Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.591
Filter
1.
J Am Chem Soc ; 146(22): 14972-14988, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38787738

ABSTRACT

Caspases are a highly conserved family of cysteine-aspartyl proteases known for their essential roles in regulating apoptosis, inflammation, cell differentiation, and proliferation. Complementary to genetic approaches, small-molecule probes have emerged as useful tools for modulating caspase activity. However, due to the high sequence and structure homology of all 12 human caspases, achieving selectivity remains a central challenge for caspase-directed small-molecule inhibitor development efforts. Here, using mass spectrometry-based chemoproteomics, we first identify a highly reactive noncatalytic cysteine that is unique to caspase-2. By combining both gel-based activity-based protein profiling (ABPP) and a tobacco etch virus (TEV) protease activation assay, we then identify covalent lead compounds that react preferentially with this cysteine and afford a complete blockade of caspase-2 activity. Inhibitory activity is restricted to the zymogen or precursor form of monomeric caspase-2. Focused analogue synthesis combined with chemoproteomic target engagement analysis in cellular lysates and in cells yielded both pan-caspase-reactive molecules and caspase-2 selective lead compounds together with a structurally matched inactive control. Application of this focused set of tool compounds to stratify the functions of the zymogen and partially processed (p32) forms of caspase-2 provide evidence to support that caspase-2-mediated response to DNA damage is largely driven by the partially processed p32 form of the enzyme. More broadly, our study highlights future opportunities for the development of proteoform-selective caspase inhibitors that target nonconserved and noncatalytic cysteine residues.


Subject(s)
Caspase 2 , Caspase Inhibitors , Proteomics , Humans , Caspase 2/metabolism , Caspase 2/chemistry , Proteomics/methods , Caspase Inhibitors/pharmacology , Caspase Inhibitors/chemistry , Caspase Inhibitors/metabolism , Molecular Structure , Cysteine Endopeptidases
2.
Retrovirology ; 21(1): 8, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38693565

ABSTRACT

The study of HIV infection and pathogenicity in physical reservoirs requires a biologically relevant model. The human immune system (HIS) mouse is an established model of HIV infection, but defects in immune tissue reconstitution remain a challenge for examining pathology in tissues. We utilized exogenous injection of the human recombinant FMS-like tyrosine kinase 3 ligand (rFLT-3 L) into the hematopoietic stem cell (HSC) cord blood HIS mouse model to significantly expand the total area of lymph node (LN) and the number of circulating human T cells. The results enabled visualization and quantification of HIV infectivity, CD4 T cell depletion and other measures of pathogenesis in the secondary lymphoid tissues of the spleen and LN. Treatment with the Caspase-1/4 inhibitor VX-765 limited CD4+ T cell loss in the spleen and reduced viral load in both the spleen and axillary LN. In situ hybridization further demonstrated a decrease in viral RNA in both the spleen and LN. Transcriptomic analysis revealed that in vivo inhibition of caspase-1/4 led to an upregulation in host HIV restriction factors including SAMHD1 and APOBEC3A. These findings highlight the use of rFLT-3 L to augment human immune system characteristics in HIS mice to support investigations of HIV pathogenesis and test host directed therapies, though further refinements are needed to further augment LN architecture and cellular populations. The results further provide in vivo evidence of the potential to target inflammasome pathways as an avenue of host-directed therapy to limit immune dysfunction and virus replication in tissue compartments of HIV+ persons.


Subject(s)
CD4-Positive T-Lymphocytes , Disease Models, Animal , HIV Infections , HIV-1 , Animals , Mice , HIV Infections/immunology , HIV Infections/virology , HIV Infections/drug therapy , HIV-1/physiology , HIV-1/drug effects , Humans , CD4-Positive T-Lymphocytes/immunology , Lymphoid Tissue/virology , Lymphoid Tissue/immunology , Viral Load/drug effects , Spleen/virology , Spleen/immunology , Lymph Nodes/immunology , Lymph Nodes/virology , Caspases/metabolism , Caspase Inhibitors/pharmacology , Anti-Retroviral Agents/therapeutic use
3.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791499

ABSTRACT

The activation of caspases is a crucial event and an indicator of programmed cell death, also known as apoptosis. These enzymes play a central role in cancer biology and are considered one promising target for current and future advancements in therapeutic interventions. Traditional methods of measuring caspase activity such as antibody-based methods provide fundamental insights into their biological functions, and are considered essential tools in the fields of cell and cancer biology, pharmacology and toxicology, and drug discovery. However, traditional methods, though extensively used, are now recognized as having various shortcomings. In addition, these methods fall short of providing solutions to and matching the needs of the rapid and expansive progress achieved in studying caspases. For these reasons, there has been a continuous improvement in detection methods for caspases and the network of pathways involved in their activation and downstream signaling. Over the past decade, newer methods based on cutting-edge state-of-the-art technologies have been introduced to the biomedical community. These methods enable both the temporal and spatial monitoring of the activity of caspases and their downstream substrates, and with enhanced accuracy and precision. These include fluorescent-labeled inhibitors (FLIs) for live imaging, single-cell live imaging, fluorescence resonance energy transfer (FRET) sensors, and activatable multifunctional probes for in vivo imaging. Recently, the recruitment of mass spectrometry (MS) techniques in the investigation of these enzymes expanded the repertoire of tools available for the identification and quantification of caspase substrates, cleavage products, and post-translational modifications in addition to unveiling the complex regulatory networks implicated. Collectively, these methods are enabling researchers to unravel much of the complex cellular processes involved in apoptosis, and are helping generate a clearer and comprehensive understanding of caspase-mediated proteolysis during apoptosis. Herein, we provide a comprehensive review of various assays and detection methods as they have evolved over the years, so to encourage further exploration of these enzymes, which should have direct implications for the advancement of therapeutics for cancer and other diseases.


Subject(s)
Caspases , Caspases/metabolism , Humans , Animals , Apoptosis , Fluorescence Resonance Energy Transfer/methods , Neoplasms/diagnosis , Neoplasms/metabolism , Caspase Inhibitors/pharmacology , Fluorescent Dyes/chemistry
4.
Acta Pharmacol Sin ; 45(7): 1381-1392, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38514863

ABSTRACT

Status epilepticus (SE), a serious and often life-threatening medical emergency, is characterized by abnormally prolonged seizures. It is not effectively managed by present first-line anti-seizure medications and could readily develop into drug resistance without timely treatment. In this study, we highlight the therapeutic potential of CZL80, a small molecule that inhibits caspase-1, in SE termination and its related mechanisms. We found that delayed treatment of diazepam (0.5 h) easily induces resistance in kainic acid (KA)-induced SE. CZL80 dose-dependently terminated diazepam-resistant SE, extending the therapeutic time window to 3 h following SE, and also protected against neuronal damage. Interestingly, the effect of CZL80 on SE termination was model-dependent, as evidenced by ineffectiveness in the pilocarpine-induced SE. Further, we found that CZL80 did not terminate KA-induced SE in Caspase-1-/- mice but partially terminated SE in IL1R1-/- mice, suggesting the SE termination effect of CZL80 was dependent on the caspase-1, but not entirely through the downstream IL-1ß pathway. Furthermore, in vivo calcium fiber photometry revealed that CZL80 completely reversed the neuroinflammation-augmented glutamatergic transmission in SE. Together, our results demonstrate that caspase-1 inhibitor CZL80 terminates diazepam-resistant SE by blocking glutamatergic transmission. This may be of great therapeutic significance for the clinical treatment of refractory SE.


Subject(s)
Anticonvulsants , Caspase 1 , Mice, Inbred C57BL , Status Epilepticus , Animals , Status Epilepticus/drug therapy , Caspase 1/metabolism , Mice , Male , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Kainic Acid/pharmacology , Mice, Knockout , Glutamic Acid/metabolism , Caspase Inhibitors/pharmacology , Caspase Inhibitors/therapeutic use , Diazepam/pharmacology , Diazepam/therapeutic use , Synaptic Transmission/drug effects
5.
Eur J Immunol ; 54(5): e2350515, 2024 May.
Article in English | MEDLINE | ID: mdl-38361219

ABSTRACT

Caspase-1 location in cells has been studied with fluorochrome-labeled inhibitors of caspase-1 (FLICA reagents). We report that FLICA reagents have limited cell-membrane permeability. This impacts experimental design as cells with intact membranes, including caspase-1 knockout cells, are not appropriate controls for cells with inflammasome-induced gasdermin D membrane pores.


Subject(s)
Caspase 1 , Caspase Inhibitors , Cell Membrane Permeability , Fluorescent Dyes , Inflammasomes , Macrophages , Caspase 1/metabolism , Animals , Macrophages/immunology , Macrophages/metabolism , Cell Membrane Permeability/drug effects , Mice , Inflammasomes/metabolism , Caspase Inhibitors/pharmacology , Mice, Knockout , Phosphate-Binding Proteins/metabolism , Humans
6.
J Virol ; 98(2): e0197523, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38294249

ABSTRACT

The highly pathogenic arenavirus, Junín virus (JUNV), expresses three truncated alternative isoforms of its nucleoprotein (NP), i.e., NP53kD, NP47kD, and NP40kD. While both NP47kD and NP40kD have been previously shown to be products of caspase cleavage, here, we show that expression of the third isoform NP53kD is due to alternative in-frame translation from M80. Based on this information, we were able to generate recombinant JUNVs lacking each of these isoforms. Infection with these mutants revealed that, while all three isoforms contribute to the efficient control of caspase activation, NP40kD plays the predominant role. In contrast to full-length NP (i.e., NP65kD), which is localized to inclusion bodies, where viral RNA synthesis takes place, the loss of portions of the N-terminal coiled-coil region in these isoforms leads to a diffuse cytoplasmic distribution and a loss of function in viral RNA synthesis. Nonetheless, NP53kD, NP47kD, and NP40kD all retain robust interferon antagonistic and 3'-5' exonuclease activities. We suggest that the altered localization of these NP isoforms allows them to be more efficiently targeted by activated caspases for cleavage as decoy substrates, and to be better positioned to degrade viral double-stranded (ds)RNA species that accumulate in the cytoplasm during virus infection and/or interact with cytosolic RNA sensors, thereby limiting dsRNA-mediated innate immune responses. Taken together, this work provides insight into the mechanism by which JUNV leverages apoptosis during infection to generate biologically distinct pools of NP and contributes to our understanding of the expression and biological relevance of alternative protein isoforms during virus infection.IMPORTANCEA limited coding capacity means that RNA viruses need strategies to diversify their proteome. The nucleoprotein (NP) of the highly pathogenic arenavirus Junín virus (JUNV) produces three N-terminally truncated isoforms: two (NP47kD and NP40kD) are known to be produced by caspase cleavage, while, here, we show that NP53kD is produced by alternative translation initiation. Recombinant JUNVs lacking individual NP isoforms revealed that all three isoforms contribute to inhibiting caspase activation during infection, but cleavage to generate NP40kD makes the biggest contribution. Importantly, all three isoforms retain their ability to digest double-stranded (ds)RNA and inhibit interferon promoter activation but have a diffuse cytoplasmic distribution. Given the cytoplasmic localization of both aberrant viral dsRNAs, as well as dsRNA sensors and many other cellular components of innate immune activation pathways, we suggest that the generation of NP isoforms not only contributes to evasion of apoptosis but also robust control of the antiviral response.


Subject(s)
Caspases , Cytoplasm , Hemorrhagic Fever, American , Host-Pathogen Interactions , Immunity, Innate , Junin virus , Nucleoproteins , Protein Biosynthesis , Humans , Apoptosis , Caspase Inhibitors/metabolism , Caspases/metabolism , Cytoplasm/metabolism , Cytoplasm/virology , Enzyme Activation , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , Interferons/genetics , Interferons/immunology , Junin virus/genetics , Junin virus/metabolism , Junin virus/pathogenicity , Nucleoproteins/biosynthesis , Nucleoproteins/genetics , Nucleoproteins/metabolism , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Viral/biosynthesis , RNA, Viral/genetics , Virus Replication
7.
J Pharmacol Exp Ther ; 388(2): 367-375, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37918856

ABSTRACT

Organophosphates cause hyperstimulation of the central nervous system, leading to extended seizures, convulsions, and brain damage. Sarin is a highly toxic organophosphate nerve agent that has been employed in several terrorist attacks. The prolonged toxicity of sarin may be enhanced by the neuroinflammatory response initiated by the inflammasome, caspase involvement, and generation/release of proinflammatory cytokines. Since neurodegeneration and neuroinflammation are prevalent in sarin-exposed animals, we were interested in evaluating the capacity of quinolyl-valyl-O-methylaspartyl-[-2,6-difluorophenoxy]-methyl ketone (Q-VD-OPh), a pan caspase inhibitor to attenuate neuroinflammation following sarin exposure. To test this hypothesis, sarin-exposed C57BL/6 mice were treated with Q-VD-OPh or negative control quinolyl-valyl-O-methylglutamyl-[-2,6-difluorophenoxy]-methyl ketone, sacrificed at 2- and 14-day time points, followed by removal of the amygdala and hippocampus. A Bio-Rad 23-Plex cytokine analysis was completed on each tissue. The results suggest that exposure to sarin induced a dramatic increase in interleukin-1ß and 6 other cytokines and a decrease in 2 of the 23 cytokines at 2 days in the amygdala compared with controls. Q-VD-OPh attenuated these changes at the 2-day time point. At 14 days, six of these cytokines were still significantly different from controls. Hippocampus was less affected at both time points. Diazepam, a neuroprotective drug against nerve agents, caused an increase in several cytokines but did not have a synergistic effect with Q-VD-OPh. Treatment of sarin exposure with apoptosis inhibitors appears to be a worthwhile approach for further testing as a comprehensive counteragent against organophosphate exposure. SIGNIFICANCE STATEMENT: A pan inhibitor of caspases (Q-VD-OPh) was proposed as a potential antidote for sarin-induced neuroinflammation by reducing the level of inflammation via inflammasome caspase inhibition. Q-VD-OPh added at 30 minutes post-sarin exposure attenuated the inflammatory response of a number of cytokines and chemokines in the amygdala and hippocampus, two brain regions sensitive to organophosphate exposure. Apoptotic marker reduction at 2 and 14 days further supports further testing of inhibitors of apoptosis as a means to lessen extended organophosphate toxicity in the brain.


Subject(s)
Amino Acid Chloromethyl Ketones , Nerve Agents , Quinolines , Sarin , Mice , Animals , Sarin/toxicity , Caspase Inhibitors/pharmacology , Caspase Inhibitors/therapeutic use , Neuroinflammatory Diseases , Inflammasomes , Mice, Inbred C57BL , Seizures/chemically induced , Seizures/drug therapy , Brain , Cytokines , Nerve Agents/pharmacology , Caspases , Inflammation/chemically induced , Inflammation/drug therapy , Organophosphates/pharmacology , Ketones/adverse effects
8.
Sci Rep ; 13(1): 21868, 2023 12 10.
Article in English | MEDLINE | ID: mdl-38072861

ABSTRACT

High transfection efficiency is the most important point for experiments of DNA and RNA introduction into cells. Decrease of cell viability during the transfection procedure is a crucial issue, resulting in transfection failure. However, the mechanism underlying cell growth inhibition has not been fully elucidated. Lipofection is frequently used for transfection experiments, whereases, depending on cell type, it causes a decrease in cell viability. The present study demonstrates here that a potent pan-caspase inhibitor Q-VD-OPh blocked cell death during the lipofection, indicating apoptosis was induced in lipofection. Moreover, Q-VD-OPh drastically increased transfected cells. This method provides easier and more effective transfection system of lipofection and may be useful for transfection of not only cell lines but also clinical uses such as gene therapy and nucleic acids vaccine.


Subject(s)
Caspases , Liposomes , Caspases/genetics , Transfection , Liposomes/pharmacology , Apoptosis , Caspase Inhibitors/pharmacology
9.
Eur J Med Chem ; 261: 115861, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37857145

ABSTRACT

Caspase-1 is a critical mediator of the inflammatory process by activating various pro-inflammatory cytokines such as pro-IL-1ß, IL-18 and IL-33. Uncontrolled activation of caspase-1 leads to various cytokines-mediated diseases. Thus, inhibition of Caspase-1 is considered therapeutically beneficial to halt the progression of such diseases. Currently, rilonacept, canakinumab and anakinra are in use for caspase-1-mediated autoinflammatory diseases. However, the poor pharmacokinetic profile of these peptides limits their use as therapeutic agents. Therefore, several peptidomimetic inhibitors have been developed, but only a few compounds (VX-740, VX-765) have advanced to clinical trials; because of their toxic profile. Several small molecule inhibitors have also been progressing based on the three-dimensional structure of caspase-1. However there is no successful candidate available clinically. In this perspective, we highlight the mechanism of caspase-1 activation, its therapeutic potential as a disease target and potential therapeutic strategies targeting caspase-1 with their limitations.


Subject(s)
Cytokines , Enzyme Inhibitors , Caspase 1 , Interleukin-1beta , Caspase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology
10.
Eur J Med Chem ; 259: 115632, 2023 Nov 05.
Article in English | MEDLINE | ID: mdl-37453329

ABSTRACT

Recent Alzheimer's research has shown increasing interest in the caspase-2 (Casp2) enzyme. However, the available Casp2 inhibitors, which have been pentapeptides or peptidomimetics, face challenges for use as CNS drugs. In this study, we successfully screened a 1920-compound chloroacetamide-based, electrophilic fragment library from Enamine. Our two-point dose screen identified 64 Casp2 hits, which were further evaluated in a ten-point dose-response study to assess selectivity over Casp3. We discovered compounds with inhibition values in the single-digit micromolar and sub-micromolar range, as well as up to 32-fold selectivity for Casp2 over Casp3. Target engagement analysis confirmed the covalent-irreversible binding of the selected fragments to Cys320 at the active site of Casp2. Overall, our findings lay a strong foundation for the future development of small-molecule Casp2 inhibitors.


Subject(s)
Caspase 2 , Caspase Inhibitors , Caspase 2/metabolism , Caspase 3/metabolism , Catalytic Domain , Caspase Inhibitors/chemistry
11.
Front Immunol ; 14: 1138920, 2023.
Article in English | MEDLINE | ID: mdl-37346039

ABSTRACT

Introduction: Inhibitors of the ATR kinase act as radiosensitizers through abrogating the G2 checkpoint and reducing DNA repair. Recent studies suggest that ATR inhibitors can also increase radiation-induced antitumor immunity, but the underlying immunomodulating mechanisms remain poorly understood. Moreover, it is poorly known how such immune effects relate to different death pathways such as caspase-dependent apoptosis. Here we address whether ATR inhibition in combination with irradiation may increase the presentation of hallmark factors of immunogenic cell death (ICD), and to what extent caspase activation regulates this response. Methods: Human lung cancer and osteosarcoma cell lines (SW900, H1975, H460, U2OS) were treated with X-rays and ATR inhibitors (VE822; AZD6738) in the absence and presence of a pan-caspase inhibitor. The ICD hallmarks HMGB1 release, ATP secretion and calreticulin surface-presentation were assessed by immunoblotting of growth medium, the CellTiter-Glo assay and an optimized live-cell flow cytometry assay, respectively. To obtain accurate measurement of small differences in the calreticulin signal by flow cytometry, we included normalization to a barcoded control sample. Results: Extracellular release of HMGB1 was increased in all the cell lines at 72 hours after the combined treatment with radiation and ATR inhibitors, relative to mock treatment or cells treated with radiation alone. The HMGB1 release correlated largely - but not strictly - with loss of plasma membrane integrity, and was suppressed by addition of the caspase inhibitor. However, one cell line showed HMGB1 release despite caspase inhibition, and in this cell line caspase inhibition induced pMLKL, a marker for necroptosis. ATP secretion occurred already at 48 hours after the co-treatment and did clearly not correlate with loss of plasma membrane integrity. Addition of pan-caspase inhibition further increased the ATP secretion. Surface-presentation of calreticulin was increased at 24-72 hours after irradiation, but not further increased by either ATR or caspase inhibition. Conclusion: These results show that ATR inhibition can increase the presentation of two out of three ICD hallmark factors from irradiated human cancer cells. Moreover, caspase activation distinctly affects each of the hallmark factors, and therefore likely plays a dual role in tumor immunogenicity by promoting both immunostimulatory and -suppressive effects.


Subject(s)
Caspases , HMGB1 Protein , Humans , Caspases/metabolism , HMGB1 Protein/metabolism , Calreticulin/metabolism , Caspase Inhibitors , Immunogenic Cell Death , Cell Line, Tumor , Protein Kinase Inhibitors , Adenosine Triphosphate , Ataxia Telangiectasia Mutated Proteins/metabolism
12.
J Vet Med Sci ; 85(4): 515-522, 2023 Apr 22.
Article in English | MEDLINE | ID: mdl-36889691

ABSTRACT

Bupivacaine, levobupivacaine and ropivacaine are potent, long acting, amide-type local anesthetics that have several clinical applications including intra-articular administration. The objectives of this study were to evaluate their in vitro effects on cell viability and caspase activity to elucidate whether they activate the extrinsic or intrinsic pathways of apoptosis in canine articular chondrocytes. Chondrocytes in monolayer culture were treated with culture medium as the control, or with 0.062% (0.62 mg/mL) bupivacaine, 0.062% levobupivacaine, and 0.062% ropivacaine for 24 hr. Cell viability was evaluated using the live/dead, 3-(4,5-dimehylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT), and Cell Counting Kit-8 (CCK-8) assays. Evaluation of caspase-3, caspase-8, and caspase-9 activity was performed using colorimetric assays. The MTT and CCK-8 assays were used to evaluate the effect of caspase inhibitors on local anesthetic chondrotoxicity. All three local anesthetics decreased chondrocyte viability after 24 hr (P<0.001). Apoptosis was induced through both the extrinsic and intrinsic pathways. Bupivacaine increased caspase-3, caspase-8, and caspase-9 activity (P<0.001). Levobupivacaine increased caspase-3 (P=0.03) while ropivacaine did not significantly upregulate activity for all three caspases. Caspase inhibition did not suppress bupivacaine chondrotoxicity whereas inhibition of caspase-8 and caspase-9 decreased ropivacaine chondrotoxicity and mildly attenuated levobupivacaine chondrotoxicity. In summary, the level of chondrotoxicity, the type of caspase activated, the level of caspase activation, and the response to caspase inhibitors was dependent on the type of local anesthetic. Therefore, ropivacaine may be a safer choice for intra-articular administration compared to levobupivacaine and bupivacaine.


Subject(s)
Anesthetics, Local , Bupivacaine , Animals , Dogs , Ropivacaine/toxicity , Chondrocytes , Levobupivacaine/pharmacology , Caspase 3 , Caspase 9/pharmacology , Caspase 8 , Caspase Inhibitors/pharmacology , Caspases
13.
Gut Microbes ; 15(1): 2163838, 2023.
Article in English | MEDLINE | ID: mdl-36656595

ABSTRACT

Conflicting evidence exists on the association between consumption of non-steroidal anti-inflammatory drugs (NSAIDs) and symptomatic worsening of inflammatory bowel disease (IBD). We hypothesized that the heterogeneous prevalence of pathobionts [e.g., adherent-invasive Escherichia coli (AIEC)], might explain this inconsistent NSAIDs/IBD correlation. Using IL10-/- mice, we found that NSAID aggravated colitis in AIEC-colonized animals. This was accompanied by activation of the NLRP3 inflammasome, Caspase-8, apoptosis, and pyroptosis, features not seen in mice exposed to AIEC or NSAID alone, revealing an AIEC/NSAID synergistic effect. Inhibition of NLRP3 or Caspase-8 activity ameliorated colitis, with reduction in NLRP3 inflammasome activation, cell death markers, activated T-cells and macrophages, improved histology, and increased abundance of Clostridium cluster XIVa species. Our findings provide new insights into how NSAIDs and an opportunistic gut-pathobiont can synergize to worsen IBD symptoms. Targeting the NLRP3 inflammasome or Caspase-8 could be a potential therapeutic strategy in IBD patients with gut inflammation, which is worsened by NSAIDs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Animals , Mice , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Caspase 8/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , Inflammasomes , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Caspase Inhibitors/pharmacology , Escherichia coli/pathogenicity
14.
Ecotoxicol Environ Saf ; 249: 114359, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36508797

ABSTRACT

Silicosis is a diffuse fibrotic lung disease in which excessive inflammatory responses are triggered by silica exposure. Pyroptosis, a pro-inflammatory mode of programmed cell death, is mediated by gasdermin and may play a pivotal role in the development of silicosis. The caspase-1 inhibitor, VX-765, was used in vivo and in vitro to investigate the effects of silica-induced early inflammatory injury and later lung fibrosis. Our findings show that VX-765 reduces inflammatory lung injury by inhibiting silica-induced pyroptosis of alveolar macrophages in a silicosis mouse model. VX-765 limits the infiltration of inflammatory M1 alveolar macrophages, decreasing expression of inflammatory cytokines, including IL-1ß, TNF-α, IL-6, CCL2, and CCL3, and down-regulating endogenous DAMPs and inflammatory immune-related cell pattern recognition receptors TLR4 and NLRP3. Furthermore, VX-765 alleviates fibrosis by down-regulating α-smooth muscle actin (α-SMA), collagen, and fibronectin. In this study, we illustrate that Alveolar macrophages pyroptosis occur in the early stages of silicosis, and VX-765 can alleviate the development of silicosis by inhibiting the pyroptosis signaling pathway. These results may provide new insight into the prevention and treatment of early-stage silicosis.


Subject(s)
Caspase Inhibitors , Lung Injury , Pulmonary Fibrosis , Pyroptosis , Silicosis , Animals , Mice , Lung Injury/chemically induced , Lung Injury/drug therapy , Lung Injury/pathology , Macrophages, Alveolar/drug effects , Pyroptosis/drug effects , Silicon Dioxide/toxicity , Silicosis/drug therapy , Caspase Inhibitors/pharmacology , Caspase Inhibitors/therapeutic use , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy
15.
Front Immunol ; 13: 1050354, 2022.
Article in English | MEDLINE | ID: mdl-36505441

ABSTRACT

Porcine sapelovirus (PSV) is an emerging pathogen associated with symptoms of enteritis, pneumonia, polioencephalomyelitis and reproductive disorders in swine, resulting in significant economic losses. Although PSV is reported to trigger cell apoptosis, its specific molecular mechanism is unclear. In this research, the cell apoptosis induced by PSV infection and its underlying mechanisms were investigated. The morphologic features of apoptosis include nuclear condensation and fragmentation, were observed after PSV infection. The cell apoptosis was confirmed by analyzing the apoptotic rates, caspase activation, and PARP1 cleavage. Caspase inhibitors inhibited the PSV-induced intrinsic apoptosis pathway and reduced viral replication. Among the proteins encoded by PSV, 2A is an important factor in inducing the mitochondrial apoptotic pathway. The conserved residues H48, D91, and C164 related to protease activity in PSV 2A were crucial for 2A-induced apoptosis. In conclusion, our results provide insights into how PSV induces host cell apoptosis.


Subject(s)
Apoptosis , Mitochondria , Swine , Animals , Caspase Inhibitors , Proteolysis , Protein Processing, Post-Translational
16.
Front Immunol ; 13: 1067767, 2022.
Article in English | MEDLINE | ID: mdl-36561752

ABSTRACT

The establishment of a latency reservoir is the major obstacle for a cure of HIV-1. The shock-and-kill strategy aims to reactivate HIV-1 replication in HIV -1 latently infected cells, exposing the HIV-1-infected cells to cytotoxic lymphocytes. However, none of the latency reversal agents (LRAs) tested so far have shown the desired effect in people living with HIV-1. We observed that NK cells stimulated with a pan-caspase inhibitor induced latency reversal in co-cultures with HIV-1 latently infected cells. Synergy in HIV-1 reactivation was observed with LRAs prostratin and JQ1. The supernatants of the pan-caspase inhibitor-treated NK cells activated the HIV-1 LTR promoter, indicating that a secreted factor by NK cells was responsible for the HIV-1 reactivation. Assessing changes in the secreted cytokine profile of pan-caspase inhibitor-treated NK cells revealed increased levels of the HIV-1 suppressor chemokines MIP1α (CCL3), MIP1ß (CCL4) and RANTES (CCL5). However, these cytokines individually or together did not induce LTR promoter activation, suggesting that CCL3-5 were not responsible for the observed HIV-1 reactivation. The cytokine profile did indicate that pan-caspase inhibitors induce NK cell activation. Altogether, our approach might be-in combination with other shock-and-kill strategies or LRAs-a strategy for reducing viral latency reservoirs and a step forward towards eradication of functionally active HIV-1 in infected individuals.


Subject(s)
Caspase Inhibitors , HIV Infections , HIV-1 , Killer Cells, Natural , Virus Latency , Humans , Caspase Inhibitors/pharmacology , CD4-Positive T-Lymphocytes/immunology , Cytokines/pharmacology , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/physiology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Virus Latency/immunology
17.
Int Immunopharmacol ; 113(Pt A): 109275, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36274488

ABSTRACT

PURPOSE: To explore the role of caspase-8 in mediating the transition between different death modes in fungal keratitis. METHODS: The expression of caspase-8 in Aspergillus fumigatus (A. fumigatus) keratitis was detected using western blotting and immunofluorescence. After subconjunctival injection of Z-IETD-FMK (caspase-8 inhibitor) or VX765 (caspase-1 inhibitor), the mice corneas of A. fumigatus keratitis were observed and scored under a slit lamp. Colony plate count, immunofluorescence staining, western blotting and qRT-PCR experiments were used to detect fungal load, inflammatory cells, and the production of related mRNAs and proteins. In vitro experiments, the LDH release test, Cell Count Kit-8(CCK-8) assay, ELISA, qRT-PCR and western blotting were used to detect cell viability, related mRNAs and proteins. RESULTS: The caspase-8 protein was upregulated following fungal infection. Compared with the A. fumigatus keratitis group, the mice treated with Z-IETD-FMK had heavier corneal turbidity, higher clinical scores, more fungal load and fewer inflammatory cells. The expression of NLRP3, cleaved-caspase-1, N-GSDMD, and IL-1ß in the fungal infection group after Z-IETD-FMK pretreatment were downregulated, while RIPK3 and p-MLKL were upregulated. In the fungal infection group after VX765 pretreatment, the expression of cleaved-caspase-8 was up-regulated, while N-GSDMD was downregulated. CONCLUSIONS: Caspase-8 is involved in the early immune defense response of A. fumigatus keratitis. It is essential for the recruitment of inflammatory cells and the clearance of the fungus. In A. fumigatus keratitis, activated caspase-8 promoted the caspase-1/GSDMD signaling pathway to participate in pyroptosis, inhibited RIPK3/MLKL signaling pathway-mediated necroptosis, and promoted IL-1ß maturation and release by activating the NLRP3 inflammasomes.


Subject(s)
Aspergillosis , Caspase 8 , Keratitis , Animals , Mice , Aspergillus fumigatus , Caspase 1/metabolism , Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Keratitis/microbiology , Mice, Inbred C57BL , Necroptosis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis
18.
Am J Physiol Gastrointest Liver Physiol ; 323(5): G439-G460, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36165492

ABSTRACT

DNA sensor pathways can initiate inflammasome, cell death, and type I interferon (IFN) signaling in immune-mediated inflammatory diseases (IMIDs), including type I interferonopathies. We investigated the involvement of these pathways in the pathogenesis of ulcerative colitis (UC) by analyzing the expression of DNA sensor, inflammasome, and type I IFN biomarker genes in colonic mucosal biopsy tissue from control (n = 31), inactive UC (n = 31), active UC (n = 33), and a UC single-cell RNA-Seq dataset. The effects of type I IFN (IFN-ß), IFN-γ, and TNF-α on gene expression, cytokine production, and cell death were investigated in human colonic organoids. In organoids treated with cytokines alone, or in combination with NLR family pyrin domain-containing 3 (NLRP3), caspase, or JAK inhibitors, cell death was measured, and supernatants were assayed for IL-1ß/IL-18/CXCL10. The expression of DNA sensor pathway genes-PYHIN family members [absent in melanoma 2 (AIM2), IFI16, myeloid cell nuclear differentiation antigen (MNDA), and pyrin and HIN domain family member 1 (PYHIN1)- as well as Z-DNA-binding protein 1 (ZBP1), cyclic GMP-AMP synthase (cGAS), and DDX41 was increased in active UC and expressed in a cell type-restricted pattern. Inflammasome genes (CASP1, IL1B, and IL18), type I IFN inducers [stimulator of interferon response cGAMP interactor 1 (STING), TBK1, and IRF3), IFNB1, and type I IFN biomarker genes (OAS2, IFIT2, and MX2) were also increased in active UC. Cotreatment of organoids with IFN-ß or IFN-γ in combination with TNFα increased expression of IFI16, ZBP1, CASP1, cGAS, and STING induced cell death and IL-1ß/IL-18 secretion. This inflammatory cell death was blocked by the JAK inhibitor tofacitinib but not by inflammasome or caspase inhibitors. Increased type I IFN activity may drive elevated expression of DNA sensor genes and JAK-dependent but inflammasome-independent inflammatory cell death of colonic epithelial cells in UC.NEW & NOTEWORTHY This study found that patients with active UC have significantly increased colonic gene expression of cytosolic DNA sensor, inflammasome, STING, and type I IFN signaling pathways. The type I IFN, IFN-ß, in combination with TNF-α induced JAK-dependent but NLRP3 and inflammasome-independent inflammatory cell death of colonic organoids. This novel inflammatory cell death phenotype is relevant to UC immunopathology and may partially explain the efficacy of the JAKinibs tofacitinib and upadacitinib in patients with UC.


Subject(s)
Colitis, Ulcerative , Interferon Type I , Janus Kinase Inhibitors , Humans , Inflammasomes/metabolism , Interleukin-18 , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Tumor Necrosis Factor-alpha , Caspase Inhibitors , Organoids/metabolism , Pyrin , Caspase 1/metabolism , Nucleotidyltransferases/metabolism , DNA , Cell Death , DNA-Binding Proteins/metabolism , Antigens, Differentiation
19.
Molecules ; 27(16)2022 Aug 15.
Article in English | MEDLINE | ID: mdl-36014431

ABSTRACT

Derivates of natural products have been wildly utilized in the treatment of malignant tumors. Isorhamnetin (ISO), a most important active ingredient derived from flavonoids, shows great potential in tumor therapy. However, the therapeutic effects of ISO on gastric cancer (GC) remain unclear. Here, we demonstrate that ISO treatment dramatically inhibited the proliferation of two types of GC cells (AGS-1 and HGC-27) both in vitro and in vivo in time- and dose-dependent manners. These results are consistent with the transcriptomic analysis of ISO-treated GC cells, which yielded hundreds of differentially expressed genes that were enriched with cell growth and apoptosis. Mechanically, ISO treatment initiated the activation of caspase-3 cascade and elevated the expression of mitochondria-associated Bax/Bcl-2, cytosolic cytochrome c, followed by the activation of the cleavage of caspase-3 as well as poly ADP-ribose polymerase (PARP), resulting in the severe reduction of the mitochondrial potential and the accumulation of reactive oxygen species (ROS), while pre-treatment of the caspase-3 inhibitor could block the anti-tumor effect. Therefore, these results indicate that ISO treatment induces the apoptosis of GC cells through the mitochondria-dependent apoptotic pathway, providing a potential strategy for clinical GC therapy.


Subject(s)
Stomach Neoplasms , Apoptosis , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Cell Proliferation , Humans , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Quercetin/analogs & derivatives , Reactive Oxygen Species/metabolism , Stomach Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...