Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Arch Toxicol ; 98(6): 1705-1716, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38555326

ABSTRACT

Amanita phalloides is the primary species responsible for fatal mushroom poisoning, as its main toxin, α-amanitin, irreversibly and potently inhibits eukaryotic RNA polymerase II (RNAP II), leading to cell death. There is no specific antidote for α-amanitin, which hinders its clinical application. However, with the advancement of precision medicine in oncology, including the development of antibody-drug conjugates (ADCs), the potential value of various toxic small molecules has been explored. These ADCs ingeniously combine the targeting precision of antibodies with the cytotoxicity of small-molecule payloads to precisely kill tumor cells. We searched PubMed for studies in this area using these MeSH terms "Amanitins, Alpha-Amanitin, Therapeutic use, Immunotherapy, Immunoconjugates, Antibodies" and did not limit the time interval. Recent studies have conducted preclinical experiments on ADCs based on α-amanitin, showing promising therapeutic effects and good tolerance in primates. The current challenges include the not fully understood toxicological mechanism of α-amanitin and the lack of clinical studies to evaluate the therapeutic efficacy of ADCs developed based on α-amanitin. In this article, we will discuss the role and therapeutic efficacy of α-amanitin as an effective payload in ADCs for the treatment of various cancers, providing background information for the research and application strategies of current and future drugs.


Subject(s)
Alpha-Amanitin , Immunoconjugates , Neoplasms , Humans , Neoplasms/drug therapy , Animals , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , RNA Polymerase II/metabolism , Mushroom Poisoning/drug therapy
2.
Burns Trauma ; 11: tkad022, 2023.
Article in English | MEDLINE | ID: mdl-37334140

ABSTRACT

Sepsis is the main cause of death in critically ill patients and gut microbiota dysbiosis plays a crucial role in sepsis. On the one hand, sepsis leads to the destruction of gut microbiota and induces and aggravates terminal organ dysfunction. On the other hand, the activation of pathogenic gut flora and the reduction in beneficial microbial products increase the susceptibility of the host to sepsis. Although probiotics or fecal microbiota transplantation preserve gut barrier function on multiple levels, their efficacy in sepsis with intestinal microbiota disruptions remains uncertain. Postbiotics consist of inactivated microbial cells or cell components. They possess antimicrobial, immunomodulatory, antioxidant and antiproliferative activities. Microbiota-targeted therapy strategies, such as postbiotics, may reduce the incidence of sepsis and improve the prognosis of patients with sepsis by regulating gut microbial metabolites, improving intestinal barrier integrity and changing the composition of the gut microbiota. They offer a variety of mechanisms and might even be superior to more conventional 'biotics' such as probiotics and prebiotics. In this review, we present an overview of the concept of postbiotics and summarize what is currently known about postbiotics and their prospective utility in sepsis therapy. Overall, postbiotics show promise as a viable adjunctive therapy option for sepsis.

3.
Front Pharmacol ; 14: 1101240, 2023.
Article in English | MEDLINE | ID: mdl-36713827

ABSTRACT

Sepsis-induced cardiomyopathy (SIC) is an important manifestation of sepsis, and abnormal cardiac function affects the development of sepsis. Notoginsenoside R1 (NG-R1) is a unique bioactive component of Panax notoginseng with anti-inflammatory and antioxidant effects. However, the effects and possible mechanisms of NG-R1 on SIC are not clear. The purpose of this study was to identify the potential targets and regulatory mechanisms of the action of NG-R1 on SIC. To investigate the potential mechanism, we used network pharmacology, molecular docking, qRT-PCR, and immunofluorescence. The results showed that NG-R1 ameliorated myocardial fibrosis in septic mice. Validation of network pharmacology and molecular docking results revealed that NG-R1 reduced tumor necrosis factor-Alpha (TNF-α) expression in myocardial tissues and AC16 cardiomyocytes in mice, as well as inflammatory factor release in AC16 cells, so TNF-α may be a potential target of NG-R1 against SIC. The present study demonstrated that NG-R1 could protect against SIC and by regulating the expression of TNF-α inflammatory factors, providing a new idea for sepsis drug development.

4.
Immunopharmacol Immunotoxicol ; 45(3): 277-286, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36263912

ABSTRACT

Objective: Sepsis is life-threatening organ dysfunction caused by the dysregulated host response to infection. Endoplasmic reticulum stress (ERS)-mediated inositol-requiring enzyme 1 α (IRE1α) inflammatory signaling pathway is involved in sepsis. NLRP3 inflammasome plays a key role in the activation of caspase-1 and the maturation of IL-1ß and IL-18, and finally enhances the inflammatory response. More and more evidences show that ERS is an endogenous trigger of NLRP3 inflammasome. Thioredoxin-1 (Trx-1) is a small ubiquitous thiol-1 protein with redox/inflammation modulatory properties relevant to sepsis pathogenesis. In this study, we investigated the role of Trx-1 in ERS mediated IRE1α/NLRP3 signaling pathway in Raw 264.7 cells.Methods: Raw 264.7 cells stimulated by LPS were used to construct an inflammation model of sepsis in vitro, and the expression of proteins related to the IRE1α/NLRP3 pathway was detected through using western blot and RT-PCR. The expression of IL-18 and IL-1ß in cell supernatant was also measured by ELISA, and caspase 1 activity and ROS expression in cells were detected by kits.Results: Our study shows that IRE1α signaling pathway related to endoplasmic reticulum stress in sepsis can activate inflammation related genes, and stimulate to produce a large number of pro-IL-1ß. At the same time, IRE1α can activate NLRP3 inflammasome and promote activation and maturation of pro-IL-1ß. Finally leads to excessive inflammatory response and ROS release, and promotes the progress of sepsis.Conclusions: Trx-1 may inhibit NLRP3 activity and pro-IL-Iß production by inhibit IRE1α pathway of ER stress. So as to inhibit inflammatory response and ROS of cells, and play a protective role in sepsis.


Subject(s)
Inflammasomes , Sepsis , Thioredoxins , Animals , Humans , Mice , Endoribonucleases/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism
5.
Arch Toxicol ; 97(1): 121-131, 2023 01.
Article in English | MEDLINE | ID: mdl-36271256

ABSTRACT

Amanita poisoning has a high mortality rate. The α-amanitin toxin in Amanita is the main lethal toxin. There is no specific detoxification drug for α-amanitin, and the clinical treatment mainly focuses on symptomatic and supportive therapy. The pathogenesis of α-amanitin mainly includes: α-amanitin can inhibit the activity of RNA polymeraseII in the nucleus, including the inhibition of the largest subunit of RNA polymeraseII, RNApb1, bridge helix, and trigger loop. In addition, α-amanitin acts in vivo through the enterohepatic circulation and transport system. α-Amanitin can cause the cell death. The existing mechanisms of cell damage mainly focus on apoptosis, oxidative stress, and autophagy. In addition to the pathogenic mechanism, α-amanitin also has a role in cancer treatment, which is the focus of current research. The mechanism of action of α-amanitin on the body is still being explored.


Subject(s)
Alpha-Amanitin , Mushroom Poisoning , Humans , Amanitins/metabolism , Mushroom Poisoning/drug therapy , Mushroom Poisoning/metabolism , Amanita , RNA
6.
Inflamm Res ; 71(10-11): 1191-1202, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35969260

ABSTRACT

PURPOSE: Gasdermin D (GSDMD) is a cytoplasmic protein that is encoded by the gasdermin family GSDMD gene and is the ultimate executor of pyroptosis. Pyroptosis is a mode of lysis and inflammation that regulates cell death, ultimately leading to cell swelling and rupture. In sepsis, a dysregulated host response to infection frequently results in hyperinflammatory responses and immunosuppression, eventually leading to multiple organ dysfunction. Pyroptosis regulates innate immune defenses and plays an important role in the process of inflammatory cell death, and the absence of any link in the entire pathway from GSDMD to pyroptosis causes bacterial clearance to be hampered. Under normal conditions, the process of pyroptosis occurs much faster than apoptosis, and the threat to the body is also much greater. MATERIALS AND METHODS: We conducted a systematic review of relevant reviews and experimental articles using the keywords sepsis, Gasdermin D, and Pyroptosis in the PubMed, Scopus, Google Scholar, and Web of Science databases. CONCLUSION: Combined with the pathogenesis of sepsis, it is not difficult to find that pyroptosis plays a key role in bacterial inflammation and sepsis. Therefore, GSDMD inhibitors may be used as targeted drugs to treat sepsis by reducing the occurrence of pyroptosis. This review mainly discusses the key role of GSDMD in sepsis.


Subject(s)
Intracellular Signaling Peptides and Proteins , Sepsis , Humans , Phosphate-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Pyroptosis
7.
Front Immunol ; 13: 1063543, 2022.
Article in English | MEDLINE | ID: mdl-36713461

ABSTRACT

Objective: Sepsis is the leading cause of death in critically ill patients. The gastrointestinal tract has long been thought to play an important role in the pathophysiology of sepsis. Antibiotic therapy can reduce a patient's commensal bacterial population and raise their risk of developing subsequent illnesses, where gut microbiota dysbiosis may be a key factor. Methods: In this study, we analyzed the 16S rRNA of fecal samples from both healthy people and patients with sepsis to determine if alterations in gut bacteria are associated with sepsis. Then, we developed a mouse model of sepsis using cecal ligation and puncture (CLP) in order to examine the effects of fecal microbiota transplantation (FMT) and short-chain fatty acids (SCFAs) on survival rate, systemic inflammatory response, gut microbiota, and mucosal barrier function. Results: Sepsis patients' gut microbiota composition significantly differed from that of healthy people. At the phylum level, the amount of Proteobacteria in the intestinal flora of sepsis patients was much larger than that of the control group, whereas the number of Firmicutes was significantly lower. Mice with gut microbiota disorders (ANC group) were found to have an elevated risk of death, inflammation, and organ failure as compared to CLP mice. However, all of these could be reversed by FMT and SCFAs. FMT and SCFAs could regulate the abundance of bacteria such as Firmicutes, Proteobacteria, Escherichia Shigella, and Lactobacillus, restoring them to levels comparable to those of healthy mice. In addition, they increased the expression of the Occludin protein in the colon of mice with sepsis, downregulated the expression of the NLRP3 and GSDMD-N proteins, and reduced the release of the inflammatory factors IL-1ß and IL-18 to inhibit cell pyroptosis, ultimately playing a protective role in sepsis. Disccusion: FMT and SCFAs provide a microbe-related survival benefit in a mouse model of sepsis, suggesting that they may be a viable treatment for sepsis.


Subject(s)
Gastrointestinal Microbiome , Sepsis , Mice , Animals , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , RNA, Ribosomal, 16S/genetics , Anti-Bacterial Agents/therapeutic use , Sepsis/therapy , Sepsis/microbiology , Fatty Acids, Volatile
8.
Life Sci ; 261: 118461, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32961227

ABSTRACT

AIMS: Parkinson's disease (PD) is a multifactorial neurodegenerative disorder. Its molecular mechanism is still unclear. Endoplasmic reticulum (ER) stress has been highlighted in PD. Transient receptor potential vanilloid 4 (TRPV4) is a kind of nonselective calcium cation channel. A defined role for TRPV4 in PD has not been reported. The purpose of the present research was to investigate the molecular mechanisms by which TRPV4 regulates ER stress induced by the 1-methyl-4-phenylpyridinium ion (MPP+) in PC12 cells. MAIN METHODS: PC12 cells were pretreated with the TRPV4-specific antagonist HC067047 or transfected with TRPV4 siRNA followed by treatment with MPP+. Cell viability was measured by the CCK-8 Assay. The expression of TRPV4, sarco/endoplasmic reticulum Ca2+-ATPase 2 (SERCA2), glucose-regulated protein 78 (GRP78), glucose-regulated protein 94 (GRP94), C/EBP homologous protein (CHOP), procaspase-12, and tyrosine hydroxylase (TH) was detected by western blot and RT-PCR. KEY FINDINGS: The expression of TRPV4 was upregulated, while cell viability was decreased by MPP+, which was reversed by HC067047. The ER stress common molecular signature SERCA2 was depressed by MPP+. Moreover, MPP+ induced upregulation of GRP78, GRP94, CHOP, and decrease in procaspase-12 and TH. HC067047 and TRPV4 siRNA reversed MPP+-induced ER stress and restored TH production. SIGNIFICANCE: TRPV4 functions upstream of ER stress induced by MPP+ and holds promise as a prospective pharmacotherapy target for PD.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Parkinson Disease, Secondary/pathology , TRPV Cation Channels/metabolism , 1-Methyl-4-phenylpyridinium , Animals , Cell Survival , PC12 Cells , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/metabolism , Rats , TRPV Cation Channels/analysis , TRPV Cation Channels/genetics , Up-Regulation
9.
Immunopharmacol Immunotoxicol ; 42(3): 280-285, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32326777

ABSTRACT

Background: Sepsis is life-threatening organ dysfunction caused by a dysregulated host response to infection. Inflammatory response and oxidative stress play an important role in the pathophysiological process of sepsis. Thioredoxin-1 (Trx-1) is a small ubiquitous thiol protein with redox/inflammation modulatory properties relevant to the pathogenesis of sepsis. We therefore investigated the expression level and significance of Trx-1, inflammatory factors and oxidative stress in peripheral blood of sepsis patients, and to explore Trx-1 relationship with inflammatory factors and oxidative stress.Methods: Plasma samples were collected from patients with sepsis and those with healthy control. Enzyme-linked immunosorbent assays (ELISA) were used to detect for interleukin (IL-1ß), IL-6, tumor necrosis factor (TNF-α), E-selectin, endothelin-1 (ET-1), thioredoxin-1, C-reactive protein (CRP), procalcitonin (PCT) for human plasma samples; RT-PCR detection of Trx-1 and thioredoxin-interacting protein (TXNIP) mRNA levels. Colorimetric assay for glutathione (GSH) and malondialdehyde (MDA) expression level in peripheral blood of patients with sepsis; Disease severity was assessed as APACHE II.Results: The expression levels of Trx-1, inflammatory factors and oxidative stress in plasma of patients with sepsis were significantly increased, TXNIP opposite.Conclusion: Our results show that Trx-1 play important role in inflammation and oxidative stress in sepsis patients. Trx-1 may be a potential therapeutic target in sepsis.


Subject(s)
Cytokines/genetics , Gene Expression , Oxidative Stress/immunology , Sepsis/blood , Thioredoxins/genetics , Case-Control Studies , Cohort Studies , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression/immunology , Humans , Male , Middle Aged , Oxidative Stress/genetics , Real-Time Polymerase Chain Reaction , Sepsis/immunology , Thioredoxins/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...