Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.880
Filter
1.
Mol Med ; 30(1): 155, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300342

ABSTRACT

BACKGROUND: Macrophage pyroptosis is a pivotal inflammatory mechanism in sepsis-induced lung injury, however, the underlying mechanisms remain inadequately elucidated. METHODS: Lipopolysaccharides (LPS)/adenosine triphosphate (ATP)-stimulated macrophages and cecal ligation and puncture (CLP)-induced mouse model for sepsis were established. The levels of key molecules were examined by qRT-PCR, Western blotting, immunohistochemistry (IHC) and ELISA assay. The subcellular localization of circMAPK1 was detected by RNA fluorescence in situ hybridization (FISH). Cell viability, LDH release and caspase-1 activity were monitored by CCK-8, LDH assays, and flow cytometry. The bindings between KDM2B/H3K36me2 and WNK1 promoter was detected by chromatin immunoprecipitation (ChIP) assay and luciferase assay, and associations among circMAPK1, UPF1 and KDM2B mRNA were assessed by RNA pull-down or RNA immunoprecipitation (RIP) assays. The pathological injury of lung tissues was evaluated by lung wet/dry weight ratio and hematoxylin and eosin (H&E) staining. RESULTS: CircMAPK1 was elevated in patients with septic lung injury. Knockdown of circMAPK1 protected against LPS/ATP-impaired cell viability and macrophage pyroptosis via WNK1/NLRP3 axis. Mechanistically, loss of circMAPK1 enhanced the association between KDM2B and WNK1 promoter to promote the demethylation of WNK1 and increase its expression. CircMAPK1 facilitated KDM2B mRNA decay by recruiting UPF1. Functional experiments showed that silencing of KDM2B or WNK1 counteracted circMAPK1 knockdown-suppressed macrophage pyroptosis. In addition, silencing of circMAPK1 alleviated CLP-induced lung injury in mice via KDM2B/WNK1/NLRP3 axis. CONCLUSION: CircMAPK1 exacerbates sepsis-induced lung injury by destabilizing KDM2B mRNA to suppress WNK1 expression, thus facilitating NLRP3-driven macrophage pyroptosis.


Subject(s)
Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases , Pyroptosis , Sepsis , WNK Lysine-Deficient Protein Kinase 1 , Animals , Pyroptosis/genetics , Sepsis/complications , Sepsis/genetics , Sepsis/metabolism , Mice , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Male , WNK Lysine-Deficient Protein Kinase 1/metabolism , WNK Lysine-Deficient Protein Kinase 1/genetics , Humans , RNA Stability , Lung Injury/etiology , Lung Injury/metabolism , Lung Injury/genetics , Disease Models, Animal , Female , Macrophages/metabolism , Mice, Inbred C57BL , F-Box Proteins
2.
Ren Fail ; 46(2): 2403649, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39311631

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) frequently occurs as a complication of sepsis. PANoptosis refers to a type of inflammatory programmed cell death that exhibits key characteristics of apoptosis, necroptosis, and pyroptosis. Here, we evaluated the role of absent in melanoma 2 (AIM2) and eukaryotic translation initiation factor 2 alpha kinase 2 (EIF2AK2) in septic AKI. METHODS: A septic AKI model was created through cecal ligation and puncture (CLP), while an in vitro model was developed using lipopolysaccharide (LPS)-stimulated HK2 cells. Hematoxylin and eosin (HE), Periodic acid-Schiff (PAS), and TUNEL staining were conducted to assess kidney injury in mice. Levels of serum creatinine (Scr) and blood urea nitrogen (BUN) were detected by kits. Gene expression was detected utilizing RT-qPCR, and Western blot was used to test protein levels. Immunofluorescence was employed to measure EIF2AK2 and AIM2 expression in mouse kidney tissue. Lactate dehydrogenase (LDH) activity assay was conducted to evaluate cytotoxicity. Co-immunoprecipitation (Co-IP) was performed to verify the binding relationship between EIF2AK2 and AIM2. RESULTS: AIM2 expression was increased in the renal tissue of mice subjected to CLP. Activation of the inflammasome and PANoptosis were observed in the renal tissue of CLP mice. AIM2 depletion attenuated PANoptosis in LPS-treated HK-2 cells. Additionally, EIF2AK2 could directly target AIM2, leading to a positive regulation of AIM2 expression. Notably, EIF2AK2 induced PANoptosis through upregulating AIM2 in HK-2 cells stimulated by LPS. CONCLUSIONS: Our results revealed the important role of EIF2AK2-induced AIM2 upregulation in the activation of PANoptosis during septic AKI.


Renal tissue from CLP mice exhibited an increase in AIM2 expression.Renal tissue from CLP mice demonstrated inflammasome activation and PANoptosis.AIM2 silencing reduced PANoptosis in LPS-treated HK-2 cells.EIF2AK2 directly targeted AIM2 and positively regulated its expression.EIF2AK2 promoted PANoptosis via AIM2 in LPS-triggered HK-2 cells.


Subject(s)
Acute Kidney Injury , Disease Models, Animal , Sepsis , eIF-2 Kinase , Animals , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Sepsis/complications , Sepsis/metabolism , Mice , Humans , eIF-2 Kinase/metabolism , Male , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Lipopolysaccharides , Kidney/pathology , Kidney/metabolism , Cell Line , Mice, Inbred C57BL , Inflammasomes/metabolism , Necroptosis , Apoptosis , Pyroptosis
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(6): 890-902, 2024 Jun 28.
Article in English, Chinese | MEDLINE | ID: mdl-39311785

ABSTRACT

OBJECTIVES: Inflammation in the central nervous system plays a crucial role in the occurrence and development of sepsis-associated encephalopathy. This study aims to explore the effects of maresin 1 (MaR1), an anti-inflammatory and pro-resolving lipid mediator, on sepsis-induced neuroinflammation and cognitive impairment. METHODS: Mice were randomly assigned to 4 groups: A sham group (sham operation+vehicle), a cecal ligation and puncture (CLP) group (CLP operation+vehicle), a MaR1-LD group (CLP operation+1 ng MaR1), and a MaR1-HD group (CLP operation+10 ng MaR1). MaR1 or vehicle was intraperitoneally administered starting 1 h before CLP operation, then every other day for 7 days. Survival rates were monitored, and serum inflammatory cytokines [tumor necrosis factor alpha (TNF-α), interleukin (IL)-1ß, and IL-6] were measured 24 h after operation using enzyme-linked immunosorbent assay (ELISA). Cognitive function was assessed 7 days after operation using the Morris water maze (MWM) test and novel object recognition (NOR) task. The mRNA expression of TNF-α, IL-1ß, IL-6, inducible nitric oxide synthase (iNOS), IL-4, IL-10, and arginase 1 (Arg1) in cortical and hippocampal tissues was determined by real-time reverse transcription PCR (RT-PCR). Western blotting was used to determine the protein expression of iNOS, Arg1, signal transducer and activator of transcription 6 (STAT6), peroxisome proliferator-activated receptor gamma (PPARγ), and phosphorylated STAT6 (p-STAT6) in hippocampal tissue. Microglia activation was visualized via immunofluorescence. Mice were also treated with the PPARγ antagonist GW9662 to confirm the involvement of this pathway in MaR1's effects. RESULTS: CLP increased serum levels of TNF-α, IL-1ß, and IL-6, and reduced body weight and survival rates (all P<0.05). Both 1 ng and 10 ng doses of MaR1 significantly reduced serum TNF-α, IL-1ß, and IL-6 levels, improved body weight, and increased survival rates (all P<0.05). No significant difference in efficacy was observed between the 2 doses (all P>0.05). MWM test and NOR task indicated that CLP impaired spatial learning, which MaR1 mitigated. However, GW9662 partially reversed MaR1's protective effects. Real-time RT-PCR results demonstrated that, compared to the sham group, mRNA expression of TNF-α, IL-1ß, and iNOS significantly increased in hippocampal tissues following CLP (all P<0.05), while IL-4, IL-10, and Arg1 showed a slight decrease, though the differences were not statistically significant (all P>0.05). Compared to the CLP group, both 1 ng and 10 ng MaR1 decreased TNF-α, IL-1ß, and iNOS mRNA expression in hippocampal tissues and increased IL-4, IL-10, and Arg1 mRNA expression (all P<0.05). Immunofluorescence results indicated a significant increase in Iba1-positive microglia in the hippocampus after CLP compared to the sham group (P<0.05). Administration of 1 ng and 10 ng MaR1 reduced the percentage area of Iba1-positive cells in the hippocampus compared to the CLP group (both P<0.05). Western blotting results showed that, compared to the CLP group, both 1 ng and 10 ng MaR1 down-regulated the iNOS expression, while up-regulated the expression of Arg1, PPARγ, and p-STAT6 (all P<0.05). However, the inclusion of GW9662 counteracted the MaR1-induced upregulation of Arg1 and PPARγ compared to the MaR1-LD group (all P<0.05). CONCLUSIONS: MaR1 inhibits the classical activation of hippocampal microglia, promotes alternative activation, reduces sepsis-induced neuroinflammation, and improves cognitive decline.


Subject(s)
Cecum , Cognitive Dysfunction , Disease Models, Animal , Docosahexaenoic Acids , Sepsis , Tumor Necrosis Factor-alpha , Animals , Mice , Cognitive Dysfunction/etiology , Cognitive Dysfunction/drug therapy , Sepsis/complications , Sepsis/metabolism , Ligation , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Cecum/surgery , Male , Interleukin-6/metabolism , Interleukin-1beta/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Hippocampus/metabolism , Hippocampus/drug effects , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/genetics , Arginase/metabolism , Punctures/adverse effects , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology
4.
Funct Integr Genomics ; 24(5): 173, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39320434

ABSTRACT

Septic cardiomyopathy is a secondary myocardial injury caused by sepsis. N6-methyl-adenosine (m6A) modification is involved in the pathological progression of septic cardiomyopathy; however, the pathological mechanism remains unclear. In this study, we identified the overall m6A modification pattern in septic myocardial injury and determined its potential interactions with differentially expressed genes (DEGs). A sepsis mouse model exhibiting septic symptoms and myocardial tissue damage was induced by lipopolysaccharide (LPS). LPS-induced septic myocardial tissues and control myocardial tissues were subjected to methylated RNA immunoprecipitation sequencing and RNA sequencing to screen for differentially expressed m6A peaks and DEGs. We identified 859 significantly m6A-modified genes in septic myocardial tissues, including 432 upregulated and 427 downregulated genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to explore the biological importance of differentially expressed m6A methylated genes and DEGs. Differentially expressed m6A methylated genes were enriched in immune- and inflammation-related pathways. Conjoint analysis revealed co-expression of differentially expressed m6A genes and DEGs, including genes that were upregulated or downregulated and those showing opposite trends. High expression of m6A-related genes (WTAP and IGF2BP2), interleukin-17, and interleukin-17 pathway-related genes (MAPK11 and TRAF3IP2) was verified using reverse transcription-quantitative PCR. We confirmed the presence of m6A modification of the transcriptome and m6A-mediated gene expression in septic myocardial tissues.


Subject(s)
Adenosine , Myocardium , Sepsis , Animals , Mice , Sepsis/genetics , Sepsis/metabolism , Myocardium/metabolism , Myocardium/pathology , Methylation , Adenosine/metabolism , Adenosine/analogs & derivatives , Male , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Transcriptome , Mice, Inbred C57BL , Lipopolysaccharides
5.
Cell Biol Toxicol ; 40(1): 82, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39320524

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2), a crucial element of the renin-angiotensin system (RAS), metabolizes angiotensin II into Ang (1-7), which then combines with the Mas receptor (MasR) to fulfill its protective role in various diseases. Nevertheless, the involvement of ACE2 in sepsis-induced cardiomyopathy (SIC) is still unexplored. In this study, our results revealed that CLP surgery dramatically impaired cardiac function accompanied with disruption of the balance between ACE2-Ang (1-7) and ACE-Ang II axis in septic heart tissues. Moreover, ACE2 knockin markedly alleviated sepsis induced RAS disorder, cardiac dysfunction and improved survival rate in mice, while ACE2 knockout significantly exacerbates these outcomes. Adoptive transfer of bone marrow cells and in vitro experiments showed the positive role of myeloid ACE2 by mitigating oxidative stress, inflammatory response, macrophage polarization and cardiomyocyte apoptosis by blocking NF-κB and STAT1 signals. However, the beneficial impacts were nullified by MasR antagonist A779. Collectively, these findings showed that ACE2 alleviated SIC by inhibiting M1 macrophage via activating the Ang (1-7)-MasR axis, highlight that ACE2 might be a promising target for the management of sepsis and SIC patients.


Subject(s)
Angiotensin-Converting Enzyme 2 , Cardiomyopathies , Macrophages , NF-kappa B , STAT1 Transcription Factor , Sepsis , Signal Transduction , Animals , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Sepsis/complications , Sepsis/metabolism , NF-kappa B/metabolism , Cardiomyopathies/metabolism , Mice , STAT1 Transcription Factor/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Apoptosis/drug effects , Renin-Angiotensin System/drug effects , Receptors, G-Protein-Coupled/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Angiotensin I/metabolism , Angiotensin I/pharmacology , Proto-Oncogene Mas , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptidyl-Dipeptidase A/metabolism , Peptidyl-Dipeptidase A/genetics
6.
Signal Transduct Target Ther ; 9(1): 245, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39300122

ABSTRACT

Pyroptosis is a type of programmed cell death characterized by cell swelling and osmotic lysis, resulting in cytomembrane rupture and release of immunostimulatory components, which play a role in several pathological processes. Significant cellular responses to various stimuli involve the formation of inflammasomes, maturation of inflammatory caspases, and caspase-mediated cleavage of gasdermin. The function of pyroptosis in disease is complex but not a simple angelic or demonic role. While inflammatory diseases such as sepsis are associated with uncontrollable pyroptosis, the potent immune response induced by pyroptosis can be exploited as a therapeutic target for anti-tumor therapy. Thus, a comprehensive review of the role of pyroptosis in disease is crucial for further research and clinical translation from bench to bedside. In this review, we summarize the recent advancements in understanding the role of pyroptosis in disease, covering the related development history, molecular mechanisms including canonical, non-canonical, caspase 3/8, and granzyme-mediated pathways, and its regulatory function in health and multiple diseases. Moreover, this review also provides updates on promising therapeutic strategies by applying novel small molecule inhibitors and traditional medicines to regulate pyroptosis. The present dilemmas and future directions in the landscape of pyroptosis are also discussed from a clinical perspective, providing clues for scientists to develop novel drugs targeting pyroptosis.


Subject(s)
Pyroptosis , Pyroptosis/genetics , Humans , Inflammasomes/metabolism , Inflammasomes/genetics , Inflammasomes/immunology , Granzymes/genetics , Granzymes/metabolism , Sepsis/genetics , Sepsis/pathology , Sepsis/metabolism , Sepsis/immunology , Caspase 8/genetics , Caspase 8/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/immunology , Neoplasms/drug therapy , Signal Transduction
7.
Biomed Pharmacother ; 179: 117359, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39236479

ABSTRACT

Sepsis, a frequently fatal condition, emerges from an exaggerated inflammatory response to infection, resulting in multi-organ dysfunction and alarmingly high mortality rates. Despite the urgent need for effective treatments, current therapeutic options remain limited to antibiotics, with no other efficacious alternatives available. Echinatin (Ecn), a potent bioactive compound extracted from the roots and rhizomes of licorice, has gained significant attention for its broad pharmacological properties, particularly its ability to combat oxidative stress. Recent research highlights the crucial role that oxidative stress plays in the onset and progression of sepsis further emphasizing the potential therapeutic value of Ecn in this context. In this study, we explored the protective effects of Ecn in a murine model of sepsis induced by cecal ligation and puncture (CLP). Ecn demonstrated a significant reduction in the levels of inflammatory cytokines and reactive oxygen species (ROS) in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Network pharmacology analysis identified 41 targets and top 15 pathways involved in the Ecn-mediated signaling network, revealing that Ecn might exert its effects through key targets including the NF-κB and MAPK signaling pathways. Molecular docking studies suggested a strong affinity between Ecn and MEK, with kinetic simulations and binding energy calculations confirming a stable interaction. Mechanistically, Ecn treatment inhibited NF-κB and the MEK/ERK signaling pathway, as evidenced by decreased phosphorylation of IκBα and nuclear p65, along with reduced phosphorylation of MEK and ERK in both LPS-stimulated RAW 264.7 macrophages and septic mice. Furthermore, the administration of MEK signaling agonists reversed the anti-inflammatory effects of Ecn, indicating the involvement of this signaling pathway in Ecn's protective mechanism. Notably, our investigation revealed that Ecn did not affect bacterial proliferation either in vivo or in vitro, underscoring its specific immunomodulatory effects rather than direct antimicrobial activity. In summation, our findings underscored the potential of Ecn as an innovative therapeutic remedy for sepsis-induced injury, particularly through the regulation of the NF-κB and MEK/ERK signaling pathway. This exploration unveiled a promising therapeutic approach for treating sepsis, supplementing existing interventions and addressing their constraints.


Subject(s)
MAP Kinase Signaling System , NF-kappa B , Sepsis , Animals , Sepsis/drug therapy , Sepsis/metabolism , NF-kappa B/metabolism , Mice , RAW 264.7 Cells , MAP Kinase Signaling System/drug effects , Male , Reactive Oxygen Species/metabolism , Molecular Docking Simulation , Lipopolysaccharides , Mice, Inbred C57BL , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Signal Transduction/drug effects , Disease Models, Animal , Chalcones
8.
Crit Care ; 28(1): 293, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39227889

ABSTRACT

INTRODUCTION: Experimental studies in animals have yielded conflicting results on the role of Tumor Necrosis Factor (TNF) in sepsis and endotoxemia, with some reporting adaptive and others inappropriate effects. A meta-analysis of the available literature was performed to determine the factors explaining this discrepancy. METHODS: The study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. The protocol was registered with PROSPERO (CRD42020167384) prior to data collection. PubMed and Embase were the databases queried. Risk of bias was evaluated using the SYRCLE Risk of Bias Tool. All animal studies investigating sepsis-related mortality and modified TNF signaling were considered eligible. The exclusion criteria were: lack of mortality data, 7-day mortality rates below 10% in both wild type and TNF-altered pathway animals, and absence of an English abstract. To determine the role of TNF according to the experimental protocol, three approaches were used: first an approach based on the statistical significance of each experiment, then the pooled mortality was calculated, and finally the weighted risk ratio for mortality was assessed. RESULTS: A total of 175 studies were included in the analysis, comprising a total of 760 experiments and involving 19,899 animals. The main species used were mice (77%) and rats (21%). The most common method of TNF pathway modulation was TNF pathway inactivation that was primarily associated with an inappropriate secretion of TNF. At the opposite, TNF injection was associated with an adaptive role of TNF. Lipopolysaccharide (LPS) injection was the most used stimulus to establish an infectious model (42%) and was strongly associated with an inappropriate role of TNF. Conversely, live bacterial models, especially the cecal ligation and puncture (CLP) model, pneumonia, meningitis, and gastrointestinal infection, were associated with an adaptive role. This was particularly evident for Listeria monocytogenes, Streptococcus pneumoniae. CONCLUSION: The role of TNF during infection varies depending on the experimental model used. Models that mimic clinical conditions, based on virulent bacteria that cause high mortality even at low inocula, demonstrated an adaptive role of TNF. Conversely, models based on LPS or low-pathogenic live bacteria, administered at doses well above physiological thresholds and combined with early antibiotic therapy, were associated with an inappropriate role.


Subject(s)
Sepsis , Tumor Necrosis Factor-alpha , Animals , Disease Models, Animal , Sepsis/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
9.
CNS Neurosci Ther ; 30(9): e70021, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39258790

ABSTRACT

BACKGROUND: Sepsis-associated encephalopathy (SAE) is a neuronal injury with poor prognosis. Mitochondrial dysfunction is critical in SAE development, and hydrogen gas (H2) has a protective effect on septic mice. This study aimed to investigate the effect of high concentration (67%) of H2 on SAE and whether it is related to mitochondrial biogenesis and mitochondrial dynamics. METHODS: A mouse sepsis model was induced by cecal ligation and puncture. The mice inhalated 67% H2 for 1 h at 1 and 6 h post-surgery, respectively. The 7-day survival rate was recorded. Cognitive function was assessed using the Y-maze test and Morris water maze test. Serum inflammatory factors, antioxidant enzymes, as well as mitochondrial function indexes including mitochondrial membrane potential (MMP) and ATP in the hippocampal tissue were evaluated 24 h after surgery. Mitochondrial dynamic proteins (DRP1 and MFN2) and biosynthetic proteins (PGC-1α, NRF2, and TFAM) in the hippocampal tissue were detected. Moreover, the morphology of mitochondria was observed by transmission electron microscopy. RESULTS: Inhalation of 67% H2 improved the 7-day survival rates and recognition memory function of septic mice, alleviated brain antioxidant enzyme activity (SOD and CAT), and reduced serum proinflammatory cytokine levels. H2 inhalation also enhanced the expression of MFN2 and mitochondrial biogenesis-related factors (PGC-1α, NRF2, and TFAM) and decreased the expression of fission protein (DRP1), leading to improvement in mitochondrial function, as evidenced by MMP and ATP levels. CONCLUSIONS: Inhalation of high concentration (67%) of H2 in septic mice improved the survival rate and reduced neuronal injury. Its mechanism might be mediated by enhancing mitochondrial biogenesis and mitochondrial dynamics.


Subject(s)
Hydrogen , Mitochondrial Dynamics , Sepsis-Associated Encephalopathy , Animals , Sepsis-Associated Encephalopathy/drug therapy , Mice , Hydrogen/pharmacology , Hydrogen/administration & dosage , Hydrogen/therapeutic use , Mitochondrial Dynamics/drug effects , Male , Administration, Inhalation , Hippocampus/drug effects , Hippocampus/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , Mice, Inbred C57BL , Membrane Potential, Mitochondrial/drug effects , Maze Learning/drug effects
10.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273360

ABSTRACT

Sepsis is caused by a dysregulated host response to an infection that leads to cascading cell death and eventually organ failure. In this study, the role of inflammatory response serum secretory phospholipase A2 (sPLA2) and albumin in sepsis was investigated by determining the activities of the two proteins in serial serum samples collected on different days from patients with sepsis after enrollment in the permissive underfeeding versus standard enteral feeding protocols in an intensive care unit. Serum sPLA2 and albumin showed an inverse relationship with increasing sPLA2 activity and decreasing albumin membrane-binding activity in patients with evolving complications of sepsis. The activities of sPLA2 and albumin returned to normal values more rapidly in the permissive underfeeding group than in the standard enteral feeding group. The inverse sPLA2-albumin activity relationship suggests a complex interplay between these two proteins and a regulatory mechanism underlying cell membrane phospholipid homeostasis in sepsis. The decreased albumin-membrane binding activity in patients' serum was due to its fatty acid-binding sites occupied by pre-bound fatty acids that might alter albumin's structure, binding capacities, and essential functions. The sPLA2-albumin dual serum assays may be useful in determining whether nutritional intervention effectively supports the more rapid recovery of appropriate immune responses in critically ill patients with sepsis.


Subject(s)
Phospholipases A2, Secretory , Sepsis , Humans , Sepsis/blood , Sepsis/metabolism , Phospholipases A2, Secretory/metabolism , Phospholipases A2, Secretory/blood , Male , Female , Middle Aged , Serum Albumin/metabolism , Aged , Enteral Nutrition
11.
J Extracell Vesicles ; 13(9): e12506, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39300768

ABSTRACT

Sepsis following burn trauma is a global complication with high mortality, with ∼60% of burn patient deaths resulting from infectious complications. Diagnosing sepsis is complicated by confounding clinical manifestations of the burn injury, and current biomarkers lack the sensitivity and specificity required for prompt treatment. There is a strong rationale to assess circulating extracellular vesicles (EVs) from patient liquid biopsy as sepsis biomarkers due to their release by pathogens from bacterial biofilms and roles in the subsequent immune response. This study applies Raman spectroscopy to patient plasma-derived EVs for rapid, sensitive, and specific detection of sepsis in burn patients, achieving 97.5% sensitivity and 90.0% specificity. Furthermore, spectral differences between septic and non-septic burn patient EVs could be traced to specific glycoconjugates of bacterial strains associated with sepsis morbidity. This work illustrates the potential application of EVs as biomarkers in clinical burn trauma care and establishes Raman analysis as a fast, label-free method to specifically identify features of bacterial EVs relevant to infection amongst the host background.


Subject(s)
Biomarkers , Burns , Extracellular Vesicles , Sepsis , Spectrum Analysis, Raman , Humans , Burns/complications , Burns/metabolism , Spectrum Analysis, Raman/methods , Extracellular Vesicles/metabolism , Sepsis/metabolism , Sepsis/blood , Biomarkers/blood , Biomarkers/metabolism , Female , Male , Adult , Middle Aged
12.
Sci Adv ; 10(36): eadl6162, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39241063

ABSTRACT

Carbapenem-resistant Klebsiella pneumoniae (CRKP) causes Gram-negative lung infections and fatal pneumonic sepsis for which limited therapeutic options are available. The lungs are densely innervated by nociceptor sensory neurons that mediate breathing, cough, and bronchoconstriction. The role of nociceptors in defense against Gram-negative lung pathogens is unknown. Here, we found that lung-innervating nociceptors promote CRKP pneumonia and pneumonic sepsis. Ablation of nociceptors in mice increased lung CRKP clearance, suppressed trans-alveolar dissemination of CRKP, and protected mice from hypothermia and death. Furthermore, ablation of nociceptors enhanced the recruitment of neutrophils and Ly6Chi monocytes and cytokine induction. Depletion of Ly6Chi monocytes, but not of neutrophils, abrogated lung and extrapulmonary CRKP clearance in ablated mice, suggesting that Ly6Chi monocytes are a critical cellular population to regulate pneumonic sepsis. Further, neuropeptide calcitonin gene-related peptide suppressed the induction of reactive oxygen species in Ly6Chi monocytes and their CRKP-killing abilities. Targeting nociceptor signaling could be a therapeutic approach for treating multidrug-resistant Gram-negative infection and pneumonic sepsis.


Subject(s)
Calcitonin Gene-Related Peptide , Carbapenems , Klebsiella Infections , Klebsiella pneumoniae , Lung , Nociceptors , Sepsis , Animals , Klebsiella pneumoniae/physiology , Mice , Klebsiella Infections/microbiology , Sepsis/metabolism , Sepsis/microbiology , Lung/microbiology , Lung/metabolism , Carbapenems/pharmacology , Calcitonin Gene-Related Peptide/metabolism , Nociceptors/metabolism , Monocytes/metabolism , Sensory Receptor Cells/metabolism , Neutrophils/metabolism , Disease Models, Animal , Antigens, Ly/metabolism , Reactive Oxygen Species/metabolism , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/pathology , Mice, Inbred C57BL
13.
Int J Biol Sci ; 20(11): 4314-4340, 2024.
Article in English | MEDLINE | ID: mdl-39247818

ABSTRACT

Background: Cellular senescence has emerged as a pivotal focus in cardiovascular research. This study investigates the previously unrecognized role of cellular senescence in septic cardiomyopathy (SCM) and evaluates senomorphic therapy using ruxolitinib (Rux) as a potential treatment option. Methods: We employed lipopolysaccharide (LPS)-induced neonatal rat cardiomyocytes (NRCMs) and two mouse models-LPS-induced and cecal ligation and puncture (CLP)-induced SCM models-to assess Rux's effects. RNA sequencing, western blotting (WB), quantitative polymerase chain reaction (qPCR), immunofluorescence, immunohistochemistry, senescence-associated ß-galactosidase (SA-ß-gal) assay, and other techniques were utilized to investigate underlying mechanisms. Results: Senescence-associated secretory phenotype (SASP) and cellular senescence markers were markedly elevated in LPS-induced NRCMs and SCM animal models, confirmed by the SA-ß-gal assay. Rux treatment attenuated SASP in vitro and in vivo, alongside downregulation of senescence markers. Moreover, Rux-based senomorphic therapy mitigated mitochondrial-mediated apoptosis, improved cardiac function in SCM mice, restored the balance of antioxidant system, and reduced reactive oxygen species (ROS) levels. Rux treatment restored mitochondrial membrane potential, mitigated mitochondrial morphological damage, and upregulated mitochondrial complex-related gene expression, thereby enhancing mitochondrial function. Additionally, Rux treatment ameliorated SCM-induced mitochondrial dynamic dysfunction and endoplasmic reticulum stress. Mechanistically, Rux inhibited JAK2-STAT3 signaling activation both in vitro and in vivo. Notably, low-dose Rux and ABT263 showed comparable efficacy in mitigating SCM. Conclusions: This study highlighted the potential significance of cellular senescence in SCM pathogenesis and suggested Rux-based senomorphic therapy as a promising therapeutic approach for SCM.


Subject(s)
Cardiomyopathies , Cellular Senescence , Janus Kinase 2 , Myocytes, Cardiac , Nitriles , Pyrazoles , Pyrimidines , STAT3 Transcription Factor , Signal Transduction , Animals , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Cellular Senescence/drug effects , Signal Transduction/drug effects , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Cardiomyopathies/metabolism , Cardiomyopathies/drug therapy , Nitriles/therapeutic use , Nitriles/pharmacology , Rats , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Male , Mice, Inbred C57BL , Sepsis/metabolism , Sepsis/drug therapy , Rats, Sprague-Dawley , Lipopolysaccharides , Disease Models, Animal
14.
Mol Med ; 30(1): 140, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251905

ABSTRACT

BACKGROUND: Sepsis-induced pulmonary injury (SPI) is a common complication of sepsis with a high rate of mortality. N4-acetylcytidine (ac4C) is mediated by the ac4C "writer", N-acetyltransferase (NAT)10, to regulate the stabilization of mRNA. This study aimed to investigate the role of NAT10 in SPI and the underlying mechanism. METHODS: Twenty-three acute respiratory distress syndrome (ARDS) patients and 27 non-ARDS volunteers were recruited. A sepsis rat model was established. Reverse transcription-quantitative polymerase chain reaction was used to detect the expression of NAT10 and transferrin receptor (TFRC). Cell viability was detected by cell counting kit-8. The levels of Fe2+, glutathione, and malondialdehyde were assessed by commercial kits. Lipid reactive oxygen species production was measured by flow cytometric analysis. Western blot was used to detect ferroptosis-related protein levels. Haematoxylin & eosin staining was performed to observe the pulmonary pathological symptoms. RESULTS: The results showed that NAT10 was increased in ARDS patients and lipopolysaccharide-treated human lung microvascular endothelial cell line-5a (HULEC-5a) cells. NAT10 inhibition increased cell viability and decreased ferroptosis in HULEC-5a cells. TFRC was a downstream regulatory target of NAT10-mediated ac4C acetylation. Overexpression of TFRC decreased cell viability and promoted ferroptosis. In in vivo study, NAT10 inhibition alleviated SPI. CONCLUSION: NAT10-mediated ac4C acetylation of TFRC aggravated SPI through promoting ferroptosis.


Subject(s)
Ferroptosis , Receptors, Transferrin , Sepsis , Sepsis/metabolism , Sepsis/complications , Sepsis/etiology , Acetylation , Animals , Humans , Rats , Male , Receptors, Transferrin/metabolism , Receptors, Transferrin/genetics , Female , Lung Injury/metabolism , Lung Injury/etiology , Lung Injury/pathology , Disease Models, Animal , Acetyltransferases/metabolism , Acetyltransferases/genetics , Middle Aged , Antigens, CD/metabolism , Antigens, CD/genetics , Cytidine/analogs & derivatives , Cytidine/pharmacology , Cell Line , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology , Rats, Sprague-Dawley , Cell Survival
15.
Exp Lung Res ; 50(1): 146-159, 2024.
Article in English | MEDLINE | ID: mdl-39243130

ABSTRACT

PURPOSE OF THE STUDY: To observe the dynamic changes in monocyte subsets during septic lung injury and to assess the anti-inflammatory role of the sulfotransferase homolog 2 (ST2) receptor. MATERIALS AND METHODS: Dynamic changes of monocyte subsets from patients with septic lung injury and mice post-cecal ligation and puncture (CLP) were monitored. ST2 receptors on mice monocytes and concentrations of IL-33, IL-1ß, IL-12, and IL-27 from peripheral blood or culture supernatant were detected. RESULTS: CD14lowCD16- (Mo0) and CD14++CD16+ (Mo2) monocyte subsets were significantly expanded in patients with sepsis-related acute respiratory distress syndrome. In sepsis model mice, monocyte counts, particularly of Ly6Cint and CDLy6Cint+hi monocytes, were significantly increased. The mean optical density value of TNF-α after CLP mainly increased after 24 h, whereas that of IL-6 was significantly increased at all time points assessed after CLP. The levels of IL-1ß, IL-12, IL-27, and IL-33 increased to variable degrees at 6, 12, 24, and 48h after CLP, and ST2+ monocytes were significantly expanded in sepsis model mice compared to sham-operated mice. ST2 receptor blockade suppressed IL-1ß and IL-12 production in cell culture. CONCLUSIONS: Changes in monocyte subsets expressing the ST2 receptor play an important role in septic lung injury by modulating inflammatory cytokine secretion.


Subject(s)
Cytokines , Monocytes , Sepsis , Animals , Monocytes/metabolism , Mice , Sepsis/metabolism , Male , Humans , Cytokines/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Female , Mice, Inbred C57BL , Middle Aged , Interleukin-33/metabolism , Lung Injury/metabolism , Acute Lung Injury/metabolism , Disease Models, Animal , Interleukin-1beta/metabolism , Aged , Interleukin-27/metabolism
16.
Ren Fail ; 46(2): 2369342, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39230047

ABSTRACT

Sepsis represents an organ dysfunction resulting from the host's maladjusted response to infection, and can give rise to acute kidney injury (AKI), which significantly increase the morbidity and mortality of septic patients. This study strived for identifying a novel therapeutic strategy for patients with sepsis-induced AKI (SI-AKI). Rat tubular epithelial NRK-52E cells were subjected to lipopolysaccharide (LPS) exposure for induction of in-vitro SI-AKI. The expressions of E1A binding protein p300 (EP300) and methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) in NRK-52E cells were assessed by western blot and qRT-PCR, and their interaction was explored by chromatin immunoprecipitation performed with antibody for H3K27 acetylation (H3K27ac). The effect of them on SI-AKI-associated mitochondrial dysfunction of tubular epithelial cells was investigated using transfection, MTT assay, TUNEL staining, 2',7'-Dichlorodihydrofluorescein diacetate probe assay, Mitosox assay, and JC-1 staining. MTHFD2 and EP300 were upregulated by LPS exposure in NRK-52E cells. LPS increased the acetylation of H3 histone in the MTHFD2 promoter region, and EP300 suppressed the effect of LPS. EP300 ablation inhibited the expression of MTHFD2. MTHFD2 overexpression antagonized LPS-induced viability reduction, apoptosis promotion, reactive oxygen species overproduction, and mitochondrial membrane potential collapse of NRK-52E cells. By contrast, MTHFD2 knockdown and EP300 ablation brought about opposite consequences. Furthermore, MTHFD2 overexpress and EP300 ablation counteracted each other's effect in LPS-exposed NRK-52E cells. EP300-mediated H3 acetylation elevates MTHFD2 expression to reduce mitochondrial dysfunction of tubular epithelial cells in SI-AKI.


Subject(s)
Acute Kidney Injury , E1A-Associated p300 Protein , Epithelial Cells , Lipopolysaccharides , Methylenetetrahydrofolate Dehydrogenase (NADP) , Mitochondria , Animals , Rats , Acetylation , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , E1A-Associated p300 Protein/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Epithelial Cells/metabolism , Mitochondria/metabolism , Cell Line , Histones/metabolism , Apoptosis , Sepsis/metabolism , Kidney Tubules/pathology , Kidney Tubules/metabolism , Up-Regulation
17.
Front Immunol ; 15: 1461051, 2024.
Article in English | MEDLINE | ID: mdl-39234245

ABSTRACT

As a grave and highly lethal clinical challenge, sepsis, along with its consequent multiorgan dysfunction, affects millions of people worldwide. Sepsis is a complex syndrome caused by a dysregulated host response to infection, leading to fatal organ dysfunction. An increasing body of evidence suggests that the pathogenesis of sepsis is both intricate and rapid and involves various cellular responses and signal transductions mediated by post-translational modifications (PTMs). Hence, a comprehensive understanding of the mechanisms and functions of PTMs within regulatory networks is imperative for understanding the pathological processes, diagnosis, progression, and treatment of sepsis. In this review, we provide an exhaustive and comprehensive summary of the relationship between PTMs and sepsis-induced organ dysfunction. Furthermore, we explored the potential applications of PTMs in the treatment of sepsis, offering a forward-looking perspective on the understanding of infectious diseases.


Subject(s)
Multiple Organ Failure , Protein Processing, Post-Translational , Sepsis , Humans , Sepsis/metabolism , Multiple Organ Failure/metabolism , Multiple Organ Failure/etiology , Multiple Organ Failure/immunology , Animals , Signal Transduction
18.
Sci Rep ; 14(1): 20400, 2024 09 02.
Article in English | MEDLINE | ID: mdl-39223234

ABSTRACT

To select the core target (RAB13) in sepsis patients' peripheral blood and investigate its molecular functions and possible mechanisms. The peripheral blood of sepsis patients (n = 21) and healthy individuals (n = 9) within 24 h after admission were collected for RNA-seq, and differential gene screening was performed by iDEP online analysis software (P < 0.01; log2FC ≥ 2) and enrichment analysis, the potential core target RAB13 was screened out. The association between RAB13 expression and sepsis severity was explored using multiple datasets in the GEO database, and survival analysis was conducted. Subsequently, peripheral blood mononuclear cells (PBMCs) from sepsis and control groups were isolated, and 10 × single-cell sequencing was used to identify the main RAB13-expressing cell types. Finally, LPS was used to stimulate THP1 cells to construct a sepsis model to explore the function and possible mechanism of RAB13. We found that RAB13 was a potential core target, and RAB13 expression level was positively associated with sepsis severity and negatively correlated with survival based on multiple public datasets. A single-cell sequencing indicated that RAB13 is predominantly localized in monocytes. Cell experiments validated that RAB13 is highly expressed in sepsis, and the knockdown of RAB13 promotes the polarization of macrophages towards the M2 phenotype. This mechanism may be associated with the ECM-receptor interaction signaling pathway. The upregulation of RAB13 in sepsis patients promotes the polarization of M2-like macrophages and correlates positively with the severity of sepsis.


Subject(s)
Macrophages , Sepsis , rab GTP-Binding Proteins , Humans , Sepsis/metabolism , Sepsis/genetics , Sepsis/pathology , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Macrophages/metabolism , Male , Female , Middle Aged , THP-1 Cells , Aged , Case-Control Studies , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology
19.
Sci Rep ; 14(1): 20313, 2024 09 02.
Article in English | MEDLINE | ID: mdl-39218968

ABSTRACT

Sepsis-induced cardiomyopathy (SIC) is described as a reversible myocardial depression that occurs in patients with septic shock. Increasing evidence shows that microRNA-194-5p (miR-194-5p) participates in the regulation of oxidative stress, mitochondrial dysfunction, and apoptosis and its expression is associated with the occurrence and progression of cardiovascular disease; however, the effects of miR-194-5p in SIC are still unclear. This study explores whether miR-194-5p could modulate SIC by affecting oxidative stress, mitochondrial function, and apoptosis. Experimental septic mice were induced by intraperitoneal injection of lipopolysaccharide (LPS) in C57BL/6J mice. The biological role of miR-194-5p in SIC in vivo was investigated using cardiac echocardiography, ELISA, western blot, qRT-PCR, transmission electron microscopy, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, bioinformatics analysis, and dual-luciferase reporter gene assay. Our major finding is that miR-194-5p antagomir mitigates sepsis-induced cardiac dysfunction, inflammation, oxidative stress, apoptosis and mitochondrial dysfunction in the hearts of septic mice, while miR-194-5p agomir triggers the opposite effects. Furthermore, dual-specificity phosphatase 9 (DUSP9) is a direct target of miR-194-5p and the cardioprotective effects of miR-194-5p antagomir on cardiac dysfunction, inflammation, apoptosis, mitochondrial dysfunction and oxidative stress are abolished through inhibiting DUSP9. Therefore, miR-194-5p inhibition could mitigate SIC via DUSP9 in vivo and the novel miR-194-5p/DUSP9 axis might be the potential treatment targets for SIC patients.


Subject(s)
Apoptosis , Cardiomyopathies , Dual-Specificity Phosphatases , Mice, Inbred C57BL , MicroRNAs , Oxidative Stress , Sepsis , Animals , Male , Mice , Antagomirs/pharmacology , Antagomirs/metabolism , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/genetics , Disease Models, Animal , Down-Regulation , Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/genetics , Lipopolysaccharides , MicroRNAs/genetics , MicroRNAs/metabolism , Sepsis/complications , Sepsis/metabolism , Sepsis/genetics
20.
Int J Mol Sci ; 25(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39273234

ABSTRACT

The clinical effectiveness of Oxiris®, particularly in reducing cytokines, remains uncertain due to the limited data provided. This study explored and analyzed the application value of Oxiris® endotoxin adsorption technology in a large animal model. Pigs received an intravenous LPS infusion. Six animals were treated 2 h after the infusion with an Oxiris® hemadsorption using a pumpless extracorporeal technique for 6 h. Five animals served as controls. Cardiocirculatory parameters, hyperspectral analysis, and a panel of cytokines were measured. The lipopolysaccharide infusion induced sepsis-like inflammation with tachycardia, elevated pulmonary pressure, elevated lactate level, as well as elevated pro-inflammatory cytokines like interferon (IFN)-γ, interleukin (IL)-1ß, IL-2, IL-6, IL-8, IL-12 and tumor necrosis factor alpha (TNF-α). In addition, increases of anti-inflammatory cytokines like IL-1ra and IL-10 were found. After 3 and 6 h in both groups, pro-inflammatory cytokines were significantly reduced. No differences between the intervention and the control group could be detected after 3 and 6 h for IL-1ß, IL-2, IL-6, IL-8, IL-12 and TNF-α, suggesting no effect of the Oxiris® filter on the elimination of elevated cytokines with a pumpless extracorporeal hemadsorption technique. The presented large animal model may be a promising option for studying the effects of hemadsorption techniques.


Subject(s)
Cytokines , Lipopolysaccharides , Animals , Cytokines/metabolism , Swine , Hemadsorption , Disease Models, Animal , Sepsis/drug therapy , Sepsis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL