Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53.930
Filtrar
1.
Nat Commun ; 15(1): 6501, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39090081

RESUMEN

The role of developmental cell death in the formation of brain circuits is not well understood. Cajal-Retzius cells constitute a major transient neuronal population in the mammalian neocortex, which largely disappears at the time of postnatal somatosensory maturation. In this study, we used mouse genetics, anatomical, functional, and behavioral approaches to explore the impact of the early postnatal death of Cajal-Retzius cells in the maturation of the cortical circuit. We find that before their death, Cajal-Retzius cells mainly receive inputs from layer 1 neurons, which can only develop their mature connectivity onto layer 2/3 pyramidal cells after Cajal-Retzius cells disappear. This developmental connectivity progression from layer 1 GABAergic to layer 2/3 pyramidal cells regulates sensory-driven inhibition within, and more so, across cortical columns. Here we show that Cajal-Retzius cell death prevention leads to layer 2/3 hyper-excitability, delayed learning and reduced performance in a multi-whisker-dependent texture discrimination task.


Asunto(s)
Muerte Celular , Células Piramidales , Corteza Somatosensorial , Animales , Corteza Somatosensorial/fisiología , Corteza Somatosensorial/citología , Ratones , Células Piramidales/fisiología , Células Piramidales/metabolismo , Neocórtex/citología , Neocórtex/fisiología , Neuronas GABAérgicas/fisiología , Neuronas GABAérgicas/metabolismo , Masculino , Vibrisas/fisiología , Femenino , Ratones Endogámicos C57BL , Inhibición Neural/fisiología , Neuronas/fisiología , Neuronas/metabolismo
2.
Cell Death Dis ; 15(8): 556, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39090114

RESUMEN

Reactive oxygen species (ROS) are highly reactive oxygen-containing molecules generated as natural byproducts during cellular processes, including metabolism. Under normal conditions, ROS play crucial roles in diverse cellular functions, including cell signaling and immune responses. However, a disturbance in the balance between ROS production and cellular antioxidant defenses can lead to an excessive ROS buildup, causing oxidative stress. This stress damages essential cellular components, including lipids, proteins, and DNA, potentially culminating in oxidative cell death. This form of cell death can take various forms, such as ferroptosis, apoptosis, necroptosis, pyroptosis, paraptosis, parthanatos, and oxeiptosis, each displaying distinct genetic, biochemical, and signaling characteristics. The investigation of oxidative cell death holds promise for the development of pharmacological agents that are used to prevent tumorigenesis or treat established cancer. Specifically, targeting key antioxidant proteins, such as SLC7A11, GCLC, GPX4, TXN, and TXNRD, represents an emerging approach for inducing oxidative cell death in cancer cells. This review provides a comprehensive summary of recent progress, opportunities, and challenges in targeting oxidative cell death for cancer therapy.


Asunto(s)
Muerte Celular , Neoplasias , Estrés Oxidativo , Especies Reactivas de Oxígeno , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Animales , Oxidación-Reducción , Antioxidantes/metabolismo , Antioxidantes/farmacología , Transducción de Señal , Apoptosis , Ferroptosis/efectos de los fármacos
3.
Commun Biol ; 7(1): 982, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39134806

RESUMEN

Transglutaminase 2 (TG2) is a GTP-binding, protein-crosslinking enzyme that has been investigated as a therapeutic target for Celiac disease, neurological disorders, and aggressive cancers. TG2 has been suggested to adopt two conformational states that regulate its functions: a GTP-bound, closed conformation, and a calcium-bound, crosslinking-active open conformation. TG2 mutants that constitutively adopt an open conformation are cytotoxic to cancer cells. Thus, small molecules that bind and stabilize the open conformation of TG2 could offer a new therapeutic strategy. Here, we investigate TG2, using static and time-resolved small-angle X-ray scattering (SAXS) and single-particle cryoelectron microscopy (cryo-EM), to determine the conformational states responsible for conferring its biological effects. We also describe a newly developed TG2 inhibitor, LM11, that potently kills glioblastoma cells and use SAXS to investigate how LM11 affects the conformational states of TG2. Using SAXS and cryo-EM, we show that guanine nucleotides bind and stabilize a monomeric closed conformation while calcium binds to an open state that can form higher order oligomers. SAXS analysis suggests how a TG2 mutant that constitutively adopts the open state binds nucleotides through an alternative mechanism to wildtype TG2. Furthermore, we use time resolved SAXS to show that LM11 increases the ability of calcium to bind and stabilize an open conformation, which is not reversible by guanine nucleotides and is cytotoxic to cancer cells. Taken together, our findings demonstrate that the conformational dynamics of TG2 are more complex than previously suggested and highlight how conformational stabilization of TG2 by LM11 maintains TG2 in a cytotoxic conformational state.


Asunto(s)
Supervivencia Celular , Proteínas de Unión al GTP , Conformación Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas , Proteína Glutamina Gamma Glutamiltransferasa 2/metabolismo , Humanos , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Transglutaminasas/metabolismo , Transglutaminasas/química , Transglutaminasas/genética , Supervivencia Celular/efectos de los fármacos , Microscopía por Crioelectrón , Línea Celular Tumoral , Muerte Celular/efectos de los fármacos , Dispersión del Ángulo Pequeño , Difracción de Rayos X , Calcio/metabolismo
4.
J Biochem Mol Toxicol ; 38(8): e23808, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39132830

RESUMEN

Oxidative stress is a hallmark of secondary injury of spinal cord injuries. Controlling oxidative stress is crucial for mitigating secondary injury and promoting functional recovery after spinal cord injuries. Calycosin is an O-methylated isoflavone with antioxidant activity. To evaluate the effect of calycosin on spinal cord neurons under oxidative stress and clarify the molecular mechanism underlying the effect, we tested the neuroprotective activity of calycosin in a primary spinal cord neuron culture model. We found that calycosin protected neurons from H2O2-induced neuronal death in a dose-dependent manner. Further experiments revealed that calycosin decreased H2O2-induced mitochondrial fragmentation and mitochondrial membrane potential loss, and subsequently reduced H2O2-triggered release of mitochondrial cytochrome c into the cytoplasm. In addition, calycosin inhibited H2O2-induced reactive oxygen species generation and activation of NF-κB signaling in spinal cord neurons. Furthermore, the expression of several antioxidant enzymes such as HO-1, NQO1, GCLC, GCLM, TrxR1, and Trx1 was significantly promoted by calycosin. More importantly, we revealed that the Nrf2/Keap1 signal is crucial for the effect of calycosin, because calycosin increased the amount of nuclear Nrf2 while decreasing the amount of cytoplasmic Nrf2. Nrf2 knockdown with siRNA transfection abolished the neuroprotective effect of calycosin. Taken together, this study disclosed a novel mechanism by which calycosin combats oxidative stress. Our study thus sheds light on the potential clinical application of calycosin in SCI treatment.


Asunto(s)
Peróxido de Hidrógeno , Isoflavonas , Proteína 1 Asociada A ECH Tipo Kelch , Mitocondrias , Factor 2 Relacionado con NF-E2 , Neuronas , Transducción de Señal , Médula Espinal , Isoflavonas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Animales , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/toxicidad , Médula Espinal/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Transducción de Señal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Ratas , Fármacos Neuroprotectores/farmacología
5.
PLoS One ; 19(8): e0308635, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39110748

RESUMEN

To date, apart from moderate hypothermia, there are almost no adequate interventions available for neuroprotection in cases of brain damage due to cardiac arrest. Affected persons often have severe limitations in their quality of life. The aim of this study was to investigate protective properties of the active compound of dimethyl fumarate, monomethyl fumarate (MMF), on distinct regions of the central nervous system after ischemic events. Dimethyl fumarate is an already established drug in neurology with known anti-inflammatory and antioxidant properties. In this study, we chose organotypic slice cultures of rat cerebellum and hippocampus as an ex vivo model. To simulate cardiac arrest and return of spontaneous circulation we performed oxygen-glucose-deprivation (OGD) followed by treatments with different concentrations of MMF (1-30 µM in cerebellum and 5-30 µM in hippocampus). Immunofluorescence staining with propidium iodide (PI) and 4',6-diamidine-2-phenylindole (DAPI) was performed to analyze PI/DAPI ratio after imaging with a spinning disc confocal microscope. In the statistical analysis, the relative cell death of the different groups was compared. In both, the cerebellum and hippocampus, the MMF-treated group showed a significantly lower PI/DAPI ratio compared to the non-treated group after OGD. Thus, we showed for the first time that both cerebellar and hippocampal slice cultures treated with MMF after OGD are significantly less affected by cell death.


Asunto(s)
Cerebelo , Fumaratos , Glucosa , Hipocampo , Fármacos Neuroprotectores , Animales , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/patología , Glucosa/metabolismo , Ratas , Fármacos Neuroprotectores/farmacología , Fumaratos/farmacología , Técnicas de Cultivo de Órganos , Oxígeno/metabolismo , Ratas Wistar , Muerte Celular/efectos de los fármacos
6.
Int J Med Sci ; 21(10): 1840-1851, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113898

RESUMEN

Pulmonary arterial hypertension (PAH) is a severe pulmonary vascular disease characterized by increased pulmonary vascular resistance because of vascular remodeling and vasoconstriction. Subsequently, PAH leads to right ventricular hypertrophy and heart failure. Cell death mechanisms play a significant role in development and tissue homeostasis, and regulate the balance between cell proliferation and differentiation. Several basic and clinical studies have demonstrated that multiple mechanisms of cell death, including pyroptosis, apoptosis, autophagy, ferroptosis, anoikis, parthanatos, and senescence, are closely linked with the pathogenesis of PAH. This review summarizes different cell death mechanisms involved in the death of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs), the primary target cells in PAH. This review summarizes the role of these cell death mechanisms, associated signaling pathways, unique effector molecules, and various pro-survival or reprogramming mechanisms. The aim of this review is to summarize the currently known molecular mechanisms underlying PAH. Further investigations of the cell death mechanisms may unravel new avenues for the prevention and treatment of PAH.


Asunto(s)
Células Endoteliales , Miocitos del Músculo Liso , Hipertensión Arterial Pulmonar , Arteria Pulmonar , Transducción de Señal , Humanos , Células Endoteliales/patología , Miocitos del Músculo Liso/patología , Hipertensión Arterial Pulmonar/fisiopatología , Hipertensión Arterial Pulmonar/patología , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Muerte Celular , Animales , Apoptosis , Autofagia/fisiología , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología
7.
Plant Signal Behav ; 19(1): 2388443, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39116108

RESUMEN

Most studies on anesthesia focus on the nervous system of mammals due to their interest in medicine. The fact that any life form can be anaesthetised is often overlooked although anesthesia targets ion channel activities that exist in all living beings. This study examines the impact of lidocaine on rice (Oryza sativa). It reveals that the cellular responses observed in rice are analogous to those documented in animals, encompassing direct effects, the inhibition of cellular responses, and the long-distance transmission of electrical signals. We show that in rice cells, lidocaine has a cytotoxic effect at a concentration of 1%, since it induces programmed reactive oxygen species (ROS) and caspase-like-dependent cell death, as already demonstrated in animal cells. Additionally, lidocaine causes changes in membrane ion conductance and induces a sharp reduction in electrical long-distance signaling following seedlings leaves burning. Finally, lidocaine was shown to inhibit osmotic stress-induced cell death and the regulation of Ca2+ homeostasis. Thus, lidocaine treatment in rice and tobacco (Nicotiana benthamiana) seedlings induces not only cellular but also systemic effects similar to those induced in mammals.


Asunto(s)
Lidocaína , Oryza , Especies Reactivas de Oxígeno , Oryza/efectos de los fármacos , Oryza/metabolismo , Lidocaína/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Presión Osmótica/efectos de los fármacos , Anestésicos/farmacología
8.
Nutrients ; 16(15)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39125297

RESUMEN

Ovarian cancer is the most fatal of all the reproductive cancers within the female population, mainly due to its late diagnosis that limits surgery and medical treatment. Classically, ovarian cancer therapy has included conventional chemotherapy, and other therapeutic approaches are now being used to treat these patients, but the outcomes of the disease are still poor. Therefore, new strategies are needed to improve life expectancy and life quality of ovarian cancer patients. Considering that, we investigated the effect of the nutritional supplement Ocoxin Oral Solution (OOS) in ovarian cancer models. OOS contains several nutritional supplements, some of them with demonstrated antitumoral action. In vitro studies showed that OOS inhibited the proliferation of several ovarian cancer cell lines, especially of those representative of the endometrioid subtype, in a time- and dose-dependent manner. A fast cell death induction after OOS treatment was observed, and when the molecular mechanisms leading to this effect were investigated, an activation of the DNA damage checkpoint was detected, as shown by activation (phosphorylation) of CHK1 and CHK2 kinases that was followed by the phosphorylation of the target protein histone H2AX. When tested in animal models of ovarian cancer, OOS reduced tumor growth without any observed secondary effects. Moreover, such reduction in tumor proliferation was caused by the induction of DNA damage as corroborated by the in vivo phosphorylation of CHK2 and Histone H2AX. Finally, OOS potentiated the action of carboplatin or olaparib, the standard of care treatments used in ovarian clinics, opening the possibility of including OOS in combination with those standard of care agents in patients with ovarian cancer.


Asunto(s)
Proliferación Celular , Daño del ADN , Neoplasias Ováricas , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Humanos , Daño del ADN/efectos de los fármacos , Línea Celular Tumoral , Animales , Proliferación Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Piridoxina/farmacología , Ratones , Ácido Fólico/farmacología , Ácido Fólico/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Suplementos Dietéticos , Antineoplásicos/farmacología , Administración Oral , Vitamina B 6/farmacología , Vitamina B 6/administración & dosificación , Histonas/metabolismo , Sulfato de Zinc , Vitamina B 12 , Extractos Vegetales , Ácido Pantoténico , Ácido Ascórbico
9.
Int J Mol Sci ; 25(15)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39125694

RESUMEN

Diabetic neuropathy (DN) is a common complication of diabetes, affecting over 50% of patients, leading to significant pain and a burden. Currently, there are no effective treatments available. Cell death is considered a key factor in promoting the progression of DN. This article reviews how cell death is initiated in DN, emphasizing the critical roles of oxidative stress, mitochondrial dysfunction, inflammation, endoplasmic reticulum stress, and autophagy. Additionally, we thoroughly summarize the mechanisms of cell death that may be involved in the pathogenesis of DN, including apoptosis, autophagy, pyroptosis, and ferroptosis, among others, as well as potential therapeutic targets offered by these death mechanisms. This provides potential pathways for the prevention and treatment of diabetic neuropathy in the future.


Asunto(s)
Muerte Celular , Neuropatías Diabéticas , Estrés Oxidativo , Humanos , Neuropatías Diabéticas/patología , Neuropatías Diabéticas/terapia , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/etiología , Animales , Autofagia , Estrés del Retículo Endoplásmico , Apoptosis , Ferroptosis , Mitocondrias/metabolismo , Mitocondrias/patología
10.
Sci Rep ; 14(1): 17874, 2024 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-39090256

RESUMEN

Acute myeloid leukemia (AML) exhibits pronounced heterogeneity and chemotherapy resistance. Aberrant programmed cell death (PCD) implicated in AML pathogenesis suggests PCD-related signatures could serve as biomarkers to predict clinical outcomes and drug response. We utilized 13 PCD pathways, including apoptosis, pyroptosis, ferroptosis, autophagy, necroptosis, cuproptosis, parthanatos, entotic cell death, netotic cell death, lysosome-dependent cell death, alkaliptosis, oxeiptosis, and disulfidptosis to develop predictive models based on 73 machine learning combinations from 10 algorithms. Bulk RNA-sequencing, single-cell RNA-sequencing transcriptomic data, and matched clinicopathological information were obtained from the TCGA-AML, Tyner, and GSE37642-GPL96 cohorts. These datasets were leveraged to construct and validate the models. Additionally, in vitro experiments were conducted to substantiate the bioinformatics findings. The machine learning approach established a 6-gene pan-programmed cell death-related genes index (PPCDI) signature. Validation in two external cohorts showed high PPCDI associated with worse prognosis in AML patients. Incorporating PPCDI with clinical variables, we constructed several robust prognostic nomograms that accurately predicted prognosis of AML patients. Multi-omics analysis integrating bulk and single-cell transcriptomics revealed correlations between PPCDI and immunological features, delineating the immune microenvironment landscape in AML. Patients with high PPCDI exhibited resistance to conventional chemotherapy like doxorubicin but retained sensitivity to dasatinib and methotrexate (FDA-approved drugs for other leukemias), suggesting the potential of PPCDI to guide personalized therapy selection in AML. In summary, we developed a novel PPCDI model through comprehensive analysis of diverse programmed cell death pathways. This PPCDI signature demonstrates great potential in predicting clinical prognosis and drug sensitivity phenotypes in AML patients.


Asunto(s)
Biomarcadores de Tumor , Leucemia Mieloide Aguda , Aprendizaje Automático , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Pronóstico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Femenino , Masculino , Muerte Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Persona de Mediana Edad , Transcriptoma
11.
FASEB J ; 38(15): e23869, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39102006

RESUMEN

EXPRESSION OF CONCERN: G. Chen, K. A. Bower, C. Ma, S. Fang, C. J. Thiele, and J. Luo, "Glycogen Synthase Kinase 3ß (GSK3ß) Mediates 6-Hydroxydopamine-induced Neuronal Death," The FASEB Journal 18, no. 10 (2004): 1162-1164, https://doi.org/10.1096/fj.04-1551fje. This Expression of Concern for the above article published online on May 07, 2004 in Wiley Online Library (wileyonlinelibrary.com), has been published by agreement between the journal Editor-in-Chief, Loren E. Wold; the Federation of American Societies for Experimental Biology; and Wiley Periodicals LLC. The Expression of Concern has been agreed following concerns raised regarding duplication of the western blot bands presented in Figures 1a and 3a. Further, duplications of bands were also observed within Figure 5c. Due to the length of time that has elapsed since the publication of this article, the original data are no longer available to the authors. The journal has decided to issue an Expression of Concern to inform the readers of these concerns while the authors are repeating the experiments.


Asunto(s)
Muerte Celular , Glucógeno Sintasa Quinasa 3 beta , Neuronas , Oxidopamina , Oxidopamina/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Animales , Neuronas/metabolismo , Ratas , Humanos
12.
J Med Virol ; 96(8): e29854, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39135475

RESUMEN

Severe fever with thrombocytopenia syndrome (SFTS) has a high mortality rate compared to other infectious diseases. SFTS is particularly associated with a high risk of mortality in immunocompromised individuals, while most patients who die of SFTS exhibit symptoms of severe encephalitis before death. However, the region of brain damage and mechanisms by which the SFTS virus (SFTSV) causes encephalitis remains unknown. Here, we revealed that SFTSV infects the brainstem and spinal cord, which are regions of the brain associated with respiratory function, and motor nerves in IFNAR1-/- mice. Further, we show that A1-reactive astrocytes are activated, causing nerve cell death, in infected mice. Primary astrocytes of SFTSV-infected IFNAR1-/- mice also induced neuronal cell death through the activation of A1-reactive astrocytes. Herein, we showed that SFTSV induces fatal neuroinflammation in the brain regions important for respiratory function and motor nerve, which may underlie mortality in SFTS patients. This study provides new insights for the treatment of SFTS, for which there is currently no therapeutic approach.


Asunto(s)
Astrocitos , Infecciones por Bunyaviridae , Ratones Noqueados , Phlebovirus , Receptor de Interferón alfa y beta , Animales , Astrocitos/virología , Astrocitos/patología , Ratones , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/deficiencia , Phlebovirus/genética , Phlebovirus/fisiología , Phlebovirus/patogenicidad , Infecciones por Bunyaviridae/virología , Infecciones por Bunyaviridae/patología , Infecciones por Bunyaviridae/inmunología , Encéfalo/virología , Encéfalo/patología , Encéfalo/inmunología , Médula Espinal/virología , Médula Espinal/patología , Modelos Animales de Enfermedad , Neuronas/virología , Neuronas/patología , Ratones Endogámicos C57BL , Tronco Encefálico/virología , Tronco Encefálico/patología , Muerte Celular
13.
Cell Commun Signal ; 22(1): 379, 2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-39068453

RESUMEN

Copper is an important metal micronutrient, required for the balanced growth and normal physiological functions of human organism. Copper-related toxicity and dysbalanced metabolism were associated with the disruption of intracellular respiration and the development of various diseases, including cancer. Notably, copper-induced cell death was defined as cuproptosis which was also observed in malignant cells, representing an attractive anti-cancer instrument. Excess of intracellular copper leads to the aggregation of lipoylation proteins and toxic stress, ultimately resulting in the activation of cell death. Differential expression of cuproptosis-related genes was detected in normal and malignant tissues. Cuproptosis-related genes were also linked to the regulation of oxidative stress, immune cell responses, and composition of tumor microenvironment. Activation of cuproptosis was associated with increased expression of redox-metabolism-regulating genes, such as ferredoxin 1 (FDX1), lipoic acid synthetase (LIAS), lipoyltransferase 1 (LIPT1), dihydrolipoamide dehydrogenase (DLD), drolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1), and pyruvate dehydrogenase E1 subunit beta (PDHB)). Accordingly, copper-activated network was suggested as an attractive target in cancer therapy. Mechanisms of cuproptosis and regulation of cuproptosis-related genes in different cancers and tumor microenvironment are discussed in this study. The analysis of current findings indicates that therapeutic regulation of copper signaling, and activation of cuproptosis-related targets may provide an effective tool for the improvement of immunotherapy regimens.


Asunto(s)
Muerte Celular , Cobre , Inmunoterapia , Oxidación-Reducción , Humanos , Cobre/metabolismo , Neoplasias Torácicas/patología , Neoplasias Torácicas/genética , Animales
14.
Curr Protoc ; 4(7): e1112, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39073015

RESUMEN

The innate immune system is the first line of host defense. Innate immune activation utilizes pattern recognition receptors to detect pathogens, pathogen-associated and damage-associated molecular patterns (PAMPs and DAMPs), and homeostatic alterations and drives inflammatory signaling pathways and regulated cell death. Cell death activation is critical to eliminate pathogens and aberrant or damaged cells, while excess activation can be linked to inflammation, tissue damage, and disease. Therefore, there is increasing interest in studying cell death mechanisms to understand the underlying biology and identify therapeutic strategies. However, there are significant technical challenges, as many cell death pathways share key molecules with each other, and genetic models where these cell death molecules are deleted remain the gold standard for evaluation. Furthermore, extensive crosstalk has been identified between the cell death pathways pyroptosis, apoptosis, necroptosis, and the more recently characterized PANoptosis, which is defined as a prominent, unique innate immune, lytic, and inflammatory cell death pathway initiated by innate immune sensors and driven by caspases and RIPKs through PANoptosomes. PANoptosomes are multi-protein complexes assembled by innate immune sensor(s) in response to pathogens, PAMPs, DAMPs, cytokines, and homeostatic changes that drive PANoptosis. In this article, we provide methods for molecularly defining distinct cell death pathways, including PANoptosis, using both genetic and chemical approaches through western blot, LDH assay, and microscopy readouts. This procedure allows for the assessment of cell death on the cell population and single-cell levels even without access to genetic models. Having this comprehensive workflow that is more accessible to all labs will improve our ability as a scientific community to accelerate discovery. Using these protocols will help identify new innate immune sensors that drive PANoptosis and define the molecular mechanisms and regulators involved to establish new targets for clinical translation. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Induction and quantification of cell death using live cell imaging Alternate Protocol 1: Quantification of cell death using LDH Alternate Protocol 2: Assessment of cell death complexes in single cells using immunofluorescence staining Basic Protocol 2: Analysis of cell death mechanisms by immunoblots (western blots).


Asunto(s)
Muerte Celular , Inmunidad Innata , Humanos , Animales , Necroptosis/inmunología , Ratones
15.
Cell Biol Toxicol ; 40(1): 51, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38958792

RESUMEN

The implementation of Zinc oxide nanoparticles (ZnO NPs) raises concerns regarding their potential toxic effects on human health. Although more and more researches have confirmed the toxic effects of ZnO NPs, limited attention has been given to their impact on the early embryonic nervous system. This study aimed to explore the impact of exposure to ZnO NPs on early neurogenesis and explore its underlying mechanisms. We conducted experiments here to confirm the hypothesis that exposure to ZnO NPs causes neural tube defects in early embryonic development. We first used mouse and chicken embryos to confirm that ZnO NPs and the Zn2+ they release are able to penetrate the placental barrier, influence fetal growth and result in incomplete neural tube closure. Using SH-SY5Y cells, we determined that ZnO NPs-induced incomplete neural tube closure was caused by activation of various cell death modes, including ferroptosis, apoptosis and autophagy. Moreover, dissolved Zn2+ played a role in triggering widespread cell death. ZnO NPs were accumulated within mitochondria after entering cells, damaging mitochondrial function and resulting in the over production of reactive oxygen species, ultimately inducing cellular oxidative stress. The N-acetylcysteine (NAC) exhibits significant efficacy in mitigating cellular oxidative stress, thereby alleviating the cytotoxicity and neurotoxicity brought about by ZnO NPs. These findings indicated that the exposure of ZnO NPs in early embryonic development can induce cell death through oxidative stress, resulting in a reduced number of cells involved in early neural tube closure and ultimately resulting in incomplete neural tube closure during embryo development. The findings of this study could raise public awareness regarding the potential risks associated with the exposure and use of ZnO NPs in early pregnancy.


Asunto(s)
Desarrollo Embrionario , Defectos del Tubo Neural , Tubo Neural , Estrés Oxidativo , Especies Reactivas de Oxígeno , Óxido de Zinc , Óxido de Zinc/toxicidad , Animales , Estrés Oxidativo/efectos de los fármacos , Embrión de Pollo , Desarrollo Embrionario/efectos de los fármacos , Ratones , Tubo Neural/efectos de los fármacos , Tubo Neural/embriología , Tubo Neural/metabolismo , Humanos , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/patología , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Femenino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanopartículas del Metal/toxicidad , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Nanopartículas/toxicidad
16.
Sci Immunol ; 9(97): eadn0178, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38996010

RESUMEN

Virus-induced cell death is a key contributor to COVID-19 pathology. Cell death induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is well studied in myeloid cells but less in its primary host cell type, angiotensin-converting enzyme 2 (ACE2)-expressing human airway epithelia (HAE). SARS-CoV-2 induces apoptosis, necroptosis, and pyroptosis in HAE organotypic cultures. Single-cell and limiting-dilution analysis revealed that necroptosis is the primary cell death event in infected cells, whereas uninfected bystanders undergo apoptosis, and pyroptosis occurs later during infection. Mechanistically, necroptosis is induced by viral Z-RNA binding to Z-DNA-binding protein 1 (ZBP1) in HAE and lung tissues from patients with COVID-19. The Delta (B.1.617.2) variant, which causes more severe disease than Omicron (B1.1.529) in humans, is associated with orders of magnitude-greater Z-RNA/ZBP1 interactions, necroptosis, and disease severity in animal models. Thus, Delta induces robust ZBP1-mediated necroptosis and more disease severity.


Asunto(s)
COVID-19 , Necroptosis , Piroptosis , Proteínas de Unión al ARN , Mucosa Respiratoria , SARS-CoV-2 , Humanos , SARS-CoV-2/inmunología , COVID-19/inmunología , COVID-19/patología , Necroptosis/inmunología , Animales , Mucosa Respiratoria/virología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Ratones , Muerte Celular/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Apoptosis/inmunología
17.
Sci Immunol ; 9(97): eadp8170, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38996011

RESUMEN

Upon SARS-CoV-2 infection, infected cells undergo necroptosis, whereas delayed apoptosis and pyroptosis occur in uninfected, bystander cells, thus providing a plausible explanation for the extensive injury among myriad uninfected cells.


Asunto(s)
COVID-19 , Necroptosis , Piroptosis , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/patología , SARS-CoV-2/inmunología , Piroptosis/inmunología , Necroptosis/inmunología , Apoptosis/inmunología , Muerte Celular/inmunología , Animales
18.
Niger J Clin Pract ; 27(7): 827-836, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39082907

RESUMEN

BACKGROUND: Docetaxel (DOC) is the main chemotherapeutic agent for the treatment of advanced metastatic prostate cancer. Docetaxel shows anticancer effects by preventing the depolymerization of microtubules in the cell, therefore preventing cell division. However, the low survival effect of docetaxel has prompted researchers to search for novel therapeutic agents. Fucoidan (FUC) is a sulfated polysaccharide derived from brown algae. It has many bioactivities which makes fucoidan a promising anticancer agent. In this study, the potential anti-tumorigenic and preventive effects of fucoidan with or without docetaxel in prostate cancer were investigated by analyzing different cell death modalities. METHODS: The in-vivo six groups (n = 8) were conducted; preventive (Pt), docetaxel treated after preventive (Pt-D), control, fucoidan (FUC), docetaxel (DOC), and FUC and DOC (FUC+DOC) combination. Apoptotic, necroptotic, and autophagic cell death-related protein expressions were assessed in tumor tissues by using immunohistochemical staining. Oxidative stress-related lipid peroxidation, glutathione peroxidase, and glutathione levels were also determined in tumor tissues. RESULTS: Although apoptotic, necroptotic, and autophagic cell deaths were significantly induced in agent-treated groups compared to the control. Apoptotic cell death was more significantly induced in FUC and FUC+DOC-treated groups. Necroptotic cell death was increased considerably by inducing MLKL protein expression in all treatment groups. In the FUC, Pt, and DOC groups, LC3A/B expressions were significantly increased. DOC, FUC+DOC, and Pt-D treatments caused a significant increase in Beclin-1 expression. Oxidative stress-related MDA, GPX, and GSH levels significantly decreased with FUC treatment. The anti-tumorigenic effects of FUC and DOC were also demonstrated through tumor size reduction. CONCLUSION: According to the findings of this study, FUC inhibited tumor growth temporally and dimensionally, especially in preventive applications. FUC and FUC+DOC combinations in both treatment groups showed anti-tumorigenic effects. The results of this study suggest that fucoidan is a promising anticancer agent against prostate cancer. FUC can be considered as a preventive or treatment agent in prostate cancer therapy with DOC. Further studies are needed to fully elucidate the mechanism of action of fucoidan in metastatic prostate cancer.


Asunto(s)
Apoptosis , Docetaxel , Polisacáridos , Neoplasias de la Próstata , Masculino , Polisacáridos/farmacología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Docetaxel/farmacología , Apoptosis/efectos de los fármacos , Humanos , Antineoplásicos/farmacología , Animales , Estrés Oxidativo/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Autofagia/efectos de los fármacos , Ratones
19.
Redox Rep ; 29(1): 2382946, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39074442

RESUMEN

OBJECTIVES: To explore the effects and mechanisms of bilirubin on mitochondrial function and type of macrophage cell death after exposure to cigarette smoke extract (CSE). METHODS: RAW264.7 macrophages were treated with different concentrations of CSE and bilirubin solutions and divided into four groups: control, CSE, bilirubin, and bilirubin + CSE groups. The necrotic and apoptotic states of the macrophages were determined using an Annexin V-fluorescein 5-isothiocyanate/propidium iodide (FITC/PI) staining kit. Cytoplasmic NOD-like receptor family, pyrin domain containing 3 (NLRP3) expression in macrophages was detected by immunofluorescence and the levels of IL-1ß and IL-18 in the supernatants of culture medium were detected by enzyme linked immunosorbent assay (ELISA) test. A JC-1 mitochondrial membrane potential detection kit was used to assess mitochondrial membrane damage and the adenosine triphosphate (ATP) assay kit was used to determine intracellular ATP levels. After the macrophages were stained with reactive oxygen species (ROS) specific dye, 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA), the fluorescence intensity and proportion of ROS-positive macrophages were measured using flow cytometry. RESULTS: We observed that compared with those of 0 µM (control group), concentrations of 5, 10, or 20 µΜ bilirubin significantly decreased cell viability, which was increased by bilirubin exposure below 1 µM. The effect of CSE on macrophage viability was concentration- and time-dependent. Bilirubin of 0.2 µM could alleviate the inhibition of macrophage viability caused by 5% CSE. In addition, bilirubin intervention could reduce the occurrence of necrosis and pyroptosis to a certain extent. CONCLUSIONS: CSE could cause mitochondrial dysfunction in macrophages, as demonstrated by a decrease in mitochondrial membrane potential and intracellular ATP levels and an increase in ROS production, while bilirubin could relieve mitochondrial dysfunction caused by CSE.


Asunto(s)
Bilirrubina , Macrófagos , Mitocondrias , Especies Reactivas de Oxígeno , Animales , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Nicotiana/efectos adversos , Nicotiana/química , Humo/efectos adversos , Apoptosis/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
20.
Cell Biol Toxicol ; 40(1): 61, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075259

RESUMEN

Advancements in the CRISPR technology, a game-changer in experimental research, have revolutionized various fields of life sciences and more profoundly, cancer research. Cell death pathways are among the most deregulated in cancer cells and are considered as critical aspects in cancer development. Through decades, our knowledge of the mechanisms orchestrating programmed cellular death has increased substantially, attributed to the revolution of cutting-edge technologies. The heroic appearance of CRISPR systems have expanded the available screening platform and genome engineering toolbox to detect mutations and create precise genome edits. In that context, the precise ability of this system for identification and targeting of mutations in cell death signaling pathways that result in cancer development and therapy resistance is an auspicious choice to transform and accelerate the individualized cancer therapy. The concept of personalized cancer therapy stands on the identification of molecular characterization of the individual tumor and its microenvironment in order to provide a precise treatment with the highest possible outcome and minimum toxicity. This study explored the potential of CRISPR technology in precision cancer treatment by identifying and targeting specific cell death pathways. It showed the promise of CRISPR in finding key components and mutations involved in programmed cell death, making it a potential tool for targeted cancer therapy. However, this study also highlighted the challenges and limitations that need to be addressed in future research to fully realize the potential of CRISPR in cancer treatment.


Asunto(s)
Sistemas CRISPR-Cas , Muerte Celular , Neoplasias , Medicina de Precisión , Humanos , Neoplasias/genética , Neoplasias/terapia , Sistemas CRISPR-Cas/genética , Medicina de Precisión/métodos , Muerte Celular/genética , Transducción de Señal/genética , Edición Génica/métodos , Apoptosis/genética , Mutación/genética , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA