Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Poult Sci ; 103(5): 103634, 2024 May.
Article in English | MEDLINE | ID: mdl-38537409

ABSTRACT

Moderate cold stimulation regulates the thymus's growth and function and facilitates cold acclimatization in broilers. However, the underlying mechanism remains unknown. To explore the possible mechanism of the thymus in cold-acclimated broilers against cold stress, 240 one-day-old Arbor Acres (AA) broilers were assigned to 2 groups randomly. The control group (C) was housed at conventional temperatures. The temperature during the first week was 33°C to 34°C. Between the ages of 8 and 32 d, the temperature was lowered by 1°C every 2 d, i.e., gradually from 32°C to 20°C, and then maintained at 20°C until 42 d of age. The cold-acclimated group (C-3) was housed at the same temperature as C from 1 to 7 d after birth. Between 8 and 42 d, the temperature of C-3 was 3°C colder than C. After 24 h exposure to acute cold stress (ACS) at 42 d, C and C-3 were named as S and S-3. The results showed that ACS was able to induce oxidation stress, modulate PI3K/AKT signal, and cause necroptosis and apoptosis in broiler thymus. By contrast, cold acclimation could alleviate apoptosis and necroptosis induced by cold stress via alleviating oxidative stress, efficiently activating the PI3K/AKT signal, as well as decreasing apoptotic and necrotic genes' levels. This study offers a novel theoretical basis for cold acclimation to improve the body's cold tolerance.


Subject(s)
Acclimatization , Apoptosis , Chickens , Cold Temperature , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Thymus Gland , Animals , Chickens/physiology , Thymus Gland/physiology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Necroptosis/physiology , Signal Transduction , Avian Proteins/metabolism , Avian Proteins/genetics , Random Allocation , Cold-Shock Response , Male
2.
Cancer Lett ; 585: 216693, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38301909

ABSTRACT

Necroptosis is a regulated necrotic cell death mechanism and plays a crucial role in the progression of cancers. However, the potential role and mechanism of necroptosis in colorectal cancer (CRC) has not been fully elucidated. In this study, we found that nuclear receptor subfamily 4 group A member 1 (NR4A1) was highly expressed in CRC cells treated with TNF-α, Smac mimetic, and z-VAD-FMK (TSZ). The depletion of NR4A1 significantly enhanced the sensitivity of CRC cells to TSZ-induced necroptosis, while NR4A1 overexpression suppressed these effects, as evidenced by the LDH assay, flow cytometry analysis of cell death, PI staining, and expression analysis of necrosome complexes (RIPK1, RIPK3, and MLKL). Moreover, NR4A1 deficiency made HT29 xenograft tumors sensitive to necroptotic cell death in vivo. Mechanistically, NR4A1 depletion promoted necroptosis activation in CRC through the RIG-I-like receptor pathway by interacting with DDX3. Importantly, the RIG-I pathway agonist poly(I:C) or inhibitor cFP abolished the effects of NR4A1 overexpression or suppression on necroptosis in CRC cells. Moreover, we observed that NR4A1 was highly expressed in CRC tissues and was associated with a poor prognosis. In conclusion, our results suggest that NR4A1 plays a critical role in modulating necroptosis in CRC cells and provide a new therapeutic target for CRC.


Subject(s)
Colorectal Neoplasms , Protein Kinases , Humans , Protein Kinases/metabolism , Necroptosis/physiology , Cell Death , Necrosis , Colorectal Neoplasms/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Apoptosis , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
3.
Cell Death Dis ; 15(1): 100, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38286985

ABSTRACT

Necroptosis, a type of lytic cell death executed by the pseudokinase Mixed Lineage Kinase Domain-Like (MLKL) has been implicated in the detrimental inflammation caused by SARS-CoV-2 infection. We minimally and extensively passaged a single clinical SARS-CoV-2 isolate to create models of mild and severe disease in mice allowing us to dissect the role of necroptosis in SARS-CoV-2 disease pathogenesis. We infected wild-type and MLKL-deficient mice and found no significant differences in viral loads or lung pathology. In our model of severe COVID-19, MLKL-deficiency did not alter the host response, ameliorate weight loss, diminish systemic pro-inflammatory cytokines levels, or prevent lethality in aged animals. Our in vivo models indicate that necroptosis is dispensable in the pathogenesis of mild and severe COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Mice , SARS-CoV-2/metabolism , Necroptosis/physiology , Protein Kinases/metabolism , Disease Models, Animal , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
4.
Immunol Rev ; 321(1): 52-70, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37897080

ABSTRACT

Necroptosis is generally considered as an inflammatory cell death form. The core regulators of necroptotic signaling are receptor-interacting serine-threonine protein kinases 1 (RIPK1) and RIPK3, and the executioner, mixed lineage kinase domain-like pseudokinase (MLKL). Evidence demonstrates that necroptosis contributes profoundly to inflammatory respiratory diseases that are common public health problem. Necroptosis occurs in nearly all pulmonary cell types in the settings of inflammatory respiratory diseases. The influence of necroptosis on cells varies depending upon the type of cells, tissues, organs, etc., which is an important factor to consider. Thus, in this review, we briefly summarize the current state of knowledge regarding the biology of necroptosis, and focus on the key molecular mechanisms that define the necroptosis status of specific cell types in inflammatory respiratory diseases. We also discuss the clinical potential of small molecular inhibitors of necroptosis in treating inflammatory respiratory diseases, and describe the pathological processes that engage cross talk between necroptosis and other cell death pathways in the context of respiratory inflammation. The rapid advancement of single-cell technologies will help understand the key mechanisms underlying cell type-specific necroptosis that are critical to effectively treat pathogenic lung infections and inflammatory respiratory diseases.


Subject(s)
Protein Kinases , Respiratory Tract Diseases , Humans , Protein Kinases/metabolism , Necroptosis/physiology , Cell Death , Signal Transduction , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Apoptosis
5.
Bioorg Chem ; 142: 106964, 2024 01.
Article in English | MEDLINE | ID: mdl-37976678

ABSTRACT

Necroptosis is one of the modes of cell death, and its occurrence and development are associated with the development of numerous diseases. To prevent the progression of necroptosis, it is crucial to inhibit the phosphorylation of three proteins: receptor-interacting protein kinase 1 (RIP1), RIP3, and mixed lineage kinase domain-like protein (MLKL). Through virtual and experimental screening approaches, we have identified 8 small molecular inhibitors with potent antinecroptotic activity and binding affinity to RIP1. Among these compounds, SY-1 demonstrated the most remarkable antinecroptotic activity (EC50 = 105.6 ± 9.6 nM) and binding affinity (RIP1 Kd = 49 nM). It effectively blocked necroptosis and impeded the formation of necrosomes by inhibiting the phosphorylations of the RIP1/RIP3/MLKL pathway triggered by TSZ (TNFα, Smac mimetic and Z-VAD-fmk). Furthermore, SY-1 exhibited a protective effect against tumor necrosis factor (TNF)-induced hypothermia in mice and significantly improved the survival rate (100 %, 30 mg/kg) of mice with systemic inflammatory response syndrome (SIRS) in a dose-dependent manner. Pharmacokinetic parameters of SY-1 were also collected in vitro and in vivo. These results strongly suggest that SY-1 and its derivatives warrant further investigation for their potential therapeutic applications.


Subject(s)
Necroptosis , Protein Kinases , Animals , Mice , Protein Kinases/metabolism , Necroptosis/physiology , Cell Death , Phosphorylation , Transcription Factors/metabolism , Apoptosis
6.
Int J Mol Sci ; 24(19)2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37834141

ABSTRACT

The mechanism of long-term cognitive impairment after neonatal sepsis remains poorly understood, although long-lasting neuroinflammation has been considered the primary contributor. Necroptosis is actively involved in the inflammatory process, and in this study, we aimed to determine whether neonatal sepsis-induced long-term cognitive impairment was associated with activation of necroptosis. Rat pups on postnatal day 3 (P3) received intraperitoneal injections of lipopolysaccharide (LPS, 1 mg/kg) to induce neonatal sepsis. Intracerebroventricular injection of IL-1ß-siRNA and necrostatin-1 (NEC1) were performed to block the production of IL-1ß and activation of necroptosis in the brain, respectively. The Morris water maze task and fear conditioning test were performed on P28-P32 and P34-P35, respectively. Enzyme-linked immunosorbent assay (ELISA), quantitative real-time PCR (RT-PCR), and Western blotting were used to examine the expression levels of proinflammatory cytokines and necroptosis-associated proteins, such as receptor-interacting protein 1 (RIP1) and receptor-interacting protein 3 (RIP3). Sustained elevation of IL-1ß level was observed in the brain after initial neonatal sepsis, which would last for at least 32 days. Sustained necroptosis activation was also observed in the brain. Knockdown of IL-1ß expression in the brain alleviated necroptosis and improved long-term cognitive function. Direct inhibition of necroptosis also improved neurodevelopment and cognitive performance. This research indicated that sustained activation of necroptosis via IL-1ß contributed to long-term cognitive dysfunction after neonatal sepsis.


Subject(s)
Neonatal Sepsis , Sepsis , Rats , Animals , Necroptosis/physiology , Brain/metabolism , Sepsis/complications , Sepsis/metabolism
7.
Int J Mol Sci ; 24(20)2023 Oct 18.
Article in English | MEDLINE | ID: mdl-37894970

ABSTRACT

Apoptosis has historically been considered the primary form of programmed cell death (PCD) and is responsible for regulating cellular processes during development, homeostasis, and disease. Conversely, necrosis was considered uncontrolled and unregulated. However, recent evidence has unveiled the significance of necroptosis, a regulated form of necrosis, as an important mechanism of PCD alongside apoptosis. The activation of necroptosis leads to cellular membrane disruption, inflammation, and vascularization. This process is crucial in various pathological conditions, including intervertebral disc degeneration (IVDD), neurodegeneration, inflammatory diseases, multiple cancers, and kidney injury. In recent years, extensive research efforts have shed light on the molecular regulation of the necroptotic pathway. Various stimuli trigger necroptosis, and its regulation involves the activation of specific proteins such as receptor-interacting protein kinase 1 (RIPK1), RIPK3, and the mixed lineage kinase domain-like (MLKL) pseudokinase. Understanding the intricate mechanisms governing necroptosis holds great promise for developing novel therapeutic interventions targeting necroptosis-associated IVDD. The objective of this review is to contribute to the growing body of scientific knowledge in this area by providing a comprehensive overview of necroptosis and its association with IVDD. Ultimately, these understandings will allow the development of innovative drugs that can modulate the necroptotic pathway, offering new therapeutic avenues for individuals suffering from necroptosis.


Subject(s)
Intervertebral Disc Degeneration , Protein Kinases , Humans , Protein Kinases/metabolism , Necroptosis/physiology , Apoptosis , Necrosis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
8.
Cell Death Dis ; 14(10): 673, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828052

ABSTRACT

AF6, a known polarity protein, contributes to the maintenance of homeostasis while ensuring tissue architecture, repair, and integrity. Mice that lack AF6 display embryonic lethality owing to cell-cell junction disruption. However, we show AF6 promotes necroptosis via regulating the ubiquitination of RIPK1 by directly interact with the intermediate domain of RIPK1, which was mediated by the deubiquitylase enzyme USP21. Consistently, while injection of mice with an adenovirus providing AF6 overexpression resulted in accelerated TNFα-induced necroptosis-mediated mortality in vivo, we observed that mice with hepatocyte-specific deletion of AF6 prevented hepatocytes from necroptosis and the subsequent inflammatory response in various liver diseases model, including non-alcoholic steatohepatitis (NASH) and the systemic inflammatory response syndrome (SIRS).Together, these data suggest that AF6 represents a novel regulator of RIPK1-RIPK3 dependent necroptotic pathway. Thus, the AF6-RIPK1-USP21 axis are potential therapeutic targets for treatment of various liver injuries and metabolic diseases.


Subject(s)
Liver Diseases , Necroptosis , Animals , Mice , Apoptosis/physiology , Hepatocytes/metabolism , Liver Diseases/genetics , Necroptosis/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Systemic Inflammatory Response Syndrome , Ubiquitination
9.
J Stomatol Oral Maxillofac Surg ; 124(6S): 101565, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37459966

ABSTRACT

Necroptosis is a type of caspase independent 'programmed or regulated' necrotic cell death that has a morphological resemblance to necrosis and mechanistic analogy to apoptosis. This type of cell death requires RIPK1, RIPK3, MLKL, death receptors, toll like receptors, interferons, and various other proteins. Necroptosis is implicated in plethora of diseases like rheumatoid arthritis, Alzheimer's disease, Crohn's disease, and head and neck cancers including oral squamous cell carcinoma. Oral carcinomas show dysregulation or mutation of necroptotic proteins, mediate antitumoral immunity, activate immune response and control tumor progression. Necroptosis is known to play a dual role (pro tumorigenic and anti-tumorigenic) in cancer progression and targeting this pathway could be an effective approach in cancer therapy. Necroptosis based chemotherapy has been proposed in malignancies, highlighting the importance of necroptotic pathway to overcome apoptosis resistance and serve as a "fail-safe" pathway to modulate cancer initiation, progression, and metastasis. However, there is dearth of information regarding the use of necroptotic cell death mechanism in the treatment of oral squamous cell carcinoma. In this review, we summarise molecular mechanism of necroptosis, and its protumorigenic and antitumorigenic role in cancers to shed light on the possible therapeutic significance of necroptosis in oral squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/therapy , Squamous Cell Carcinoma of Head and Neck , Necroptosis/physiology , Protein Kinases/genetics , Protein Kinases/metabolism , Mouth Neoplasms/therapy , Necrosis
10.
J Periodontal Res ; 58(5): 919-931, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37334934

ABSTRACT

OBJECTIVE: To explore the mechanism of receptor-interacting protein 1 (RIP1)-mediated necroptosis during periodontitis progression. BACKGROUND: RIP3 and mixed lineage kinase domain-like protein (MLKL) have been detected to be upregulated in periodontitis models. Because RIP1 is involved in necroptosis, it might also play a role in the progression of periodontitis. METHODS: An experimental periodontitis model in BALB/c mice was established by inducing oral bacterial infection. Western blotting and immunofluorescence analyses were used to detect RIP1 expression in the periodontal ligament. Porphyromonas gingivalis was used to stimulate L929 and MC3T3-E1. RIP1 was inhibited using small-interfering RNA. Western blotting, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and enzyme-linked immunosorbent assay (ELISA) analyses were used to detect the effect of necroptosis inhibition on the expression of damage-associated molecular patterns and inflammatory cytokines. Necrostatin-1 (Nec-1) was intraperitoneally injected to inhibit RIP1 expression in mice. Necroptosis activation and inflammatory cytokine expression in periodontal tissue were verified. Tartrate-resistant acid phosphatase staining was applied to observe osteoclasts in the bone tissues of different groups. RESULTS: RIP1-mediated necroptosis was activated in mice with periodontitis. P. gingivalis induced RIP1-mediated necroptosis in L929 and MC3T3-E1 cells. After RIP1 inhibition, the expression levels of high mobility group protein B1 (HMGB1) and inflammatory cytokines were downregulated. After inhibiting RIP1 with Nec-1 in vivo, necroptosis was also inhibited, the expression levels of HMGB1 and inflammatory cytokines were downregulated, and osteoclast counts in the periodontal tissue decreased. CONCLUSION: RIP1-mediated necroptosis plays a role in the pathological process of periodontitis in mice. Nec-1 inhibited necroptosis, alleviated inflammation in periodontal tissue, and reduced bone resorption in periodontitis.


Subject(s)
HMGB1 Protein , Periodontitis , Mice , Animals , HMGB1 Protein/pharmacology , Necroptosis/physiology , Periodontitis/metabolism , Cytokines , Apoptosis
11.
Cell Death Dis ; 14(3): 227, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36991017

ABSTRACT

Diabetic retinopathy (DR) is a leading cause of blindness that poses significant public health concerns worldwide. Increasing evidence suggests that neuroinflammation plays a key role in the early stages of DR. Microglia, long-lived immune cells in the central nervous system, can become activated in response to pathological insults and contribute to retinal neuroinflammation. However, the molecular mechanisms of microglial activation during the early stages of DR are not fully understood. In this study, we used in vivo and in vitro assays to investigate the role of microglial activation in the early pathogenesis of DR. We found that activated microglia triggered an inflammatory cascade through a process called necroptosis, a newly discovered pathway of regulated cell death. In the diabetic retina, key components of the necroptotic machinery, including RIP1, RIP3, and MLKL, were highly expressed and mainly localized in activated microglia. Knockdown of RIP3 in DR mice reduced microglial necroptosis and decreased pro-inflammatory cytokines. Additionally, blocking necroptosis with the specific inhibitor GSK-872 improved retinal neuroinflammation and neurodegeneration, as well as visual function in diabetic mice. RIP3-mediated necroptosis was activated and contributed to inflammation in BV2 microglia under hyperglycaemic conditions. Our data demonstrate the importance of microglial necroptosis in retinal neuroinflammation related to diabetes and suggest that targeting necroptosis in microglia may be a promising therapeutic strategy for the early stages of DR.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Animals , Mice , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Microglia/metabolism , Necroptosis/physiology , Neuroinflammatory Diseases , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
12.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(2): 242-251, 2023 Feb 28.
Article in English, Chinese | MEDLINE | ID: mdl-36999471

ABSTRACT

Necroptosis is one of the regulated cell death, which involves receptor interacting protein kinase (RIPK) 1/RIPK3/mixed lineage kinase domain like protein (MLKL) signaling pathway. Among them, MLKL is the final execution of necroptosis. The formation of RIPK1/RIPK3/MLKL necrosome induces the phosphorylated MLKL, and the activated MLKL penetrates into the membrane bilayer to form membrane pores, which damages the integrity of the membrane and leads to cell death. In addition to participating in necroptosis, MLKL is also closely related to other cell death, such as NETosis, pyroptosis, and autophagy. Therefore, MLKL is involved in the pathological processes of various diseases related to abnormal cell death pathways (such as cardiovascular diseases, neurodegenerative diseases and cancer), and may be a therapeutic target of multiple diseases. Understanding the role of MLKL in different cell death can lay a foundation for seeking various MLKL-related disease targets, and also guide the development and application of MLKL inhibitors.


Subject(s)
Necroptosis , Protein Kinases , Apoptosis , Necroptosis/physiology , Protein Kinases/metabolism , Pyroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction , Humans
13.
Ageing Res Rev ; 86: 101855, 2023 04.
Article in English | MEDLINE | ID: mdl-36681250

ABSTRACT

Necroptosis, a regulated form of cell death, is involved in the genesis and development of various life-threatening diseases, including cancer, neurological disorders, cardiac myopathy, and diabetes. Necroptosis initiates with the formation and activation of a necrosome complex, which consists of RIPK1, RIPK2, RIPK3, and MLKL. Emerging studies has demonstrated the regulation of the necroptosis cell death pathway through the implication of numerous post-translational modifications, namely ubiquitination, acetylation, methylation, SUMOylation, hydroxylation, and others. In addition, the negative regulation of the necroptosis pathway has been shown to interfere with brain homeostasis through the regulation of axonal degeneration, mitochondrial dynamics, lysosomal defects, and inflammatory response. Necroptosis is controlled by the activity and expression of signaling molecules, namely VEGF/VEGFR, PI3K/Akt/GSK-3ß, c-Jun N-terminal kinases (JNK), ERK/MAPK, and Wnt/ß-catenin. Herein, we briefly discussed the implication and potential of necrosome activation in the pathogenesis and progression of neurological manifestations, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, traumatic brain injury, and others. Further, we present a detailed picture of natural compounds, micro-RNAs, and chemical compounds as therapeutic agents for treating neurological manifestations.


Subject(s)
Nervous System Diseases , Protein Kinases , Humans , Protein Kinases/metabolism , Necroptosis/physiology , Glycogen Synthase Kinase 3 beta , Phosphatidylinositol 3-Kinases , Apoptosis
14.
Life Sci ; 314: 121338, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36592788

ABSTRACT

BACKGROUND AND AIM: Many attempts to control acute kidney injury (AKI) have failed due to a lack of understanding of its pathophysiological key components. Macrophages are a crucial determinant of AKI, which can be categorized functionally as M1 pro-inflammatory and M2 anti-inflammatory macrophages. Low-intensity pulsed ultrasound (LIPUS) is currently being investigated as an immune modulator. The present study aimed to explore the potential effects of LIPUS on the polarization of renal macrophages, as well as the possible interplay between macrophage polarization and necroptosis in gentamicin-induced acute kidney injury. METHOD: All rats were randomly allocated into one of four groups: control, LIPUS-treated control, gentamicin acute kidney (GM-AKI), and LIPUS-treated GM-AKI. Renal functions, macrophage polarization, necroptosis, and heat shock protein-70 (HSP70) were analyzed using real-time reverse-transcriptase-polymerase chain reaction (rT-PCR), Western Blot, Enzyme-linked immunosorbent assay (ELISA) as well as immunohistological analysis. RESULTS: we found that LIPUS markedly inhibited the expressions of M1 macrophage-related genes and promoted significantly the expression of M2 macrophages related genes. This was accompanied by an inhibition of necroptosis and a marked reduction of HSP-70, resulting in a reversal of gentamicin-induced renal alteration. CONCLUSION: Functional switching of macrophage responses from M1 into M2 seems to be a potential approach to ameliorate necroptosis as well as HSP-70 by low pulsed ultrasound waves in GM-AKI.


Subject(s)
Acute Kidney Injury , Necroptosis , Ultrasonic Waves , Animals , Rats , Acute Kidney Injury/chemically induced , Acute Kidney Injury/therapy , Acute Kidney Injury/metabolism , Macrophages/metabolism , Necroptosis/physiology , Phenotype , HSP70 Heat-Shock Proteins/metabolism
15.
Neurotoxicology ; 94: 71-86, 2023 01.
Article in English | MEDLINE | ID: mdl-36347329

ABSTRACT

Neuronal cell death as a prominent pathological feature contributes to cognitive decline and memory loss in Alzheimer's disease. We investigated the role of two forms of cell death pathways, ferroptosis and necroptosis, and their interactions following entorhinal cortex (EC) amyloidopathy. The Aß25-35 was bilaterally injected into the rat's EC, and Morris Water Maze was applied to determine spatial performance one week after Aß injection. For evaluation of ferroptosis and necroptosis involvement in Aß induced pathology, ferroptosis inhibitor, Ferrostatin (Fer-1), and necroptosis inhibitor, Necrostatin (Nec-1), were injected into the EC during training days of behavioral test. Our behavioral and histological assessment showed spatial learning and memory impairment, along with neuropathology changes such as cell survival and intracellular Aß deposits in response to EC amyloidopathy, which were ameliorated by treatment with Fer-1 or Nec-1. The expression of ferroptosis key factors GPX4 and SLC7A11 were decreased and the level of TfR was increased following Aß toxicity. Also, Necroptosis pathway related factors RIP1, RIP3, and MLKL were modulated by Aß neurotoxicity. However, application of Fer-1 or Nec-1 could inhibit the hippocampal ferroptosis and necroptosis pathways due to EC amyloidopathy. Our data also demonstrated that Aß-induced necroptosis suppressed by Fer-1, although Nec-1 had no effect on ferroptosis, indicating that ferroptosis pathway is upstream of necroptosis process in the Aß neurotoxicity. Moreover, Aß induced hippocampal mGLUR5 overexpression and reduced level of STIM1/2 recovered by Fer-1 or Nec-1. According to our findings ferroptosis and necroptosis pathways are involved in Aß neurotoxicity through modulation of mGLUR5 and STIM1/2 signaling.


Subject(s)
Alzheimer Disease , Ferroptosis , Rats , Animals , Amyloid beta-Peptides/toxicity , Necroptosis/physiology , Cell Death
16.
Article in English | WPRIM (Western Pacific) | ID: wpr-971391

ABSTRACT

Necroptosis is one of the regulated cell death, which involves receptor interacting protein kinase (RIPK) 1/RIPK3/mixed lineage kinase domain like protein (MLKL) signaling pathway. Among them, MLKL is the final execution of necroptosis. The formation of RIPK1/RIPK3/MLKL necrosome induces the phosphorylated MLKL, and the activated MLKL penetrates into the membrane bilayer to form membrane pores, which damages the integrity of the membrane and leads to cell death. In addition to participating in necroptosis, MLKL is also closely related to other cell death, such as NETosis, pyroptosis, and autophagy. Therefore, MLKL is involved in the pathological processes of various diseases related to abnormal cell death pathways (such as cardiovascular diseases, neurodegenerative diseases and cancer), and may be a therapeutic target of multiple diseases. Understanding the role of MLKL in different cell death can lay a foundation for seeking various MLKL-related disease targets, and also guide the development and application of MLKL inhibitors.


Subject(s)
Protein Kinases/metabolism , Necroptosis/physiology , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction , Pyroptosis , Apoptosis
17.
Front Immunol ; 14: 1297408, 2023.
Article in English | MEDLINE | ID: mdl-38164133

ABSTRACT

The mammalian target of rapamycin (mTOR), an evolutionarily highly conserved serine/threonine protein kinase, plays a prominent role in controlling gene expression, metabolism, and cell death. Programmed cell death (PCD) is indispensable for maintaining homeostasis by removing senescent, defective, or malignant cells. Necroptosis, a type of PCD, relies on the interplay between receptor-interacting serine-threonine kinases (RIPKs) and the membrane perforation by mixed lineage kinase domain-like protein (MLKL), which is distinguished from apoptosis. With the development of necroptosis-regulating mechanisms, the importance of mTOR in the complex network of intersecting signaling pathways that govern the process has become more evident. mTOR is directly responsible for the regulation of RIPKs. Autophagy is an indirect mechanism by which mTOR regulates the removal and interaction of RIPKs. Another necroptosis trigger is reactive oxygen species (ROS) produced by oxidative stress; mTOR regulates necroptosis by exploiting ROS. Considering the intricacy of the signal network, it is reasonable to assume that mTOR exerts a bifacial effect on necroptosis. However, additional research is necessary to elucidate the underlying mechanisms. In this review, we summarized the mechanisms underlying mTOR activation and necroptosis and highlighted the signaling pathway through which mTOR regulates necroptosis. The development of therapeutic targets for various diseases has been greatly advanced by the expanding knowledge of how mTOR regulates necroptosis.


Subject(s)
Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Reactive Oxygen Species/metabolism , Necroptosis/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Apoptosis/physiology , TOR Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Mammals/metabolism
18.
Front Immunol ; 13: 968639, 2022.
Article in English | MEDLINE | ID: mdl-36059491

ABSTRACT

Acinar cell death and inflammatory response are two important events which determine the severity of acute pancreatitis (AP). Endoplasmic reticulum (ER) stress and necroptosis are involved in this process, but the relationships between them remain unknown. Here, we analyzed the interaction between ER stress and necroptosis and the underlying mechanisms during AP. Experimental pancreatitis was induced in Balb/C mice by caerulein (Cae) and lipopolysaccharide (LPS) or L-arginine (L-Arg) in vivo, and pancreatic acinar cells were also used to follow cellular mechanisms during cholecystokinin (CCK) stimulation in vitro. AP severity was assessed by serum amylase, lipase levels and histological examination. Changes in ER stress, trypsinogen activation and necroptosis levels were analyzed by western blotting, enzyme-linked immunosorbent assay (ELISA), adenosine triphosphate (ATP) analysis or lactate dehydrogenase (LDH) assay. The protein kinase C (PKC)α -mitogen-activated protein kinase (MAPK) -cJun pathway and cathepsin B (CTSB) activation were evaluated by western blotting. Activating protein 1 (AP-1) binding activity was detected by electrophoretic mobility shift assay (EMSA). We found that ER stress is initiated before necroptosis in CCK-stimulated acinar cells in vitro. Inhibition of ER stress by 4-phenylbutyrate (4-PBA) can significantly alleviate AP severity both in two AP models in vivo. 4-PBA markedly inhibited ER stress and necroptosis of pancreatic acinar cells both in vitro and in vivo. Mechanistically, we found that 4-PBA significantly reduced CTSB maturation and PKCα-JNK-cJun pathway -mediated AP-1 activation during AP. Besides, CTSB inhibitor CA074Me markedly blocked PKCα-JNK-cJun pathway -mediated AP-1 activation and necroptosis in AP. However, pharmacologic inhibition of trypsin activity with benzamidine hydrochloride had no effect on PKCα-JNK-cJun pathway and necroptosis in CCK-stimulated pancreatic acinar cells. Furthermore, SR11302, the inhibitor of AP-1, significantly lowered tumor necrosis factor (TNF) α levels, and its subsequent receptor interacting protein kinases (RIP)3 and phosphorylated mixed lineagekinase domain-like (pMLKL) levels, ATP depletion and LDH release rate in CCK-stimulated pancreatic acinar cells. To sum up, all the results indicated that during AP, ER stress promoted pancreatic acinar cell necroptosis through CTSB maturation, thus induced AP-1 activation and TNFα secretion via PKCα-JNK-cJun pathway, not related with trypsin activity. These findings provided potential therapeutic target and treatment strategies for AP or other cell death-related diseases.


Subject(s)
Acinar Cells , Cathepsin B , Endoplasmic Reticulum Stress , Necroptosis , Pancreatitis , Transcription Factor AP-1 , Acinar Cells/metabolism , Acinar Cells/pathology , Acute Disease , Adenosine Triphosphate/metabolism , Animals , Cathepsin B/genetics , Cathepsin B/metabolism , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/physiology , Mice , Mice, Inbred BALB C , Necroptosis/genetics , Necroptosis/physiology , Pancreatitis/genetics , Pancreatitis/metabolism , Pancreatitis/pathology , Protein Kinase C-alpha/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Trypsin/metabolism
19.
Biochem J ; 479(5): 609-628, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35244141

ABSTRACT

Two years after the emergence of SARS-CoV-2, our understanding of COVID-19 disease pathogenesis is still incomplete. Despite unprecedented global collaborative scientific efforts and rapid vaccine development, an uneven vaccine roll-out and the emergence of novel variants of concern such as omicron underscore the critical importance of identifying the mechanisms that contribute to this disease. Overt inflammation and cell death have been proposed to be central drivers of severe pathology in COVID-19 patients and their pathways and molecular components therefore present promising targets for host-directed therapeutics. In our review, we summarize the current knowledge on the role and impact of diverse programmed cell death (PCD) pathways on COVID-19 disease. We dissect the complex connection of cell death and inflammatory signaling at the cellular and molecular level and identify a number of critical questions that remain to be addressed. We provide rationale for targeting of cell death as potential COVID-19 treatment and provide an overview of current therapeutics that could potentially enter clinical trials in the near future.


Subject(s)
COVID-19/etiology , COVID-19/pathology , Antiviral Agents , Apoptosis/drug effects , Apoptosis/physiology , Humans , Inflammasomes/physiology , Interferons/metabolism , Necroptosis/physiology , Neutrophils/pathology , Neutrophils/virology , Pyroptosis/physiology , SARS-CoV-2/pathogenicity , COVID-19 Drug Treatment
20.
Cell Prolif ; 55(3): e13193, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35083817

ABSTRACT

Chronic alcohol consumption can cause alcoholic liver disease (ALD), leading to morbidity and mortality worldwide. Complex disease progression of ALD varies from alcoholic fatty liver to alcoholic steatohepatitis, eventually contributing to fibrosis and cirrhosis. Accumulating evidence revealed that necroptosis, a way of programmed cell death different from apoptosis and traditional necrosis, is involved in the underlying pathogenic molecular mechanism of ALD. Receptor-interacting protein kinase 1 (RIPK1), RIPK3 and mixed-lineage kinase domain-like pseudokinase have been implicated as key mediators to execute necroptosis. Also, necroptosis has gained increasing attention due to its potential association with primary pathological hallmarks of ALD, including oxidative stress, hepatic steatosis and inflammation. This review summarizes the recent progress on the roles and mechanisms of necroptosis and focuses on the crosstalk between necroptosis and the other pathogenesis of ALD, providing a theoretical basis for targeting necroptosis as a novel treatment for ALD.


Subject(s)
Hepatocytes/pathology , Liver Diseases, Alcoholic/pathology , Liver/pathology , Necroptosis/physiology , Animals , Hepatocytes/metabolism , Humans , Liver Diseases, Alcoholic/metabolism , Necrosis/metabolism , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...