Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Int Immunopharmacol ; 132: 111870, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38547771

ABSTRACT

Extracellular histones have been determined as important mediators of sepsis, which induce excessive inflammatory responses in macrophages and impair innate immunity. Magnesium (Mg2+), one of the essential nutrients of the human body, contributes to the proper regulation of immune function. However, no reports indicate whether extracellular histones affect survival and bacterial phagocytosis in macrophages and whether Mg2+ is protective against histone-induced macrophage damage. Our clinical data revealed a negative correlation between circulating histone and monocyte levels in septic patients, and in vitro experiments confirmed that histones induced mitochondria-associated apoptosis and defective bacterial phagocytosis in macrophages. Interestingly, our clinical data also indicated an association between lower serum Mg2+ levels and reduced monocyte levels in septic patients. Moreover, in vitro experiments demonstrated that Mg2+ attenuated histone-induced apoptosis and defective bacterial phagocytosis in macrophages through the PLC/IP3R/STIM-mediated calcium signaling pathway. Importantly, further animal experiments proved that Mg2+ significantly improved survival and attenuated histone-mediated lung injury and macrophage damage in histone-stimulated mice. Additionally, in a cecal ligation and puncture (CLP) + histone-induced injury mouse model, Mg2+ inhibited histone-mediated apoptosis and defective phagocytosis in macrophages and further reduced bacterial load. Overall, these results suggest that Mg2+ supplementation may be a promising treatment for extracellular histone-mediated macrophage damage in sepsis.


Subject(s)
Apoptosis , Calcium Signaling , Histones , Macrophages , Magnesium , Mice, Inbred C57BL , Phagocytosis , Sepsis , Animals , Phagocytosis/drug effects , Apoptosis/drug effects , Magnesium/metabolism , Histones/metabolism , Humans , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Sepsis/immunology , Sepsis/drug therapy , Sepsis/metabolism , Mice , Male , Calcium Signaling/drug effects , Female , Middle Aged , RAW 264.7 Cells
2.
J Control Release ; 369: 215-230, 2024 May.
Article in English | MEDLINE | ID: mdl-38508529

ABSTRACT

In the progression of acute inflammation, the activation and recruitment of macrophages and neutrophils are mutually reinforcing, leading to amplified inflammatory response and severe tissue damage. Therefore, to regulate the axis of neutrophils and macrophages is essential to avoid tissue damage induced from acute inflammatory. Apoptotic neutrophils can regulate the anti-inflammatory activity of macrophages through the efferocytosis. The strategy of in situ targeting and inducing neutrophil apoptosis has the potential to modulate macrophage activity and transfer anti-inflammatory drugs. Herein, a natural glycyrrhiza protein nanoparticle loaded with dexamethasone (Dex@GNPs) was constructed, which could simultaneously regulate neutrophil and macrophage function during acute inflammation treatment by combining in situ neutrophil apoptosis and macrophage efferocytosis. Dex@GNPs can be rapidly and selectively internalized by neutrophils and subsequently induce neutrophils apoptosis through a ROS-dependent mechanism. The efferocytosis of apoptotic neutrophils not only promoted the polarization of macrophages into anti-inflammatory state, but also facilitated the transfer of Dex@GNPs to macrophages. This enabled dexamethasone to further modulate macrophage function. In mouse models of acute respiratory distress syndrome and sepsis, Dex@GNPs significantly ameliorated the disordered immune microenvironment and alleviated tissue injury. This study presents a novel strategy for drug delivery and inflammation regulation to effectively treat acute inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents , Apoptosis , Dexamethasone , Glycyrrhiza , Inflammation , Macrophages , Nanoparticles , Neutrophils , Animals , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Apoptosis/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Nanoparticles/chemistry , Macrophages/drug effects , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Glycyrrhiza/chemistry , Mice, Inbred C57BL , Male , Mice , Phagocytosis/drug effects , Humans , Sepsis/drug therapy , Sepsis/immunology , Respiratory Distress Syndrome/drug therapy , RAW 264.7 Cells , Efferocytosis
3.
Thorax ; 78(4): 383-393, 2023 04.
Article in English | MEDLINE | ID: mdl-35354645

ABSTRACT

BACKGROUND: One hallmark of sepsis is the reduced number of lymphocytes, termed lymphopenia, that occurs from decreased lymphocyte proliferation or increased cell death contributing to immune suppression. Histone modification enzymes regulate immunity by their epigenetic and non-epigenetic functions; however, the role of these enzymes in lymphopenia remains elusive. METHODS: We used molecular biological approaches to investigate the high expression and function of a chromatin modulator protein arginine N-methyltransferase 4 (PRMT4)/coactivator-associated arginine methyltransferase 1 in human samples from septic patients and cellular and animal septic models. RESULTS: We identified that PRMT4 is elevated systemically in septic patients and experimental sepsis. Gram-negative bacteria and their derived endotoxin lipopolysaccharide (LPS) increased PRMT4 in B and T lymphocytes and THP-1 monocytes. Single-cell RNA sequencing results indicate an increase of PRMT4 gene expression in activated T lymphocytes. Augmented PRMT4 is crucial for inducing lymphocyte apoptosis but not monocyte THP-1 cells. Ectopic expression of PRMT4 protein caused substantial lymphocyte death via caspase 3-mediated cell death signalling, and knockout of PRMT4 abolished LPS-mediated lymphocyte death. PRMT4 inhibition with a small molecule compound attenuated lymphocyte death in complementary models of sepsis. CONCLUSIONS: These findings demonstrate a previously uncharacterised role of a key chromatin modulator in lymphocyte survival that may shed light on devising therapeutic modalities to lessen the severity of septic immunosuppression.


Subject(s)
Lymphopenia , Protein-Arginine N-Methyltransferases , Sepsis , Animals , Humans , Arginine/genetics , Caspase 3/genetics , Caspase 3/immunology , Chromatin , Lipopolysaccharides/pharmacology , Lymphopenia/etiology , Lymphopenia/genetics , Lymphopenia/immunology , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Sepsis/complications , Sepsis/genetics , Sepsis/immunology
4.
Int J Mol Sci ; 23(3)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35163625

ABSTRACT

In addition to being a leading cause of morbidity and mortality worldwide, sepsis is also the most common cause of acute kidney injury (AKI). When sepsis leads to the development of AKI, mortality increases dramatically. Since the cardinal feature of sepsis is a dysregulated host response to infection, a disruption of kidney-immune crosstalk is likely to be contributing to worsening prognosis in sepsis with acute kidney injury. Since immune-mediated injury to the kidney could disrupt its protein manufacturing capacity, an investigation of molecules mediating this crosstalk not only helps us understand the sepsis immune response, but also suggests that their supplementation could have a therapeutic effect. Erythropoietin, vitamin D and uromodulin are known to mediate kidney-immune crosstalk and their disrupted production could impact morbidity and mortality in sepsis with acute kidney injury.


Subject(s)
Acute Kidney Injury/etiology , Immune System , Kidney , Sepsis/complications , Animals , Erythropoietin , Humans , Sepsis/immunology , Sepsis/physiopathology , Uromodulin , Vitamin D
5.
Int J Mol Sci ; 22(23)2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34884728

ABSTRACT

S100A9, a pro-inflammatory alarmin, is up-regulated in inflamed tissues. However, the role of S100A9 in regulating neutrophil activation, inflammation and lung damage in sepsis is not known. Herein, we hypothesized that blocking S100A9 function may attenuate neutrophil recruitment in septic lung injury. Male C57BL/6 mice were pretreated with the S100A9 inhibitor ABR-238901 (10 mg/kg), prior to cercal ligation and puncture (CLP). Bronchoalveolar lavage fluid (BALF) and lung tissue were harvested for analysis of neutrophil infiltration as well as edema and CXC chemokine production. Blood was collected for analysis of membrane-activated complex-1 (Mac-1) expression on neutrophils as well as CXC chemokines and IL-6 in plasma. Induction of CLP markedly increased plasma levels of S100A9. ABR-238901 decreased CLP-induced neutrophil infiltration and edema formation in the lung. In addition, inhibition of S100A9 decreased the CLP-induced up-regulation of Mac-1 on neutrophils. Administration of ABR-238901 also inhibited the CLP-induced increase of CXCL-1, CXCL-2 and IL-6 in plasma and lungs. Our results suggest that S100A9 promotes neutrophil activation and pulmonary accumulation in sepsis. Targeting S100A9 function decreased formation of CXC chemokines in circulation and lungs and attenuated sepsis-induced lung damage. These novel findings suggest that S100A9 plays an important pro-inflammatory role in sepsis and could be a useful target to protect against the excessive inflammation and lung damage associated with the disease.


Subject(s)
Acute Lung Injury/prevention & control , Calgranulin B/metabolism , Neutrophil Infiltration/drug effects , Sepsis/complications , Sulfonamides/therapeutic use , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Animals , Chemokines, CXC/metabolism , Drug Evaluation, Preclinical , Interleukin-6/metabolism , Lung/metabolism , Male , Mice, Inbred C57BL , Sepsis/immunology , Sepsis/metabolism , Sulfonamides/pharmacology
6.
Front Immunol ; 12: 778830, 2021.
Article in English | MEDLINE | ID: mdl-34777396

ABSTRACT

Pathogenic inflammation and immuno-suppression are cardinal features of exhausted monocytes increasingly recognized in septic patients and murine models of sepsis. However, underlying mechanisms responsible for the generation of exhausted monocytes have not been addressed. In this report, we examined the generation of exhausted primary murine monocytes through prolonged and repetitive challenges with high dose bacterial endotoxin lipopolysaccharide (LPS). We demonstrated that repetitive LPS challenges skew monocytes into the classically exhausted Ly6Chi population, and deplete the homeostatic non-classical Ly6Clo population, reminiscent of monocyte exhaustion in septic patients. scRNAseq analyses confirmed the expansion of Ly6Chi monocyte cluster, with elevation of pathogenic inflammatory genes previously observed in human septic patients. Furthermore, we identified CD38 as an inflammatory mediator of exhausted monocytes, associated with a drastic depletion of cellular NAD+; elevation of ROS; and compromise of mitochondria respiration, representative of septic monocytes. Mechanistically, we revealed that STAT1 is robustly elevated and sustained in LPS-exhausted monocytes, dependent upon the TRAM adaptor of the TLR4 pathway. TRAM deficient monocytes are largely resistant to LPS-mediated exhaustion, and retain the non-classical homeostatic features. Together, our current study addresses an important yet less-examined area of monocyte exhaustion, by providing phenotypic and mechanistic insights regarding the generation of exhausted monocytes.


Subject(s)
Immunologic Memory , Inflammation/immunology , Monocytes/immunology , Sepsis/immunology , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Cells, Cultured , Immunologic Memory/drug effects , Inflammation/genetics , Inflammation/metabolism , Kruppel-Like Factor 4/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Phenotype , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , STAT1 Transcription Factor/metabolism , Sepsis/genetics , Sepsis/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism
7.
Biomed Pharmacother ; 144: 112337, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34688080

ABSTRACT

The growing number of deaths related to sepsis has become a major concern for past few years. Sepsis is a complex pathological reactions that is explained by series of host response to microbial insult. The resulted systemic reactions are manifested by early appearance of proinflammatory cytokines leading to hyperinflammatory phase which is followed by septic shock and death of the patient. The present study has revealed that antibiotics are not self-sufficient to control the complex mechanism of sepsis. Moreover prolonged and unnecessary administration of antibiotics may lead to antibiotic resistance to pathogens. In addition to this, immunosuppressive medications are selective and have targeted approach to certain study population. Drugs from herbal origin have shown to possess a mammoth of immunomodulatory potential by suppressing proinflammatory and anti-inflammatory cytokines exhibiting no or minimal unwanted secondary responses. Concomitantly, herbal plants tend to modulate oxidative stress level and haematological imbalance during inflammatory diseased conditions. Natural compounds have gained much attention for the treatment of several clinical complications. Considering the promising responses of medicinal plants with less/no side effects and easy procurement, comprehensive research on herbal plants to treat sepsis should be contemplated.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Immunomodulating Agents/therapeutic use , Phytotherapy , Plant Extracts/therapeutic use , Plants, Medicinal , Sepsis/drug therapy , Animals , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Antioxidants/therapeutic use , Humans , Immunomodulating Agents/isolation & purification , Plant Extracts/isolation & purification , Plants, Medicinal/chemistry , Sepsis/immunology , Sepsis/metabolism , Sepsis/microbiology
8.
Cells ; 10(9)2021 08 27.
Article in English | MEDLINE | ID: mdl-34571869

ABSTRACT

Sepsis remains a common cause of death in intensive care units, accounting for approximately 20% of total deaths worldwide. Its pathogenesis is partly attributable to dysregulated inflammatory responses to bacterial endotoxins (such as lipopolysaccharide, LPS), which stimulate innate immune cells to sequentially release early cytokines (such as tumor necrosis factor (TNF) and interferons (IFNs)) and late mediators (such as high-mobility group box 1, HMGB1). Despite difficulties in translating mechanistic insights into effective therapies, an improved understanding of the complex mechanisms underlying the pathogenesis of sepsis is still urgently needed. Here, we review recent progress in elucidating the intricate mechanisms underlying the regulation of HMGB1 release and action, and propose a few potential therapeutic candidates for future clinical investigations.


Subject(s)
Cytokines/immunology , HMGB1 Protein/immunology , Lipopolysaccharides/immunology , Sepsis/immunology , Animals , Humans
9.
J Immunol Res ; 2021: 9998517, 2021.
Article in English | MEDLINE | ID: mdl-34285925

ABSTRACT

Patients who survive the acute phase of sepsis can progress to persistent inflammation, immunosuppression, and catabolism syndrome (PICS), which usually results in extended recovery periods and multiple complications. Alpinetin is a flavonoid isolated from Alpinia katsumadai Hayata that has been demonstrated to have anti-inflammatory, antibacterial, and antioxidant activities. The aim of this study was to investigate whether the administration of alpinetin could attenuate PICS in a septic mouse model. Mice were randomly divided into four groups: the (1) sham-operated group, (2) sham+alpinetin (1 mg/kg intravenously infused for once per day after sham operation), (3) cecal ligation and puncture (CLP), and (4) CLP+alpinetin (50 mg/kg intravenously infused for once per day after CLP). Eight days after sham operation or CLP surgery, mice were euthanized for subsequent examination. Alpinetin significantly improved the survival of septic mice. Also, it attenuated the CLP-induced persistent inflammation, immunosuppression, and catabolism syndrome. The level of plasma proinflammatory cytokines and apoptosis of T lymphocytes were obviously decreased by alpinetin as well. Moreover, oxidative stress in the organs was compelling lower in the alpinetin-treated CLP mice. In this clinically relevant model of sepsis, alpinetin ameliorates CLP-induced organ dysfunction and improves the likelihood of survival, possibly through suppressing the inflammatory response, oxidative stress, and apoptosis. These findings suggested that alpinetin could be a potential novel therapeutic approach to prevent sepsis-induced PICS.


Subject(s)
Flavanones/therapeutic use , Immune Tolerance/drug effects , Sepsis/drug therapy , Systemic Inflammatory Response Syndrome/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Flavanones/pharmacology , Humans , Male , Mice , Oxidative Stress/drug effects , Oxidative Stress/immunology , Sepsis/complications , Sepsis/immunology , Systemic Inflammatory Response Syndrome/immunology
10.
Front Immunol ; 12: 711939, 2021.
Article in English | MEDLINE | ID: mdl-34305952

ABSTRACT

Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Recently was been found that pyroptosis is a unique form of proinflammatory programmed death, that is different from apoptosis. A growing number of studies have investigated pyroptosis and its relationship with sepsis, including the mechanisms, role, and relevant targets of pyroptosis in sepsis. While moderate pyroptosis in sepsis can control pathogen infection, excessive pyroptosis can lead to a dysregulated host immune response and even organ dysfunction. This review provides an overview of the mechanisms and potential therapeutic targets underlying pyroptosis in sepsis identified in recent decades, looking forward to the future direction of treatment for sepsis.


Subject(s)
Pyroptosis/physiology , Sepsis/immunology , Alarmins/physiology , Apoptosis/physiology , Caspases/metabolism , Cytokines/physiology , Disseminated Intravascular Coagulation , Drugs, Chinese Herbal/pharmacology , Heart/physiopathology , Humans , Lung/physiopathology , Pyroptosis/drug effects , Pyroptosis/immunology , Sepsis/blood , Sepsis/drug therapy , Sepsis/physiopathology
11.
Infect Immun ; 89(10): e0016221, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34310884

ABSTRACT

Extremely drug-resistant (XDR) Acinetobacter baumannii is a notorious and frequently encountered pathogen demanding novel therapeutic interventions. An initial monoclonal antibody (MAb), C8, raised against A. baumannii capsule, proved a highly effective treatment against a minority of clinical isolates. To overcome this limitation, we broadened coverage by developing a second antibody for use in a combination regimen. We sought to develop an additional anti-A. baumannii MAb through hybridoma technology by immunizing mice with sublethal inocula of virulent, XDR clinical isolates not bound by MAb C8. We identified a new antibacterial MAb, 65, which bound to strains in a pattern distinct from and complementary to that of MAb C8. MAb 65 enhanced macrophage opsonophagocytosis of targeted strains and markedly improved survival in lethal bacteremic sepsis and aspiration pneumonia murine models of A. baumannii infection. MAb 65 was also synergistic with colistin, substantially enhancing protection compared to monotherapy. Treatment with MAb 65 significantly reduced blood bacterial density, ameliorated cytokine production (interleukin-1ß [IL-1ß], IL-6, IL-10, and tumor necrosis factor), and sepsis biomarkers. We describe a novel MAb targeting A. baumannii that broadens immunotherapeutic strain coverage, is highly potent and effective, and synergistically improves outcomes in combination with antibiotics.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Antibodies, Monoclonal/immunology , Acinetobacter Infections/blood , Acinetobacter Infections/microbiology , Animals , Anti-Bacterial Agents/immunology , Antibodies, Bacterial/immunology , Biomarkers/blood , Colistin/immunology , Cytokines/blood , Cytokines/immunology , Drug Resistance, Multiple, Bacterial/immunology , Mice , Microbial Sensitivity Tests/methods , Sepsis/blood , Sepsis/immunology , Sepsis/microbiology
12.
Phytomedicine ; 86: 153567, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33940332

ABSTRACT

BACKGROUND: Sepsis is a life-threatening condition caused by a dysregulated host response to infection. Several studies have indicated that flavonoids exhibit a wide variety of biological actions including free radical scavenging and antioxidant activities. Quercetin, one of the most extensively distributed flavonoids in the vegetables and fruits, presents various biological activities including modulation of oxidative stress, anti-infectious, anti-inflammatory, and neuroprotective activities. METHODS: The present systematic review was conducted according to the guidelines of the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) statements. We searched Web of Sciences, Google Scholar, PubMed, Scopus, and Embase databases up to February 2021 by using the relevant keywords. RESULTS: Out of 672 records screened, 35 articles met the study criteria. The evidence reviewed here indicates that quercetin supplementation may exert beneficial effects on sepsis by attenuating inflammation and oxidative stress, downregulating the mRNA expression of toll-like receptors (TLRs), modulating the immune response, and alleviating sepsis-related organ dysfunctions. CONCLUSION: Due to the promising therapeutic effects of quercetin on sepsis complications and the lack of clinical trials in this regard, future human randomized clinical trials are warranted.


Subject(s)
Quercetin/pharmacology , Sepsis/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/pharmacology , Dietary Supplements , Humans , Inflammation/drug therapy , Oxidative Stress/drug effects , Quercetin/therapeutic use , Sepsis/immunology , Sepsis/physiopathology
13.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33658363

ABSTRACT

Blood pH is tightly maintained between 7.35 and 7.45, and acidosis (pH <7.3) indicates poor prognosis in sepsis, wherein lactic acid from anoxic tissues overwhelms the buffering capacity of blood. Poor sepsis prognosis is also associated with low zinc levels and the release of High mobility group box 1 (HMGB1) from activated and/or necrotic cells. HMGB1 added to whole blood at physiological pH did not bind leukocyte receptors, but lowering pH with lactic acid to mimic sepsis conditions allowed binding, implying the presence of natural inhibitor(s) preventing binding at normal pH. Testing micromolar concentrations of divalent cations showed that zinc supported the robust binding of sialylated glycoproteins with HMGB1. Further characterizing HMGB1 as a sialic acid-binding lectin, we found that optimal binding takes place at normal blood pH and is markedly reduced when pH is adjusted with lactic acid to levels found in sepsis. Glycan array studies confirmed the binding of HMGB1 to sialylated glycan sequences typically found on plasma glycoproteins, with binding again being dependent on zinc and normal blood pH. Thus, HMGB1-mediated hyperactivation of innate immunity in sepsis requires acidosis, and micromolar zinc concentrations are protective. We suggest that the potent inflammatory effects of HMGB1 are kept in check via sequestration by plasma sialoglycoproteins at physiological pH and triggered when pH and zinc levels fall in late stages of sepsis. Current clinical trials independently studying zinc supplementation, HMGB1 inhibition, or pH normalization may be more successful if these approaches are combined and perhaps supplemented by infusions of heavily sialylated molecules.


Subject(s)
Acidosis/blood , HMGB1 Protein/blood , Sepsis/blood , Sialoglycoproteins/blood , Zinc/blood , Acidosis/immunology , Acidosis/metabolism , Acidosis/pathology , Carrier Proteins , HMGB1 Protein/pharmacology , Humans , Hydrogen-Ion Concentration , Immunity, Innate , Lipopolysaccharides/pharmacology , Polysaccharides/chemistry , Sepsis/immunology , Sepsis/pathology , Sialic Acids/chemistry , Sialoglycoproteins/chemistry , Zinc/metabolism
14.
Sci Rep ; 11(1): 2942, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536477

ABSTRACT

Sepsis is caused by organ dysfunction initiated by an unrestrained host immune response to infection. The emergence of antibiotic-resistant bacteria has rapidly increased in the last decades and has stimulated a firm research platform to combat infections caused by antibiotic-resistant bacteria that cannot be eradicated with conventional antibiotics. Strategies like epigenetic regulators such as lysine demethylase (Kdm) has received attention as a new target. Thus, we sought to investigate the epigenetic mechanisms in sepsis pathophysiology with the aim of discovering new concepts for treatment. A transcriptome analysis of dendritic cells during their inflammatory state identified Kdm as a critical molecule in sepsis regulation. Next, 8-hydroxyquinoline-5-carboxylic acid (IOX1) ability to control endotoxemia induced by Lipopolysaccharide and bacterial sepsis was demonstrated. IOX1 has been shown to regulate endotoxemia and sepsis caused by Escherichia coli and carbapenem-resistant Acinetobacter baumannii and has also contributed to the suppression of multidrug-resistant bacterial growth through the inhibition of DNA Gyrase. These findings show that IOX1 could be a component agent against bacterial sepsis by functioning as a broad-spectrum antibiotic with dual effects.


Subject(s)
Acinetobacter Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/drug therapy , Hydroxyquinolines/pharmacology , Sepsis/drug therapy , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Animals , Anti-Bacterial Agents/therapeutic use , DNA Gyrase/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Humans , Hydroxyquinolines/therapeutic use , Mice , Microbial Sensitivity Tests , Molecular Docking Simulation , Sepsis/immunology , Sepsis/microbiology
15.
Mol Cell Biochem ; 476(6): 2337-2344, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33586093

ABSTRACT

Sepsis is described as a systemic immune response of the body to an infectious process that might result in dysfunctional organs that may lead to death. In clinical practice, sepsis is considered a medical emergency. The initial event in sepsis caused by a deregulated host response towards harmful microorganisms that leads to an aggravated systemic inflammatory response syndrome (SIRS) to tackle with pathogen invasion and a compensatory anti-inflammatory response syndrome (CARS) that lasts for several days. The inflammatory response and the cellular damage as well as the risk of an organ dysfunction are in direct proportion. Even though, the pathogenesis of sepsis remains unclear, many studies have shown evidence of role of oxidants and antioxidants in sepsis. The altered innate and adaptive immune cell and upregulated production and release of cytokines and chemokines most probably due to involvement of JAK-STAT pathway, disturbance in redox homeostasis due to low clearance of lactate and other oxidative stressors, contributes to sepsis process to organ dysfunction which contribute to increase rates of mortality among these patients. Hence, the treatment strategies for sepsis include antibiotics, ventilator and blood glucose management and other strategies for resuscitation are rapidly progressing. In the current review, we mainly concentrate on throwing light on the main molecular aspects and chemico-biological interactions that shows involvement in pathways manipulating alteration in physiology of immune cells (innate and adaptive) that change the bioenergetics/cellular metabolism to organ dysfunction and correlation of these altered pathway, improve the understating for new therapeutic target for sepsis.


Subject(s)
Cytokines/immunology , Energy Metabolism/immunology , Multiple Organ Failure/immunology , Oxidative Stress/immunology , Sepsis/immunology , Humans , Multiple Organ Failure/pathology , Sepsis/pathology , Translational Research, Biomedical
16.
Hum Exp Toxicol ; 40(2): 342-354, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32840384

ABSTRACT

Hydroxytyrosol (HT) is among the main bioactive ingredients isolated from olive tree with a variety of biological and pharmacological activities. In the current study, the antioxidative and anti-inflammatory activities of HT were distinguished in the splenic tissue following lipopolysaccharide (LPS)-mediated septic response. Thirty-five Swiss mice were divided into five groups (n = 7): control, HT (40 mg/kg), LPS (10 mg/kg), HT 20 mg+LPS and HT 40 mg+LPS. HT was administered for 10 days, while a single LPS dose was applied. The obtained findings demonstrate that HT administration enhanced the survival rate and decreased lactate dehydrogenase level in LPS-challenged mice. Treatment with HT inhibited the incidence of oxidative damage in splenic tissue through decreasing lipoperoxidation and increasing antioxidant molecules, namely glutathione, superoxide dismutase and catalase. HT also decreased total leukocytes count, C-reactive protein, monocyte chemoattractant protein-1, and myeloperoxidase levels. Additionally, HT suppressed the production levels of tumor necrosis factor-α, interleukin-1ß, and interleukin-6. Moreover, mRNA expression of inducible nitric oxide synthase and nitric oxide production were increased after HT administration. Furthermore, HT supplementation resulted in a downregulation of p38 mitogen-activated protein kinase, inhibited the activation of the nuclear factor kappa-B from the nucleus to the cytoplasm, and attenuated infiltration of activated immune cells and tissue injury following LPS injection. Collectively, these findings demonstrate the antioxidative and anti-inflammatory properties of HT against LPS-mediated inflammation and sepsis. Therefore, HT could be applied as an alternative anti-inflammatory agent to minimize or prevent the development of systemic inflammatory response associated with septic shock.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Phenylethyl Alcohol/analogs & derivatives , Sepsis/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , C-Reactive Protein/analysis , Catalase/metabolism , Cytokines/genetics , Glutathione/metabolism , Leukocyte Count , Lipopolysaccharides , Male , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Oxidative Stress/drug effects , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/therapeutic use , Sepsis/genetics , Sepsis/immunology , Sepsis/pathology , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Superoxide Dismutase/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
17.
Adv Wound Care (New Rochelle) ; 10(9): 477-489, 2021 09.
Article in English | MEDLINE | ID: mdl-33066719

ABSTRACT

Objective: The incidence of severe infectious complications after burn injury increases mortality by 40%. However, traditional approaches for managing burn infections are not always effective. High-voltage, pulsed electric field (PEF) treatment shortly after a burn injury has demonstrated an antimicrobial effect in vivo; however, the working parameters and long-term effects of PEF treatment have not yet been investigated. Approach: Nine sets of PEF parameters were investigated to optimize the applied voltage, pulse duration, and frequency or pulse repetition for disinfection of Pseudomonas aeruginosa infection in a stable mouse burn wound model. The bacterial load after PEF administration was monitored for 3 days through bioluminescence imaging. Histological assessments and inflammation response analyses were performed at 1 and 24 h after the therapy. Results: Among all tested PEF parameters, the best disinfection efficacy of P. aeruginosa infection was achieved with a combination of 500 V, 100 µs, and 200 pulses delivered at 3 Hz through two plate electrodes positioned 1 mm apart for up to 3 days after the injury. Histological examinations revealed fewer inflammatory signs in PEF-treated wounds compared with untreated infected burns. Moreover, the expression levels of multiple inflammatory-related cytokines (interleukin [IL]-1α/ß, IL-6, IL-10, leukemia inhibitory factor [LIF], and tumor necrosis factor-alpha [TNF-α]), chemokines (macrophage inflammatory protein [MIP]-1α/ß and monocyte chemoattractant protein-1 [MCP-1]), and inflammation-related factors (vascular endothelial growth factor [VEGF], macrophage colony-stimulating factor [M-CSF], and granulocyte-macrophage colony-stimulating factor [G-CSF]) were significantly decreased in the infected burn wound after PEF treatment. Innovation: We showed that PEF treatment on infected wounds reduces the P. aeruginosa load and modulates inflammatory responses. Conclusion: The data presented in this study suggest that PEF treatment is a potent candidate for antimicrobial therapy for P. aeruginosa burn infections.


Subject(s)
Burns/therapy , Disinfection/methods , Electric Stimulation Therapy/methods , Pseudomonas Infections/therapy , Wound Infection/therapy , Animals , Burns/complications , Burns/microbiology , Disease Models, Animal , Electrophoresis, Gel, Pulsed-Field , Inflammation , Pseudomonas aeruginosa , Sepsis/etiology , Sepsis/immunology , Tachycardia , Vascular Endothelial Growth Factor A , Wound Infection/microbiology
18.
Biomaterials ; 267: 120389, 2021 01.
Article in English | MEDLINE | ID: mdl-33130319

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new strain of coronavirus not previously identified in humans. Globally, the number of confirmed cases and mortality rates of coronavirus disease 2019 (COVID-19) have risen dramatically. Currently, there are no FDA-approved antiviral drugs and there is an urgency to develop treatment strategies that can effectively suppress SARS-CoV-2-mediated cytokine storms, acute respiratory distress syndrome (ARDS), and sepsis. As symptoms progress in patients with SARS-CoV-2 sepsis, elevated amounts of cell-free DNA (cfDNA) are produced, which in turn induce multiple organ failure in these patients. Furthermore, plasma levels of DNase-1 are markedly reduced in SARS-CoV-2 sepsis patients. In this study, we generated recombinant DNase-1-coated polydopamine-poly(ethylene glycol) nanoparticulates (named long-acting DNase-1), and hypothesized that exogenous administration of long-acting DNase-1 may suppress SARS-CoV-2-mediated neutrophil activities and the cytokine storm. Our findings suggest that exogenously administered long-acting nanoparticulate DNase-1 can effectively reduce cfDNA levels and neutrophil activities and may be used as a potential therapeutic intervention for life-threatening SARS-CoV-2-mediated illnesses.


Subject(s)
COVID-19/complications , Cytokine Release Syndrome/drug therapy , DNA/blood , Deoxyribonuclease I/therapeutic use , Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Neutrophils/drug effects , SARS-CoV-2 , Sepsis/drug therapy , Animals , COVID-19/blood , COVID-19/immunology , Cytokine Release Syndrome/etiology , Deoxyribonuclease I/administration & dosage , Dexamethasone/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Extracellular Traps/drug effects , Humans , Indoles , Male , Mice , Mice, Inbred C57BL , Multiple Organ Failure/blood , Multiple Organ Failure/etiology , Multiple Organ Failure/prevention & control , NF-kappa B/blood , Neutrophils/enzymology , Peroxidase/blood , Polyethylene Glycols , Polyglactin 910 , Polymers , Sepsis/etiology , Sepsis/immunology
19.
Biochem Pharmacol ; 182: 114254, 2020 12.
Article in English | MEDLINE | ID: mdl-33010213

ABSTRACT

The circadian clock is a collection of endogenous oscillators with a periodicity of ~ 24 h. Recently, our understanding of circadian rhythms and their regulation at genomic and physiologic scales has grown significantly. Knowledge of the circadian influence on biological processes has provided new possibilities for novel pharmacological strategies. Directly targeting the biological clock or its downstream targets, and/or using timing as a variable in drug therapy are now important pharmacological considerations. The circadian machinery mediates many aspects of the inflammatory response and, reciprocally, an inflammatory environment can disrupt circadian rhythms. Therefore, intense interest exists in leveraging circadian biology as a means to treat chronic inflammatory diseases such as sepsis, asthma, rheumatoid arthritis, osteoarthritis, and cardiovascular disease, which all display some type of circadian signature. The purpose of this review is to evaluate the crosstalk between circadian rhythms, inflammatory diseases, and their pharmacological treatment. Evidence suggests that carefully rationalized application of chronotherapy strategies - alone or in combination with small molecule modulators of circadian clock components - can improve efficacy and reduce toxicity, thus warranting further investigation and use.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Chronotherapy/methods , Circadian Clocks/physiology , Circadian Rhythm/physiology , Inflammation Mediators/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Asthma/drug therapy , Asthma/immunology , Asthma/metabolism , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Chronic Disease , Chronotherapy/trends , Circadian Clocks/drug effects , Circadian Rhythm/drug effects , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/antagonists & inhibitors , Sepsis/drug therapy , Sepsis/immunology , Sepsis/metabolism , Treatment Outcome
20.
J Tradit Chin Med ; 40(5): 820-826, 2020 10.
Article in English | MEDLINE | ID: mdl-33000583

ABSTRACT

OBJECTIVE: To detect antioxidant and antiinflammatory efficacy of Curcumin (Cur) on lung tissue in rats with sepsis. METHODS: Totally 32 rats were divided into 4 groups; the rats in Group 1 (control group) had abdominal incision under sterile conditions following anesthesia and the abdomen was sutured. Abdominal incision was performed in the rats in Group 2 (Cur group) under sterile conditions following anesthesia and the abdomen was closed. Cur was given to this group after dissolving within dimethylsulphoxide as 100 mg/kg through oral gavage and started for 3 d before surgical procedure. Group 3 (CLP group) had caecal ligation and punction (CLP) under sterile conditions to create sepsis following anesthesia and the abdomen was sutured. CLP was performed in the rats in Group 4 (CLP + Cur group) under sterile conditions following anesthesia to create a sepsis model and the abdomen was closed. Cur was also given to this group after dissolving within dimethylsulphoxide as 100 mg/kg through oral gavage and started for 3 d before surgical procedure. All the rats were sacrificed through blood aspiration from the heart under sterile conditions following anesthesia and lung tissues were removed after 24 h following the surgical procedures. The tissue samples were homogenizated for biochemical analyses; and malondialdehyde (MDA), nitric okxide (NO), myeloperoxidase (MPO), superoxidedysmutase (SOD) nd catalase (CAT) were analyzed through spectrophotometric method, immunhistochemical iNOS staining was performed to assess the inflammation; and histopathological differences between the groups were evaluated. RESULTS: A statistically significant decrease was detected in the CLP + Cur group when compared with the CLP group of which Cur was not given in terms of MDA, MPO and NO levels (P < 0.05) whereas a statistically significant elevation was fpund in the CLP + Cur group when compared with the CLP group in terms of SOD and CAT levels (P < 0.05). CONCLUSION: The study outcomes revealed that supplementation of Cur presents an antioxidant effect by reducing the free radical level and increasing the antioxidant enzyme levels; and an antiinflammatory effect by reducing iNOS level.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Curcumin/administration & dosage , Lung/drug effects , Sepsis/drug therapy , Animals , Humans , Lung/immunology , Male , Malondialdehyde/immunology , Nitric Oxide/immunology , Oxidative Stress/drug effects , Peroxidase/immunology , Rats , Rats, Wistar , Sepsis/genetics , Sepsis/immunology , Superoxide Dismutase/genetics , Superoxide Dismutase/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL