C3H/HeJ mouse model. To better define the role of individual JAKs in the pathogenesis of AA, in this study, we tested and compared the efficacy of several next-generation JAK-selective inhibitors in the C3H/HeJ mouse model of AA, using both systemic and topical delivery. We found that JAK1-selective inhibitors as well as JAK3-selective inhibitors robustly induced hair regrowth and decreased AA-associated inflammation, whereas several JAK2-selective inhibitors failed to restore hair growth in treated C3H/HeJ mice with AA. Unlike JAK1, which is broadly expressed in many tissues, JAK3 expression is largely restricted to hematopoietic cells. Our study demonstrates inhibiting JAK3 signaling is sufficient to prevent and reverse disease in the preclinical model of AA.


Subject(s)
Alopecia Areata/drug therapy , Janus Kinase 3/metabolism , Protein Kinase Inhibitors/pharmacology , Administration, Topical , Alopecia Areata/metabolism , Alopecia Areata/prevention & control , Animals , Azetidines/administration & dosage , Azetidines/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Isonicotinic Acids/administration & dosage , Isonicotinic Acids/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Janus Kinase 3/antagonists & inhibitors , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C3H , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nitriles/pharmacology , Piperidines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacology , Triazoles/pharmacology
6.
Biochem Biophys Res Commun ; 554: 71-75, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33780862

ABSTRACT

Histone lysine demethylase 4D (KDM4D), also known as JMJD2D, plays an important role in cell proliferation and survival and has been associated with several tumor types. KDM4D has emerged as a potential target for the treatment of human cancer. Here, we reported crystal complex structures for two KDM4D inhibitors, OWS [2-(1H-pyrazol-3-yl)isonicotinic acid] and 10r (5-hydroxy-2-methylpyrazolo[1,5-a]pyrido[3,2-e]pyrimidine-3-carbonitrile), which were both determined to 2.0 Å. OWS is a newly discovered KDM4D inhibitor (IC50 = 4.28 µM) and the critical pharmacophores of this compound are confirmed by the complex structure. Compound 10r is a KDM4D inhibitor reported by us previously. To clarify the binding mode in more detail, the crystal structure was determined and the comparison analysis revealed unique interactions that had never been observed before. Overall, our data provide new structural insights for rational design and offer an opportunity for optimization of KDM4D inhibitors.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Isonicotinic Acids/chemistry , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/chemistry , Pyrazoles/chemistry , Antineoplastic Agents/pharmacology , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/pharmacology , Humans , Isonicotinic Acids/pharmacology , Jumonji Domain-Containing Histone Demethylases/isolation & purification , Jumonji Domain-Containing Histone Demethylases/metabolism , Models, Molecular , Protein Structural Elements , Pyrazoles/pharmacology , Structure-Activity Relationship
7.
Molecules ; 26(5)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33652837

ABSTRACT

In search of anti-inflammatory compounds, novel scaffolds containing isonicotinoyl motif were synthesized via an efficient strategy. The compounds were screened for their in vitro anti-inflammatory activity. Remarkably high activities were observed for isonicotinates 5-6 and 8a-8b. The compound 5 exhibits an exceptional IC50 value (1.42 ± 0.1 µg/mL) with 95.9% inhibition at 25 µg/mL, which is eight folds better than the standard drug ibuprofen (11.2 ± 1.9 µg/mL). To gain an insight into the mode of action of anti-inflammatory compounds, molecular docking studies were also performed. Decisively, further development and fine tuning of these isonicotinates based scaffolds for the treatment of various aberrations is still a wide-open field of research.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Inflammation/drug therapy , Isonicotinic Acids/chemical synthesis , Reactive Oxygen Species/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Ibuprofen/chemistry , Isonicotinic Acids/chemistry , Isonicotinic Acids/pharmacology , Molecular Docking Simulation , Reactive Oxygen Species/chemistry , Structure-Activity Relationship
8.
Biochem Pharmacol ; 186: 114430, 2021 04.
Article in English | MEDLINE | ID: mdl-33556338

ABSTRACT

Colorectal cancer (CRC) is one of the most malignant cancers in the world. A major cause of death in CRC patients is the limited therapeutic options in its advanced stages. The Farnesoid X receptor (FXR) is a member of the nuclear superfamily, which is effective in slowing the progression of colorectal cancer in addition to its extraordinary role in regulating metabolic disorders. Due to the systemic side-effects caused by non-selective agonists, the intestine-restricted FXR agonists can induce a whole-body benefit without activating the hepatic FXR, suggesting intestinal FXR activation as a potentially safer therapy in the treatment of CRC. This review highlights the effects of FXR on the disturbed bile acid circulation and the carcinogenesis of CRC and with a specific emphasis on listing the functions of several intestinal-restricted FXR agonists.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Intestinal Mucosa/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Antineoplastic Agents/pharmacology , Azetidines/pharmacology , Azetidines/therapeutic use , Benzene Derivatives/pharmacology , Benzene Derivatives/therapeutic use , Bile Acids and Salts/metabolism , Colorectal Neoplasms/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Isonicotinic Acids/pharmacology , Isonicotinic Acids/therapeutic use , Receptors, Cytoplasmic and Nuclear/metabolism
9.
Respir Physiol Neurobiol ; 284: 103563, 2021 02.
Article in English | MEDLINE | ID: mdl-33053424

ABSTRACT

The pontine Kölliker-Fuse nucleus (KFn) is a core nucleus of respiratory network that mediates the inspiratory-expiratory phase transition and gates eupneic motor discharges in the vagal and hypoglossal nerves. In the present study, we investigated whether the same KFn circuit may also gate motor activities that control the resistance of the nasal airway, which is of particular importance in rodents. To do so, we simultaneously recorded phrenic, facial, vagal and hypoglossal cranial nerve activity in an in situ perfused brainstem preparation before and after bilateral injection of the GABA-receptor agonist isoguvacine (50-70 nl, 10 mM) into the KFn (n = 11). Our results show that bilateral inhibition of the KFn triggers apneusis (prolonged inspiration) and abolished pre-inspiratory discharge of facial, vagal and hypoglossal nerves as well as post-inspiratory discharge in the vagus. We conclude that the KFn plays a critical role for the eupneic regulation of naso-pharyngeal airway patency and the potential functions of the KFn in regulating airway patency and orofacial behavior is discussed.


Subject(s)
Facial Nerve/physiology , Hypoglossal Nerve/physiology , Kolliker-Fuse Nucleus/physiology , Motor Activity/physiology , Nerve Net/physiology , Phrenic Nerve/physiology , Respiration , Vagus Nerve/physiology , Animals , Facial Nerve/drug effects , Female , GABA Agonists/pharmacology , Hypoglossal Nerve/drug effects , Isonicotinic Acids/pharmacology , Kolliker-Fuse Nucleus/drug effects , Male , Motor Activity/drug effects , Nerve Net/drug effects , Phrenic Nerve/drug effects , Rats , Rats, Sprague-Dawley , Respiration/drug effects , Respiratory Center , Respiratory Rate/drug effects , Respiratory Rate/physiology , Vagus Nerve/drug effects
10.
Osteoarthritis Cartilage ; 29(2): 248-256, 2021 02.
Article in English | MEDLINE | ID: mdl-33246158

ABSTRACT

OBJECTIVE: This study aimed to examine the temporal activation of NF-κB and its relationship to the development of pain-related sensitivity and behavioral changes in a non-invasive murine knee loading model of PTOA. METHOD: Following knee injury NF-κB activity was assessed longitudinally via in vivo imaging in FVB. Cg-Tg (HIV-EGFP,luc)8Tsb/J mice. Measures of pain-related sensitivity and behavior were also assessed longitudinally for 16 weeks. Additionally, we antagonized NF-κB signaling via intra-articular delivery of an IκB kinase two antagonist to understand how local NF-κB inhibition might alter disease progression. RESULTS: Following joint injury NF-κB signaling within the knee joint was transiently increased and peaked on day 3 with an estimated 1.35 p/s/cm2/sr (95% CI 0.913.1.792 p/s/cm2/sr) fold increase in signaling when compared to control joints. Furthermore, injury resulted in the long-term development of hindpaw allodynia. Hyperalgesia withdrawal thresholds were reduced at injured knee joints, with the largest reduction occurring 2 days following injury (estimate of between group difference 129.1 g with 95% CI 60.9,197.4 g), static weight bearing on injured limbs was also reduced. Local delivery of an NF-κB inhibitor following joint injury reduced chondrocyte death and influenced the development of pain-related sensitivity but did not reduce long-term cartilage degeneration. CONCLUSION: These findings underscore the development of behavioral changes in this non-invasive loading model of PTOA and their relationships to NF-κB activation and pathology. They also highlight the potential chondroprotective effects of NF-κB inhibition shortly following joint injury despite limitations in preventing the long-term development of joint degeneration in this model of PTOA.


Subject(s)
Cartilage, Articular/metabolism , NF-kappa B/metabolism , Osteoarthritis/metabolism , Stifle/metabolism , Weight-Bearing , Animals , Behavior, Animal , Biomechanical Phenomena , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Disease Models, Animal , Hyperalgesia , I-kappa B Kinase/antagonists & inhibitors , Indazoles/pharmacology , Isonicotinic Acids/pharmacology , Knee Injuries/complications , Luminescent Measurements , Mice , Mice, Transgenic , NF-kappa B/drug effects , Osteoarthritis/etiology , Osteoarthritis, Knee/etiology , Osteoarthritis, Knee/metabolism , Stifle/drug effects , Stifle/injuries
11.
J Med Chem ; 63(23): 14724-14739, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33205981

ABSTRACT

Aromatic aldehydes elicit their antisickling effects primarily by increasing the affinity of hemoglobin (Hb) for oxygen (O2). However, challenges related to weak potency and poor pharmacokinetic properties have hampered their development to treat sickle cell disease (SCD). Herein, we report our efforts to enhance the pharmacological profile of our previously reported compounds. These compounds showed enhanced effects on Hb modification, Hb-O2 affinity, and sickling inhibition, with sustained pharmacological effects in vitro. Importantly, some compounds exhibited unusually high antisickling activity despite moderate effects on the Hb-O2 affinity, which we attribute to an O2-independent antisickling activity, in addition to the O2-dependent activity. Structural studies are consistent with our hypothesis, which revealed the compounds interacting strongly with the polymer-stabilizing αF-helix could potentially weaken the polymer. In vivo studies with wild-type mice demonstrated significant pharmacologic effects. Our structure-based efforts have identified promising leads to be developed as novel therapeutic agents for SCD.


Subject(s)
Antisickling Agents/pharmacology , Benzaldehydes/pharmacology , Isonicotinic Acids/pharmacology , Nicotinic Acids/pharmacology , Picolinic Acids/pharmacology , Animals , Antisickling Agents/chemical synthesis , Antisickling Agents/metabolism , Benzaldehydes/chemical synthesis , Benzaldehydes/metabolism , Crystallography, X-Ray , Hemoglobins/metabolism , Isonicotinic Acids/chemical synthesis , Isonicotinic Acids/metabolism , Mice, Inbred C57BL , Molecular Structure , Nicotinic Acids/chemical synthesis , Nicotinic Acids/metabolism , Oxygen/metabolism , Picolinic Acids/chemical synthesis , Picolinic Acids/metabolism , Protein Binding , Structure-Activity Relationship
12.
Clin Cancer Res ; 26(23): 6299-6309, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32998963

ABSTRACT

PURPOSE: T cells engineered to express a chimeric antigen receptor (CAR) are a promising cancer immunotherapy. Such targeted therapies have shown long-term relapse-free survival in patients with B-cell leukemia and lymphoma. However, cytokine release syndrome (CRS) represents a serious, potentially life-threatening side effect often associated with CAR T-cell therapy. CRS manifests as a rapid (hyper)immune reaction driven by excessive inflammatory cytokine release, including IFNγ and IL6. EXPERIMENTAL DESIGN: Many cytokines implicated in CRS are known to signal through the JAK-STAT pathway. Here we study the effect of blocking JAK pathway signaling on CAR T-cell proliferation, antitumor activity, and cytokine levels in in vitro and in vivo models. RESULTS: We report that itacitinib, a potent, selective JAK1 inhibitor, was able to significantly and dose-dependently reduce levels of multiple cytokines implicated in CRS in several in vitro and in vivo models. Importantly, we also report that at clinically relevant doses that mimic human JAK1 pharmacologic inhibition, itacitinib did not significantly inhibit proliferation or antitumor killing capacity of three different human CAR T-cell constructs (GD2, EGFR, and CD19). Finally, in an in vivo model, antitumor activity of CD19-CAR T cells adoptively transferred into CD19+ tumor-bearing immunodeficient animals was unabated by oral itacitinib treatment. CONCLUSIONS: Together, these data suggest that itacitinib has potential as a prophylactic agent for the prevention of CAR T cell-induced CRS, and a phase II clinical trial of itacitinib for prevention of CRS induced by CAR T-cell therapy has been initiated (NCT04071366).


Subject(s)
Azetidines/pharmacology , Cytokine Release Syndrome/drug therapy , Cytokines/antagonists & inhibitors , Immunotherapy, Adoptive/adverse effects , Isonicotinic Acids/pharmacology , Janus Kinase 1/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Apoptosis , Cell Proliferation , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Bioorg Med Chem Lett ; 30(23): 127558, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32961321

ABSTRACT

A sustainable synthesis of new 3,5-[(sub)phenyl]-1H-pyrazole bearing N1-isonicotinoyl derivatives from substituted chalcones and isoniazid by using sulfamic acid and their pharmacological activity evaluation is reported. An anti-oxidant study is performed by using DPPH assay. In vitro anti-mycobacterial activity of compounds bearing R/R' = 4-CH3/4-F and 3-OCH3/4-Cl showed complete inhibition (99%) at the MIC of 31 and 34 µM respectively. Antibacterial screening of compounds bearing R/R' = 4-CH3/4-F; 4-OCH3/4-Br; and 4-OCH3/4-Cl has shown noticeable inhibition (27 mm) against Staphylococcus aureus. The anti-cancer bioassay demonstrated that the five compounds were active on human breast cancer cell line MCF-7; however on HeLa cervical cancer cells only two compounds are active in comparison to standard drug Doxorubicin. Higher inhibitory effects observed in this study appear to be dependent on the chloro, bromo, fluoro and methoxy functionality present on the aromatic nucleus. The structures of all the compounds are established using NMR (1H and 13C), FT-IR, Mass and elemental analysis.


Subject(s)
Antineoplastic Agents/pharmacology , Antitubercular Agents/pharmacology , Isonicotinic Acids/pharmacology , Pyrazoles/pharmacology , Sulfonic Acids/chemistry , Antineoplastic Agents/chemical synthesis , Antitubercular Agents/chemical synthesis , Catalysis , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Isonicotinic Acids/chemical synthesis , Microbial Sensitivity Tests , Molecular Structure , Pyrazoles/chemical synthesis , Structure-Activity Relationship
14.
Eur J Pharmacol ; 885: 173505, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32861662

ABSTRACT

Pharmacological modulation of the Janus kinase (JAK) family has achieved clinically meaningful therapeutic outcomes for the treatment of inflammatory and hematopoietic diseases. Several JAK1 selective compounds are being investigated clinically to determine their anti-inflammatory potential. We used recombinant enzymes and primary human lymphocytes to assess the JAK1 specificity of itacitinib (INCB039110) and study inhibition of signal transducers and activators of transcription (STAT) signaling. Rodent models of arthritis and inflammatory bowel disease were subsequently explored to elucidate the efficacy of orally administered itacitinib on inflammatory pathogenesis. Itacitinib is a potent and selective JAK1 inhibitor when profiled against the other JAK family members. Upon oral administration in rodents, itacitinib achieved dose-dependent pharmacokinetic exposures that highly correlated with STAT3 pharmacodynamic pathway inhibition. Itacitinib ameliorated symptoms and pathology of established experimentally-induced arthritis in a dose-dependent manner. Furthermore, itacitinib effectively delayed disease onset, reduced symptom severity, and accelerated recovery in three distinct mouse models of inflammatory bowel disease. Low dose itacitinib administered via cannula directly into the colon was highly efficacious in TNBS-induced colitis but with minimal systemic drug exposure, suggesting localized JAK1 inhibition is sufficient for disease amelioration. Itacitinib treatment in an acute graft-versus-host disease (GvHD) model rapidly reduced inflammatory markers within lymphocytes and target tissue, resulting in a marked improvement in disease symptoms. This is the first manuscript describing itacitinib as a potent and selective JAK1 inhibitor with anti-inflammatory activity across multiple preclinical disease models. These data support the scientific rationale for ongoing clinical trials studying itacitinib in select GvHD patient populations.


Subject(s)
Azetidines/pharmacology , Inflammation/drug therapy , Isonicotinic Acids/pharmacology , Janus Kinase 1/antagonists & inhibitors , Animals , Arthritis, Experimental/drug therapy , Azetidines/pharmacokinetics , Azetidines/therapeutic use , Chemokine CCL2/drug effects , Colitis/chemically induced , Colitis/drug therapy , Dose-Response Relationship, Drug , Graft vs Host Disease/drug therapy , Humans , Inflammatory Bowel Diseases/drug therapy , Isonicotinic Acids/pharmacokinetics , Isonicotinic Acids/therapeutic use , Lymphocytes/drug effects , Mice , Mice, Inbred BALB C , Primary Cell Culture , Rats , Rats, Inbred Lew , STAT Transcription Factors/drug effects , STAT3 Transcription Factor/drug effects , Signal Transduction/drug effects , T-Lymphocytes/drug effects
15.
Hepatology ; 72(1): 58-71, 2020 07.
Article in English | MEDLINE | ID: mdl-32115759

ABSTRACT

BACKGROUND AND AIMS: We evaluated the safety and efficacy of cilofexor (formerly GS-9674), a small-molecule nonsteroidal agonist of farnesoid X receptor, in patients with nonalcoholic steatohepatitis (NASH). APPROACH AND RESULTS: In this double-blind, placebo-controlled, phase 2 trial, 140 patients with noncirrhotic NASH, diagnosed by magnetic resonance imaging-proton density fat fraction (MRI-PDFF) ≥8% and liver stiffness ≥2.5 kPa by magnetic resonance elastography (MRE) or historical liver biopsy, were randomized to receive cilofexor 100 mg (n = 56), 30 mg (n = 56), or placebo (n = 28) orally once daily for 24 weeks. MRI-PDFF, liver stiffness by MRE and transient elastography, and serum markers of fibrosis were measured at baseline and week 24. At baseline, median MRI-PDFF was 16.3% and MRE-stiffness was 3.27 kPa. At week 24, patients receiving cilofexor 100 mg had a median relative decrease in MRI-PDFF of -22.7%, compared with an increase of 1.9% in those receiving placebo (P = 0.003); the 30-mg group had a relative decrease of -1.8% (P = 0.17 vs. placebo). Declines in MRI-PDFF of ≥30% were experienced by 39% of patients receiving cilofexor 100 mg (P = 0.011 vs. placebo), 14% of those receiving cilofexor 30 mg (P = 0.87 vs. placebo), and 13% of those receiving placebo. Serum gamma-glutamyltransferase, C4, and primary bile acids decreased significantly at week 24 in both cilofexor treatment groups, whereas significant changes in Enhanced Liver Fibrosis scores and liver stiffness were not observed. Cilofexor was generally well-tolerated. Moderate to severe pruritus was more common in patients receiving cilofexor 100 mg (14%) than in those receiving cilofexor 30 mg (4%) and placebo (4%). CONCLUSIONS: Cilofexor for 24 weeks was well-tolerated and provided significant reductions in hepatic steatosis, liver biochemistry, and serum bile acids in patients with NASH. ClinicalTrials.gov No. NCT02854605.


Subject(s)
Azetidines/pharmacology , Isonicotinic Acids/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Adolescent , Adult , Aged , Azetidines/therapeutic use , Double-Blind Method , Female , Humans , Isonicotinic Acids/therapeutic use , Male , Middle Aged , Treatment Outcome , Young Adult
16.
Front Immunol ; 11: 344, 2020.
Article in English | MEDLINE | ID: mdl-32194562

ABSTRACT

Background: Cutaneous lupus erythematosus (CLE) is an interferon (IFN) -driven autoimmune skin disease characterized by an extensive cytotoxic lesional inflammation with activation of different innate immune pathways. Aim of our study was to investigate the specific role of Janus kinase 1 (JAK1) activation in this disease and the potential benefit of selective JAK1 inhibitors as targeted therapy in a preclinical CLE model. Methods: Lesional skin of patients with different CLE subtypes and healthy controls (N = 31) were investigated on JAK1 activation and expression of IFN-associated mediators via immunohistochemistry and gene expression analyses. The functional role of JAK1 and efficacy of inhibition was evaluated in vitro using cultured keratinocytes stimulated with endogenous nucleic acids. Results were confirmed in vivo using an established lupus-prone mouse model. Results: Proinflammatory immune pathways, including JAK/STAT signaling, are significantly upregulated within inflamed CLE skin. Here, lesional keratinocytes and dermal immune cells strongly express activated phospho-JAK1. Selective pharmacological JAK1 inhibition significantly reduces the expression of typical proinflammatory mediators such as CXCL chemokines, BLyS, TRAIL, and AIM2 in CLE in vitro models and also improves skin lesions in lupus-prone TREX1-/- -mice markedly. Conclusion: IFN-associated JAK/STAT activation plays a crucial role in the pathophysiology of CLE. Selective inhibition of JAK1 leads to a decrease of cytokine expression, reduced immune activation, and decline of keratinocyte cell death. Topical treatment with a JAK1-specific inhibitor significantly improves CLE-like skin lesions in a lupus-prone TREX1-/- -mouse model and appears to be a promising therapeutic approach for CLE patients.


Subject(s)
Azetidines/therapeutic use , Enzyme Inhibitors/therapeutic use , Isonicotinic Acids/therapeutic use , Janus Kinase 1/antagonists & inhibitors , Lupus Erythematosus, Cutaneous/drug therapy , Animals , Azetidines/pharmacology , Cell Line , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Induction , Enzyme Inhibitors/pharmacology , Exodeoxyribonucleases/deficiency , Gene Expression Regulation , Humans , Isonicotinic Acids/pharmacology , Janus Kinase 1/biosynthesis , Janus Kinase 1/genetics , Keratinocytes/drug effects , Keratinocytes/enzymology , Lichen Planus/enzymology , Lupus Erythematosus, Cutaneous/enzymology , Lupus Erythematosus, Discoid/enzymology , Mice , Mice, Inbred C57BL , Models, Immunological , Phosphoproteins/deficiency , Specific Pathogen-Free Organisms
17.
Cell ; 178(4): 867-886.e24, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398341

ABSTRACT

Somatosensory over-reactivity is common among patients with autism spectrum disorders (ASDs) and is hypothesized to contribute to core ASD behaviors. However, effective treatments for sensory over-reactivity and ASDs are lacking. We found distinct somatosensory neuron pathophysiological mechanisms underlie tactile abnormalities in different ASD mouse models and contribute to some ASD-related behaviors. Developmental loss of ASD-associated genes Shank3 or Mecp2 in peripheral mechanosensory neurons leads to region-specific brain abnormalities, revealing links between developmental somatosensory over-reactivity and the genesis of aberrant behaviors. Moreover, acute treatment with a peripherally restricted GABAA receptor agonist that acts directly on mechanosensory neurons reduced tactile over-reactivity in six distinct ASD models. Chronic treatment of Mecp2 and Shank3 mutant mice improved body condition, some brain abnormalities, anxiety-like behaviors, and some social impairments but not memory impairments, motor deficits, or overgrooming. Our findings reveal a potential therapeutic strategy targeting peripheral mechanosensory neurons to treat tactile over-reactivity and select ASD-related behaviors.


Subject(s)
Autism Spectrum Disorder/metabolism , GABA Agonists/pharmacology , Isonicotinic Acids/pharmacology , Phenotype , Sensory Receptor Cells/drug effects , Touch/drug effects , Action Potentials/drug effects , Animals , Anxiety/drug therapy , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/genetics , Behavior, Animal/drug effects , Brain/drug effects , Disease Models, Animal , Female , GABA Agonists/therapeutic use , Isonicotinic Acids/therapeutic use , Male , Maze Learning/drug effects , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/genetics , Prepulse Inhibition/drug effects , Sensory Receptor Cells/metabolism
18.
FASEB J ; 33(5): 6099-6114, 2019 05.
Article in English | MEDLINE | ID: mdl-30753095

ABSTRACT

Our objective was to investigate the effect of cholesterol [hypercholesterolemia and 7-ketocholesterol (7K)] on endoglin (Eng) expression and regulation with respect to endothelial or vascular dysfunction in vivo and in vitro. In vivo experiments were performed in 2-mo-old atherosclerosis-prone apolipoprotein E-deficient/LDL receptor-deficient (ApoE-/-/LDLR-/-) female mice and their wild-type C57BL/6J littermates. In in vitro experiments, human aortic endothelial cells (HAECs) were treated with 7K. ApoE-/-/LDLR-/- mice developed hypercholesterolemia accompanied by increased circulating levels of P-selectin and Eng and a disruption of NO metabolism. Functional analysis of the aorta demonstrated impaired vascular reactivity, and Western blot analysis revealed down-regulation of membrane Eng/Smad2/3/eNOS signaling in ApoE-/-/LDLR-/- mice. 7K increased Eng expression via Krüppel-like factor 6 (KLF6), liver X nuclear receptor, and NF-κB in HAECs. 7K-induced Eng expression was prevented by the treatment with 2-hydroxypropyl-ß-cyclodextrin; 8-{[5-chloro-2-(4-methylpiperazin-1-yl) pyridine-4-carbonyl] amino}-1-(4-fluorophenyl)-4, 5-dihydrobenzo[g]indazole-3-carboxamide; or by KLF6 silencing. 7K induced increased adhesion and transmigration of monocytic human leukemia promonocytic cell line cells and was prevented by Eng silencing. We concluded that hypercholesterolemia altered Eng expression and signaling, followed by endothelial or vascular dysfunction before formation of atherosclerotic lesions in ApoE-/-/LDLR-/- mice. By contrast, 7K increased Eng expression and induced inflammation in HAECs, which was followed by an increased adhesion and transmigration of monocytes via endothelium, which was prevented by Eng inhibition. Thus, we propose a relevant role for Eng in endothelial or vascular dysfunction or inflammation when exposed to cholesterol.-Vicen, M., Vitverova, B., Havelek, R., Blazickova, K., Machacek, M., Rathouska, J., Najmanová, I., Dolezelova, E., Prasnicka, A., Sternak, M., Bernabeu, C., Nachtigal, P. Regulation and role of endoglin in cholesterol-induced endothelial and vascular dysfunction in vivo and in vitro.


Subject(s)
Endoglin/metabolism , Endothelium, Vascular/metabolism , Hypercholesterolemia/metabolism , Plaque, Atherosclerotic/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Cells, Cultured , Cholesterol/metabolism , Endoglin/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Indazoles/pharmacology , Isonicotinic Acids/pharmacology , Kruppel-Like Factor 6/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , P-Selectin/metabolism , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/genetics , Receptors, LDL/genetics , Smad Proteins/metabolism , beta-Cyclodextrins/pharmacology
19.
Vet Parasitol ; 265: 19-23, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30638516

ABSTRACT

Pharmacological options to treat canine babesiosis caused by Babesia gibsoni, are limited. To address this challenge, screening for novel drug candidates and drug targets against B. gibsoni is urgently needed. In this study, we explored the inhibitory effects of two phytohormone inhibitors, fluridone (FLU) and inabenfide (INA), against B. gibsoni in vitro. The half-maximal inhibitory concentration (IC50) values of FLU and INA against B. gibsoni were 60.6 ± 3.4 and 4.3 ± 0.3 µM, respectively. Parasitemia and viability at 24, 48, and 72 h after FLU and INA treatments were significantly lower than those in the control group. The cytotoxicity of FLU and INA was evaluated using the dog-derived Madin-Darby canine kidney (MDCK) cell line; both FLU and INA were less toxic to the MDCK cells than to the control cells. The selectivity index of FLU and INA were higher than 16.5 and 232.6, respectively. In summary, the present study demonstrated that FLU and INA were effective against B. gibsoni infection in vitro and that these compounds might have potential as candidate drugs for the treatment of B. gibsoni.


Subject(s)
Antiprotozoal Agents/pharmacology , Babesia/drug effects , Isonicotinic Acids/pharmacology , Pyridones/pharmacology , Animals , Antiprotozoal Agents/chemistry , Isonicotinic Acids/chemistry , Pyridones/chemistry
20.
Environ Entomol ; 47(6): 1560-1564, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30346518

ABSTRACT

The onion thrips, Thrips tabaci Lindeman (Thysanoptera: Thripidae), is a polyphagous pest that causes serious damage to agricultural crops, vegetables, and ornamental plants worldwide. Farmers rely on the extensive usage of synthetic chemical insecticides to control T. tabaci. There is a dire need to develop alternative control strategies to overcome the problems posed by chemical insecticides. Efficient traps would allow sensitive monitoring and possibly mass trapping. A field experiment was conducted to evaluate the potential of three plant compounds with known release rates (ranging from 6-30 mg/d); eugenol (Eug), 1, 8-cineole (eucalyptol), and linalool in all possible combinations with a thrips attractant, ethyl iso-nicotinate (EI). A combination of EI with Eug increased the effect of EI by attracting 100% more thrips (effect size, 1.95) as compared to the control of EI alone. Catches in remaining treatments were lower and or not significantly different from EI alone. The results from our study could be used to develop improved volatile blends to be used for monitoring traps. Our data suggests that these traps could be effective even at very low populations.


Subject(s)
Eugenol/pharmacology , Isonicotinic Acids/pharmacology , Monoterpenes/pharmacology , Pheromones/pharmacology , Thysanoptera/drug effects , Acyclic Monoterpenes , Animals , Drug Synergism
SELECTION OF CITATIONS
SEARCH DETAIL
...