Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 116
1.
Biochem Pharmacol ; 224: 116232, 2024 Jun.
Article En | MEDLINE | ID: mdl-38648905

Heart failure (HF) is a major global health problem afflicting millions worldwide. Despite the significant advances in therapies and prevention, HF still carries very high morbidity and mortality, requiring enormous healthcare-related expenditure, and the search for new weapons goes on. Following initial treatment strategies targeting inotropism and congestion, attention has focused on offsetting the neurohormonal overactivation and three main therapies, including angiotensin-converting enzyme inhibitors or angiotensin II type 1 receptor antagonists, ß-adrenoceptor antagonists, and mineralocorticoid receptor antagonists, have been the foundation of standard treatment for patients with HF. Recently, a paradigm shift, including angiotensin receptor-neprilysin inhibitor, sodium glucose co-transporter 2 inhibitor, and ivabradine, has been added. Moreover, soluble guanylate cyclase stimulator, elamipretide, and omecamtiv mecarbil have come out as a next-generation therapeutic agent for patients with HF. Although these pharmacologic therapies have been significantly successful in relieving symptoms, there is still no complete cure for HF. We may be currently entering a new era of treatment for HF with animal experiments and human clinical trials assessing the value of antibody-based immunotherapy and gene therapy as a novel therapeutic strategy. Such tempting therapies still have some challenges to be addressed but may become a weighty option for treatment of HF. This review article will compile the paradigm shifts in HF treatment over the past dozen years or so and illustrate current landscape of antibody-based immunotherapy and gene therapy as a new therapeutic algorithm for patients with HF.


Heart Failure , Humans , Heart Failure/drug therapy , Heart Failure/therapy , Animals , Angiotensin-Converting Enzyme Inhibitors/therapeutic use
2.
Nihon Yakurigaku Zasshi ; 159(2): 100, 2024.
Article Ja | MEDLINE | ID: mdl-38432916
3.
Nihon Yakurigaku Zasshi ; 159(2): 101-106, 2024.
Article Ja | MEDLINE | ID: mdl-38432917

Sepsis is defined as the body's overwhelming and life-threatening response to infection that can lead to tissue damage, organ failure, and death. Since bacterial infection is one of the main causes of sepsis, appropriate antimicrobial therapy remains the cornerstone of sepsis and septic shock management. However, since sepsis is a multifaceted chaos involving inflammation and anti-inflammation disbalance leading to the unregulated widespread release of inflammatory mediators, cytokines, and pathogen-related molecules leading to system-wide organ dysfunction, the whole body control to prevent the progression of organ dysfunction is needed. In sepsis and septic shock, pathogen-associated molecular patterns (PAMPs), such as bacterial exotoxins, cause direct cellular damage and/or trigger an immune response in the host. PAMPs are recognized by pattern recognizing receptors (PRRs) expressed on immune-reactive cells. PRRs are also activated by host nuclear, mitochondrial, and cytosolic proteins, known as damage-associated molecular patterns (DAMPs) that are released from cells during sepsis. Thus, most PRRs respond to PAMPs or DAMPs by triggering activation of transcriptional factors, NF-κB, AP1, and STAT-3. On the other hand, sepsis leads to immune (lymphocytes and macrophages) and nonimmune (endothelial and epithelial cells) cell death. Apoptosis has been the major focus of research on cell death in sepsis, but autophagy, necrosis, necroptosis, pyroptosis, NETosis, and ferroptosis may also play an important role in this critical situation. The recent development in our understanding regarding the cellular pathogenesis of sepsis will help in developing new treatment of sepsis.


Sepsis , Shock, Septic , Humans , Multiple Organ Failure/etiology , Pathogen-Associated Molecular Pattern Molecules , Apoptosis , Alarmins
4.
Life Sci ; 330: 122008, 2023 Oct 01.
Article En | MEDLINE | ID: mdl-37549828

AIM: Sepsis-induced cardiac dysfunction is the leading cause of higher morbidity and mortality with poor prognosis in septic patients. Our recent previous investigation provides evidence of the hallmarks of signal transducer and activator of transcription3 (STAT3) activation in sepsis and targeting of STAT3 with Stattic, a small-molecule inhibitor of STAT3, has beneficial effects in various septic tissues. We investigated the possible cardioprotective effects of Stattic on cardiac inflammation and dysfunction in mice with cecal ligation and puncture (CLP)-induced sepsis. MAIN METHODS: A polymicrobial sepsis model was induced by CLP in mice and Stattic (25 mg/kg) was intraperitoneally given at one and twelve hours after CLP operation. The cecum was exposed in sham-control mice without CLP. After 18 h of surgery, electrocardiogram (ECG) for anaesthized mice was registered followed by collecting of samples of blood and tissues for bimolecular and histopathological assessments. Myeloperoxidase, a marker of neutrophil infiltration, was assessed immunohistochemically. KEY FINDINGS: CLP profoundly impaired cardiac functions as evidenced by ECG changes in septic mice as well as elevation of cardiac enzymes, and inflammatory markers with myocardial histopathological and immunohistochemical alterations. While, Stattic markedly reversed the CLP-induced cardiac abnormalities and restored the cardiac function by its anti-inflammatory activities. SIGNIFICANCE: Stattic treatment had potential beneficial effects against sepsis-induced cardiac inflammation, dysfunction and damage. Its cardioprotective effects were possibly attributed to its anti-inflammatory activities by targeting STAT3 and downregulation of IL-6 and gp130. Our investigations suggest that Stattic could be a promising target for management of cardiac sepsis and inflammation-related cardiac damage.


Heart Injuries , Sepsis , Animals , Mice , Anti-Inflammatory Agents/pharmacology , Cecum/surgery , Cecum/injuries , Cytokine Receptor gp130/metabolism , Inflammation/etiology , Interleukin-6/metabolism , Ligation/adverse effects , Punctures/adverse effects , Sepsis/drug therapy , Signal Transduction
5.
Naunyn Schmiedebergs Arch Pharmacol ; 396(1): 35-42, 2023 01.
Article En | MEDLINE | ID: mdl-36282300

The history of hitherto existing pharmacology in Japan presented here is authored in commemoration of the 150th anniversary of Naunyn-Schmiedeberg's Archives of Pharmacology. After the publication of the new book of anatomy "Anatomische Tabellen" translated into Japanese in 1774, the foundation of understanding the medical science was gradually formed in Japan under seclusion policy, and, since the Meiji Restoration of 1868, the modernization of Japanese medicine was rapidly fostered on the basis of German medicine. Thus, the Japanese government officially adopted German medicine, and the philosophy and practice of German medical schools were incorporated. Most of the medical texts used in Japan were of German origins, often in Dutch translations, and many Japanese physicians and medical researchers studied abroad in Germany. The start of experimental pharmacology in Japan was also made up by Japanese disciples of Oswald Schmiedeberg, who was the one of founders of the Archives in 1873. Additionally, it was customary for professor candidates in charge of pharmacology in medical faculties in Japan to go to Germany and study pharmacology. Through such historical circumstances, the Japanese Pharmacology Society has been established to fulfill the responsibility for contributing internationally to world-class research achievements in the field of medical sciences by supplying numerous talented pharmacologists. During the course of the development of experimental pharmacology in Japan, the Archives has provided an excellent stage for many Japanese pharmacologists to publish their research outcomes to proliferate them internationally. Without German medicine influence, Japanese pharmacology would not have been what it is today.


Pharmacology , Physicians , Humans , Germany , History, 20th Century , Japan
7.
Nihon Yakurigaku Zasshi ; 157(4): 226-231, 2022.
Article Ja | MEDLINE | ID: mdl-35781449

The acute respiratory distress syndrome (ARDS) is an important cause of respiratory failure in critically ill patients and may become a life-threatening condition where inflammation of the lungs may begin in one lung but eventually affects both, leading to damage to the alveoli and surrounding small blood vessels. ARDS is particularly characterized by noncardiogenic pulmonary edema caused by an increase in pulmonary capillary permeability. Several clinical disorders can precipitate in ARDS, including pneumonia, sepsis, aspiration of gastric contents, and major trauma. The most common cause of ARDS is sepsis, which is a serious and widespread infection of the bloodstream and is now defined as life-threatening organ dysfunction due to a dysregulated reponse of the host to infection. In sepsis, a number of vascular hyperpermeable factors, such as histamine, nitric oxide, thromboxane A2, and vascular endothelial growth factor, can be overproducted and contribute to the development of pulmonary edema. Given that sepsis can be regarded as a gene-related disorder, the nucleic-acid based gene therapeutic strategy to regulate some transcription factors involved in expression of vascular hyperpermeable genes may be considered to be a promising novel approach for treatment of ARDS in sepsis.


Pulmonary Edema , Respiratory Distress Syndrome , Sepsis , Humans , Lung , Pulmonary Edema/etiology , Respiratory Distress Syndrome/etiology , Sepsis/complications , Vascular Endothelial Growth Factor A
8.
Biochem Pharmacol ; 197: 114909, 2022 03.
Article En | MEDLINE | ID: mdl-35021044

Vascular endothelial cells are major participants in and regulators of immune responses and inflammation. Vascular endotheliitis is regarded as a host immune-inflammatory response of the endothelium forming the inner surface of blood vessels in association with a direct consequence of infectious pathogen invasion. Vascular endotheliitis and consequent endothelial dysfunction can be a principle determinant of microvascular failure, which would favor impaired perfusion, tissue hypoxia, and subsequent organ failure. Emerging evidence suggests the role of vascular endotheliitis in the pathogenesis of coronavirus disease 2019 (COVID-19) and its related complications. Thus, once initiated, vascular endotheliitis and resultant cytokine storm cause systemic hyperinflammation and a thrombotic phenomenon in COVID-19, leading to acute respiratory distress syndrome and widespread organ damage. Vascular endotheliitis also appears to be a contributory factor to vasculopathy and coagulopathy in sepsis that is defined as life-threatening organ dysfunction due to a dysregulated response of the host to infection. Therefore, protecting endothelial cells and reversing vascular endotheliitis may be a leading therapeutic goal for these diseases associated with vascular endotheliitis. In this review, we outline the etiological and pathogenic importance of vascular endotheliitis in infection-related inflammatory diseases, including COVID-19, and possible mechanisms leading to vascular endotheliitis. We also discuss pharmacological agents which may be now considered as potential endotheliitis-based treatment modalities for those diseases.


COVID-19/pathology , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Vascular Diseases/pathology , COVID-19/complications , COVID-19/immunology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Sepsis/drug therapy , Sepsis/etiology , Sepsis/immunology , Sepsis/pathology , Vascular Diseases/drug therapy , Vascular Diseases/etiology , Vascular Diseases/immunology , COVID-19 Drug Treatment
9.
Naunyn Schmiedebergs Arch Pharmacol ; 394(10): 2129-2139, 2021 10.
Article En | MEDLINE | ID: mdl-34402957

Cumulative evidence has established that macrophages orchestrate inflammatory responses that crucially contribute to the pathogenesis of insulin-resistant obesity and type 2 diabetes. In the present study, we examined the impact of hyperglycemia on macrophage pro-inflammatory responses under an inflammatory stimulus. To conduct this study, RAW264.7 macrophages were cultured under normal- (5.5 mM) or high-glucose (22 or 40 mM) conditions for 7 days and stimulated with lipopolysaccharide (LPS). Long-term exposure to high glucose significantly enhanced the increase in the production of pro-inflammatory cytokines, including tumor necrosis-α, interleukin (IL)-1ß, and IL-6, when macrophages were stimulated with LPS. The LPS-induced increases in inducible nitric oxide (NO) synthase (iNOS) expression and NO production were also significantly enhanced by long-term exposure of macrophages to high glucose. Treatment with N-acetyl-L-cysteine, a widely used thiol-containing antioxidant, blunted the enhancement of the LPS-induced upregulation of pro-inflammatory cytokine production, iNOS expression, and NO production in macrophages. When intracellular reactive oxygen species (ROS) were visualized using the fluorescence dye 5-(and-6)-chloromethyl-2',7'-dichlorofluorescein diacetate, acetyl ester, a significant increase in ROS generation was found after stimulation of macrophages with LPS, and this increased ROS generation was exacerbated under long-term high-glucose conditions. LPS-induced translocation of phosphorylated nuclear factor-κB (NF-κB), a transcription factor regulating many pro-inflammatory genes, into the nucleus was promoted under long-term high-glucose conditions. Altogether, the present results indicate that a long-term high-glucose environment can enhance activation of NF-κB in LPS-stimulated macrophages possibly due to excessive ROS production, thereby leading to increased macrophage pro-inflammatory responses.


Glucose/pharmacology , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Animals , Cytokines/metabolism , Heme Oxygenase-1/metabolism , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Membrane Proteins/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells
10.
Intern Med ; 60(5): 755-759, 2021 Mar 01.
Article En | MEDLINE | ID: mdl-33028772

Keishibukuryogan is a Kampo medicine that induces vasodilation and improves the blood flow velocity in subcutaneous blood vessels. We herein report two cases in which keishibukuryogan completely diminished subcutaneous hematoma after cardiac resynchronization therapy pacemaker implantation and defibrillator battery replacement within a month. Keishibukuryogan can be a good option for treating or preventing subcutaneous hematoma after surgical procedures for devices.


Cardiac Resynchronization Therapy , Defibrillators, Implantable , Drugs, Chinese Herbal , Pacemaker, Artificial , Defibrillators, Implantable/adverse effects , Hematoma/diagnostic imaging , Hematoma/etiology , Humans , Pacemaker, Artificial/adverse effects
11.
Sci Rep ; 10(1): 15316, 2020 09 17.
Article En | MEDLINE | ID: mdl-32943679

Sepsis is a major clinical challenge with unacceptably high mortality. The signal transducers and activators of transcription (STAT) family of transcription factors is known to activate critical mediators of cytokine responses, and, among this family, STAT3 is implicated to be a key transcription factor in both immunity and inflammatory pathways. We investigated whether in vivo introduction of synthetic double-stranded STAT3 decoy oligodeoxynucleotides (ODNs) can provide benefits for reducing organ injury and mortality in mice with cecal ligation and puncture (CLP)-induced polymicrobial sepsis. We found that STAT3 was rapidly activated in major end-organ tissues following CLP, which was accompanied by activation of the upstream kinase JAK2. Transfection of STAT3 decoy ODNs downregulated pro-inflammatory cytokine/chemokine overproduction in CLP mice. Moreover, STAT3 decoy ODN transfection significantly reduced the increases in tissue mRNAs and proteins of high mobility group box 1 (HMGB1) and strongly suppressed the excessive elevation in serum HMGB1 levels in CLP mice. Finally, STAT3 decoy ODN administration minimized the development of sepsis-driven major end-organ injury and led to a significant survival advantage in mice after CLP. Our results suggest a critical role of STAT3 in the sepsis pathophysiology and the potential usefulness of STAT3 decoy ODNs for sepsis gene therapy.


Cecum/metabolism , Oligodeoxyribonucleotides/metabolism , STAT3 Transcription Factor/metabolism , Sepsis/metabolism , Animals , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/physiology , HMGB1 Protein/metabolism , Inflammation/metabolism , Janus Kinase 2/metabolism , Ligation/methods , Male , Mice , Mice, Inbred BALB C , Punctures/methods , RNA, Messenger/metabolism , Transfection/methods
12.
Naunyn Schmiedebergs Arch Pharmacol ; 393(12): 2365-2374, 2020 12.
Article En | MEDLINE | ID: mdl-32696151

Vascular endothelial growth factor (VEGF) is a prime regulator of vascular permeability. Acute lung injury (ALI) is characterized by high-permeability pulmonary edema in addition to refractory hypoxemia and diffuse pulmonary infiltrates. In this study, we examined whether VEGF can be implicated as a pulmonary vascular permeability factor in sepsis-associated ALI. We found that a great increase in lung vascular leak occurred in mice instilled intranasally with lipopolysaccharide (LPS), as assessed by IgM levels in bronchoalveolar lavage fluid. Treatment with the VEGF-neutralizing monoclonal antibody bevacizumab significantly reduced this hyperpermeability response, suggesting active participation of VEGF in non-cardiogenic lung edema associated with LPS-induced ALI. However, this was not solely attributable to excessive levels of intrapulmonary VEGF. Expression levels of VEGF were significantly reduced in lung tissues from mice with both intranasal LPS administration and cecal ligation and puncture (CLP)-induced sepsis, which may stem from decreases in non-endothelial cells-dependent VEGF production in the lungs. In support of this assumption, stimulation with LPS and interferon-γ (IFN-γ) significantly increased VEGF in human pulmonary microvascular endothelial cells (HPMECs) at mRNA and protein levels. Furthermore, a significant rise in plasma VEGF levels was observed in CLP-induced septic mice. The increase in VEGF released from HPMECs after LPS/IFN-γ challenge was completely blocked by either specific inhibitor of mitogen-activated protein kinase (MAPK) subgroups. Taken together, our results indicate that VEGF can contribute to the development of non-cardiogenic lung edema in sepsis-associated ALI due to increased VEGF secretion from pulmonary vascular endothelial cells through multiple MAPK-dependent pathways.


Acute Lung Injury/blood , Capillary Permeability/physiology , Lung/blood supply , Lung/metabolism , Sepsis/blood , Vascular Endothelial Growth Factor A/blood , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Cell Line, Transformed , Humans , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Sepsis/complications , Sepsis/pathology
13.
Front Pharmacol ; 10: 59, 2019.
Article En | MEDLINE | ID: mdl-30778300

G protein-coupled receptor kinase 2 (GRK2) is a ubiquitous member of the GRK family that restrains cellular activation by G protein-coupled receptor (GPCR) phosphorylation leading to receptor desensitization and internalization, but has been identified to regulate a variety of signaling molecules, among which may be associated with inflammation. In this study, we attempted to establish the regulatory role of GRK2 in the Toll-like receptor (TLR) signaling pathway for inducible nitric oxide synthase (iNOS) expression in microglial cells. When mouse MG6 cells were stimulated with the TLR4 ligands lipopolysaccharide (LPS) and paclitaxel, we found that interferon regulatory factor 1 (IRF1) protein expression and activation was upregulated, transcription of interferon-ß (IFN-ß) was accelerated, induction/activation of STAT1 and activation of STAT3 were promoted, and subsequently iNOS expression was upregulated. The ablation of GRK2 by small interfering RNAs (siRNAs) not only eliminated TLR4-mediated upregulation of IRF1 protein expression and nuclear translocation but also suppressed the activation of the STAT pathway, resulting in negating the iNOS upregulation. The TLR3-mediated changes in IRF1 and STAT1/3, leading to iNOS induction, were also abrogated by siRNA knockdown of GRK2. Furthermore, transfection of GRK2 siRNA blocked the exogenous IFN-ß supplementation-induced increases in phosphorylation of STAT1 as well as STAT3 and abrogated the augmentation of iNOS expression in the presence of exogenous IFN-ß. Taken together, our results show that GRK2 regulates the activation of IRF1 as well as the activation of the STAT pathway, leading to upregulated transcription of iNOS in activated microglial cells. Modulation of the TLR signaling pathway via GRK2 in microglia may be a novel therapeutic target for treatment of neuroinflammatory disorders.

15.
Immunobiology ; 223(12): 777-785, 2018 12.
Article En | MEDLINE | ID: mdl-30115376

Pulmonary endothelial injury is central in the pathogenesis of acute lung injury (ALI). The MAPK signaling cascades are generally thought to be involved in the molecular mechanism underlying the ALI development, but their roles in pulmonary endothelial injury is poorly understood. We thus examined the involvement of the MAPK family member in inflammatory responses of human pulmonary microvascular endothelial cells (HPMVECs) stimulated with LPS and IFN-γ. HPMVECs were found to exhibit the upregulation of expression of Toll-like receptor 4 by IFN-γ, resulting in potentiation of inflammatory cytokine release by LPS stimulation. All MAPKs, ERK1/2, JNK, and p38, were activated by simultaneous stimulation with LPS/IFN-γ. JNK activation in cells stimulated with LPS/IFN-γ was significantly potentiated by the two different p38 inhibitors, SB203580 and RWJ67657, suggesting the negative regulation of JNK activation by p38 in HPMVECs. The mRNA and protein expression levels of ICAM-1 were eliminated by the JNK inhibitor, suggesting that ICAM-1 expression is positively regulated by JNK. The p38 inhibitor significantly enhanced ICAM-1 expression. ERK1/2 activation was not responsible for the LPS/IFN-γ-induced ICAM-1 upregulation in HPMVECs. THP-1 monocyte adhesion to HPMVECs under LPS/IFN-γ stimulation was inhibited by the JNK inhibitor and enhanced by the p38 inhibitor. We conclude that, in HPMVECs stimulated with LPS/IFN-γ, JNK mediates ICAM-1 expression that can facilitate leukocyte adherence and transmigration, while p38 MAPK negatively regulates the upregulation of ICAM-1 through inhibition of JNK activation.


Endothelial Cells/metabolism , Interferon-gamma/metabolism , Lipopolysaccharides/immunology , Lung/immunology , Lung/metabolism , Mitogen-Activated Protein Kinases/metabolism , Biomarkers , Cell Adhesion/immunology , Cell Line , Cytokines/immunology , Cytokines/metabolism , Endothelial Cells/drug effects , Gene Expression , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Monocytes/immunology , Monocytes/metabolism , Pneumonia/etiology , Pneumonia/metabolism , Pneumonia/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Int Immunopharmacol ; 62: 120-131, 2018 Sep.
Article En | MEDLINE | ID: mdl-30005227

Cilostazol, a selective inhibitor of phosphodiesterase type III with anti-platelet, anti-mitogenic, and vasodilating properties, is widely used to treat ischemic symptoms of peripheral vascular disease. Ample evidence has suggested that cilostazol also exhibits an anti-inflammatory effect, but its anti-inflammatory mechanism is not fully understood. Here, we showed that cilostazol specifically inhibited expression of cytokines, which are induced by nuclear factor-κB (NF-κB) activation, in RAW264.7 macrophage cells stimulated with different Toll-like receptor (TLR) ligands. Cilostazol was found to significantly reduce TLR-4 and TLR-3 ligands-stimulated NF-κB transcriptional activity, which was quantified by luciferase reporter assays. However, cilostazol was without effect on IκBα degradation and NF-κB p65 phosphorylation and nuclear translocation after challenge with the TLR-4 ligand lipopolysaccharide (LPS). Cilostazol did not also prevent the LPS-induced increase in phosphorylated levels of the mitogen-activated protein kinase (MAPK) family. On the other hand, using chromatin immunoprecipitation assays, we demonstrated that cilostazol reduced the LPS-induced transcriptional activities of interleukin-6 and tumor necrosis factor-α by preventing the recruitment of NF-κB p65 to these gene promoters. When cilostazol was given to mice by oral gavage daily for 7 days, LPS-induced aberrant pro-inflammatory cytokine production and end-organ tissue injury were significantly reduced. The results of this study suggest that cilostazol is capable of directly interrupting DNA binding activity of NF-κB proteins from the TLR signaling pathways. The therapy to specifically intervene in this pathway may be potentially beneficial for the prevention of different inflammatory disorders.


Anti-Inflammatory Agents/pharmacology , Cilostazol/pharmacology , DNA/metabolism , NF-kappa B/metabolism , Toll-Like Receptors/metabolism , Animals , Cytokines/genetics , Mice , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/genetics , RAW 264.7 Cells , Signal Transduction , Transcription, Genetic/drug effects
17.
Nihon Yakurigaku Zasshi ; 152(1): 10-15, 2018.
Article Ja | MEDLINE | ID: mdl-29998946

Sepsis is the leading cause of death in critically ill patients, and its incidence continues to rise. Sepsis is now defined as life-threatening organ dysfunction due to a dysregulated host response to infection. Histamine assumes a critical role as a major mediator of many pathologic disorders with inflammation and immune reactions. However, direct evidence has not been provided showing the involvement of histamine in the development of multiple organ dysfunction or failure in sepsis. We have found that sepsis-induced major end-organ (lung, liver, and kidney) injury is attenuated in histidine decarboxylase (HDC) gene knockout mice. H1/H2-receptor gene-double knockout mice apparently behave similar to HDC knockout mice in reducing sepsis-related pathologic changes. Here we provide an overview on the role of endogenous histamine as an aggregating mediator that could contribute to the development of major end-organ injury in sepsis.


Histamine/genetics , Multiple Organ Failure/etiology , Sepsis/complications , Animals , Histidine Decarboxylase/genetics , Mice , Mice, Knockout , Receptors, Histamine H1 , Receptors, Histamine H2/genetics
18.
Eur J Pharmacol ; 833: 403-410, 2018 Aug 15.
Article En | MEDLINE | ID: mdl-29935173

Despite advances in overall medical care, sepsis and its sequelae continue to be an embarrassing clinical entity with an unacceptably high mortality rate. The central reason for high morbidity and high mortality of sepsis and its sequelae is the lack of an effective treatment. Previous clinical trials have largely failed to identify an effective therapeutic target to improve clinical outcomes in sepsis. Thus, the key goal favoring the outcome of septic patients is to devise innovative and evolutionary therapeutic strategies. Gene therapy can be considered as one of the most promising novel therapeutic approaches for nasty disorders. Since a number of transcription factors, such as nuclear factor-κB (NF-κB) and activator protein-1 (AP-1), play a pivotal role in the pathophysiology of sepsis that can be characterized by the induction of multiple genes and their products, sepsis may be regarded as a gene-related disorder and gene therapy may be considered a promising novel therapeutic approach for treatment of sepsis. In this review article, we provide an up-to-date summary of the gene-targeting approaches, which have been developed in animal models of sepsis. Our review sheds light on the molecular basis of sepsis pathology for the development of novel gene therapy approaches and leads to the conclusion that future research efforts may fully take into account gene therapy for the treatment of sepsis.


Genetic Therapy/methods , MicroRNAs/genetics , NF-kappa B/genetics , Sepsis/therapy , Transcription Factor AP-1/genetics , Animals , Apoptosis/genetics , Gene Expression Regulation/genetics , Humans , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/therapeutic use , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Sepsis/epidemiology , Sepsis/genetics , Treatment Outcome
19.
Naunyn Schmiedebergs Arch Pharmacol ; 391(9): 1021-1032, 2018 09.
Article En | MEDLINE | ID: mdl-29922941

Levosimendan and milrinone may be used in place of dobutamine to increase cardiac output in septic patients with a low cardiac output due to impaired cardiac function. The effects of the two inotropic agents on cardiac inflammation and left ventricular (LV) performance were examined in mice with cecal ligation and puncture (CLP)-induced sepsis. CLP mice displayed significant cardiac inflammation, as indicated by highly increased pro-inflammatory cytokines and neutrophil infiltration in myocardial tissues. When continuously given, levosimendan prevented but milrinone exaggerated cardiac inflammation, but they significantly reduced the elevations in plasma cardiac troponin-I and heart-type fatty acid-binding protein, clinical markers of cardiac injury. Echocardiographic assessment of cardiac function showed that the effect of levosimendan, given by an intravenous bolus injection, on LV performance was impaired in CLP mice, whereas milrinone produced inotropic responses equally in sham-operated and CLP mice. A lesser effect of levosimendan on LV performance after CLP was also found in spontaneously beating Langendorff-perfused hearts. In ventricular myocytes isolated from control and CLP mice, levosimendan, but not milrinone, caused a large increase in the L-type calcium current. This study represents that levosimendan and milrinone have cardioprotective properties but provide different advantages and drawbacks to cardiac inflammation/dysfunction in sepsis.


Cardiotonic Agents/therapeutic use , Milrinone/therapeutic use , Sepsis/drug therapy , Simendan/therapeutic use , Animals , Calcium/physiology , Cecum/surgery , Cytokines/genetics , Ligation , Male , Mice, Inbred BALB C , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Sepsis/etiology , Sepsis/genetics , Sepsis/physiopathology , Ventricular Function, Left/drug effects , Wounds, Penetrating/complications
20.
Nihon Yakurigaku Zasshi ; 151(3): 111-116, 2018.
Article Ja | MEDLINE | ID: mdl-29526919

Sepsis is the leading cause of death in critically ill patients, and its incidence continues to rise. Sepsis was defined as a systemic inflammatory response syndrome with an identifiable focus of infection, but therapeutic strategies aimed at eliminating the inflammatory response have only modest clinical benefit. The development of a failure of one or more organs poses a major threat to the survival of patients with sepsis, and mortality in sepsis is most often attributed to multiple organ dysfunction. Accordingly, sepsis has been recently redefined as life-threatening organ dysfunction due to a dysregulated host response to infection. Cardiac dysfunction is a well-recognized important component of septic multiple organ failure and can compromise the balance between oxygen supply and demand, ultimately leading to the development of multiple organ failure. The existence of cardiac dysfunction in sepsis is associated with much higher mortality when compared with septic patients without heart problems. Dobutamine, a ß1-selective adrenoceptor agonist, has been used in septic shock for many years as an only inotrope, but limited clinical outcome measures have been provided as to advisability of the usefulness of dobutamine in septic shock management. Here we provide an overview on the possible mechanisms underlying intrinsic myocardial depression during sepsis and discuss the perspective of several inotropes for sepsis-associated cardiac dysfunction.


Cardiomyopathies/diet therapy , Cardiomyopathies/metabolism , Shock, Septic/metabolism , Animals , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Humans , Molecular Targeted Therapy , Shock, Septic/etiology
...