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










Publication year range
1.
Free Radic Biol Med ; 195: 47-57, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36566798

ABSTRACT

Doxorubicin (Dox), an anthracycline antibiotic, is an anticancer drug that inhibits DNA replication and cellular metabolic processes in cancer cells with high proliferative potential. However, Dox causes severe side effects, including myocardial damage and heart failure, but the molecular mechanism underlying Dox-induced myocardial injury remains uncertain. In the present study, we evaluated the effects of Dox on the mitochondrial quality control system and regulation of mitochondrial respiration and autophagy in an in vitro rat myoblast H9c2 cell culture model using western blotting, immunohistochemistry, the Seahorse XF24 system, and flow cytometry. Our results showed that Dox did not impair the initiation of autophagic flux or the functions of lysosomes; however, Dox affected the mitochondrial quality control system, leading to a fission-dominant morphology and impaired regulation of mitochondrial respiration, thereby increasing oxidative stress and inhibited progression of autophagy, particularly the fusion of autophagosomes with lysosomes. This inhibition caused a significant decrease in the formation of autolysosomes and was responsible for the accumulation of dysfunctional mitochondria and subsequent increase in oxidative stress, eventually leading to increased myocardial cell death.


Subject(s)
Doxorubicin , Myocytes, Cardiac , Rats , Animals , Myocytes, Cardiac/metabolism , Doxorubicin/adverse effects , Autophagy , Mitochondria/metabolism , Antibiotics, Antineoplastic/pharmacology , Oxidative Stress , Apoptosis
2.
Mol Pharmacol ; 96(4): 441-451, 2019 10.
Article in English | MEDLINE | ID: mdl-31383768

ABSTRACT

The G protein-coupled receptor (GPCR) signaling pathways mediated by trimeric G proteins have been extensively elucidated, but their associated regulatory mechanisms remain unclear. Parathyroid hormone (PTH)/PTH-related protein receptor (PTHR) is a GPCR coupled with Gs and Gq Gs activates adenylyl cyclases (ACs), which produces cAMP to regulate various cell fates. We previously showed that cell surface expression of PTHR was increased by its direct interaction with a subcortical cytoskeletal protein, 4.1G, whereas PTHR-mediated Gs/AC/cAMP signaling was suppressed by 4.1G through an unknown mechanism in human embryonic kidney (HEK)293 cells. In the present study, we found that AC type 6 (AC6), one of the major ACs activated downstream of PTHR, interacts with 4.1G in HEK293 cells, and the N-terminus of AC6 (AC6-N) directly and selectively binds to the 4.1/ezrin/radixin/moesin (FERM) domain of 4.1G (4.1G-FERM) in vitro. AC6-N was distributed at the plasma membrane, which was disturbed by knockdown of 4.1G. An AC6-N mutant, AC6-N-3A, in which three consecutive arginine residues are mutated to alanine residues, altered both binding to 4.1G-FERM and its plasma membrane distribution in vivo. Further, we overexpressed AC6-N to competitively inhibit the interaction of endogenous AC6 and 4.1G in cells. cAMP production induced by forskolin, an adenylyl cyclase activator, and PTH-(1-34) was enhanced by AC6-N expression and 4.1G-knockdown. In contrast, AC6-N-3A had no impact on forskolin- and PTH-(1-34)-induced cAMP productions. These data provide a novel regulatory mechanism that AC6 activity is suppressed by the direct binding of 4.1G to AC6-N, resulting in attenuation of PTHR-mediated Gs/AC6/cAMP signaling.


Subject(s)
Adenylyl Cyclases/chemistry , Adenylyl Cyclases/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Adenylyl Cyclases/genetics , Binding Sites , Cell Membrane/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Enzyme Activation , HEK293 Cells , Humans , Mutation , Protein Binding , Signal Transduction
3.
Redox Biol ; 13: 407-417, 2017 10.
Article in English | MEDLINE | ID: mdl-28683400

ABSTRACT

Highly active anti-retrovirus therapy (HAART) has been used to block the progression and symptoms of human immunodeficiency virus infection. Although it decreases morbidity and mortality, clinical use of HAART has also been linked to various adverse effects such as severe cardiomyopathy resulting from compromised mitochondrial functioning. However, the mechanistic basis for these effects remains unclear. Here, we demonstrate that a key component of HAART, 3ꞌ-azido-3ꞌ-deoxythymidine (AZT), particularly, its active metabolite AZT-triphosphate (AZT-TP), caused mitochondrial dysfunction, leading to induction of cell death in H9c2 cells derived from rat embryonic myoblasts, which serve as a model for cardiomyopathy. Specifically, treatment with 100µM AZT for 48h disrupted the mitochondrial tubular network via accumulation of AZT-TP. The mRNA expression of dynamin-related protein (Drp)1 and the Drp1 receptor mitochondrial fission factor (Mff) was upregulated whereas that of optic atrophy 1 (Opa1) was downregulated following AZT treatment. Increased mitochondrial translocation of Drp1, Mff upregulation, and decreased functional Opa1 expression induced by AZT impaired the balance of mitochondrial fission vs. fusion. These data demonstrate that AZT-TP causes cell death by altering mitochondrial dynamics.


Subject(s)
Anti-HIV Agents/toxicity , Mitochondria/drug effects , Mitochondrial Dynamics , Zidovudine/toxicity , Animals , Anti-HIV Agents/adverse effects , Cardiotoxicity/etiology , Cell Line , Dynamins/genetics , Dynamins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Rats , Zidovudine/adverse effects
4.
EBioMedicine ; 20: 27-38, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28579242

ABSTRACT

Mitochondrial dysfunction increases oxidative stress and depletes ATP in a variety of disorders. Several antioxidant therapies and drugs affecting mitochondrial biogenesis are undergoing investigation, although not all of them have demonstrated favorable effects in the clinic. We recently reported a therapeutic mitochondrial drug mitochonic acid MA-5 (Tohoku J. Exp. Med., 2015). MA-5 increased ATP, rescued mitochondrial disease fibroblasts and prolonged the life span of the disease model "Mitomouse" (JASN, 2016). To investigate the potential of MA-5 on various mitochondrial diseases, we collected 25 cases of fibroblasts from various genetic mutations and cell protective effect of MA-5 and the ATP producing mechanism was examined. 24 out of the 25 patient fibroblasts (96%) were responded to MA-5. Under oxidative stress condition, the GDF-15 was increased and this increase was significantly abrogated by MA-5. The serum GDF-15 elevated in Mitomouse was likewise reduced by MA-5. MA-5 facilitates mitochondrial ATP production and reduces ROS independent of ETC by facilitating ATP synthase oligomerization and supercomplex formation with mitofilin/Mic60. MA-5 reduced mitochondria fragmentation, restores crista shape and dynamics. MA-5 has potential as a drug for the treatment of various mitochondrial diseases. The diagnostic use of GDF-15 will be also useful in a forthcoming MA-5 clinical trial.


Subject(s)
Indoleacetic Acids/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Phenylbutyrates/pharmacology , Protein Multimerization/drug effects , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Cell Line , Cell Survival/drug effects , DNA, Mitochondrial , Disease Models, Animal , Fibroblast Growth Factors/metabolism , Fibroblasts/metabolism , Growth Differentiation Factor 15/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/genetics , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/genetics , Mitochondrial Dynamics/drug effects , Mitochondrial Proton-Translocating ATPases/chemistry , Multiprotein Complexes/metabolism , Mutation , Organelle Biogenesis , Prognosis , Protective Agents , Protein Binding
5.
EMBO Rep ; 18(8): 1460-1472, 2017 08.
Article in English | MEDLINE | ID: mdl-28607034

ABSTRACT

The primary cilium is a plasma membrane-protruding sensory organelle that undergoes regulated assembly and resorption. While the assembly process has been studied extensively, the cellular machinery that governs ciliary resorption is less well understood. Previous studies showed that the ciliary pocket membrane is an actin-rich, endocytosis-active periciliary subdomain. Furthermore, Tctex-1, originally identified as a cytoplasmic dynein light chain, has a dynein-independent role in ciliary resorption upon phosphorylation at Thr94. Here, we show that the remodeling and endocytosis of the ciliary pocket membrane are accelerated during ciliary resorption. This process depends on phospho(T94)Tctex-1, actin, and dynamin. Mechanistically, Tctex-1 physically and functionally interacts with the actin dynamics regulators annexin A2, Arp2/3 complex, and Cdc42. Phospho(T94)Tctex-1 is required for Cdc42 activation before the onset of ciliary resorption. Moreover, inhibiting clathrin-dependent endocytosis or suppressing Rab5GTPase on early endosomes effectively abrogates ciliary resorption. Taken together with the epistasis functional assays, our results support a model in which phospho(T94)Tctex-1-regulated actin polymerization and periciliary endocytosis play an active role in orchestrating the initial phase of ciliary resorption.


Subject(s)
Actins/physiology , Cilia/physiology , Dyneins/metabolism , Cell Line , Clathrin/physiology , Dynamins , Dyneins/genetics , Endocytosis , Epithelial Cells , Humans , Phosphorylation , Protein Multimerization , Retina/cytology
7.
J Am Soc Nephrol ; 27(7): 1925-32, 2016 07.
Article in English | MEDLINE | ID: mdl-26609120

ABSTRACT

Mitochondrial dysfunction causes increased oxidative stress and depletion of ATP, which are involved in the etiology of a variety of renal diseases, such as CKD, AKI, and steroid-resistant nephrotic syndrome. Antioxidant therapies are being investigated, but clinical outcomes have yet to be determined. Recently, we reported that a newly synthesized indole derivative, mitochonic acid 5 (MA-5), increases cellular ATP level and survival of fibroblasts from patients with mitochondrial disease. MA-5 modulates mitochondrial ATP synthesis independently of oxidative phosphorylation and the electron transport chain. Here, we further investigated the mechanism of action for MA-5. Administration of MA-5 to an ischemia-reperfusion injury model and a cisplatin-induced nephropathy model improved renal function. In in vitro bioenergetic studies, MA-5 facilitated ATP production and reduced the level of mitochondrial reactive oxygen species (ROS) without affecting activity of mitochondrial complexes I-IV. Additional assays revealed that MA-5 targets the mitochondrial protein mitofilin at the crista junction of the inner membrane. In Hep3B cells, overexpression of mitofilin increased the basal ATP level, and treatment with MA-5 amplified this effect. In a unique mitochondrial disease model (Mitomice with mitochondrial DNA deletion that mimics typical human mitochondrial disease phenotype), MA-5 improved the reduced cardiac and renal mitochondrial respiration and seemed to prolong survival, although statistical analysis of survival times could not be conducted. These results suggest that MA-5 functions in a manner differing from that of antioxidant therapy and could be a novel therapeutic drug for the treatment of cardiac and renal diseases associated with mitochondrial dysfunction.


Subject(s)
Indoleacetic Acids/pharmacology , Kidney Tubules/cytology , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Smooth Muscle/drug effects , Phenylbutyrates/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL
8.
Biochem Biophys Res Commun ; 469(4): 897-902, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26713362

ABSTRACT

Genetic analyses have revealed an important association between A-kinase anchoring proteins (AKAPs) and the intracellular calcium modulating system. AKAP5, also known as AKAP79/150, is an anchoring protein between PKA and voltage-dependent calcium channels, ryanodine receptor-2, phospholamban and other molecules. The aim of the present study was to elucidate the physiological importance of AKAP5 in the creation of cardiac rhythm using AKAP5-null mice. ECG analysis showed a normal sinus rhythm and a decreased responsiveness to isoproterenol in AKAP5-null mice compared with wild-type mice. Analysis of heart rate variability revealed that the R-R interval was unstable in AKAP5-null mutants and that the low-frequency components had decreased, indicating that the tonus of the sympathetic nervous system was affected. Furthermore, the atrium of the AKAP5-null mice showed a decreased positive inotropic response to isoproterenol, indicating the involvement of AKAP5 in a PKA-dependent pathway. Thus, our present study revealed that AKAP5 plays a significant role in the regulation of sympathetic nerve activities.


Subject(s)
A Kinase Anchor Proteins/metabolism , Brain/physiology , Heart Rate/physiology , Heart/innervation , Heart/physiology , Sympathetic Nervous System/physiology , A Kinase Anchor Proteins/genetics , Animals , Mice , Mice, Knockout
9.
Physiol Rep ; 3(10)2015 Oct.
Article in English | MEDLINE | ID: mdl-26462747

ABSTRACT

The anti-inflammatory effects of macrolides may be associated with a reduced frequency of exacerbation of chronic obstructive pulmonary disease (COPD). However, because the long-term use of antibiotics may promote the growth of drug-resistant bacteria, the development of a treatment to prevent COPD exacerbation with macrolides that do not exert anti-bacterial effects is necessary. Additionally, the inhibitory effects of nonantibiotic macrolides on the replication of rhinovirus (RV), which is the major cause of COPD exacerbation, have not been demonstrated. Primary cultures of human tracheal epithelial cells and nasal epithelial cells were pretreated with the nonantibiotic macrolide EM900 for 72 h prior to infection with a major group RV type 14 rhinovirus (RV14) and were further treated with EM900 after infection. Treatment with EM900 before and after infection reduced RV14 titers in the supernatants and viral RNA within the cells. Moreover, cytokine levels, including interleukin (IL)-1ß and IL-6, were reduced in the supernatants following RV14 infection. Treatment with EM900 before and after infection also reduced the mRNA and protein expression of intercellular adhesion molecule-1 (ICAM-1), which is the receptor for RV14, after infection and reduced the activation of the nuclear factor kappa-B protein p50 in nuclear extracts after infection. Pretreatment with EM900 reduced the number and fluorescence intensity of the acidic endosomes through which RV RNA enters the cytoplasm. Thus, pretreatment with EM900 may inhibit RV infection by reducing the ICAM-1 levels and acidic endosomes and thus modulate the airway inflammation associated with RV infections.

10.
Methods Mol Biol ; 1317: 55-67, 2015.
Article in English | MEDLINE | ID: mdl-26072401

ABSTRACT

Suicide gene therapy of cancer (SGTC) entails the introduction of a cDNA sequence into tumor cells whose polypeptide product is capable of either directly activating apoptotic pathways itself or facilitating the activation of pharmacologic agents that do so. The latter class of SGTC approaches is of the greater utility in cancer therapy owing to the ability of some small, activated cytotoxic compounds to diffuse from their site of activation into neighboring malignant cells, where they can also mediate destruction. This phenomenon, termed "bystander killing", can be highly advantageous in driving significant tumor regression in vivo without the requirement of transduction of each and every tumor cell with the suicide gene. We have developed a robust suicide gene therapy enzyme/prodrug system based on an engineered variant of the human thymidylate kinase (TMPK), which has been endowed with the ability to drive azidothymidine (AZT) activation. Delivery of this suicide gene sequence into tumors by means of recombinant lentivirus-mediated transduction embodies an SGTC strategy that successfully employs bystander cell killing as a mechanism to achieve significant ablation of solid tumors in vivo. Thus, this engineered TMPK/AZT suicide gene therapy axis holds great promise for clinical application in the treatment of inoperable solid tumors in the neoadjuvant setting. Here we present detailed procedures for the preparation of recombinant TMPK-based lentivirus, transduction of target cells, and various approaches for the evaluation of bystander cell killing effects in SGCT in both in vitro and in vivo models.


Subject(s)
Bystander Effect/drug effects , Genes, Transgenic, Suicide , Genetic Therapy/methods , Neoplasms/genetics , Neoplasms/therapy , Nucleoside-Phosphate Kinase/genetics , Prodrugs/pharmacology , Zidovudine/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation , Colorimetry , Gene Expression , Genetic Engineering , HEK293 Cells , Humans , Lentivirus/metabolism , Male , Mice, Inbred NOD , Mice, SCID , Nucleoside-Phosphate Kinase/therapeutic use , Recombinant Proteins/metabolism , Transduction, Genetic , Transgenes
11.
Tohoku J Exp Med ; 236(3): 225-32, 2015 07.
Article in English | MEDLINE | ID: mdl-26118651

ABSTRACT

Mitochondria are key organelles implicated in a variety of processes related to energy and free radical generation, the regulation of apoptosis, and various signaling pathways. Mitochondrial dysfunction increases cellular oxidative stress and depletes ATP in a variety of inherited mitochondrial diseases and also in many other metabolic and neurodegenerative diseases. Mitochondrial diseases are characterized by the dysfunction of the mitochondrial respiratory chain, caused by mutations in the genes encoded by either nuclear DNA or mitochondrial DNA. We have hypothesized that chemicals that increase the cellular ATP levels may ameliorate the mitochondrial dysfunction seen in mitochondrial diseases. To search for the potential drugs for mitochondrial diseases, we screened an in-house chemical library of indole-3-acetic-acid analogs by measuring the cellular ATP levels in Hep3B human hepatocellular carcinoma cells. We have thus identified mitochonic acid 5 (MA-5), 4-(2,4-difluorophenyl)-2-(1H-indol-3-yl)-4-oxobutanoic acid, as a potential drug for enhancing ATP production. MA-5 is a newly synthesized derivative of the plant hormone, indole-3-acetic acid. Importantly, MA-5 improved the survival of fibroblasts established from patients with mitochondrial diseases under the stress-induced condition, including Leigh syndrome, MELAS (myopathy encephalopathy lactic acidosis and stroke-like episodes), Leber's hereditary optic neuropathy, and Kearns-Sayre syndrome. The improved survival was associated with the increased cellular ATP levels. Moreover, MA-5 increased the survival of mitochondrial disease fibroblasts even under the inhibition of the oxidative phosphorylation or the electron transport chain. These data suggest that MA-5 could be a therapeutic drug for mitochondrial diseases that exerts its effect in a manner different from anti-oxidant therapy.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Survival/drug effects , Drug Discovery , Fibroblasts/drug effects , Indoleacetic Acids/chemistry , Indoleacetic Acids/pharmacology , Mitochondrial Diseases/drug therapy , Phenylbutyrates/pharmacology , Analysis of Variance , Cell Line, Tumor , Cell Survival/physiology , Fibroblasts/physiology , Humans , Oxidative Phosphorylation , Phenylbutyrates/chemistry , Small Molecule Libraries
12.
Biochem Biophys Res Commun ; 461(2): 200-5, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25892515

ABSTRACT

Genetic analyses have revealed an important association between P/Q-type calcium channel activities and hereditary neurological disorders. The P/Q-type channels are composed principally of heterologous multimeric subunits including CaV2.1 and CaVß4. Of these, the ß4 subunit is thought to play a significant role in channel physiology, because a mouse line mutant in that subunit (the lethargic mouse: lh) exhibits a severe ataxic phenotype. The aim of the present study was to elucidate the physiological importance of the ß4 subunit. ECG analysis showed that the T wave was high in 8-week-old lh mutants; this may be associated with hyperkalemia. Upon pharmacological ECG analysis, 2-3-week-old lh mutants exhibited reduced responses to a ß-blocker and a muscarinic receptor antagonist. Analysis of heart rate variability revealed that the R-R interval was unstable in lh mutants and that both the low- and high-frequency components had increased in extent, indicating that the tonus of both the sympathetic and parasympathetic nervous systems was modified. Thus, our present study revealed that the ß4 subunit played a significant role in regulation of sympathetic and parasympathetic nerve activities.


Subject(s)
Calcium Channels, N-Type/genetics , Heart/innervation , Heart/physiology , Mutation , Parasympathetic Nervous System/physiology , Sympathetic Nervous System/physiology , Animals , Base Sequence , Calcium Channels, N-Type/metabolism , Genotype , Heart Rate , Mice , Molecular Sequence Data , Protein Subunits/genetics , Protein Subunits/metabolism
13.
Biochem Biophys Res Commun ; 460(4): 1076-81, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25849891

ABSTRACT

Orexin, also known as hypocretin, is a secreted neuropeptide implicated in the regulation of sleep and food intake. In the present study, we examined the importance of orexin in regulation of the sympathetic nervous system using an orexin/ataxin-3 transgenic (OXTg) rat, which has a minimal number of orexin neurons. RT-PCR analysis identified expression of prepro-orexin and orexin receptor-1 (OX1R) in the superior cervical ganglion (SCG), and expression of another receptor (OX2R) was marginal in the wild-type rat. The orexin/ataxin-3 transgenic rat showed increased expression of OX1R and OX2R, whereas expression of prepro-orexin was undetectable, suggesting a compensatory increase in both receptors. In the ECG recording (R-R interval), orexin/ataxin-3 transgenic rats showed decreased responsiveness to the ß-adrenergic blocker propranolol. Furthermore, OXTg rats had deteriorated R-R interval regulation, indicating involvement of the orexin system in sympathetic nerve regulation. This was accompanied by decreased baroreflex and responsiveness to ß-adrenergic blocker in blood pressure recording, also suggesting involvement of the orexin system in sympathetic nerve regulation. Histological examination revealed hypotrophic changes in the transgenic heart, suggesting involvement of the orexin system in cardiac development. Taken together, our present results indicate involvement of the orexin system in sympathetic nerve control.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Sympathetic Nervous System/physiology , Animals , Blood Pressure/physiology , Electrocardiography , Gene Expression Profiling , Intracellular Signaling Peptides and Proteins/genetics , Neuropeptides/genetics , Orexins , Rats , Rats, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
14.
Biochem Biophys Res Commun ; 458(3): 584-589, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25680462

ABSTRACT

The histamine system is involved in the regulation of the autonomic nervous system. We used gene-targeted mice to investigate the role of histamine receptors in the regulation of the sympathetic nervous system. Reverse transcription-polymerase chain reaction (RT-PCR) analysis revealed histamine H1, H2, and H3 receptor expression in the superior cervical ganglion, which contains sympathetic nerve cell bodies. We measured the heart rate variability (HRV), the changes in the beat-to-beat heart rate, which is widely used to assess autonomic activity in the heart. H1 blockade attenuated the baroreflex-mediated changes in heart rate in wild-type (WT) mice, whereas the heart rate response to H2- and H3-specific blockers was unaffected. l-Histidine decarboxylase (HDC) expression in the superior cervical ganglion of H1R-null mice was higher than that in WT controls, whereas the enzyme levels in H2R- and H3R-null mice were not significantly different from those in the WT. All mutant mice (H1R-, H2R-, and H3R-null mice) showed normal electrocardiogram (ECG) patterns with little modification in ECG parameters and the expected response to the ß-adrenergic blocker propranolol. Similar to our findings in WT mice, H1 blockade attenuated the baroreflex-mediated heart rate change in H1R-null mice, whereas the heart rate response was unaffected in H2R- and H3R-null mice. The HRV analysis revealed relatively unstable RR intervals, an increased standard deviation of the interbeat interval (SDNN), and low-frequency (LF) component in H1R-null mice compared with the other groups, suggesting that sympathetic nerve activity was altered in H1R-null mice. Taken together, our findings indicate that H1 receptors play a major role in the regulation of sympathetic nerve activity.


Subject(s)
Receptors, Histamine H1/metabolism , Sympathetic Nervous System/physiology , Animals , Electrocardiography , Gene Deletion , Heart Rate , Histamine/metabolism , Histidine Decarboxylase/genetics , Mice , Receptors, Histamine/genetics , Receptors, Histamine/metabolism , Receptors, Histamine H1/genetics , Up-Regulation
15.
PLoS One ; 8(10): e78711, 2013.
Article in English | MEDLINE | ID: mdl-24194950

ABSTRACT

We previously described a novel suicide (or 'cell fate control') gene therapy enzyme/prodrug system based on an engineered variant of human thymidylate kinase (TMPK) that potentiates azidothymidine (AZT) activation. Delivery of a suicide gene sequence into tumors by lentiviral transduction embodies a cancer gene therapy that could employ bystander cell killing as a mechanism driving significant tumor regression in vivo. Here we present evidence of a significant bystander cell killing in vitro and in vivo mediated by the TMPK/AZT suicide gene axis that is reliant on the formation of functional gap-junctional intercellular communications (GJICs). Potentiation of AZT activation by the engineered TMPK expressed in the human prostate cancer cell line, PC-3, resulted in effective bystander killing of PC-3 cells lacking TMPK expression--an effect that could be blocked by the GJIC inhibitor, carbenoxolone. Although GJICs are mainly formed by connexins, a new family of GJIC molecules designated pannexins has been recently identified. PC-3 cells expressed both connexin43 (Cx43) and Pannexin1 (Panx1), but Panx1 expression predominated at the plasma membrane, whereas Cx43 expression was primarily localized to the cytosol. The contribution of bystander effects to the reduction of solid tumor xenografts established by the PC-3 cell line was evaluated in an animal model. We demonstrate the contribution of bystander cell killing to tumor regression in a xenograft model relying on the delivery of expression of the TMPK suicide gene into tumors via direct intratumoral injection of recombinant therapeutic lentivirus. Taken together, our data underscore that the TMPK/AZT enzyme-prodrug axis can be effectively utilized in suicide gene therapy of solid tumors, wherein significant tumor regression can be achieved via bystander effects mediated by GJICs.


Subject(s)
Bystander Effect/physiology , Genes, Transgenic, Suicide/genetics , Genetic Therapy/methods , Nucleoside-Phosphate Kinase/genetics , Prodrugs/pharmacology , Prostatic Neoplasms/therapy , Zidovudine/pharmacology , Analysis of Variance , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Flow Cytometry , HEK293 Cells , Humans , Male , Microscopy, Confocal , Prodrugs/chemistry , Reactive Oxygen Species/metabolism , Zidovudine/chemistry
17.
J Pharmacol Sci ; 122(3): 193-204, 2013.
Article in English | MEDLINE | ID: mdl-23823934

ABSTRACT

Release of growth hormone (GH) from the somatotroph is regulated by binding GH-releasing hormone (GHRH) to its cognate receptor (GHRHR), one of the members of the G protein-coupled receptor (GPCR) superfamily. Proteins bound to the carboxy (C)-terminus of GPCR have been reported to regulate intracellular trafficking and function of the receptor; however, no functionally significant protein associated with GHRHR has been reported. We have identified a protein interacting with C-kinase 1 (PICK1) as a binding partner of GHRHR. In vitro binding assay revealed the PDZ-domain of PICK1 and the last four amino acid residues of GHRHR were prerequisite for the interaction. Further, in vivo association of these proteins was confirmed. Immunostaining data of a stable cell line expressing GHRHR with or without PICK1 suggested the C-terminus of GHRHR promoted cell surface expression of GHRHR and PICK1 affected the kinetics of the cell surface expression of GHRHR. Furthermore, cAMP production assay showed the C-terminus of GHRHR is involved in the regulation of receptor activation, and the interaction of GHRHR with PICK1 may influence intensities of the signal response after ligand stimulation. Thus, the interaction of the C-terminus of GHRHR with PICK1 has a profound role in regulating the trafficking and the signaling of GHRHR. [Supplementary Figure: available only at http://dx.doi.org/10.1254/jphs.12287FP].


Subject(s)
Carrier Proteins/physiology , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/physiology , Nuclear Proteins/physiology , PDZ Domains/physiology , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/physiology , Signal Transduction/physiology , Animals , Brain/metabolism , Carrier Proteins/metabolism , Cytoskeletal Proteins , Humans , Male , Nuclear Proteins/metabolism , Protein Binding , Protein Transport , Rats, Sprague-Dawley
18.
Cell Signal ; 25(3): 690-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23201780

ABSTRACT

It has been shown lately that activity of G protein-coupled receptors (GPCRs) is regulated by an array of proteins binding to carboxy (C)-terminus of GPCRs. Proteins of 4.1 family are subsets of subcortical cytoskeletal proteins and are known to stabilize cellular structures and proteins at the plasma membrane. One of the 4.1 family proteins, 4.1G has been shown to interact with the C-terminus of GPCRs and regulate intracellular distribution of the receptors, including parathyroid hormone (PTH)/PTH-related protein receptor (PTHR). PTHR is coupled to trimeric G proteins G(s) and G(q), which activate the adenylyl cyclase/cyclic AMP (cAMP) pathway and phospholipase C pathway, respectively. During the course of investigation of the role of 4.1G on adenylyl cyclase/cAMP signaling pathway, we found that 4.1G suppressed forskolin-induced cAMP production in cells. The cAMP accumulation induced by forskolin was decreased in HEK293 cells overexpressing 4.1G or increased in 4.1G-knockdown cells. Furthermore, PTH -(1-34)-stimulated cAMP production was also suppressed in the presence of exogenously expressed 4.1G despite its activity to increase the distribution of PTHR to the cell surface. In cells overexpressing FERM domain-deleted 4.1G, a mutant form of the protein deficient in plasma membrane distribution, neither forskolin-induced nor PTH -(1-34)-stimulated cAMP production was not altered. The suppression of the forskolin-induced cAMP production was observed even in membrane preparations of 4.1G-overexpressing cells. In 4.1G-knockdown HEK293 cells, plasma membrane distribution of adenylyl cyclase 6, one of the major subtypes of the enzyme in the cells, showed a slight decrease, in spite of the increased production of cAMP in those cells when stimulated by forskolin. Also, cytochalasin D treatment did not cause any influence on forskolin-induced cAMP production in HEK293 cells. These data indicate that plasma membrane-associated 4.1G regulates GPCR-mediated G(s) signaling by suppressing adenylyl cyclase-mediated cAMP production.


Subject(s)
Adenylyl Cyclases/metabolism , Cyclic AMP/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Colforsin/pharmacology , Cytochalasin D/pharmacology , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Parathyroid Hormone/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Radioimmunoassay , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction/drug effects , Transfection
19.
Cell Signal ; 23(11): 1843-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21749919

ABSTRACT

Functional roles of putative helix 8 in the carboxy-terminal tail of the human histamine H(3) receptor were investigated using deleted and alanine-substituted mutant receptors. While the deletion of the carboxy-terminal tail did not decrease the total expression level, surface expression, or ligand binding affinity, the agonist-stimulated cAMP response, [((35))S] GTPγS binding, and MAPK activation were totally abolished. The receptor lacking the carboxy-terminal tail also failed to respond to an inverse agonist, thioperamide, suggesting that the carboxy-terminal tail is involved in the regulation of receptor activity by changing G-protein coupling with the receptor. Site-directed mutagenesis revealed that hydrophobic amino acids in the putative helix 8 such as phenylalanines at position 419 (F7.60) and 423 (F7.64) or leucines at 426 (L7.67) and 427 (L7.68) were important for the agonist-induced activation of H(3) receptor. Substitution of F7.60 also resulted in a receptor that was less responsive to inactivation by the inverse agonist, implying the existence of an intermediate conformation that can be either activated or inactivated. Our results suggest that hydrophobic interface of putative helix 8 is important for the regulation of H(3) receptor activity, presumably by stabilizing the helix to the plasma membrane.


Subject(s)
Amino Acids/chemistry , GTP-Binding Proteins/metabolism , Histamine Agonists/pharmacology , Mutation , Receptors, Histamine H3 , Signal Transduction/genetics , Amino Acid Sequence , Amino Acids/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Flow Cytometry , HEK293 Cells , Histamine/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Mutagenesis, Site-Directed , Piperidines/pharmacology , Plasmids , Protein Binding , Protein Structure, Secondary , Receptors, Histamine H3/chemistry , Receptors, Histamine H3/genetics , Receptors, Histamine H3/metabolism , Structure-Activity Relationship , Transfection
20.
Neurosci Res ; 67(2): 126-36, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20188128

ABSTRACT

ADP-ribosylation factor 6 (ARF6) is a small GTPase that regulates neuronal morphogenesis processes such as axonal, dendritic, and spine formation possibly through the actin cytoskeleton and membrane trafficking. In an attempt to define the molecular mechanisms that regulate neuronal morphogenesis by ARF6, we identified vezatin as a novel binding partner of active GTP-bound ARF6 using yeast two-hybrid screening. Vezatin was able to bind specifically to GTP-ARF6 among the ARF family. In the adult mouse brain, vezatin exhibited widespread gene expression with high levels in the hippocampus and medial habenular nucleus. In hippocampal neurons, vezatin was localized at dendrites as well as cell bodies. Knockdown of endogenous vezatin significantly reduced total dendritic length and arborization of cultured hippocampal neurons, while overexpression of vezatin increased dendritic length. Our present study suggests that vezatin may regulate dendritic formation as a downstream effector of ARF6.


Subject(s)
ADP-Ribosylation Factors/metabolism , Carrier Proteins/metabolism , Dendrites/drug effects , Hippocampus/cytology , Membrane Proteins/metabolism , Neurons/cytology , ADP-Ribosylation Factor 6 , Animals , Animals, Newborn , Carrier Proteins/genetics , Cells, Cultured , Central Nervous System/metabolism , Chlorocebus aethiops , Dendrites/physiology , Embryo, Mammalian , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Transfection/methods , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...