Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell Death Dis ; 15(2): 123, 2024 02 09.
Article in English | MEDLINE | ID: mdl-38336804

ABSTRACT

Discovery of new small molecules that can activate distinct programmed cell death pathway is of significant interest as a research tool and for the development of novel therapeutics for pathological conditions such as cancer and infectious diseases. The small molecule raptinal was discovered as a pro-apoptotic compound that can rapidly trigger apoptosis by promoting the release of cytochrome c from the mitochondria and subsequently activating the intrinsic apoptotic pathway. As raptinal is very effective at inducing apoptosis in a variety of different cell types in vitro and in vivo, it has been used in many studies investigating cell death as well as the clearance of dying cells. While examining raptinal as an apoptosis inducer, we unexpectedly identified that in addition to its pro-apoptotic activities, raptinal can also inhibit the activity of caspase-activated Pannexin 1 (PANX1), a ubiquitously expressed transmembrane channel that regulates many cell death-associated processes. By implementing numerous biochemical, cell biological and electrophysiological approaches, we discovered that raptinal can simultaneously induce apoptosis and inhibit PANX1 activity. Surprisingly, raptinal was found to inhibit cleavage-activated PANX1 via a mechanism distinct to other well-described PANX1 inhibitors such as carbenoxolone and trovafloxacin. Furthermore, raptinal also interfered with PANX1-regulated apoptotic processes including the release of the 'find-me' signal ATP, the formation of apoptotic cell-derived extracellular vesicles, as well as NLRP3 inflammasome activation. Taken together, these data identify raptinal as the first compound that can simultaneously induce apoptosis and inhibit PANX1 channels. This has broad implications for the use of raptinal in cell death studies as well as in the development new PANX1 inhibitors.


Subject(s)
Apoptosis , Connexins , Fluorenes , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Cell Death , Connexins/antagonists & inhibitors , Connexins/metabolism , Cyclopentanes/pharmacology
2.
Biochem Soc Trans ; 51(4): 1687-1699, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37622532

ABSTRACT

Pannexin 1 (PANX1) is a widely expressed large-pore ion channel located in the plasma membrane of almost all vertebrate cells. It possesses a unique ability to act as a conduit for both inorganic ions (e.g. potassium or chloride) and bioactive metabolites (e.g. ATP or glutamate), thereby activating varying signaling pathways in an autocrine or paracrine manner. Given its crucial role in cell-cell interactions, the activity of PANX1 has been implicated in maintaining homeostasis of cardiovascular, immune, and nervous systems. Dysregulation of PANX1 has also been linked to numerous diseases, such as ischemic stroke, seizure, and inflammatory disorders. Therefore, the mechanisms underlying different modes of PANX1 activation and its context-specific channel properties have gathered significant attention. In this review, we summarize the roles of PANX1 in various physiological processes and diseases, and analyze the accumulated lines of evidence supporting diverse molecular mechanisms associated with different PANX1 activation modalities. We focus on examining recent discoveries regarding PANX1 regulations by reversible post-translational modifications, elevated intracellular calcium concentration, and protein-protein interactions, as well as by irreversible cleavage of its C-terminal tail. Additionally, we delve into the caveats in the proposed PANX1 gating mechanisms and channel open-closed configurations by critically analyzing the structural insights derived from cryo-EM studies and the unitary properties of PANX1 channels. By doing so, we aim to identify potential research directions for a better understanding of the functions and regulations of PANX1 channels.


Subject(s)
Calcium , Cell Communication , Connexins , Nerve Tissue Proteins , Cell Membrane , Chlorides , Glutamic Acid , Humans , Connexins/genetics , Nerve Tissue Proteins/genetics
3.
J Clin Immunol ; 42(8): 1730-1741, 2022 11.
Article in English | MEDLINE | ID: mdl-35947322

ABSTRACT

PURPOSE: Anti-granulocyte-macrophage colony-stimulating factor autoantibodies (anti-GM-CSF Abs) are a predisposing factor for pulmonary alveolar proteinosis (PAP) and Cryptococcus gattii cryptococcosis. This study aimed to investigate clinical manifestations in anti-GM-CSF Ab-positive patients with C. gattii cryptococcosis and analyze the properties of anti-GM-CSF Abs derived from these patients and patients with PAP. METHODS: Thirty-nine patients diagnosed with cryptococcosis (caused by C. neoformans or C. gattii) and 6 with PAP were enrolled in the present study. Clinical information was obtained from medical records. Blood samples were collected for analysis of autoantibody properties. We also explored the National Health Insurance Research Database (NHIRD) of Taiwan to investigate the epidemiology of cryptococcosis and PAP. RESULTS: High titers of neutralizing anti-GM-CSF Abs were identified in 15 patients with cryptococcosis (15/39, 38.5%). Most anti-GM-CSF Ab-positive cryptococcosis cases had central nervous system (CNS) involvement (14/15, 93.3%). Eleven out of 14 (78.6%) anti-GM-CSF Ab-positive CNS cryptococcosis patients were confirmed to be infected with C. gattii, and PAP did not occur synchronously or metachronously in a single patient from our cohort. Exploration of an association between HLA and anti-GM-CSF Ab positivity or differential properties of autoantibodies from cryptococcosis patients and PAP yielded no significant results. CONCLUSION: Anti-GM-CSF Abs can cause two diseases, C. gattii cryptococcosis and PAP, which seldom occur in the same subject. Current biological evidence regarding the properties of anti-GM-CSF Abs cannot provide clues regarding decisive mechanisms. Further analysis, including more extensive cohort studies and investigations into detailed properties, is mandatory to better understand the pathogenesis of anti-GM-CSF Abs.


Subject(s)
Cryptococcosis , Pulmonary Alveolar Proteinosis , Humans , Autoantibodies , Cryptococcosis/diagnosis , Cryptococcosis/epidemiology , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/etiology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology
4.
Immunity ; 54(8): 1715-1727.e7, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34283971

ABSTRACT

Allergic airway inflammation is driven by type-2 CD4+ T cell inflammatory responses. We uncover an immunoregulatory role for the nucleotide release channel, Panx1, in T cell crosstalk during airway disease. Inverse correlations between Panx1 and asthmatics and our mouse models revealed the necessity, specificity, and sufficiency of Panx1 in T cells to restrict inflammation. Global Panx1-/- mice experienced exacerbated airway inflammation, and T-cell-specific deletion phenocopied Panx1-/- mice. A transgenic designed to re-express Panx1 in T cells reversed disease severity in global Panx1-/- mice. Panx1 activation occurred in pro-inflammatory T effector (Teff) and inhibitory T regulatory (Treg) cells and mediated the extracellular-nucleotide-based Treg-Teff crosstalk required for suppression of Teff cell proliferation. Mechanistic studies identified a Salt-inducible kinase-dependent phosphorylation of Panx1 serine 205 important for channel activation. A genetically targeted mouse expressing non-phosphorylatable Panx1S205A phenocopied the exacerbated inflammation in Panx1-/- mice. These data identify Panx1-dependent Treg:Teff cell communication in restricting airway disease.


Subject(s)
Asthma/immunology , Cell Communication/immunology , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Cell Proliferation/physiology , Connexins/genetics , Disease Models, Animal , HEK293 Cells , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Respiratory System/immunology
5.
Nat Commun ; 12(1): 4482, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34301959

ABSTRACT

Activation of Pannexin 1 (PANX1) ion channels causes release of intercellular signaling molecules in a variety of (patho)physiological contexts. PANX1 can be activated by G protein-coupled receptors (GPCRs), including α1-adrenergic receptors (α1-ARs), but how receptor engagement leads to channel opening remains unclear. Here, we show that GPCR-mediated PANX1 activation can occur via channel deacetylation. We find that α1-AR-mediated activation of PANX1 channels requires Gαq but is independent of phospholipase C or intracellular calcium. Instead, α1-AR-mediated PANX1 activation involves RhoA, mammalian diaphanous (mDia)-related formin, and a cytosolic lysine deacetylase activated by mDia - histone deacetylase 6. HDAC6 associates with PANX1 and activates PANX1 channels, even in excised membrane patches, suggesting direct deacetylation of PANX1. Substitution of basally-acetylated intracellular lysine residues identified on PANX1 by mass spectrometry either prevents HDAC6-mediated activation (K140/409Q) or renders the channels constitutively active (K140R). These data define a non-canonical RhoA-mDia-HDAC6 signaling pathway for GαqPCR activation of PANX1 channels and uncover lysine acetylation-deacetylation as an ion channel silencing-activation mechanism.


Subject(s)
Connexins/metabolism , Histone Deacetylase 6/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Acetylation , Animals , Cells, Cultured , Connexins/genetics , Connexins/physiology , HEK293 Cells , Histone Deacetylase 6/genetics , Humans , Jurkat Cells , Lysine/genetics , Lysine/metabolism , Membrane Potentials/physiology , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Patch-Clamp Techniques , Receptors, Adrenergic, alpha-1/genetics , Signal Transduction/genetics , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
6.
Elife ; 102021 01 07.
Article in English | MEDLINE | ID: mdl-33410749

ABSTRACT

Pannexin 1 (Panx1) is a membrane channel implicated in numerous physiological and pathophysiological processes via its ability to support release of ATP and other cellular metabolites for local intercellular signaling. However, to date, there has been no direct demonstration of large molecule permeation via the Panx1 channel itself, and thus the permselectivity of Panx1 for different molecules remains unknown. To address this, we expressed, purified, and reconstituted Panx1 into proteoliposomes and demonstrated that channel activation by caspase cleavage yields a dye-permeable pore that favors flux of anionic, large-molecule permeants (up to ~1 kDa). Large cationic molecules can also permeate the channel, albeit at a much lower rate. We further show that Panx1 channels provide a molecular pathway for flux of ATP and other anionic (glutamate) and cationic signaling metabolites (spermidine). These results verify large molecule permeation directly through caspase-activated Panx1 channels that can support their many physiological roles.


Subject(s)
Adenosine Triphosphate/metabolism , Connexins/genetics , Ion Channels/genetics , Nerve Tissue Proteins/genetics , Signal Transduction , Xenopus Proteins/genetics , Animals , Caspases/metabolism , Connexins/metabolism , Humans , Ion Channels/metabolism , Nerve Tissue Proteins/metabolism , Spodoptera/genetics , Spodoptera/metabolism , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/metabolism
7.
J Microbiol Immunol Infect ; 54(3): 447-456, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32102749

ABSTRACT

BACKGROUND: This study aims to investigate the antimicrobial ability and mechanism analysis of Lactobacillus species against carbapenemase-producing Enterobacteriaceae (CPE). METHODS: Five Lactobacillus spp. strains and 18 CPE clinical isolates were collected. Their anti-CPE effects were assessed by agar well diffusion and broth microdilution assay, as well as time-kill test. Finally, the specific anti-CPE mechanism, especially for the effect of organic acids was determined using broth microdilution method. RESULTS: All of five Lactobacilli isolates displayed the potent activity against most CPE isolates with mean zones of inhibition ranging 10.2-21.1 mm. The anti-CPE activity was not affected by heating, catalase, and proteinase treatment. Under the concentration of 50% LUC0180 cell-free supernatant (CFS), lactic acid, and mix acid could totally inhibit the growth of carbapenem-resistant Klebsiella pneumoniae (CPE0011), and acetic acid could inhibit 67.8%. In contrast, succinic acid and citric acid could not inhibit the growth of CPE0011. While we decreased the concentration to 25%, only lactic acid and mix acid displayed 100% inhibition. In contrast, succinic acid, citric acid and acetic acid did not show any inhibitory effect. CONCLUSIONS: Lactobacillus strains exhibit potent anti-CPE activity, and lactic acid produced by Lactobacillus strains is the major antimicrobial mechanism.


Subject(s)
Antibiosis , Carbapenem-Resistant Enterobacteriaceae/physiology , Lactobacillus/physiology , Anti-Bacterial Agents/pharmacology , Carbapenem-Resistant Enterobacteriaceae/drug effects , Citric Acid/pharmacology , Enterobacteriaceae Infections/microbiology , Humans , In Vitro Techniques , Klebsiella pneumoniae/drug effects , Lactic Acid/pharmacology , Lactobacillus/chemistry , Microbial Sensitivity Tests , Succinic Acid/pharmacology
8.
J Med Chem ; 63(15): 8088-8113, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32551603

ABSTRACT

The serine protease factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anticoagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein, we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement factor D inhibitor and exhibited submicromolar FXIa activity and an encouraging absorption, distribution, metabolism, and excretion (ADME) profile while being devoid of a peptidomimetic architecture. Optimization of interactions in the S1, S1ß, and S1' pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with subnanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a preclinical pharmacokinetics (PK) profile consistent with bid dosing in patients.


Subject(s)
Factor XIa/antagonists & inhibitors , Factor XIa/genetics , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/chemistry , Administration, Oral , Amino Acid Sequence , Animals , Biological Availability , Dogs , Drug Evaluation, Preclinical/methods , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
9.
J Microbiol Immunol Infect ; 53(2): 191-208, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32169531

ABSTRACT

Clostridioides difficile infection (CDI) is a major enteric disease associated with antibiotic use and a leading cause of hospital-acquired infections worldwide. This is the first guideline for treatment of CDI in Taiwan, aiming to optimize medical care for patients with CDI. The target audience of this document includes all healthcare personnel who are involved in the medical care of patients with CDI. The 2018 Guidelines Recommendations for Evidence-based Antimicrobial agents use in Taiwan (GREAT) working group was formed, comprising of infectious disease specialists from 13 medical centers in Taiwan, to review the evidence and draft recommendations using the grading of recommendations assessment, development, and evaluation (GRADE) methodology. A nationwide expert panel reviewed the recommendations during a consensus meeting in March 2019. The recommendation is endorsed by the Infectious Diseases Society of Taiwan (IDST). This guideline describes the epidemiology and risk factors of CDI, and provides recommendations for treatment of CDI in both adults and children. Recommendations for treatment of the first episode of CDI, first recurrence, second and subsequent recurrences of CDI, severe CDI, fulminant CDI, and pediatric CDI are provided.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Clostridium Infections/drug therapy , Guidelines as Topic , Adult , Child , Clostridioides difficile/drug effects , Clostridium Infections/epidemiology , Cross Infection/epidemiology , Databases, Factual , Diarrhea/drug therapy , Diarrhea/microbiology , Humans , Risk Factors , Taiwan/epidemiology
10.
Blood ; 133(13): 1507-1516, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30692123

ABSTRACT

A large unmet medical need exists for safer antithrombotic drugs because all currently approved anticoagulant agents interfere with hemostasis, leading to an increased risk of bleeding. Genetic and pharmacologic evidence in humans and animals suggests that reducing factor XI (FXI) levels has the potential to effectively prevent and treat thrombosis with a minimal risk of bleeding. We generated a fully human antibody (MAA868) that binds the catalytic domain of both FXI (zymogen) and activated FXI. Our structural studies show that MAA868 traps FXI and activated FXI in an inactive, zymogen-like conformation, explaining its equally high binding affinity for both forms of the enzyme. This binding mode allows the enzyme to be neutralized before entering the coagulation process, revealing a particularly attractive anticoagulant profile of the antibody. MAA868 exhibited favorable anticoagulant activity in mice with a dose-dependent protection from carotid occlusion in a ferric chloride-induced thrombosis model. MAA868 also caused robust and sustained anticoagulant activity in cynomolgus monkeys as assessed by activated partial thromboplastin time without any evidence of bleeding. Based on these preclinical findings, we conducted a first-in-human study in healthy subjects and showed that single subcutaneous doses of MAA868 were safe and well tolerated. MAA868 resulted in dose- and time-dependent robust and sustained prolongation of activated partial thromboplastin time and FXI suppression for up to 4 weeks or longer, supporting further clinical investigation as a potential once-monthly subcutaneous anticoagulant therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Factor XI/antagonists & inhibitors , Thrombosis/drug therapy , Adolescent , Adult , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Anticoagulants/pharmacology , Female , Humans , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Macaca fascicularis , Male , Mice, Inbred C57BL , Middle Aged , Molecular Docking Simulation , Thrombosis/blood , Young Adult
11.
J Microbiol Immunol Infect ; 52(2): 273-281, 2019 Apr.
Article in English | MEDLINE | ID: mdl-27133391

ABSTRACT

BACKGROUND/PURPOSE: In vitro studies of the combination of an aminoglycoside with tigecycline or doxycycline against Klebsiella pneumoniae carbapenemase (KPC)-producing K. pneumoniae isolates are rarely published. The goal of this study was to evaluate the antibacterial activity of the combination regimens. METHODS: Thirteen genetically different KPC-producing K. pneumoniae isolates were randomly selected. Drug concentrations of amikacin, gentamicin, tigecycline, and doxycycline were adjusted to 1-, 1/2-, and 1/4-fold of respective minimum inhibitory concentrations (MICs). Each drug alone or the combinations of amikacin or gentamicin with tigecycline or doxycycline were tested by combination studies. RESULTS: Treatment with the 1× MIC concentration in combinations of amikacin or gentamicin and tigecycline or doxycycline for 24 hours resulted in bactericidal activity of 84-100% in the isolates. Treatment with 1/2× MIC combinations resulted in synergism of 69-100% in the isolates. Notably, doxycycline plus gentamicin or amikacin was synergistic for all tested isolates. However, bactericidal or synergistic effect was barely evident following 1/4× MIC combinations. There was no antagonism in any of the combination regimens. CONCLUSION: Enhanced activity was noted following treatment with doxycycline combined with gentamicin or amikacin against KPC-producing K. pneumoniae isolates, warranting further in vitro and animal investigations before clinical application.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Colistin/pharmacology , Klebsiella pneumoniae/drug effects , beta-Lactamases/biosynthesis , Amikacin/pharmacology , Bacterial Proteins/genetics , DNA, Bacterial/genetics , Doxycycline/pharmacology , Drug Combinations , Drug Resistance, Multiple, Bacterial/drug effects , Drug Synergism , Genotype , Gentamicins/pharmacology , Humans , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/genetics , Microbial Sensitivity Tests , Tigecycline/pharmacology , beta-Lactamases/genetics
12.
Infect Drug Resist ; 11: 2555-2560, 2018.
Article in English | MEDLINE | ID: mdl-30573984

ABSTRACT

New-Delhi metallo-ß-lactamase1 (NDM-1) Enterobacteriaceae are increasing worldwide. Herein, we describe a single patient who carried three unusual NDM-1 carbapenem-resistant Enterobacteriaceae - Enterobacter cloacae (E. cloacae) yielded from a urine specimen and Klebsiella pneumoniae (K. pneumoniae) and Escherichia coli (E. coli) from stool specimens. For E. cloacae, its bla NDM-1-encoding plasmid was pKP04NDM with a size of ~54 kb replicons with an IncN backbone. For K. pneumoniae, its bla NDM-1-encoding plasmid was pNDM-BTR with a size of ~59.6 kb and belonged to IncN. For E. coli, its main bla NDM-1-encoding plasmid was pIMP-HK1500, and the NDM-1 gene was obtained from a part of pNDM-BTR (8439 bp). These three clinical strains are reported for the first time and are assumed to be imported from mainland China to Taiwan. The three different plasmids were never reported in K. pneumoniae, E. coli, and Citrobacter spp before. Owing to their associated multidrug resistance, appropriate measures of periodic, targeted surveillance, and development of new antimicrobial agents are urgently needed.

14.
J Microbiol Immunol Infect ; 51(1): 76-81, 2018 Feb.
Article in English | MEDLINE | ID: mdl-27260781

ABSTRACT

BACKGROUND/PURPOSE: The aim of this study is to investigate the role of tigecycline in Vibrio vulnificus infection. METHODS: Eight randomly selected clinical V. vulnificus isolates were studied to obtain the minimal inhibitory concentrations (MICs) of minocycline, cefotaxime, and tigecycline, and the time-kill curves of tigecycline alone or in combination with other drugs. A peritonitis mouse model was used for the evaluation of the therapeutic efficacy of tigecycline alone or cefotaxime in combination with minocycline or tigecycline. RESULTS: The MIC of minocycline, cefotaxime, and tigecycline for eight clinical V. vulnificus isolates was 0.06-0.12 µg/mL, 0.03-0.06 µg/mL, and 0.03-0.06 µg/mL, respectively. In time-killing studies, at the concentration of 1 × MIC, the inhibitory effect of tigecycline persisted for 24 hours in five of eight isolates. With 2 × MIC and trough level, the inhibitory effect was noted in all isolates for 24 hours. With the combination of minocycline plus cefotaxime and tigecycline plus cefotaxime at 1/2 × MIC, the bactericidal effect was noted in 25% and 62.5% of eight isolates and synergism in 50% and 75% of isolates. With a low (1.25 × 105 CFU/mL) inoculum, all infected mice survived with tigecycline alone, tigecycline plus cefotaxime, or minocycline plus cefotaxime on the 14th day. At the inoculum of 1.25 × 106 CFU, the survival rate was 33.3% on the 14th day in the tigecycline plus cefotaxime-treated group, but none of the mice treated by tigecycline alone or minocycline plus cefotaxime survived (33.3% vs. 0%, p = 0.01 by Fisher's exact test). CONCLUSION: Our in vitro combination and animal studies indicate that tigecycline could be an option for the treatment of invasive V. vulnificus infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Minocycline/analogs & derivatives , Vibrio Infections/drug therapy , Vibrio vulnificus/drug effects , Animals , Cefotaxime/pharmacology , Colony Count, Microbial , Disease Models, Animal , Drug Combinations , Drug Synergism , Drug Therapy, Combination , Humans , Mice , Microbial Sensitivity Tests , Minocycline/pharmacology , Minocycline/therapeutic use , Peritonitis/drug therapy , Peritonitis/microbiology , Survival Rate , Taiwan , Tigecycline , Time Factors , Vibrio vulnificus/isolation & purification
15.
J Gen Physiol ; 150(1): 19-39, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29233884

ABSTRACT

Pannexin 1 (Panx1) forms plasma membrane ion channels that are widely expressed throughout the body. Panx1 activation results in the release of nucleotides such as adenosine triphosphate and uridine triphosphate. Thus, these channels have been implicated in diverse physiological and pathological functions associated with purinergic signaling, such as apoptotic cell clearance, blood pressure regulation, neuropathic pain, and excitotoxicity. In light of this, substantial attention has been directed to understanding the mechanisms that regulate Panx1 channel expression and activation. Here we review accumulated evidence for the various activation mechanisms described for Panx1 channels and, where possible, the unitary channel properties associated with those forms of activation. We also emphasize current limitations in studying Panx1 channel function and propose potential directions to clarify the exciting and expanding roles of Panx1 channels.


Subject(s)
Connexins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Connexins/chemistry , Connexins/genetics , Humans , Ion Channel Gating , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics
16.
Circ Res ; 122(4): 606-615, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29237722

ABSTRACT

RATIONALE: Resistant hypertension is a major health concern with unknown cause. Spironolactone is an effective antihypertensive drug, especially for patients with resistant hypertension, and is considered by the World Health Organization as an essential medication. Although spironolactone can act at the mineralocorticoid receptor (MR; NR3C2), there is increasing evidence of MR-independent effects of spironolactone. OBJECTIVE: Here, we detail the unexpected discovery that Panx1 (pannexin 1) channels could be a relevant in vivo target of spironolactone. METHODS AND RESULTS: First, we identified spironolactone as a potent inhibitor of Panx1 in an unbiased small molecule screen, which was confirmed by electrophysiological analysis. Next, spironolactone inhibited α-adrenergic vasoconstriction in arterioles from mice and hypertensive humans, an effect dependent on smooth muscle Panx1, but independent of the MR NR3C2. Last, spironolactone acutely lowered blood pressure, which was dependent on smooth muscle cell expression of Panx1 and independent of NR3C2. This effect, however, was restricted to steroidal MR antagonists as a nonsteroidal MR antagonist failed to reduced blood pressure. CONCLUSIONS: These data suggest new therapeutic modalities for resistant hypertension based on Panx1 inhibition.


Subject(s)
Antihypertensive Agents/pharmacology , Connexins/antagonists & inhibitors , Diuretics/pharmacology , Hypertension/drug therapy , Mineralocorticoid Receptor Antagonists/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Spironolactone/pharmacology , Animals , Antihypertensive Agents/therapeutic use , Arterioles/drug effects , Connexins/metabolism , Diuretics/therapeutic use , HEK293 Cells , Humans , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mineralocorticoid Receptor Antagonists/therapeutic use , Nerve Tissue Proteins/metabolism , Spironolactone/therapeutic use
17.
Mol Med Rep ; 17(1): 1311-1320, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115516

ABSTRACT

The signal transducer and activator of transcription 3 (STAT3) is known to be involved in hypertrophy and fibrosis in cardiac dysfunction. The activation of STAT3 via the phosphorylation of STAT3 is required for the production of functional activity. It has been established that lipopolysaccharide (LPS)­induced phosphorylation of STAT3 in cardiomyocytes primarily occurs through a direct receptor­mediated action. This effect is demonstrated to be produced rapidly. STAT3 in cardiac fibrosis of diabetes is induced by high glucose through promotion of the STAT3­associated signaling pathway. However, the time schedule for STAT3 activation between LPS and high glucose appears to be different. Therefore, the difference in STAT3 activation between LPS and hyperglycemia in cardiomyocytes requires elucidation. The present study investigated the phosphorylation of STAT3 induced by LPS and hyperglycemia in the rat cardiac cell line H9c2. Additionally, phosphorylation of STAT3 induced by erythropoietin (EPO) via receptor activation was compared. Then, the downstream signals for fibrosis, including the connective tissue growth factor (CTGF) and matrix metalloproteinase (MMP)­9, were determined using western blotting, while the mRNA levels were quantified. LPS induced a rapid elevation of STAT3 phosphorylation in H9c2 cells within 30 min, similar to that produced by EPO. However, LPS or EPO failed to modify the mRNA level of STAT3, and/or the downstream signals for fibrosis. High glucose increased STAT3 phosphorylation to be stable after a long period of incubation. Glucose incubation for 24 h may augment the STAT3 expression in a dose­dependent manner. Consequently, fibrosis­associated signals, including CTGF and MMP­9 protein, were raised in parallel. In the presence of tiron, an antioxidant, these changes by hyperglycemia were markedly reduced, demonstrating the mediation of oxidative stress. Therefore, LPS­ or EPO­induced STAT3 phosphorylation is different compared with that caused by high glucose in H9c2 cells. Sustained activation of STAT3 by hyperglycemia may promote the expression of fibrosis­associated signals, including CTGF and MMP­9, in H9c2 cells. Therefore, regarding the cardiac dysfunctions associated with diabetes and/or hyperglycemia, the identification of nuclear STAT3 may be more reliable compared with the assay of phosphorylated STAT3 in cardiac cells.


Subject(s)
Myocytes, Cardiac/metabolism , STAT3 Transcription Factor/physiology , Animals , Cell Line , Connective Tissue Growth Factor/metabolism , Erythropoietin/pharmacology , Hyperglycemia/metabolism , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 9/metabolism , Myocytes, Cardiac/immunology , Oxidative Stress , Phosphorylation , Protein Processing, Post-Translational , Rats , Signal Transduction
18.
Front Microbiol ; 8: 884, 2017.
Article in English | MEDLINE | ID: mdl-28572795

ABSTRACT

The empirical combination of both a beta-lactam and glycopeptide to counter potential staphylococcal pathogens may improve the clinical outcomes for cases of Staphylococcus aureus bacteremia. We reported comparative in vitro studies of combination effects of different cephalosporins (i.e., cefazolin, cefmetazole, cefotaxime, and cefepime) combined with glycopeptides for 34 randomly selected methicillin-resistant S. aureus (MRSA) isolates by three methods, including the checkerboard, time-killing, and combination MIC measurement methods. Thirteen SCCmec type III isolates with a cefazolin MIC of ≥ 128 µg/mL were classified as the high-cefazolin MIC (HCM) group, whereas 13 SCCmec type IV and 8 SCCmec type V isolates were classified as the low-cefazolin MIC (LCM) group. With the checkerboard method, synergism was present for vancomycin-based combinations at 30.8-69.2 and 13.6-66.7%, as well as teicoplanin-based combinations of 38.5-84.6 and 0-47.6%, of the HCM and LCM isolates, respectively. No antagonism was noted. The in vitro inhibitory activity was evident even at a low concentration of 1/512x MIC of cephalosporin combined with sub-inhibitory concentrations (1/2x MIC) of a glycopeptide. With time-killing assays, synergism was noted at 1/2x or 1x susceptible breakpoint concentrations (SBCs) of a cephalosporin combined with 1/4 or 1/2 MIC of a glycopeptide. In the presence of 1/2 SBC of a cephalosporin, vancomycin or teicoplanin MICs decreased an average of 2.0- to 6.6- or 1.6- to 5.5-fold, respectively. With 8 µg/mL cephalosporin, the decline of glycopeptide MICs was most obvious in the presence of cefmetazole. In conclusion, cephalosporin-glycopeptide combinations at clinically achievable concentrations can exhibit in vitro synergistic antibacterial activity against clinical MRSA isolates. Such combinations require more clinical data to support their application for use in human MRSA infections.

19.
Mol Cell ; 65(6): 999-1013.e7, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28306514

ABSTRACT

PARK2 is a gene implicated in disease states with opposing responses in cell fate determination, yet its contribution in pro-survival signaling is largely unknown. Here we show that PARK2 is altered in over a third of all human cancers, and its depletion results in enhanced phosphatidylinositol 3-kinase/Akt (PI3K/Akt) activation and increased vulnerability to PI3K/Akt/mTOR inhibitors. PARK2 depletion contributes to AMPK-mediated activation of endothelial nitric oxide synthase (eNOS), enhanced levels of reactive oxygen species, and a concomitant increase in oxidized nitric oxide levels, thereby promoting the inhibition of PTEN by S-nitrosylation and ubiquitination. Notably, AMPK activation alone is sufficient to induce PTEN S-nitrosylation in the absence of PARK2 depletion. Park2 loss and Pten loss also display striking cooperativity to promote tumorigenesis in vivo. Together, our findings reveal an important missing mechanism that might account for PTEN suppression in PARK2-deficient tumors, and they highlight the importance of PTEN S-nitrosylation in supporting cell survival and proliferation under conditions of energy deprivation.


Subject(s)
Energy Metabolism , Neoplasms/enzymology , Nitric Oxide/metabolism , Oxidative Stress , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitin-Protein Ligases/deficiency , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Movement , Cell Proliferation , Cell Survival , Dose-Response Relationship, Drug , Enzyme Activation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection , Tumor Burden , Ubiquitin-Protein Ligases/genetics , Ubiquitination
20.
Sci Rep ; 7: 42550, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28195232

ABSTRACT

Neuropathic pain symptoms respond poorly to available therapeutics, with most treated patients reporting unrelieved pain and significant impairment in daily life. Here, we show that Pannexin 1 (Panx1) in hematopoietic cells is required for pain-like responses following nerve injury in mice, and a potential therapeutic target. Panx1 knockout mice (Panx1-/-) were protected from hypersensitivity in two sciatic nerve injury models. Bone marrow transplantation studies show that expression of functional Panx1 in hematopoietic cells is necessary for mechanical hypersensitivity following nerve injury. Reconstitution of irradiated Panx1 knockout mice with hematopoietic Panx1-/- cells engineered to re-express Panx1 was sufficient to recover hypersensitivity after nerve injury; this rescue required expression of a Panx1 variant that can be activated by G protein-coupled receptors (GPCRs). Finally, chemically distinct Panx1 inhibitors blocked development of nerve injury-induced hypersensitivity and partially relieved this hypersensitivity after it was established. These studies indicate that Panx1 expressed in immune cells is critical for pain-like effects following nerve injury in mice, perhaps via a GPCR-mediated activation mechanism, and suggest that inhibition of Panx1 may be useful in treating neuropathic pain.


Subject(s)
Connexins/genetics , Connexins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Animals , Bone Marrow Cells/metabolism , Connexins/antagonists & inhibitors , Disease Models, Animal , Female , Gene Expression , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/antagonists & inhibitors , Neuralgia/drug therapy , Neuralgia/physiopathology , Peripheral Nerve Injuries/complications , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...