Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
PLoS One ; 19(3): e0299510, 2024.
Article in English | MEDLINE | ID: mdl-38452137

ABSTRACT

The Japanese national guidelines recommend significantly lower doses of carvedilol for heart failure with reduced ejection fraction (HFrEF) management than the US guidelines. Using real-world data, we determined whether initial and target doses of carvedilol in Japanese patients (JPNs) differ from those in US patients (USPs), especially in Asian Americans (ASA) and Caucasians (CA), and investigated differences in outcomes. We collected data from the electronic medical records, including demographics, carvedilol dosing, tolerability, cardiac functional indicators like EF, cardiovascular events including all-cause deaths, and laboratory values from the University of California, San Diego Health and Osaka University. JPNs had significantly lower doses (mg/day) of carvedilol initiation (66 USPs composed of 38 CAs and 28 ASAs, 17.1±16.2; 93 JPNs, 4.3±4.2, p<0.001) and one year after initiation (33.0±21.8; 11.2±6.5, p<0.001), and a significantly lower relative rate (RR) of dose discontinuation and reduction than USPs (RR: 0.406, 95% confidence interval (CI): 0.181-0.911, p<0.05). CAs showed the highest reduction rate (0.184), and ASAs had the highest discontinuation rate (0.107). A slight mean difference with narrow 95% CI ranges straddling zero was observed between the two regions in the change from the baseline of each cardiac functional indicator (LVEF, -0.68 [-5.49-4.12]; LVDd, -0.55 [-3.24-2.15]; LVDd index, -0.25 [-1.92-1.43]; LVDs, -0.03 [-3.84-3.90]; LVDs index, -0.04 [-2.38-2.30]; heart rate, 1.62 [-3.07-6.32]). The event-free survival showed no difference (p = 0.172) among the races. Conclusively, despite JPNs exhibiting markedly lower carvedilol doses, their dose effectiveness has the potential to be non-inferior to that in USPs. Dose de-escalation, not discontinuation, could be an option in some Asian and ASA HFrEF patients intolerable to high doses of carvedilol.


Subject(s)
Carvedilol , Heart Failure , Ventricular Dysfunction, Left , Humans , Adrenergic beta-Antagonists , Carvedilol/therapeutic use , Heart Failure/drug therapy , Japan , Stroke Volume , Treatment Outcome , Ventricular Dysfunction, Left/drug therapy
2.
Clin Pract ; 14(2): 377-387, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38525708

ABSTRACT

BACKGROUND: Avelumab, durvalumab, and atezolizumab are anti-programmed death-ligand 1 (PD-L1) antibodies approved for clinical application in Japan. Despite targeting the same molecule, avelumab elicits a different frequency of infusion-related reactions (IRRs) compared with durvalumab and atezolizumab, leading to differences in premedication recommendations. This study aimed to collect information to verify the relationship during IRRs and the characteristics of antibody molecules, by investigating the frequency of IRRs caused by three types of antibodies and the actual status of prophylactic measures. METHODS: This single-center, retrospective observational study collected the medical records of 73 patients who received avelumab, durvalumab, or atezolizumab at Osaka University Hospital. RESULTS: The frequency of IRRs was 50.0% (12/24) for avelumab, 31.0% (8/27) for durvalumab, and 18.2% (4/22) for atezolizumab. The IRRs were grade 2 in seven patients and grade 1 in five patients treated with avelumab, grade 2 in six patients and grade 1 in two patients treated with durvalumab, and grade 1 in all patients treated with atezolizumab. Among patients in whom symptoms were observed during the first administration, measures were taken to prevent IRRs for the second administration, but cases were confirmed in which symptoms reappeared, especially in patients who received durvalumab. CONCLUSION: Our findings indicate that the frequency of IRRs due to anti-PD-L1 antibodies is higher than that previously reported in clinical trials and different modifications in antibody molecules may affect the difference in IRR frequency.

3.
Sci Rep ; 12(1): 16656, 2022 10 05.
Article in English | MEDLINE | ID: mdl-36198906

ABSTRACT

Runt-related transcription factor 2 (Runx2), a regulator of osteoblast differentiation, is pathologically involved in vascular calcification; however, the significance of Runx2 in cardiac homeostasis remains unclear. Here, we investigated the roles of Runx2 in cardiac remodeling after myocardial infarction (MI). The expression of Runx2 mRNA and protein was upregulated in murine hearts after MI. Runx2 was expressed in heart-infiltrating myeloid cells, especially in macrophages, at the border zone of post-infarct myocardium. To analyze the biological functions of Runx2 in cardiac remodeling, myeloid cell-specific Runx2 deficient (CKO) mice were exposed to MI. After MI, ventricular weight/tibia length ratio was increased in CKO mice, concomitant with severe cardiac dysfunction. Cardiac fibrosis was exacerbated in CKO mice, consistent with the upregulation of collagen 1a1 expression. Mechanistically, immunohistochemical analysis using anti-CD31 antibody showed that capillary density was decreased in CKO mice. Additionally, conditioned culture media of myeloid cells from Runx2 deficient mice exposed to MI induced the tube formation of vascular endothelial cells to a lesser extent than those from control mice. RNA-sequence showed that the expression of pro-angiogenic or anti-angiogenic factors was altered in macrophages from Runx2-deficient mice. Collectively, Runx2+ myeloid cells infiltrate into post-infarct myocardium and prevent adverse cardiac remodeling, at least partially, by regulating endothelial cell function.


Subject(s)
Myocardial Infarction , Ventricular Remodeling , Animals , Collagen/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Culture Media, Conditioned/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardium/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Ventricular Remodeling/genetics
4.
Commun Biol ; 5(1): 734, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35869269

ABSTRACT

Podocyte injury is involved in the onset and progression of various kidney diseases. We previously demonstrated that the transcription factor, old astrocyte specifically induced substance (OASIS) in myofibroblasts, contributes to kidney fibrosis, as a novel role of OASIS in the kidneys. Importantly, we found that OASIS is also expressed in podocytes; however, the pathophysiological significance of OASIS in podocytes remains unknown. Upon lipopolysaccharide (LPS) treatment, there is an increase in OASIS in murine podocytes. Enhanced serum creatinine levels and tubular injury, but not albuminuria and podocyte injury, are attenuated upon podocyte-restricted OASIS knockout in LPS-treated mice, as well as diabetic mice. The protective effects of podocyte-specific OASIS deficiency on tubular injury are mediated by protein kinase C iota (PRKCI/PKCι), which is negatively regulated by OASIS in podocytes. Furthermore, podocyte-restricted OASIS transgenic mice show tubular injury and tubulointerstitial fibrosis, with severe albuminuria and podocyte degeneration. Finally, there is an increase in OASIS-positive podocytes in the glomeruli of patients with minimal change nephrotic syndrome and diabetic nephropathy. Taken together, OASIS in podocytes contributes to podocyte and/or tubular injury, in part through decreased PRKCI. The induction of OASIS in podocytes is a critical event for the disturbance of kidney homeostasis.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Podocytes , Albuminuria/genetics , Albuminuria/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Fibrosis , Homeostasis , Kidney/metabolism , Lipopolysaccharides/metabolism , Mice , Nerve Tissue Proteins/metabolism , Up-Regulation
6.
Physiol Rep ; 10(9): e15304, 2022 05.
Article in English | MEDLINE | ID: mdl-35542987

ABSTRACT

Tumor suppressor protein p53 plays crucial roles in the onset of heart failure. p53 activation results in cardiac dysfunction, at least partially by suppressing angiogenesis. Though p53 has been reported to reduce VEGF production by inhibiting hypoxia-inducible factor, the anti-angiogenic property of p53 remains to be fully elucidated in cardiomyocytes. To explore the molecular signals downstream of p53 that regulate vascular function, especially under normoxic conditions, DNA microarray was performed using p53-overexpressing rat neonatal cardiomyocytes. Among genes induced by more than 2-fold, we focused on CXCL10, an anti-angiogenic chemokine. Real-time PCR revealed that p53 upregulated the CXCL10 expression as well as p21, a well-known downstream target of p53. Since p53 is known to be activated by doxorubicin (Doxo), we examined the effects of Doxo on the expression of CXCL10 and found that Doxo enhanced the CXCL10 expression, accompanied by p53 induction. Importantly, Doxo-induced CXCL10 was abrogated by siRNA knockdown of p53, indicating that p53 activation is necessary for Doxo-induced CXCL10. Next, we examined the effect of hypoxic condition on p53-mediated induction of CXCL10. Interestingly, CXCL10 was induced by hypoxia and its induction was potentiated by the overexpression of p53. Finally, the conditioned media from cultured cardiomyocytes expressing p53 decreased the tube formation of endothelial cells compared with control, analyzed by angiogenesis assay. However, the inhibition of CXCR3, the receptor of CXCL10, restored the tube formation. These data indicate that CXCL10 is a novel anti-angiogenic factor downstream of p53 in cardiomyocytes and could contribute to the suppression of vascular function by p53.


Subject(s)
Chemokine CXCL10 , Myocytes, Cardiac , Tumor Suppressor Protein p53 , Animals , Cell Hypoxia , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Doxorubicin/pharmacology , Endothelial Cells , Myocytes, Cardiac/metabolism , Neovascularization, Pathologic/metabolism , Rats , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
J Cell Physiol ; 237(5): 2539-2549, 2022 05.
Article in English | MEDLINE | ID: mdl-35312066

ABSTRACT

Because mammalian cardiomyocytes largely cease to proliferate immediately after birth, the regenerative activity of the heart is limited. To date, much effort has been made to clarify the regulatory mechanism of cardiomyocyte proliferation because the amplification of cardiomyocytes could be a promising strategy for heart regenerative therapy. Recently, it was reported that the inhibition of glycogen synthase kinase (GSK)-3 promotes the proliferation of neonatal rat cardiomyocytes (NRCMs) and human iPS cell-derived cardiomyocytes (hiPSC-CMs). Additionally, Yes-associated protein (YAP) induces cardiomyocyte proliferation. The purpose of this study was to address the importance of YAP activity in cardiomyocyte proliferation induced by GSK-3 inhibitors (GSK-3Is) to develop a novel strategy for cardiomyocyte amplification. Immunofluorescent microscopic analysis using an anti-Ki-67 antibody demonstrated that the treatment of NRCMs with GSK-3Is, such as BIO and CHIR99021, increased the ratio of proliferative cardiomyocytes. YAP was localized in the nuclei of more than 95% of cardiomyocytes, either in the presence or absence of GSK-3Is, indicating that YAP was endogenously activated. GSK-3Is increased the expression of ß-catenin and promoted its translocation into the nucleus without influencing YAP activity. The knockdown of YAP using siRNA or pharmacological inhibition of YAP using verteporfin or CIL56 dramatically reduced GSK-3I-induced cardiomyocyte proliferation without suppressing ß-catenin activation. Interestingly, the inhibition of GSK-3 also induced the proliferation of hiPSC-CMs under sparse culture conditions, where YAP was constitutively activated. In contrast, under dense culture conditions, in which YAP activity was suppressed, the proliferative effects of GSK-3Is on hiPSC-CMs were not detected. Importantly, the activation of YAP by the knockdown of α-catenin restored the proproliferative activity of GSK-3Is. Collectively, YAP activation potentiates the GSK-3I-induced proliferation of cardiomyocytes. The blockade of GSK-3 in combination with YAP activation resulted in remarkable amplification of cardiomyocytes.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Animals , Cell Proliferation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Induced Pluripotent Stem Cells/metabolism , Mammals/metabolism , Myocytes, Cardiac/metabolism , Rats , YAP-Signaling Proteins , beta Catenin/metabolism
8.
Clin Proteomics ; 19(1): 3, 2022 Jan 11.
Article in English | MEDLINE | ID: mdl-35016606

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) has few objective symptoms, and it is difficult to make an early diagnosis by using existing methods. Therefore, new biomarkers enabling diagnosis of renal dysfunction at an early stage need to be developed. Here, we searched for new biomarkers of CKD by focusing on kidney-derived proteins that could sensitively reflect that organ's disease state. METHODS: To identify candidate marker proteins, we performed a proteomics analysis on renal influx and efflux blood collected from the same individual. RESULTS: Proteomics analysis revealed 662 proteins in influx blood and 809 in efflux. From these identified proteins, we selected complement C1q as a candidate; the plasma C1q level was significantly elevated in the renal efflux of donors. Moreover, the plasma concentration of C1q in a mouse model of diabetic nephropathy was significantly increased, in association with increases in blood glucose concentration and urinary protein content. Importantly, we demonstrated that the tendency of C1q to increase in the plasma of CKD patients was correlated with a decrease in their estimated glomerular filtration rate. CONCLUSION: Overall, our results indicate that our approach of focusing on kidney-derived proteins is useful for identifying new CKD biomarkers and that C1q has potential as a biomarker of renal function.

9.
Int Heart J ; 62(6): 1297-1304, 2021.
Article in English | MEDLINE | ID: mdl-34853223

ABSTRACT

Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) frequently induce cardiovascular adverse events, though VEGFR-TKIs contribute to the improvement of the prognosis of patients with malignancies. It is widely accepted that VEGFR-TKIs impair left ventricular systolic functions; however, their effects on diastolic functions remain to be fully elucidated. The purpose of this study was to analyze the impact of VEGFR-TKIs on left ventricular diastolic functions. This study was designed as a retrospective single-center cohort study in Japan. We assessed 24 cases who received VEGFR-TKI monotherapy (sunitinib, sorafenib, pazopanib, axitinib) with left ventricular ejection fraction (LVEF) above 50% during the therapy at the Osaka University Hospital from January 2008 to June 2019. Left ventricular diastolic functions were evaluated by the change in echocardiographic parameters before and after the VEGFR-TKI treatment. Both septal e' and lateral e's decreased after treatment (septal e': before, 6.1 ± 1.8; after, 5.0 ± 1.9; n = 21, P < 0.01; lateral e': before, 8.7 ± 2.8; after, 6.9 ± 2.3; n = 21, P < 0.01). E/A declined after VEGFR-TKIs administration, though not statistically significantly. In 20 cases with at least one risk factor for heart failure with preserved ejection fraction (HFpEF), E/A significantly decreased (0.87 ± 0.34 versus 0.68 ± 0.14; P < 0.05) as well as the septal and lateral e's. These results suggest that treatment with VEGFR-TKIs impairs left ventricular diastolic functions in patients with preserved LVEF, especially in those with risk factors for HFpEF.


Subject(s)
Diastole/drug effects , Protein Kinase Inhibitors/adverse effects , Ventricular Dysfunction, Left/chemically induced , Aged , Cohort Studies , Echocardiography , Female , Humans , Male , Neoplasms/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Retrospective Studies , Stroke Volume
11.
Biochem Biophys Res Commun ; 571: 88-95, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34311199

ABSTRACT

The number of patients with chronic kidney disease (CKD) is increasing worldwide. When kidneys are exposed to severe injury, tubular cell death occurs and kidney fibrosis progresses by activating fibroblasts and myofibroblasts (referred to as (myo)fibroblasts), leading to CKD; however, the pathological and molecular mechanisms underlying CKD, including kidney fibrosis, remain obscure. In the present study, we focused on a transcription factor PBX/Knotted Homeobox 2 (PKNOX2) in kidney fibrosis. The transcript and protein expression of PKNOX2 was upregulated in fibrotic kidneys after unilateral ureteral obstruction (UUO). Importantly, immunofluorescence microscopic analysis revealed that the number of PKNOX2-expressing myofibroblasts was increased, whereas the expression of PKNOX2 was decreased in proximal tubular epithelial cells after UUO. In (myo)fibroblasts, PKNOX2 was induced by TGF-ß1. Knockdown of PKNOX2 using shRNA lentiviral system reduced the viability of (myo)fibroblasts either in the presence or absence of TGF-ß1, accompanied by increased apoptosis. Moreover, PKNOX2 knockdown decreased TGF-ß1-induced migration of myofibroblasts and differentiation of fibroblasts into myofibroblasts. Significantly, knockdown of PKNOX2 also decreased the viability and increased apoptosis of tubular epithelial cells. Collectively, PKNOX2 regulates the function of (myo)fibroblasts and the viability of proximal tubular epithelial cells in progression of kidney fibrosis.


Subject(s)
Fibrosis/metabolism , Homeodomain Proteins/metabolism , Kidney Tubules/metabolism , Myofibroblasts/metabolism , Transcription Factors/metabolism , Ureteral Obstruction/metabolism , Animals , Cell Survival , Cells, Cultured , Fibrosis/pathology , Homeodomain Proteins/genetics , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/pathology , Transcription Factors/genetics , Ureteral Obstruction/pathology
12.
Am J Med Genet A ; 185(10): 2895-2902, 2021 10.
Article in English | MEDLINE | ID: mdl-34047014

ABSTRACT

The HECT, C2, and WW domain containing E3 ubiquitin protein ligase 2 gene (HECW2) is involved in protein ubiquitination. Several genes associated with protein ubiquitination have been linked to neurodevelopmental disorders. HECW2-related disorder has been established through the identification of de novo variants in HECW2 in patients with neurodevelopmental disorders with hypotonia, seizures, and absent language. Recently, we identified novel HECW2 variants in four Japanese patients with neurodevelopmental disorders. Regarding motor development, two of the patients cannot walk, whereas the other two can walk with an unsteady gait, owing to hypotonia. All HECW2 variants, including those that were previously reported, are missense, and no loss-of-function variants have been identified. Most of the identified variants are located around the HECT domain. These findings suggest that the dominant negative effects of missense variants around the HECT domain may be the mechanism underlying HECW2-related disorder.


Subject(s)
Muscle Hypotonia/genetics , Neurodevelopmental Disorders/genetics , Seizures/genetics , Ubiquitin-Protein Ligases/genetics , Child , Child, Preschool , Exome/genetics , Female , Humans , Infant , Infant, Newborn , Intellectual Disability/complications , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Intellectual Disability/pathology , Japan/epidemiology , Male , Muscle Hypotonia/complications , Muscle Hypotonia/diagnosis , Muscle Hypotonia/pathology , Mutation, Missense/genetics , Neurodevelopmental Disorders/complications , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/pathology , Seizures/complications , Seizures/diagnosis , Seizures/pathology
13.
Am J Physiol Cell Physiol ; 321(1): C82-C93, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34038245

ABSTRACT

The resolution of inflammation is closely linked with tissue repair. Recent studies have revealed that macrophages suppress inflammatory reactions by producing lipid mediators, called specialized proresolving mediators (SPMs); however, the biological significance of SPMs in tissue repair remains to be fully elucidated in the heart. In this study, we focused on maresin-1 (MaR1) and examined the reparative effects of MaR1 in cardiomyocytes. The treatment with MaR1 increased cell size in cultured neonatal rat cardiomyocytes. Since the expression of fetal cardiac genes was unchanged by MaR1, physiological hypertrophy was induced by MaR1. SR3335, an inhibitor of retinoic acid-related orphan receptor α (RORα), mitigated MaR1-induced cardiomyocyte hypertrophy, consistent with the recent report that RORα is one of MaR1 receptors. Importantly, in response to MaR1, cardiomyocytes produced IGF-1 via RORα. Moreover, MaR1 activated phosphoinositide 3-kinase (PI3K)/Akt signaling pathway and wortmannin, a PI3K inhibitor, or triciribine, an Akt inhibitor, abrogated MaR1-induced cardiomyocyte hypertrophy. Finally, the blockade of IGF-1 receptor by NVP-AEW541 inhibited MaR-1-induced cardiomyocyte hypertrophy as well as the activation of PI3K/Akt pathway. These data indicate that MaR1 induces cardiomyocyte hypertrophy through RORα/IGF-1/PI3K/Akt pathway. Considering that MaR1 is a potent resolving factor, MaR1 could be a key mediator that orchestrates the resolution of inflammation with myocardial repair.


Subject(s)
Cardiomegaly/genetics , Cardiotonic Agents/pharmacology , Docosahexaenoic Acids/adverse effects , Insulin-Like Growth Factor I/genetics , Myocardial Infarction/genetics , Myocytes, Cardiac/drug effects , Paracrine Communication/genetics , Animals , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Disease Models, Animal , Docosahexaenoic Acids/antagonists & inhibitors , Gene Expression Regulation , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Paracrine Communication/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rats , Ribonucleosides/pharmacology , Signal Transduction , Sulfonamides/pharmacology , Thiophenes/pharmacology , Wortmannin/pharmacology
14.
Gene ; 788: 145664, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33887371

ABSTRACT

The angiogenic gene therapy is an attractive approach for the treatment of ischemic muscle diseases, including peripheral arterial disease and ischemic heart diseases. Although a variety of gene transfer methods have been developed, the efficiency of gene transfer is still limited. We have been developing the needleless high-energy bioinjector device, Pyro-drive Jet Injector (PJI), based on pyrotechnics using a combination of ignition powder and gunpowder, however, the utility of PJI in gene transfer into muscle tissues remains unclear. pcDNA3.1 plasmid containing Flag was injected to the thigh muscles of C57BL/6J mice using PJI or needle, as a control. Histological analysis demonstrated that the protein expression of Flag was observed in a wider range in PJI group than in needle group. To assess the validity of PJI for gene therapy, pcDNA3.1-human fibroblast growth factor 2 (FGF2), which has angiogenic activity and tissue protective properties, was injected into the ischemic thigh muscles with PJI or needle. ELISA assay revealed that the protein expression of FGF2 was increased in the thigh muscle tissues by PJI-mediated gene delivery. Significantly, histological analyses revealed that muscle fiber cross-sectional area and the number of endothelial marker CD31 (+) cells was increased in ischemic hind-limb tissues of the PJI-FGF2 group but not in those of needle-FGF2 group. To expand the applicability of the PJI-mediated gene transfer, pcDNA3.1-venus plasmid was injected into murine hearts with PJI or needle. PJI method was successful in gene transfer into murine hearts, especially into cardiomyocytes, with high efficiency when compared to needle method. Collectively, the non-needle, non-liposomal and non-viral gene transfer by PJI could be a novel therapeutic approach for muscle diseases.


Subject(s)
Fibroblast Growth Factor 2/administration & dosage , Gene Transfer Techniques/instrumentation , Muscle, Skeletal/metabolism , Myocardium/metabolism , Animals , Cell Line , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Genetic Therapy/instrumentation , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacology , Hindlimb , Humans , Male , Mice , Mice, Inbred C57BL , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/therapy , Plasmids/genetics
15.
FASEB J ; 35(2): e21158, 2021 02.
Article in English | MEDLINE | ID: mdl-33150680

ABSTRACT

Prevention of kidney fibrosis is an essential requisite for effective therapy in preventing chronic kidney disease (CKD). Here, we identify Old astrocyte specifically induced substance (OASIS)/cAMP responsive element-binding protein 3-like 1 (CREB3l1), a CREB/ATF family transcription factor, as a candidate profibrotic gene that drives the final common pathological step along the fibrotic pathway in CKD. Although microarray data from diseased patient kidneys and fibrotic mouse model kidneys both exhibit OASIS/Creb3l1 upregulation, the pathophysiological roles of OASIS in CKD remains unknown. Immunohistochemistry revealed that OASIS protein was overexpressed in human fibrotic kidney compared with normal kidney. Moreover, OASIS was upregulated in murine fibrotic kidneys, following unilateral ureteral obstruction (UUO), resulting in an increase in the number of OASIS-expressing pathological myofibroblasts. In vitro assays revealed exogenous TGF-ß1 increased OASIS expression coincident with fibroblast-to-myofibroblast transition and OASIS contributed to TGF-ß1-mediated myofibroblast migration and increased proliferation. Significantly, in vivo kidney fibrosis induced via UUO or ischemia/reperfusion injury was ameliorated by systemic genetic knockout of OASIS, accompanied by reduced myofibroblast proliferation. Microarrays revealed that the transmembrane glycoprotein Bone marrow stromal antigen 2 (Bst2) expression was reduced in OASIS knockout myofibroblasts. Interestingly, a systemic anti-Bst2 blocking antibody approach attenuated kidney fibrosis in normal mice but not in OASIS knockout mice after UUO, signifying Bst2 functions downstream of OASIS. Finally, myofibroblast-restricted OASIS conditional knockouts resulted in resistance to kidney fibrosis. Taken together, OASIS in myofibroblasts promotes kidney fibrosis, at least in part, via increased Bst2 expression. Thus, we have identified and demonstrated that OASIS signaling is a novel regulator of kidney fibrosis.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Kidney/metabolism , Kidney/pathology , Nerve Tissue Proteins/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Antigens, CD/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Disease Models, Animal , Fibrosis , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/metabolism , Nerve Tissue Proteins/genetics , Signal Transduction/genetics , Transfection , Up-Regulation/genetics
16.
Pharmacol Res Perspect ; 8(2): e00590, 2020 04.
Article in English | MEDLINE | ID: mdl-32302067

ABSTRACT

BACKGROUND AND PURPOSE: In cardiovascular diseases, cardiac fibroblasts (CFs) participate in the myocardial inflammation by producing pro-inflammatory cytokines, worsening the prognosis. ß2-adrenergic receptor (AR) and ß3AR are expressed in CFs, and ß-adrenergic stimulation promotes CFs to produce pro-inflammatory cytokines. However, the mechanism of the expression of pro-inflammatory cytokines in response to ß-adrenergic stimulation remains to be fully elucidated. EXPERIMENTAL APPROACH: CFs were isolated from adult wild-type or AT-rich interactive domain-containing protein 5A (Arid5a) knockout mice. The expression of mRNA was measured by real-time RT-PCR. Interleukin (IL)-6 protein was measured by ELISA. The activity of nuclear factor-κB (NF-κB) and cyclic AMP (cAMP) response element binding protein (CREB) was assessed by ELISA-like assay or Western blotting. KEY RESULTS: The ß-adrenergic stimulation remarkably induced IL-6 mRNA and protein through ß2AR in CFs. The activation of adenylate cyclase and the enhancement of intracellular cAMP resulted in the upregulation of IL-6 mRNA expression. The induction of IL-6 transcript by ß2AR signaling was independent of NF-κB. Concomitant with IL-6, the expression of Arid5a, an IL-6 mRNA stabilizing factor, was enhanced by ß2-adrenergic stimulation and by cAMP increase. Importantly, ß2AR signaling-mediated IL-6 induction was suppressed in Arid5a knockout CFs. Finally, ß2AR stimulation phosphorylated CREB via PKA pathway, and the activation of CREB was essential for the induction of Arid5a and IL-6 mRNA. CONCLUSION AND IMPLICATIONS: ß2-adrenergic stimulation post-transcriptionally upregulates the expression of IL-6 by the induction of Arid5a through cAMP/PKA/CREB pathway in adult CFs. ß2AR/Arid5a/IL-6 axis could be a therapeutic target against cardiac inflammation.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , Interleukin-6/genetics , Receptors, Adrenergic, beta-2/metabolism , Transcription Factors/genetics , Adrenergic beta-Agonists/pharmacology , Animals , Female , Isoproterenol/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardium/cytology , RNA, Messenger/metabolism , Signal Transduction , Transcription Factor RelA/metabolism
17.
Eur J Clin Pharmacol ; 75(7): 901-911, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30852642

ABSTRACT

PURPOSE: Large inter-individual differences in warfarin maintenance dose are mostly due to the effect of genetic polymorphisms in multiple genes, including vitamin K epoxide reductase complex 1 (VKORC1), cytochromes P450 2C9 (CYP2C9), and cytochrome P450 4F2 (CYP4F2). Thus, several algorithms for predicting the warfarin dose based on pharmacogenomics data with clinical characteristics have been proposed. Although these algorithms consider these genetic polymorphisms, the formulas have different coefficient values that are critical in this context. In this study, we assessed the mutual validity among these algorithms by specifically considering racial differences. METHODS: Clinical data including actual warfarin dose (AWD) of 125 Japanese patients from our previous study (Eur J Clin Pharmacol 65(11):1097-1103, 2009) were used as registered data that provided patient characteristics, including age, sex, height, weight, and concomitant medications, as well as the genotypes of CYP2C9 and VKORC1. Genotyping for CYP4F2*3 was performed by the PCR method. Five algorithms that included these factors were selected from peer-reviewed articles. The selection covered four populations, Japanese, Chinese, Caucasian, and African-American, and the International Warfarin Pharmacogenetics Consortium (IWPC). RESULTS: For each algorithm, we calculated individual warfarin doses for 125 subjects and statistically evaluated its performance. The algorithm from the IWPC had the statistically highest correlation with the AWD. Importantly, the calculated warfarin dose (CWD) using the algorithm from African-Americans was less correlated with the AWD as compared to those using the other algorithms. The integration of CYP4F2 data into the algorithm did not improve the prediction accuracy. CONCLUSION: The racial difference is a critical factor for warfarin dose predictions based on pharmacogenomics.


Subject(s)
Algorithms , Anticoagulants/administration & dosage , Asian People/genetics , Cytochrome P-450 CYP2C9/genetics , Cytochrome P450 Family 4/genetics , Vitamin K Epoxide Reductases/genetics , Warfarin/administration & dosage , Adult , Aged , Aged, 80 and over , Female , Genotype , Humans , Male , Middle Aged , Pharmacogenetics
18.
Biochem Biophys Res Commun ; 510(1): 149-155, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30683314

ABSTRACT

Abnormal ß-adrenergic signaling plays a central role in human heart failure. In mice, chronic ß-adrenergic receptor (ßAR) stimulation elicits cardiac hypertrophy. It has been reported that cultured cardiac fibroblasts express ßAR; however, the functional in vivo requirement of ßAR signaling in cardiac fibroblasts during the development of cardiac hypertrophy remains elusive. ß2AR null mice exhibited attenuated hypertrophic responses to chronic ßAR stimulation upon continuous infusion of an agonist, isoprenaline (ISO), compared to those in wildtype controls, suggesting that ß2AR activation in the heart induces pro-hypertrophic effects in mice. Since ß2AR signaling is protective in cardiomyocytes, we focused on ß2AR signaling in cardiac myofibroblasts. To determine whether ß2AR signaling in myofibroblasts affects cardiac hypertrophy, we generated myofibroblast-specific transgenic mice (TG) with the catalytic subunit of protein kinase A (PKAcα) using Cre-loxP system. Myofibroblast-specific PKAcα overexpression resulted in enhanced heart weight normalized to body weight ratio, associated with an enlargement of cardiomyocytes at 12 weeks of age, indicating that myofibroblast-specific activation of PKA mediates cardiac hypertrophy in mice. Neonatal rat cardiomyocytes stimulated with conditioned media from TG cardiac fibroblasts likewise exhibited significantly more growth than those from controls. Thus, ß2AR signaling in myofibroblasts plays a substantial role in ISO-induced cardiac hypertrophy, possibly due to a paracrine effect. ß2AR signaling in cardiac myofibroblasts may represent a promising target for development of novel therapies for cardiac hypertrophy.


Subject(s)
Cardiomegaly/etiology , Myofibroblasts/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/adverse effects , Cyclic AMP-Dependent Protein Kinases/genetics , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Paracrine Communication , Rats
19.
Cardiovasc Res ; 115(4): 765-775, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30307485

ABSTRACT

AIMS: Accumulating evidence demonstrates that cardiomyocyte death contributes to the onset and progression of heart failure (HF) after myocardial injury. Recent studies revealed that immune/inflammatory reactions play important roles in cardiovascular diseases. However, it remains unclear whether immunosurveillance system, which eliminates cytopathic cells, including infected or malignant cancer cells, is involved in cardiomyocyte death, though cardiomyocytes are exposed to pathological stresses during post-infarct remodelling. The aim of this study is to clarify the pathophysiological significance of Natural Killer Group 2 member D (NKG2D)/NKG2D ligand (NKG2DL)-mediated cell death in HF after myocardial infarction (MI). METHODS AND RESULTS: MI was generated by ligating left anterior descending artery in mice. The expression of NKG2D, NKG2DLs, especially Retinoic acid early induced transcript-1ɛ (Rae-1ɛ), perforin and granzyme B was concomitantly up-regulated after MI. Immunohistological analysis revealed that Rae-1 was expressed on the membranes of injured cardiomyocytes in the infarct and border area. The MI-induced increase of Rae-1 expression was suppressed in p53-/- mice and Rae-1 was induced by the overexpression of p53. We identified p53-binding sites in Rae-1ɛ gene promoter, by chromatin immunoprecipitation assay, indicating that Rae-1 expression was mediated partially through p53. Flow cytometric analysis indicated that NKG2D-expressing immune cells in the post-infarct myocardium were mainly γδT cells. The co-culture with γδT cells increased the frequency of apoptotic cells in the cultured cardiomyocytes. The blockade of NKG2D/NKG2DL interaction by intraperitoneal injection of anti-Rae-1ɛ antibody after MI reduced the frequency of apoptotic cardiomyocytes, accompanied by suppression of cardiac fibrosis, attenuating cardiac dysfunction. Finally, tamoxifen-inducible cardiomyocyte-specific Rae-1ɛ overexpressing mice exhibited the susceptibility to post-infarct remodelling with increased cardiomyocyte apoptosis and severer cardiac dysfunction. CONCLUSION: The interaction between immune cells and cardiomyocytes via NKG2D/NKG2DL induces cardiomyocyte death, exacerbating cardiac remodelling after MI. The blockade of NKG2D/NKG2DL interaction could be a promising therapeutic strategy against HF.


Subject(s)
Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/metabolism , Ventricular Remodeling , Animals , Apoptosis , Cell Communication , Cell Line , Coculture Techniques , Disease Models, Animal , Granzymes/genetics , Granzymes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , NK Cell Lectin-Like Receptor Subfamily K/genetics , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Signal Transduction , T-Lymphocyte Subsets/immunology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
20.
Oncotarget ; 9(101): 37520-37533, 2018 Dec 25.
Article in English | MEDLINE | ID: mdl-30680067

ABSTRACT

The acquisition of resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs) is one of the major problems in the pharmacotherapy against non-small cell lung cancers; however, molecular mechanisms remain to be fully elucidated. Here, using a newly-established erlotinib-resistant cell line, PC9/ER, from PC9 lung cancer cells, we demonstrated that the expression of translation-related molecules, including eukaryotic translation initiation factor 3 subunit C (eIF3c), was upregulated in PC9/ER cells by proteome analyses. Immunoblot analyses confirmed that eIF3c protein increased in PC9/ER cells, compared with PC9 cells. Importantly, the knockdown of eIF3c with its siRNAs enhanced the drug sensitivity in PC9/ER cells. Mechanistically, we found that LC3B-II was upregulated in PC9/ER cells, while downregulated by the knockdown of eIF3c. Consistently, the overexpression of eIF3c increased the number of autophagosomes, proposing the causality between eIF3c expression and autophagy. Moreover, chloroquine, an autophagy inhibitor, restored the sensitivity to erlotinib. Finally, immunohistochemical analyses of biopsy samples showed that the frequency of eIF3c-positive cases was higher in the patients with EGFR-TKI resistance than those prior to EGFR-TKI treatment. Moreover, the eIF3c-positive cases exhibited poor prognosis in EGFR-TKI treatment. Collectively, the upregulation of eIF3c could impair the sensitivity to EGFR-TKI as a novel mechanism of the drug resistance.

SELECTION OF CITATIONS
SEARCH DETAIL
...