Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Sci Transl Med ; 16(749): eabp8334, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809966

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease driven by gain-of-function variants in activin receptor-like kinase 2 (ALK2), the most common variant being ALK2R206H. In FOP, ALK2 variants display increased and dysregulated signaling through the bone morphogenetic protein (BMP) pathway resulting in progressive and permanent replacement of skeletal muscle and connective tissues with heterotopic bone, ultimately leading to severe debilitation and premature death. Here, we describe the discovery of BLU-782 (IPN60130), a small-molecule ALK2R206H inhibitor developed for the treatment of FOP. A small-molecule library was screened in a biochemical ALK2 binding assay to identify potent ALK2 binding compounds. Iterative rounds of structure-guided drug design were used to optimize compounds for ALK2R206H binding, ALK2 selectivity, and other desirable pharmacokinetic properties. BLU-782 preferentially bound to ALK2R206H with high affinity, inhibiting signaling from ALK2R206H and other rare FOP variants in cells in vitro without affecting signaling of closely related homologs ALK1, ALK3, and ALK6. In vivo efficacy of BLU-782 was demonstrated using a conditional knock-in ALK2R206H mouse model, where prophylactic oral dosing reduced edema and prevented cartilage and heterotopic ossification (HO) in both muscle and bone injury models. BLU-782 treatment preserved the normal muscle-healing response in ALK2R206H mice. Delayed dosing revealed a short 2-day window after injury when BLU-782 treatment prevented HO in ALK2R206H mice, but dosing delays of 4 days or longer abrogated HO prevention. Together, these data suggest that BLU-782 may be a candidate for prevention of HO in FOP.


Subject(s)
Disease Models, Animal , Myositis Ossificans , Ossification, Heterotopic , Animals , Myositis Ossificans/drug therapy , Myositis Ossificans/metabolism , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/prevention & control , Mice , Humans , Activin Receptors, Type II/metabolism , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/antagonists & inhibitors , Signal Transduction/drug effects
2.
PLoS One ; 8(9): e72714, 2013.
Article in English | MEDLINE | ID: mdl-24023766

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) is the most common form of arthritis worldwide. Pain and reduced function are the main symptoms in this prevalent disease. There are currently no treatments for OA that modify disease progression; therefore analgesic drugs and joint replacement for larger joints are the standard of care. In light of several recent studies reporting the use of bisphosphonates for OA treatment, our work aimed to evaluate published literature to assess the effectiveness of bisphosphonates in OA treatment. METHODS: Literature databases were searched from inception to the 30th June 2012 for clinical trials of bisphosphonates to treat OA pain. Data was appraised and levels of evidence determined qualitatively using best evidence synthesis from the Cochrane Collaboration. The two largest studies were conducted with risedronate in the treatment of knee OA, for which meta-analyses were performed for pain and functional outcomes. RESULTS: Our searches found 13/297 eligible studies, which included a total of 3832 participants. The trials recruited participants with OA of the hand (n=1), knee (n=8), knee and spine (n=3), or hip (n=1). Our meta-analysis of the two largest knee studies using risedronate 15 mg showed odds ratios favouring placebo interventions for the Western Ontario and McMaster Universities Arthritis Index (WOMAC) pain (1.73), WOMAC function (2.03), and WOMAC stiffness (1.82). However, 8 trials (61.5%) reported that bisphosphonates improve pain assessed by VAS scores and 2 (38.5%) reported significant improvement in WOMAC pain scores compared to control groups. CONCLUSIONS: There is limited evidence that bisphosphonates are effective in the treatment of OA pain. Limitations of the studies we analysed included the differences in duration of bisphosphonate use, the dose and route of administration and the lack of long-term data on OA joint structure modification post-bisphosphonate therapy. Future more targeted studies are required to appreciate the value of bisphosphonates in treating osteoarthritis pain. TRIAL REGISTRATION: PROSPERO Register CRD42012002541.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Diphosphonates/therapeutic use , Osteoarthritis/drug therapy , Pain/drug therapy , Humans , Treatment Outcome
3.
J Bacteriol ; 195(10): 2244-54, 2013 May.
Article in English | MEDLINE | ID: mdl-23475976

ABSTRACT

The plasmid-encoded type three secretion system (TTSS) of Yersinia spp. is responsible for the delivery of effector proteins into cells of the innate immune system, where these effectors disrupt the target cells' activity. Successful translocation of effectors into mammalian cells requires Yersinia to both insert a translocon into the host cell membrane and sense contact with host cells. To probe the events necessary for translocation, we investigated protein-protein interactions among TTSS components of the needle-translocon complex using a chemical cross-linking-based approach. We detected extracellular protein complexes containing YscF, LcrV, and YopD that were dependent upon needle formation. The formation of these complexes was evaluated in a secretion-competent but translocation-defective mutant, the YscFD28AD46A strain (expressing YscF with the mutations D28A and D46A). We found that one of the YscF and most of the LcrV and YopD cross-linked complexes were nearly absent in this mutant. Furthermore, the YscFD28AD46A strain did not support YopB insertion into mammalian membranes, supporting the idea that the LcrV tip complex is required for YopB insertion and translocon formation. However, the YscFD28AD46A strain did secrete Yops in the presence of host cells, indicating that a translocation-competent tip complex is not required to sense contact with host cells to trigger Yop secretion. In conclusion, in the absence of cross-linkable LcrV-YscF interactions, translocon insertion is abolished, but Yersinia still retains the ability to sense cell contact.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Yersinia pseudotuberculosis/metabolism , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Biological Transport/genetics , Biological Transport/physiology , Cell Line , Host-Pathogen Interactions , Humans , Immunoprecipitation , Pore Forming Cytotoxic Proteins/genetics , Protein Binding/genetics , Protein Binding/physiology , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/physiology
4.
Am J Physiol Cell Physiol ; 304(8): C739-47, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23364266

ABSTRACT

This study investigated the molecular identity and impact of enhancing voltage-gated Na(+) (Na(V)) channels in the control of vascular tone. In rat isolated mesenteric and femoral arteries mounted for isometric tension recording, the vascular actions of the Na(V) channel activator veratridine were examined. Na(V) channel expression was probed by molecular techniques and immunocytochemistry. In mesenteric arteries, veratridine induced potent contractions (pEC(50) = 5.19 ± 0.20, E(max) = 12.0 ± 2.7 mN), which were inhibited by 1 µM TTX (a blocker of all Na(V) channel isoforms, except Na(V)1.5, Na(V)1.8, and Na(V)1.9), but not by selective blockers of Na(V)1.7 (ProTx-II, 10 nM) or Na(V)1.8 (A-80347, 1 µM) channels. The responses were insensitive to endothelium removal but were partly (~60%) reduced by chemical destruction of sympathetic nerves by 6-hydroxydopamine (2 mM) or antagonism at the α1-adrenoceptor by prazosin (1 µM). KB-R7943, a blocker of the reverse mode of the Na(+)/Ca(2+) exchanger (3 µM), inhibited veratridine contractions in the absence or presence of prazosin. T16A(inh)-A01, a Ca(2+)-activated Cl(-) channel blocker (10 µM), also inhibited the prazosin-resistant contraction to veratridine. Na(V) channel immunoreactivity was detected in freshly isolated mesenteric myocytes, with apparent colocalization with the Na(+)/Ca(2+) exchanger. Veratridine induced similar contractile effects in the femoral artery, and mRNA transcripts for Na(V)1.2 and Na(V)1.3 channels were evident in both vessel types. We conclude that, in addition to sympathetic nerves, NaV channels are expressed in vascular myocytes, where they are functionally coupled to the reverse mode of Na(+)/Ca(2+) exchanger and subsequent activation of Ca(2+)-activated Cl(-) channels, causing contraction. The TTX-sensitive Na(V)1.2 and Na(V)1.3 channels are likely involved in vascular control.


Subject(s)
NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Vasoconstriction/physiology , Animals , Male , Mesenteric Arteries/physiology , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Veratridine/pharmacology
5.
Br J Pharmacol ; 168(3): 773-84, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22946562

ABSTRACT

BACKGROUND AND PURPOSE: T16A(inh) -A01 is a recently identified inhibitor of the calcium-activated chloride channel TMEM16A. The aim of this study was to test the efficacy of T16A(inh) -A01 for inhibition of calcium-activated chloride channels in vascular smooth muscle and consequent effects on vascular tone. EXPERIMENTAL APPROACH: Single channel and whole cell patch clamp was performed on single smooth muscle cells from rabbit pulmonary artery and mouse thoracic aorta. Isometric tension studies were performed on mouse thoracic aorta and mesenteric artery as well as human abdominal visceral adipose artery. KEY RESULTS: In rabbit pulmonary artery myocytes T16A(inh) -A01 (1-30 µM) inhibited single calcium (Ca(2+) )-activated chloride (Cl(-) ) channels and whole cell currents activated by 500 nM free Ca(2+) . Similar effects were observed for single Ca(2+) -activated Cl(-) channels in mouse thoracic aorta, and in both cell types, channel activity was abolished by two antisera raised against TMEM16A but not by a bestrophin antibody. The TMEM16A potentiator, F(act) (10 µM), increased single channel and whole cell Ca(2+) -activated Cl(-) currents in rabbit pulmonary arteries. In isometric tension studies, T16A(inh) -A01 relaxed mouse thoracic aorta pre-contracted with methoxamine with an IC(50) of 1.6 µM and suppressed the methoxamine concentration-effect curve. T16A(inh) -A01 did not affect the maximal contraction produced by 60 mM KCl and the relaxant effect of 10 µM T16A(inh) -A01 was not altered by incubation of mouse thoracic aorta in a cocktail of potassium (K(+) ) channel blockers. T16A(inh) -A01 (10 µM) also relaxed human visceral adipose arteries by 88 ± 3%. CONCLUSIONS AND IMPLICATIONS: T16A(inh) -A01 blocks calcium-activated chloride channels in vascular smooth muscle cells and relaxes murine and human blood vessels.


Subject(s)
Chloride Channels/antagonists & inhibitors , Myocytes, Smooth Muscle/drug effects , Neoplasm Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Thiazoles/pharmacology , Vasodilator Agents/pharmacology , Adipose Tissue/blood supply , Adipose Tissue/physiology , Animals , Anoctamin-1 , Aorta, Thoracic/cytology , Aorta, Thoracic/physiology , Chloride Channels/physiology , Humans , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mice , Mice, Inbred BALB C , Myocytes, Smooth Muscle/physiology , Neoplasm Proteins/physiology , Pulmonary Artery/cytology , Pulmonary Artery/physiology , Rabbits
6.
Am J Physiol Cell Physiol ; 303(12): C1229-43, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23034390

ABSTRACT

Pulmonary artery smooth muscle cells (PASMCs) are more depolarized and display higher Ca(2+) levels in pulmonary hypertension (PH). Whether the functional properties and expression of Ca(2+)-activated Cl- channels (Cl(Ca)), an important excitatory mechanism in PASMCs, are altered in PH is unknown. The potential role of Cl(Ca) channels in PH was investigated using the monocrotaline (MCT)-induced PH model in the rat. Three weeks postinjection with a single dose of MCT (50 mg/kg ip), the animals developed right ventricular hypertrophy (heart weight measurements) and changes in pulmonary arterial flow (pulse-waved Doppler imaging) that were consistent with increased pulmonary arterial pressure and PH. Whole cell patch experiments revealed an increase in niflumic acid (NFA)-sensitive Ca(2+)-activated Cl(-) current [I(Cl(Ca))] density in PASMCs from large conduit and small intralobar pulmonary arteries of MCT-treated rats vs. aged-matched saline-injected controls. Quantitative RT-PCR and Western blot analysis revealed that the alterations in I(Cl(Ca)) were accompanied by parallel changes in the expression of TMEM16A, a gene recently shown to encode for Cl(Ca) channels. The contraction to serotonin of conduit and intralobar pulmonary arteries from MCT-treated rats exhibited greater sensitivity to nifedipine (1 µM), an l-type Ca(2+) channel blocker, and NFA (30 or 100 µM, with or without 10 µM indomethacin to inhibit cyclooxygenases) or T16A(Inh)-A01 (10 µM), TMEM16A/Cl(Ca) channel inhibitors, than that of control animals. In conclusion, augmented Cl(Ca)/TMEM16A channel activity is a major contributor to the changes in electromechanical coupling of PA in this model of PH. TMEM16A-encoded channels may therefore represent a novel therapeutic target in this disease.


Subject(s)
Chloride Channels/biosynthesis , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/physiopathology , Animals , Anoctamin-1 , Calcium Channel Blockers/pharmacology , Chloride Channel Agonists , Chloride Channels/antagonists & inhibitors , Chloride Channels/physiology , Cyclooxygenase Inhibitors/pharmacology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/drug therapy , Indomethacin/pharmacology , Male , Monocrotaline/toxicity , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Nifedipine/pharmacology , Niflumic Acid/pharmacology , Patch-Clamp Techniques , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pyrimidines/pharmacology , Rats , Rats, Wistar , Serotonin/pharmacology , Thiazoles/pharmacology
7.
Hypertension ; 59(4): 877-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22353613

ABSTRACT

KCNQ4-encoded voltage-dependent potassium (Kv7.4) channels are important regulators of vascular tone that are severely compromised in models of hypertension. However, there is no information as to the role of these channels in responses to endogenous vasodilators. We used a molecular knockdown strategy, as well as pharmacological tools, to examine the hypothesis that Kv7.4 channels contribute to ß-adrenoceptor-mediated vasodilation in the renal vasculature and underlie the vascular deficit in spontaneously hypertensive rats. Quantitative PCR and immunohistochemistry confirmed gene and protein expression of KCNQ1, KCNQ3, KCNQ4, KCNQ5, and Kv7.1, Kv7.4, and Kv7.5 in rat renal artery. Isoproterenol produced concentration-dependent relaxation of precontracted renal arteries and increased Kv7 channel currents in isolated smooth muscle cells. Application of the Kv7 blocker linopirdine attenuated isoproterenol-induced relaxation and current. Isoproterenol-induced relaxations were also reduced in arteries incubated with small interference RNAs targeted to KCNQ4 that produced a ≈60% decrease in Kv7.4 protein level. Relaxation to isoproterenol and the Kv7 activator S-1 were abolished in arteries from spontaneously hypertensive rats, which was associated with ≈60% decrease in Kv7.4 abundance. This study provides the first evidence that Kv7 channels contribute to ß-adrenoceptor-mediated vasodilation in the renal vasculature and that abrogation of Kv7.4 channels is strongly implicated in the impaired ß-adrenoceptor pathway in spontaneously hypertensive rats. These findings may provide a novel pathogenic link between arterial dysfunction and hypertension.


Subject(s)
Hypertension/physiopathology , KCNQ Potassium Channels/deficiency , Receptors, Adrenergic, beta/physiology , Renal Artery/physiology , Vasodilation/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Isoproterenol/pharmacology , KCNQ Potassium Channels/drug effects , KCNQ Potassium Channels/genetics , Male , RNA, Small Interfering/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Vasodilation/drug effects
8.
Br J Pharmacol ; 166(4): 1377-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22251082

ABSTRACT

BACKGROUND AND PURPOSE: The aim of this study was to characterize the functional impact of KCNQ1-encoded voltage-dependent potassium channels (K(v)7.1) in the vasculature. EXPERIMENTAL APPROACH: Mesenteric arteries, intrapulmonary arteries and thoracic aortae were isolated from adult rats. K(v)7.1 channel expression was established by fluorescence immunocytochemistry. Wire myography determined functionality of these channels in response to selective blockers and activators. Xenopus oocytes expressing K(v)7.1 channels were used to assess the effectiveness of selective K(v)7.1 channel blockers. KEY RESULTS: K(v)7.1 channels were identified in arterial myocytes by immunocytochemistry. K(v)7.1 blockers HMR1556, L-768,673 (10 µM) and JNJ39490282 (JNJ282; 1 µM) had no contractile effects in arteries, whereas the pan-K(v)7 channel blocker linopirdine (10 µM) evoked robust contractions. Application of two compounds purported to activate K(v)7.1 channels, L-364 373 (R-L3) and mefenamic acid, relaxed mesenteric arteries preconstricted by methoxamine. These responses were reversed by HMR1556 or L-768,673 but not JNJ282. Similar effects were observed in the thoracic aorta and intrapulmonary arteries. CONCLUSIONS AND IMPLICATIONS: In contrast to previous assumptions, K(v)7.1 channels expressed in arterial myocytes are functional ion channels. Although these channels do not appear to contribute to resting vascular tone, K(v)7.1 activators were effective vasorelaxants.


Subject(s)
Aorta, Thoracic/metabolism , KCNQ1 Potassium Channel/metabolism , Mesenteric Arteries/metabolism , Potassium Channel Blockers/pharmacology , Pulmonary Artery/metabolism , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Female , Humans , In Vitro Techniques , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/metabolism , KCNQ1 Potassium Channel/agonists , KCNQ1 Potassium Channel/antagonists & inhibitors , KCNQ1 Potassium Channel/genetics , Male , Membrane Potentials/drug effects , Mesenteric Arteries/cytology , Mesenteric Arteries/drug effects , Oocytes/drug effects , Oocytes/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Rats , Rats, Wistar , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Xenopus laevis
9.
Circulation ; 124(5): 602-11, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21747056

ABSTRACT

BACKGROUND: Voltage-gated potassium (K(+)) channels encoded by KCNQ genes (Kv7 channels) have been identified in various rodent and human blood vessels as key regulators of vascular tone; however, nothing is known about the functional impact of these channels in vascular disease. We ascertained the effect of 3 structurally different activators of Kv7.2 through Kv7.5 channels (BMS-204352, S-1, and retigabine) on blood vessels from normotensive and hypertensive animals. METHODS AND RESULTS: Precontracted thoracic aorta and mesenteric artery segments from normotensive rats were relaxed by all 3 Kv7 activators, with potencies of BMS-204352=S-1>retigabine. We also tested these agents in the coronary circulation using the Langendorff heart preparation. BMS-204352 and S-1 dose dependently increased coronary perfusion at concentrations between 0.1 and 10 µmol/L, whereas retigabine was effective at 1 to 10 µmol/L. In addition, S-1 increased K(+) currents in isolated mesenteric artery myocytes. The ability of these agents to relax precontracted vessels, increase coronary flow, or augment K(+) currents was impaired considerably in tissues isolated from spontaneously hypertensive rats (SHRs). Of the 5 KCNQ genes, only the expression of KCNQ4 was reduced (≈3.7 fold) in SHRs aorta. Kv7.4 protein levels were ≈50% lower in aortas and mesenteric arteries from spontaneously hypertensive rats compared with normotensive vessels. A similar attenuated response to S-1 and decreased Kv7.4 were observed in mesenteric arteries from mice made hypertensive by angiotensin II infusion compared with normotensive controls. CONCLUSIONS: In 2 different rat and mouse models of hypertension, the functional impact of Kv7 channels was dramatically downregulated.


Subject(s)
Hypertension/physiopathology , KCNQ Potassium Channels/physiology , Angiotensin II/pharmacology , Animals , Aorta, Thoracic/physiology , Blood Pressure/physiology , Carbamates/pharmacology , Down-Regulation/physiology , Hypertension/chemically induced , In Vitro Techniques , Indoles/pharmacology , KCNQ Potassium Channels/agonists , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mesenteric Arteries/physiology , Mice , Phenylenediamines/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar , Vasoconstrictor Agents/pharmacology
10.
Breast ; 20(3): 259-63, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21324695

ABSTRACT

PURPOSE: Current AJCC/UICC staging of early breast cancer defines tumor stage using the largest focus, adding the suffix "(m)" to indicate multiplicity. This method may underestimate the total tumor burden in multifocal and multicentric breast cancer (MMBC). This study examines other measures of tumor size in MMBC to determine which provides the best fit in a multivariate model for survival outcomes. PATIENTS AND METHODS: This prospective cohort study used data from the Australian Capital Territory and New South Wales Breast Cancer Treatment Group database to identify 812 women with ipsilateral invasive breast cancer; 141 of these women had MMBC. The pathology slides of all women with MMBC were reviewed and all foci of invasive breast cancer were re-measured. The measures of interest were the diameter of the largest deposit, the aggregate diameter and the aggregate volume. These measures of tumor size were included with other clinicopathological features of MMBC in a multivariate analysis to assess their relationship with progression-free survival (PFS) and overall survival (OS). RESULTS: Tumor size was associated with PFS and OS in MMBC using any of the three measures; however, the diameter of the largest deposit provided the best fit in the multivariate model for OS. CONCLUSION: Tumor size is an important prognostic factor for MMBC, and the diameter of the largest deposit provides a better fit in a multivariate model for OS than aggregate diameter and aggregate volume. Therefore, tumor size in MMBC should continue to be measured using the diameter of the largest deposit.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/mortality , Disease Progression , Female , Humans , Middle Aged , Multivariate Analysis , Neoplasm Staging , Prognosis , Prospective Studies , Survival Analysis , Tumor Burden
11.
Br J Pharmacol ; 162(1): 42-53, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20840535

ABSTRACT

BACKGROUND AND PURPOSE: KCNQ-encoded voltage-gated potassium channels (K(v) 7) have recently been identified as important anti-constrictor elements in rodent blood vessels but the role of these channels and the effects of their modulation in human arteries remain unknown. Here, we have assessed KCNQ gene expression and function in human arteries ex vivo. EXPERIMENTAL APPROACH: Fifty arteries (41 from visceral adipose tissue, 9 mesenteric arteries) were obtained from subjects undergoing elective surgery. Quantitative RT-PCR experiments using primers specific for all known KCNQ genes and immunohistochemsitry were used to show K(v) 7 channel expression. Wire myography and single cell electrophysiology assessed the function of these channels. KEY RESULTS: KCNQ4 was expressed in all arteries assessed, with variable contributions from KCNQ1, 3 and 5. KCNQ2 was not detected. K(v) 7 channel isoform-dependent staining was revealed in the smooth muscle layer. In functional studies, the K(v) 7 channel blockers, XE991 and linopirdine increased isometric tension and inhibited K(+) currents. In contrast, the K(v) 7.1-specific blocker chromanol 293B did not affect vascular tone. Two K(v) 7 channel activators, retigabine and acrylamide S-1, relaxed preconstricted arteries, actions reversed by XE991. K(v) 7 channel activators also suppressed spontaneous contractile activity in seven arteries, reversible by XE991. CONCLUSIONS AND IMPLICATIONS: This is the first study to demonstrate not only the presence of KCNQ gene products in human arteries but also their contribution to vascular tone ex vivo. LINKED ARTICLE: This article is commented on by Mani and Byron, pp. 38-41 of this issue. To view this commentary visit http://dx.doi.org/10.1111/j.1476-5381.2010.01065.x.


Subject(s)
Arteries/metabolism , KCNQ Potassium Channels/genetics , Aged , Female , Humans , Immunohistochemistry , In Vitro Techniques , KCNQ Potassium Channels/drug effects , Male , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/genetics
12.
Am J Physiol Cell Physiol ; 299(5): C948-59, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20686072

ABSTRACT

Recently, overexpression of the genes TMEM16A and TMEM16B has been shown to produce currents qualitatively similar to native Ca(2+)-activated Cl(-) currents (I(ClCa)) in vascular smooth muscle. However, there is no information about this new gene family in vascular smooth muscle, where Cl(-) channels are a major depolarizing mechanism. Qualitatively similar Cl(-) currents were evoked by a pipette solution containing 500 nM Ca(2+) in smooth muscle cells isolated from BALB/c mouse portal vein, thoracic aorta, and carotid artery. Quantitative PCR using SYBR Green chemistry and primers specific for transmembrane protein (TMEM) 16A or the closely related TMEM16B showed TMEM16A expression as follows: portal vein > thoracic aorta > carotid artery > brain. In addition, several alternatively spliced variant transcripts of TMEM16A were detected. In contrast, TMEM16B expression was very low in smooth muscle. Western blot analysis with different antibodies directed against TMEM16A revealed a number of products with a consistent band at ∼120 kDa, except portal vein, where an 80-kDa band predominated. TMEM16A protein was identified in the smooth muscle layers of 4-µm-thick slices of portal vein, thoracic aorta, and carotid artery. In isolated myocytes, fluorescence specific to a TMEM16A antibody was detected diffusely throughout the cytoplasm, as well as near the membrane. The same antibody used in Western blot analysis of lysates from vascular tissues also recognized an ∼147-kDa mouse TMEM16A-green fluorescent protein (GFP) fusion protein expressed in HEK 293 cells, which correlated to a similar band detected by a GFP antibody. Patch-clamp experiments revealed that I(ClCa) generated by transfection of TMEM16A-GFP in HEK 293 cells displayed remarkable similarities to I(ClCa) recorded in vascular myocytes, including slow kinetics, steep outward rectification, and a response similar to the pharmacological agent niflumic acid. This study shows that TMEM16A expression is robust in murine vascular smooth muscle cells, consolidating the view that this gene is a viable candidate for the native Ca(2+)-activated Cl(-) channel in this cell type.


Subject(s)
Chloride Channels/metabolism , Myocytes, Smooth Muscle/physiology , Protein Biosynthesis , Protein Isoforms/metabolism , Alternative Splicing , Animals , Anoctamin-1 , Cell Line , Chloride Channels/genetics , Gene Expression Profiling , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Myocytes, Smooth Muscle/cytology , Patch-Clamp Techniques , Protein Isoforms/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tissue Distribution
13.
Antimicrob Agents Chemother ; 54(8): 3241-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20498321

ABSTRACT

Type three secretion systems (TTSSs) are virulence factors found in many pathogenic Gram-negative species, including the family of pathogenic Yersinia spp. Yersinia pseudotuberculosis requires the translocation of a group of effector molecules, called Yops, to subvert the innate immune response and establish infection. Polarized transfer of Yops from bacteria to immune cells depends on several factors, including the presence of a functional TTSS, the successful attachment of Yersinia to the target cell, and translocon insertion into the target cell membrane. Here we employed a high-throughput screen to identify small molecules that block translocation of Yops into mammalian cells. We identified 6 compounds that inhibited translocation of effectors without affecting synthesis of TTSS components and secreted effectors, assembly of the TTSS, or secretion of effectors. One compound, C20, reduced adherence of Y. pseudotuberculosis to target cells. Additionally, the compounds caused leakage of Yops into the supernatant during infection and thus reduced polarized translocation. Furthermore, several molecules, namely, C20, C22, C24, C34, and C38, also inhibited ExoS-mediated cell rounding, suggesting that the compounds target factors that are conserved between Pseudomonas aeruginosa and Y. pseudotuberculosis. In summary, we have identified 6 compounds that specifically inhibit translocation of Yops into mammalian cells but not Yop synthesis or secretion.


Subject(s)
Bacterial Outer Membrane Proteins/antagonists & inhibitors , High-Throughput Screening Assays/methods , Protein Transport/drug effects , Small Molecule Libraries , Yersinia pseudotuberculosis/drug effects , Bacterial Adhesion/drug effects , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Cell Line , Culture Media, Conditioned , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression Regulation, Bacterial , Humans , Molecular Weight , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Yersinia pseudotuberculosis/metabolism , Yersinia pseudotuberculosis/physiology , beta-Lactamase Inhibitors , beta-Lactamases/genetics , beta-Lactamases/metabolism
14.
Mol Microbiol ; 76(1): 236-59, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20199604

ABSTRACT

Yersinia pseudotuberculosis uses a type III secretion system (T3SS) to deliver effectors into host cells. A key component of the T3SS is the needle, which is a hollow tube on the bacterial surface through which effectors are secreted, composed of the YscF protein. To study needle assembly, we performed a screen for dominant-negative yscF alleles that prevented effector secretion in the presence of wild-type (WT) YscF. One allele, yscF-L54V, prevents WT YscF secretion and needle assembly, although purified YscF-L54V polymerizes in vitro. YscF-L54V binds to its chaperones YscE and YscG, and the YscF-L54V-EG complex targets to the T3SS ATPase, YscN. We propose that YscF-L54V stalls at a binding site in the needle assembly pathway following its release from the chaperones, which blocks the secretion of WT YscF and other early substrates required for building a needle. Interestingly, YscF-L54V does not affect the activity of pre-assembled actively secreting machines, indicating that a factor and/or binding site required for YscF secretion is absent from T3SS machines already engaged in effector secretion. Thus, substrate switching may involve the removal of an early substrate-specific binding site as a mechanism to exclude early substrates from Yop-secreting machines.


Subject(s)
Bacterial Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation, Missense , Virulence Factors/metabolism , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/metabolism , Amino Acid Substitution/genetics , Macromolecular Substances/metabolism , Membrane Proteins/metabolism , Models, Molecular , Molecular Chaperones/metabolism , Protein Multimerization , Protein Structure, Tertiary
15.
Cardiovasc Res ; 87(3): 476-84, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20172862

ABSTRACT

AIMS: Calcium-activated chloride channels (CACCs) share common pharmacological properties with Kcnma1-encoded large conductance K(+) channels (BK(Ca) or K(Ca)1.1) and it has been suggested that they may co-exist in a macromolecular complex. As K(Ca)1.1 channels are known to localize to cholesterol and caveolin-rich lipid rafts (caveolae), the present study investigated whether Ca(2+)-sensitive Cl(-) currents in vascular myocytes were affected by the cholesterol depleting agent methyl-beta-cyclodextrin (M-betaCD). METHODS AND RESULTS: Calcium-activated chloride and potassium currents were recorded from single murine portal vein myocytes in whole cell voltage clamp. Western blot was undertaken following sucrose gradient ultracentrifugation using protein lysates from whole portal veins. Ca(2+)-activated Cl(-) currents were augmented by 3 mg mL(-1) M-betaCD with a rapid time course (t(0.5) = 1.8 min). M-betaCD had no effect on the bi-modal response to niflumic acid or anthracene-9-carboxylate but completely removed the inhibitory effects of the K(Ca)1.1 blockers, paxilline and tamoxifen, as well as the stimulatory effect of the K(Ca)1.1 activator NS1619. Discontinuous sucrose density gradients followed by western blot analysis revealed that the position of lipid raft markers caveolin and flotillin-2 was altered by 15 min application of 3 mg mL(-1) M-betaCD. The position of K(Ca)1.1 and the newly identified candidate for CACCs, TMEM16A, was also affected by M-betaCD. CONCLUSION: These data reveal that CACC properties are influenced by lipid raft integrity.


Subject(s)
Chloride Channels/metabolism , Cholesterol/deficiency , Membrane Microdomains/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Anoctamin-1 , Blotting, Western , Caveolins/metabolism , Centrifugation, Density Gradient , Chloride Channels/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Membrane Microdomains/drug effects , Membrane Potentials , Membrane Proteins/metabolism , Membrane Transport Modulators/pharmacology , Mice , Mice, Inbred BALB C , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neoplasm Proteins/metabolism , Patch-Clamp Techniques , Portal Vein/metabolism , Time Factors , beta-Cyclodextrins/pharmacology
16.
J Bacteriol ; 189(1): 83-97, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17071752

ABSTRACT

The trafficking of effectors, termed Yops, from Yersinia spp. into host cells is a multistep process that requires the type III secretion system (TTSS). The TTSS has three main structural parts: a base, a needle, and a translocon, which work together to ensure the polarized movement of Yops directly from the bacterial cytosol into the host cell cytosol. To understand the interactions that take place at the interface between the tip of the TTSS needle and the translocon, we developed a screen to identify mutations in the needle protein YscF that separated its function in secretion from its role in translocation. We identified 25 translocation-defective (TD) yscF mutants, which fall into five phenotypic classes. Some classes exhibit aberrant needle structure and/or reduced levels of Yop secretion, consistent with known functions for YscF. Strikingly, two yscF TD classes formed needles and secreted Yops normally but displayed distinct translocation defects. Class I yscF TD mutants showed diminished pore formation, suggesting incomplete pore insertion and/or assembly. Class II yscF TD mutants formed pores but showed nonpolar translocation, suggesting unstable needle-translocon interactions. These results indicate that YscF functions in Yop secretion and translocation can be genetically separated. Furthermore, the identification of YscF residues that are required for the assembly of the translocon and/or productive interactions with the translocon has allowed us to initiate the mapping of the needle-translocon interface.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Yersinia pseudotuberculosis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biological Transport , Cell Line, Tumor , Humans , Models, Molecular , Mutation , Yersinia pseudotuberculosis Infections/microbiology
17.
Mol Biol Cell ; 18(2): 475-86, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17122363

ABSTRACT

The import of polytopic membrane proteins into the mitochondrial inner membrane (IM) is facilitated by Tim9p/Tim10p and Tim8p/Tim13p protein complexes in the intermembrane space (IMS). These complexes are proposed to act as chaperones by transporting the hydrophobic IM proteins through the aqueous IMS and preventing their aggregation. To examine the nature of this interaction, Tim23p molecules containing a single photoreactive cross-linking probe were imported into mitochondria in the absence of an IM potential where they associated with small Tim complexes in the IMS. On photolysis and immunoprecipitation, a probe located at a particular Tim23p site (27 different locations were examined) was found to react covalently with, in most cases, only one of the small Tim proteins. Tim8p, Tim9p, Tim10p, and Tim13p were therefore positioned adjacent to specific sites in the Tim23p substrate before its integration into the IM. This specificity of binding to Tim23p strongly suggests that small Tim proteins do not function solely as general chaperones by minimizing the exposure of nonpolar Tim23p surfaces to the aqueous medium, but may also align a folded Tim23p substrate in the proper orientation for delivery and integration into the IM at the TIM22 translocon.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae/metabolism , Binding Sites , Membrane Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/metabolism , Protein Conformation , Protein Transport , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
18.
Invest New Drugs ; 21(1): 85-97, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12795533

ABSTRACT

PURPOSE: MG98 is a second generation phosphorothioate antisense oligodeoxynucleotide which is a highly specific inhibitor of translation of the mRNA for human DNA MeTase I (DNMT 1). This phase I study examined the toxicity and pharmacologic profile of MG98 administered as a continuous 21-day intravenous infusion every 4 weeks. PATIENTS AND METHODS: Fourteen patients with solid cancers received a total of 25 cycles of MG98 at doses ranging from 40 to 240 mg/m2/day. Steady-state concentrations of MG98 were measured as were several pharmacodynamic assessments including mRNA of the target gene, DNMT1, in PBMC. In addition, other potential surrogate markers of drug effects were explored, including hemoglobin F, Vimentin and GADD45. RESULTS: Dose limiting effects were drug-related reversible transaminase elevation and fatigue seen at doses of 240, 200 and 160 mg/m2/day. The dose level of 80 mg/m2/day was felt to be safe and tolerable when delivered on this schedule. No evidence of antitumor activity was observed. Although pharmacokinetic analysis revealed that at the higher dose levels, mean Css values of MG98 were approximately 10-fold times the IC50 values associated with target inhibition in vitro, the extent of MG98 penetration into target tumors in this trial was not determined. No consistent, dose-related changes in correlative markers including DNMT1 mRNA, hemoglobin F, Vimentin and GADD45, were observed. CONCLUSIONS: This schedule of MG98 given as a 21-day continuous intravenous infusion every 4 weeks was poorly tolerated in the highest doses; therefore, further disease-site specific evaluation of the efficacy of this agent will utilize a more favorable, intermittent dosing schedule. Pharmacodynamic evaluations undertaken in an attempt to explore and validate the biological mechanisms of MG98 did not show dose-related effects.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Neoplasms/drug therapy , Oligodeoxyribonucleotides/pharmacology , Oligonucleotides, Antisense/pharmacology , Adult , Aged , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fetal Hemoglobin/analysis , Humans , Infusions, Intravenous , Intracellular Signaling Peptides and Proteins , Male , Maximum Tolerated Dose , Middle Aged , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/blood , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/blood , Proteins/analysis , Proteins/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/blood , Thionucleotides , Time Factors , Treatment Outcome , Up-Regulation , Vimentin/blood , Vimentin/genetics , GADD45 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL