Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Virus Res ; 341: 199322, 2024 03.
Article in English | MEDLINE | ID: mdl-38228190

ABSTRACT

The emergence of highly infectious pathogens with their potential for triggering global pandemics necessitate the development of effective treatment strategies, including broad-spectrum antiviral therapies to safeguard human health. This study investigates the antiviral activity of emetine, dehydroemetine (DHE), and congeneric compounds against SARS-CoV-2 and HCoV-OC43, and evaluates their impact on the host cell. Concurrently, we assess the potential cardiotoxicity of these ipecac alkaloids. Significantly, our data reveal that emetine and the (-)-R,S isomer of 2,3-dehydroemetine (designated in this paper as DHE4) reduce viral growth at nanomolar concentrations (i.e., IC50 ∼ 50-100 nM), paralleling those required for inhibition of protein synthesis, while calcium channel blocking activity occurs at elevated concentrations (i.e., IC50 ∼ 40-60 µM). Our findings suggest that the antiviral mechanisms primarily involve disruption of host cell protein synthesis and is demonstrably stereoisomer specific. The prospect of a therapeutic window in which emetine or DHE4 inhibit viral propagation without cardiotoxicity renders these alkaloids viable candidates in strategies worthy of clinical investigation.


Subject(s)
Alkaloids , Emetine , Emetine/analogs & derivatives , Humans , Emetine/pharmacology , Ipecac/pharmacology , Cardiotoxicity , Antiviral Agents/toxicity
3.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33990467

ABSTRACT

Cardiac arrhythmias are the most common cause of sudden cardiac death worldwide. Lengthening the ventricular action potential duration (APD), either congenitally or via pathologic or pharmacologic means, predisposes to a life-threatening ventricular arrhythmia, Torsade de Pointes. IKs (KCNQ1+KCNE1), a slowly activating K+ current, plays a role in action potential repolarization. In this study, we screened a chemical library in silico by docking compounds to the voltage-sensing domain (VSD) of the IKs channel. Here, we show that C28 specifically shifted IKs VSD activation in ventricle to more negative voltages and reversed the drug-induced lengthening of APD. At the same dosage, C28 did not cause significant changes of the normal APD in either ventricle or atrium. This study provides evidence in support of a computational prediction of IKs VSD activation as a potential therapeutic approach for all forms of APD prolongation. This outcome could expand the therapeutic efficacy of a myriad of currently approved drugs that may trigger arrhythmias.


Subject(s)
Action Potentials/drug effects , KCNQ1 Potassium Channel/genetics , Myocytes, Cardiac/metabolism , Small Molecule Libraries/pharmacology , Action Potentials/physiology , Amino Acid Substitution , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Calcium/metabolism , Dogs , Furans/pharmacology , Gene Expression , Guinea Pigs , Heart Atria/cytology , Heart Atria/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Humans , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/metabolism , Moxifloxacin/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Phenethylamines/pharmacology , Potassium/metabolism , Primary Cell Culture , Pyridines/pharmacology , Pyrimidines/pharmacology , Sodium/metabolism , Sulfonamides/pharmacology , Transgenes , Xenopus laevis
4.
Front Physiol ; 11: 587040, 2020.
Article in English | MEDLINE | ID: mdl-33240105

ABSTRACT

It has long been known that heart rate is regulated by the autonomic nervous system. Recently, we demonstrated that the pacemaker current, I f , is regulated by phosphoinositide 3-kinase (PI3K) signaling independently of the autonomic nervous system. Inhibition of PI3K in sinus node (SN) myocytes shifts the activation of I f by almost 16 mV in the negative direction. I f in the SN is predominantly mediated by two members of the HCN gene family, HCN4 and HCN1. Purkinje fibers also possess I f and are an important secondary pacemaker in the heart. In contrast to the SN, they express HCN2 and HCN4, while ventricular myocytes, which do not normally pace, express HCN2 alone. In the current work, we investigated PI3K regulation of HCN2 expressed in HEK293 cells. Treatment with the PI3K inhibitor PI-103 caused a negative shift in the activation voltage and a dramatic reduction in the magnitude of the HCN2 current. Similar changes were also seen in cells treated with an inhibitor of the protein kinase Akt, a downstream effector of PI3K. The effects of PI-103 were reversed by perfusion of cells with phosphatidylinositol 3,4,5-trisphosphate (the second messenger produced by PI3K) or active Akt protein. We identified serine 861 in mouse HCN2 as a putative Akt phosphorylation site. Mutation of S861 to alanine mimicked the effects of Akt inhibition on voltage dependence and current magnitude. In addition, the Akt inhibitor had no effect on the mutant channel. These results suggest that Akt phosphorylation of mHCN2 S861 accounts for virtually all of the observed actions of PI3K signaling on the HCN2 current. Unexpectedly, Akt inhibition had no effect on I f in SN myocytes. This result raises the possibility that diverse PI3K signaling pathways differentially regulate HCN-induced currents in different tissues, depending on the isoforms expressed.

5.
Commun Biol ; 3(1): 385, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32678288

ABSTRACT

KCNQ family K+ channels (KCNQ1-5) in the heart, nerve, epithelium and ear require phosphatidylinositol 4,5-bisphosphate (PIP2) for voltage dependent activation. While membrane lipids are known to regulate voltage sensor domain (VSD) activation and pore opening in voltage dependent gating, PIP2 was found to interact with KCNQ1 and mediate VSD-pore coupling. Here, we show that a compound CP1, identified in silico based on the structures of both KCNQ1 and PIP2, can substitute for PIP2 to mediate VSD-pore coupling. Both PIP2 and CP1 interact with residues amongst a cluster of amino acids critical for VSD-pore coupling. CP1 alters KCNQ channel function due to different interactions with KCNQ compared with PIP2. We also found that CP1 returned drug-induced action potential prolongation in ventricular myocytes to normal durations. These results reveal the structural basis of PIP2 regulation of KCNQ channels and indicate a potential approach for the development of anti-arrhythmic therapy.


Subject(s)
KCNQ Potassium Channels/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Action Potentials , Animals , Computer Simulation , Guinea Pigs , KCNQ Potassium Channels/chemistry , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism , Oocytes , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/analogs & derivatives , Phosphatidylinositol 4,5-Diphosphate/chemistry , Protein Structure, Tertiary , Xenopus laevis
6.
J Invest Dermatol ; 140(5): 1035-1044.e7, 2020 05.
Article in English | MEDLINE | ID: mdl-31705875

ABSTRACT

Keratitis-ichthyosis-deafness (KID) syndrome is a severe, untreatable condition characterized by ocular, auditory, and cutaneous abnormalities, with major complications of infection and skin cancer. Most cases of KID syndrome (86%) are caused by a heterozygous missense mutation (c.148G>A, p.D50N) in the GJB2 gene, encoding gap junction protein Cx26, which alters gating properties of Cx26 channels in a dominant manner. We hypothesized that a mutant allele-specific small interfering RNA could rescue the cellular phenotype in patient keratinocytes (KCs). A KID syndrome cell line (KID-KC) was established from primary patient KCs with a heterozygous p.D50N mutation. This cell line displayed impaired gap junction communication and hyperactive hemichannels, confirmed by dye transfer, patch clamp, and neurobiotin uptake assays. A human-murine chimeric skin graft model constructed with KID-KCs mimicked patient skin in vivo, further confirming the validity of these cells as a model. In vitro treatment with allele-specific small interfering RNA led to robust inhibition of the mutant GJB2 allele without altering expression of the wild-type allele. This corrected both gap junction and hemichannel activity. Notably, allele-specific small interfering RNA treatment caused only low-level off-target effects in KID-KCs, as detected by genome-wide RNA sequencing. Our data provide an important proof-of-concept and model system for the potential use of allele-specific small interfering RNA in treating KID syndrome and other dominant genetic conditions.


Subject(s)
Connexins/genetics , Keratinocytes/physiology , Keratitis/genetics , Mutation, Missense/genetics , RNA, Small Interfering/genetics , Skin/metabolism , Alleles , Animals , Cell Line , Chimera , Connexin 26 , Gap Junctions/metabolism , Heterografts , Heterozygote , Humans , Keratitis/therapy , Membrane Potentials , Mice , Skin/pathology , Skin Transplantation
7.
J Gen Physiol ; 151(8): 1051-1058, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31217223

ABSTRACT

Heart rate in physiological conditions is set by the sinoatrial node (SN), the primary cardiac pacing tissue. Phosphoinositide 3-kinase (PI3K) signaling is a major regulatory pathway in all normal cells, and its dysregulation is prominent in diabetes, cancer, and heart failure. Here, we show that inhibition of PI3K slows the pacing rate of the SN in situ and in vitro and reduces the early slope of diastolic depolarization. Furthermore, inhibition of PI3K causes a negative shift in the voltage dependence of activation of the pacemaker current, I F, while addition of its second messenger, phosphatidylinositol 3,4,5-trisphosphate, induces a positive shift. These shifts in the activation of I F are independent of, and larger than, those induced by the autonomic nervous system. These results suggest that PI3K is an important regulator of heart rate, and perturbations in this signaling pathway may contribute to the development of arrhythmias.


Subject(s)
Heart Rate , Phosphatidylinositol 3-Kinases/metabolism , Second Messenger Systems , Sinoatrial Node/physiology , Action Potentials , Animals , Biological Clocks , Cells, Cultured , Dogs , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositol Phosphates/metabolism , Rabbits , Sinoatrial Node/metabolism
8.
J Nanobiotechnology ; 14: 34, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27102228

ABSTRACT

BACKGROUND: Titanium dioxide (TiO2) is one of the most common nanoparticles found in industry ranging from food additives to energy generation. Approximately four million tons of TiO2 particles are produced worldwide each year with approximately 3000 tons being produced in nanoparticulate form, hence exposure to these particles is almost certain. RESULTS: Even though TiO2 is also used as an anti-bacterial agent in combination with UV, we have found that, in the absence of UV, exposure of HeLa cells to TiO2 nanoparticles significantly increased their risk of bacterial invasion. HeLa cells cultured with 0.1 mg/ml rutile and anatase TiO2 nanoparticles for 24 h prior to exposure to bacteria had 350 and 250 % respectively more bacteria per cell. The increase was attributed to bacterial polysaccharides absorption on TiO2 NPs, increased extracellular LDH, and changes in the mechanical response of the cell membrane. On the other hand, macrophages exposed to TiO2 particles ingested 40 % fewer bacteria, further increasing the risk of infection. CONCLUSIONS: In combination, these two factors raise serious concerns regarding the impact of exposure to TiO2 nanoparticles on the ability of organisms to resist bacterial infection.


Subject(s)
Metal Nanoparticles/adverse effects , Staphylococcal Infections/chemically induced , Staphylococcus aureus/drug effects , Titanium/adverse effects , Anti-Bacterial Agents/adverse effects , Cell Line, Tumor , Cell Survival/drug effects , HeLa Cells , Humans , Particle Size
9.
FEBS Lett ; 589(12): 1340-5, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25935417

ABSTRACT

Gap junction channels can modify their activity in response to cell signaling pathways. Here, we demonstrate that Connexin50 (Cx50) coupling, but not Connexin46 (Cx46), increased when co-expressed with a constitutively active p110α subunit of PI3K in Xenopus oocytes. In addition, inhibition of PI3K signaling by blocking p110α, or Akt, significantly decreased gap junctional conductance in Cx50 transfected HeLa cells, with no effect on Cx46. Alterations in coupling levels were not a result of reduced Cx50 unitary conductance, suggesting that changes in the number of active channels were responsible. These data indicate that Cx50 is specifically regulated by the PI3K signaling pathway.


Subject(s)
Connexins/metabolism , Gap Junctions/metabolism , Oocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Second Messenger Systems , Xenopus Proteins/metabolism , Animals , Connexins/antagonists & inhibitors , Connexins/genetics , Enzyme Inhibitors/pharmacology , Female , Gap Junctions/drug effects , Gene Silencing , HeLa Cells , Humans , Membrane Potentials/drug effects , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Oocytes/drug effects , Oocytes/enzymology , Patch-Clamp Techniques , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Complementary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Second Messenger Systems/drug effects , Single-Cell Analysis , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics , Xenopus laevis
10.
Front Pharmacol ; 6: 9, 2015.
Article in English | MEDLINE | ID: mdl-25691868

ABSTRACT

4-phenylbutyrate (4-PB) has been shown to increase the protein content in a number of cells types. One such protein is Connexin43 (Cx43). We show here that 4-phenylbutyrate exposure results in significantly elevated cell to cell coupling, as determined by dual whole cell patch clamp. Incubation with 5 mM 4PB for 24 h or more nearly doubles junctional conductance. Interestingly, mRNA levels for Cx43 declined with exposure to 4-PB while western blot analysis revealed not significant change in protein levels. These data are most consistent with stabilization of the existing Cx43 pool or alterations in the number of functional channels within an existing pool of active and silent channels. These data represent a baseline for testing the efficacy of increased connexin mediated coupling in a variety of multicellular functions including erectile function.

11.
J Invest Dermatol ; 135(4): 1033-1042, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25229253

ABSTRACT

Keratitis-ichthyosis-deafness (KID) syndrome is an ectodermal dysplasia caused by dominant mutations of connexin26 (Cx26). Loss of Cx26 function causes nonsyndromic sensorineural deafness, without consequence in the epidermis. Functional analyses have revealed that a majority of KID-causing mutations confer a novel expansion of hemichannel activity, mediated by connexin channels in a nonjunctional configuration. Inappropriate Cx26 hemichannel opening is hypothesized to compromise keratinocyte integrity and epidermal homeostasis. Pharmacological modulators of Cx26 are needed to assess the pathomechanistic involvement of hemichannels in the development of hyperkeratosis in KID syndrome. We have used electrophysiological assays to evaluate small-molecule analogs of quinine for suppressive effects on aberrant hemichannel currents elicited by KID mutations. Here, we show that mefloquine (MFQ) inhibits several mutant hemichannel forms implicated in KID syndrome when expressed in Xenopus laevis oocytes (IC50∼16 µM), using an extracellular divalent cation, zinc (Zn(++)), as a nonspecific positive control for comparison (IC50∼3 µM). Furthermore, we used freshly isolated transgenic keratinocytes to show that micromolar concentrations of MFQ attenuated increased macroscopic membrane currents in primary mouse keratinocytes expressing human Cx26-G45E, a mutation that causes a lethal form of KID syndrome.


Subject(s)
Connexins/genetics , Mefloquine/pharmacology , Animals , Cations , Connexin 26 , Connexins/metabolism , Electrophysiology , Epidermis/metabolism , Homeostasis , Humans , Inhibitory Concentration 50 , Keratinocytes/cytology , Keratitis/metabolism , Mice , Mice, Transgenic , Mutation , Oocytes/cytology , Patch-Clamp Techniques , Xenopus , Xenopus laevis , Zinc/chemistry
12.
Am J Physiol Cell Physiol ; 304(12): C1150-8, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23447037

ABSTRACT

Mutations in the human gene encoding connexin 26 (Cx26 or GJB2) cause either nonsyndromic deafness or syndromic deafness associated with skin diseases. That distinct clinical disorders can be caused by different mutations within the same gene suggests that different channel activities influence the ear and skin. Here we use three different expression systems to examine the functional characteristics of two Cx26 mutations causing either mild (Cx26-D50A) or lethal (Cx26-A88V) keratitis-ichthyosis-deafness (KID) syndrome. In either cRNA-injected Xenopus oocytes, transfected HeLa cells, or transfected primary human keratinocytes, we show that both Cx26-D50A and Cx26-A88V form active hemichannels that significantly increase membrane current flow compared with wild-type Cx26. This increased membrane current accelerated cell death in low extracellular calcium solutions and was not due to increased mutant protein expression. Elevated mutant hemichannel currents could be blocked by increased extracellular calcium concentration. These results show that these two mutations exhibit a shared gain of functional activity and support the hypothesis that increased hemichannel activity is a common feature of human Cx26 mutations responsible for KID syndrome.


Subject(s)
Connexins/genetics , Deafness/genetics , Ichthyosis/genetics , Keratitis/genetics , Mutation/genetics , Animals , Connexin 26 , Deafness/metabolism , Deafness/pathology , Female , HeLa Cells , Humans , Ichthyosis/metabolism , Ichthyosis/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Keratitis/metabolism , Keratitis/pathology , Xenopus laevis
13.
BJU Int ; 110(8 Pt B): E402-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22520450

ABSTRACT

OBJECTIVES: To examine the effect of partial urethral obstruction (PUO) on bladder smooth muscle outward potassium current and the contribution of the large-conductance calcium-activated potassium (Maxi-K, BKCa) channel to this activity in smooth muscle cells isolated from bladders of sham-operated and PUO male rats using whole-cell patch clamp recording techniques. To determine the effect of PUO on the expression of the Maxi-K channel α and ß1 subunits and in vitro detrusor contractility. MATERIALS AND METHODS: Twenty adult male Sprague-Dawley rats were divided equally into two groups and subjected to surgical ligation of the urethra (PUO) or sham surgery (SHAM). After 2 weeks, the detrusors from PUO and SHAM rats were used for molecular analyses (mRNA and protein quantification of Maxi-K subunits) or organ bath contractility studies, or myocytes were isolated for conventional whole-cell patch clamp analyses. RESULTS: PUO increased bladder mass 2.5-fold and detrusor strips exhibited a more tonic-type contraction and increased contractility compared with controls (SHAM). Iberiotoxin (300 nM) sensitive Maxi-K channel current comprised about 40% of the outward whole-cell current in SHAM bladders but only about 8% in PUO bladders. Expression of the α subunit of the Maxi-K channel was significantly decreased ~40% while the expression of the ß1 subunit was increased ~2-fold at the mRNA level. The increase in ß1 expression was confirmed by Western blotting. CONCLUSIONS: Our findings show that obstruction of the rat bladder is associated with decreased Maxi-K channel activity of bladder smooth muscle cells, determined via direct current measurement. Increased expression of the ß1 subunit points to a compensatory reaction to decreased Maxi-K channel activity. Maxi-K channel openers or gene therapy may therefore provide therapeutic benefit for the overactive bladder.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/physiology , Myocytes, Smooth Muscle/physiology , Urethral Obstruction/physiopathology , Urinary Bladder/physiopathology , Animals , Cells, Cultured , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
14.
J Cardiovasc Pharmacol ; 60(1): 88-99, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22526298

ABSTRACT

The voltage-gated Na+ channel is a critical determinant of the action potential (AP) upstroke. Increasing Na+ conductance may speed AP propagation. In this study, we propose use of the skeletal muscle Na+ channel SkM1 as a more favorable gene than the cardiac isoform SCN5A to enhance conduction velocity in depolarized cardiac tissue. We used cells that electrically coupled with cardiac myocytes as a delivery platform to introduce the Na+ channels. Human embryonic kidney 293 cells were stably transfected with SkM1 or SCN5A. SkM1 had a more depolarized (18 mV shift) inactivation curve than SCN5A. We also found that SkM1 recovered faster from inactivation than SCN5A. When coupled with SkM1 expressing cells, cultured myocytes showed an increase in the dV/dtmax of the AP. Expression of SCN5A had no such effect. In an in vitro cardiac syncytium, coculture of neonatal cardiac myocytes with SkM1 expressing but not SCN5A expressing cells significantly increased the conduction velocity under both normal and depolarized conditions. In an in vitro reentry model induced by high-frequency stimulation, expression of SkM1 also enhanced angular velocity of the induced reentry. These results suggest that cells carrying a Na+ channel with a more depolarized inactivation curve can improve cardiac excitability and conduction in depolarized tissues.


Subject(s)
Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , NAV1.4 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Action Potentials , Animals , Animals, Newborn , Cell- and Tissue-Based Therapy/methods , Dogs , Female , Genetic Therapy/methods , HEK293 Cells , Heart Conduction System/metabolism , Humans , Male , NAV1.4 Voltage-Gated Sodium Channel/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Transfection
15.
Mol Biol Cell ; 22(24): 4776-86, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22031297

ABSTRACT

Mutations in the GJB2 gene (Cx26) cause deafness in humans. Most are loss-of-function mutations and cause nonsyndromic deafness. Some mutations produce a gain of function and cause syndromic deafness associated with skin disorders, such as keratitis-ichthyosis-deafness syndrome (KIDS). Cx26-G45E is a lethal mutation linked to KIDS that forms constitutively active connexin hemichannels. The pathomechanism(s) by which mutant Cx26 hemichannels perturb normal epidermal cornification are poorly understood. We created an animal model for KIDS by generating an inducible transgenic mouse expressing Cx26-G45E in keratinocytes. Cx26-G45E mice displayed reduced viability, hyperkeratosis, scaling, skin folds, and hair loss. Histopathology included hyperplasia, acanthosis, papillomatosis, increased cell size, and osteal plugging. These abnormalities correlated with human KIDS pathology and were associated with increased hemichannel currents in transgenic keratinocytes. These results confirm the pathogenic nature of the G45E mutation and provide a new model for studying the role of aberrant connexin hemichannels in epidermal differentiation and inherited connexin disorders.


Subject(s)
Connexins/biosynthesis , Deafness/metabolism , Deafness/pathology , Disease Models, Animal , Ichthyosis/metabolism , Ichthyosis/pathology , Keratitis/metabolism , Keratitis/pathology , Mutation, Missense , Amino Acid Substitution , Animals , Connexin 26 , Connexins/genetics , Deafness/genetics , Epidermis/metabolism , Epidermis/pathology , HeLa Cells , Humans , Ichthyosis/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Keratitis/genetics , Mice , Mice, Transgenic
16.
Eur Urol ; 52(4): 1229-37, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17150299

ABSTRACT

OBJECTIVES: Erectile dysfunction is a common diabetic complication. Preclinical studies have documented that the Slo gene (encoding the BK or Maxi-K channel alpha-subunit) plays a critical role in erectile function. Therefore, we determined whether diabetes induces changes in the splicing of the Slo gene relevant to erectile function. METHODS: Reverse transcriptase-polymerase chain reaction was used to compare Slo splice variant expression in corporal tissue excised from control and streptozotocin (STZ)-induced diabetic Fischer F-344 rats. Splice variants were sequenced, characterized by patch clamping, and fused to green fluorescent protein to determine cellular localization. The impact of altered Slo expression on erectile function was further evaluated in vivo. RESULTS: A novel Slo splice variant (SVcyt, with a cytoplasmic location) was predominantly expressed in corporal tissue from control rats. STZ-diabetes caused upregulation of a channel-forming transcript SV0. Preliminary results suggest that SV0 was also more prevalent in the corporal tissue of human diabetic compared with nondiabetic patients. The change in isoform expression in STZ-treated rats was partially reversed by insulin treatment. Intracorporal injection of a plasmid expressing the SV0 transcript, but not SVcyt, restored erectile function in STZ-diabetic rats. CONCLUSIONS: Alternative splicing of the Slo transcript may represent an important compensatory mechanism to increase the ease with which relaxation of corporal tissue may be triggered as a result of a diabetes-related decline in erectile capacity.


Subject(s)
Alternative Splicing , Diabetes Complications/genetics , Diabetes Mellitus, Experimental/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Animals , Base Sequence , Cell Line , Erectile Dysfunction/etiology , Erectile Dysfunction/genetics , Humans , Kidney , Male , Molecular Sequence Data , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Transfection
17.
BJU Int ; 97(5): 1076-82, 2006 May.
Article in English | MEDLINE | ID: mdl-16643495

ABSTRACT

OBJECTIVE: To establish the methods, feasibility and utility of evaluating the impact of diabetes on bladder and erectile function in the same rat, as more than half of diabetic patients have bladder dysfunction, and half of diabetic men have erectile dysfunction, but the severity of coincident disease has not been rigorously assessed. MATERIALS AND METHODS: In all, 16 F-344 rats had diabetes induced by streptozotocin (STZ), and were divided into insulin-treated (five) and untreated (11), and compared with age-matched controls (10), all assessed in parallel. All STZ rats were diabetic for 8-11 weeks. Cystometric studies were conducted on all rats, with cavernosometric studies conducted on a subset of rats. RESULTS: There were insulin-reversible increases in the following cystometric variables; bladder weight, bladder capacity, micturition volume, residual volume, micturition pressure and spontaneous activity (P < 0.05, in all, one-way analysis of variance, anova). Cavernosometry showed a diabetes-related, insulin-reversible decline in the cavernosal nerve-stimulated intracavernosal pressure (ICP) response at all levels of current stimulation (P < 0.05, in all one-way anova). Plotting erectile capacity (i.e. ICP) against bladder capacity showed no correlation between the extent of the decline in erectile capacity and the magnitude of the increase in bladder capacity. CONCLUSIONS: These studies extend previous work to indicate that the extent of diabetes-related bladder and erectile dysfunction can vary in the same rat. As such, these findings highlight the importance of evaluating the impact of diabetes on multiple organ systems in the lower urinary tract. Future studies using this model system should lead to a better understanding of the initiation, development, progression and coincidence of these common diabetic complications.


Subject(s)
Diabetes Mellitus, Experimental/complications , Erectile Dysfunction/etiology , Urinary Bladder Diseases/etiology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Electric Stimulation , Erectile Dysfunction/drug therapy , Erectile Dysfunction/physiopathology , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Male , Organ Size , Penis/innervation , Rats , Rats, Inbred F344 , Urinary Bladder Diseases/pathology , Urinary Bladder Diseases/physiopathology , Urination/physiology
18.
J Urol ; 175(1): 381-6, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16406945

ABSTRACT

PURPOSE: Intercellular communication through gap junctions was assessed in 8 to 10-week STZ diabetic rats to evaluate diabetes related effects on gap junctional conductance and permeability in short-term cultures of corporeal myocytes. MATERIALS AND METHODS: Rats were made diabetic by a single intraperitoneal injection of STZ. Eight to 10 weeks later erectile function was evaluated in vivo and corporeal tissue was harvested to isolate corporeal myocytes. Dual whole cell patch clamp studies of intercellular communication through connexin43 (Cx43) derived gap junction channels were done in short-term, ie passages 0 to 2, cultured corporeal myocytes excised from STZ diabetic rats with documented erectile impairment as well as in myocytes from age matched control rats. RESULTS: No differences in macroscopic junctional conductance, single channel conductance or open probability were detected between myocytes from age matched control and STZ diabetic rats, confirming the lack of diabetes related alterations in Cx43 gating or conductance. However, fluorescence dye transfer experiments revealed a marked 3-fold increase in Cx43 mediated junctional permeability in the absence of any detectable change in Cx43 protein expression. CONCLUSIONS: These data suggest that an alteration in the selectivity filter of Cx43 in diabetic animals affects the permeability of specifically sized and charged solutes. To our knowledge these studies provide the first evidence of a diabetes related increase in intercellular permselectivity in corporeal myocytes and, thus, they may have important implications for diabetes related erectile dysfunction.


Subject(s)
Cell Membrane Permeability , Connexin 43/physiology , Diabetes Mellitus, Experimental/metabolism , Gap Junctions/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Penis/blood supply , Animals , Cells, Cultured , Electrophysiology , Male , Rats , Time Factors
19.
Adv Exp Med Biol ; 539(Pt A): 239-54, 2003.
Article in English | MEDLINE | ID: mdl-15088908

ABSTRACT

There is now considerable experimental and clinical evidence supporting the supposition that overactivity of the bladder is associated with detectable alterations in the electrical properties of the detrusor smooth muscle cells. The preliminary data described in this report indicates that intercellular communication through gap junctions might play an important role in this process. Moreover, alterations in Cx43 mRNA expression may represent a tissue response to a physiologic insult (i.e., increased after load) in an attempt to further increase the syncytial nature and force of detrusor contractility to compensate for an increased pressure load. Finally, this report elucidates the rationale for suspecting that intercellular communication through gap junctions may play a role in normal bladder physiology and the pathophysiology of urinary incontinence caused by partial outlet obstruction.


Subject(s)
Cell Communication/physiology , Urinary Bladder Neck Obstruction/physiopathology , Urinary Bladder/cytology , Urinary Bladder/physiology , Urinary Incontinence/physiopathology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...