Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Publication year range
1.
Nat Rev Mol Cell Biol ; 18(2): 127-136, 2017 02.
Article in English | MEDLINE | ID: mdl-27999438

ABSTRACT

In the early 2000s, receptor-interacting serine/threonine protein kinase 1 (RIPK1), a molecule already recognized as an important regulator of cell survival, inflammation and disease, was attributed an additional function: the regulation of a novel cell death pathway that came to be known as necroptosis. Subsequently, the related kinase RIPK3 and its substrate mixed-lineage kinase domain-like protein (MLKL) were also implicated in the necroptotic pathway, and links between this pathway and apoptosis were established. In this Timeline article, we outline the discoveries that have helped to identify the roles of RIPK1, RIPK3, MLKL and other regulators of necroptosis, and how they interact to determine cell fate.


Subject(s)
Apoptosis/physiology , Inflammation/pathology , Necrosis/pathology , Animals , Caspase 8/metabolism , Cell Death , Disease Models, Animal , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Necrosis/physiopathology , Protein Kinases/genetics , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
2.
Cell ; 157(5): 1189-202, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24813850

ABSTRACT

Receptor-interacting protein kinase (RIPK)-1 is involved in RIPK3-dependent and -independent signaling pathways leading to cell death and/or inflammation. Genetic ablation of ripk1 causes postnatal lethality, which was not prevented by deletion of ripk3, caspase-8, or fadd. However, animals that lack RIPK1, RIPK3, and either caspase-8 or FADD survived weaning and matured normally. RIPK1 functions in vitro to limit caspase-8-dependent, TNFR-induced apoptosis, and animals lacking RIPK1, RIPK3, and TNFR1 survive to adulthood. The role of RIPK3 in promoting lethality in ripk1(-/-) mice suggests that RIPK3 activation is inhibited by RIPK1 postbirth. Whereas TNFR-induced RIPK3-dependent necroptosis requires RIPK1, cells lacking RIPK1 were sensitized to necroptosis triggered by poly I:C or interferons. Disruption of TLR (TRIF) or type I interferon (IFNAR) signaling delayed lethality in ripk1(-/-)tnfr1(-/-) mice. These results clarify the complex roles for RIPK1 in postnatal life and provide insights into the regulation of FADD-caspase-8 and RIPK3-MLKL signaling by RIPK1.


Subject(s)
Caspase 8/metabolism , Genes, Lethal , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Apoptosis , Caspase 8/genetics , Cell Death , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/metabolism , Inflammation/metabolism , Interferons/metabolism , Mice , Mice, Inbred C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factors/metabolism
3.
Gut ; 72(10): 1927-1941, 2023 10.
Article in English | MEDLINE | ID: mdl-37230755

ABSTRACT

OBJECTIVE: To better understand the immune microenvironment of pancreatic ductal adenocarcinomas (PDACs), here we explored the relevance of T and B cell compartmentalisation into tertiary lymphoid structures (TLSs) for the generation of local antitumour immunity. DESIGN: We characterised the functional states and spatial organisation of PDAC-infiltrating T and B cells using single-cell RNA sequencing (scRNA-seq), flow cytometry, multicolour immunofluorescence, gene expression profiling of microdissected TLSs, as well as in vitro assays. In addition, we performed a pan-cancer analysis of tumour-infiltrating T cells using scRNA-seq and sc T cell receptor sequencing datasets from eight cancer types. To evaluate the clinical relevance of our findings, we used PDAC bulk RNA-seq data from The Cancer Genome Atlas and the PRINCE chemoimmunotherapy trial. RESULTS: We found that a subset of PDACs harbours fully developed TLSs where B cells proliferate and differentiate into plasma cells. These mature TLSs also support T cell activity and are enriched with tumour-reactive T cells. Importantly, we showed that chronically activated, tumour-reactive T cells exposed to fibroblast-derived TGF-ß may act as TLS organisers by producing the B cell chemoattractant CXCL13. Identification of highly similar subsets of clonally expanded CXCL13 + tumour-infiltrating T cells across multiple cancer types further indicated a conserved link between tumour-antigen recognition and the allocation of B cells within sheltered hubs in the tumour microenvironment. Finally, we showed that the expression of a gene signature reflecting mature TLSs was enriched in pretreatment biopsies from PDAC patients with longer survival after receiving different chemoimmunotherapy regimens. CONCLUSION: We provided a framework for understanding the biological role of PDAC-associated TLSs and revealed their potential to guide the selection of patients for future immunotherapy trials.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Tertiary Lymphoid Structures , Humans , Tertiary Lymphoid Structures/metabolism , Tertiary Lymphoid Structures/pathology , Pancreatic Neoplasms/genetics , Carcinoma, Pancreatic Ductal/genetics , Immunity , Tumor Microenvironment , Pancreatic Neoplasms
4.
Immunity ; 41(6): 947-59, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25500368

ABSTRACT

Nonresolving inflammation expands a heterogeneous population of myeloid suppressor cells capable of inhibiting T cell function. This heterogeneity has confounded the functional dissection of individual myeloid subpopulations and presents an obstacle for antitumor immunity and immunotherapy. Using genetic manipulation of cell death pathways, we found the monocytic suppressor-cell subset, but not the granulocytic subset, requires continuous c-FLIP expression to prevent caspase-8-dependent, RIPK3-independent cell death. Development of the granulocyte subset requires MCL-1-mediated control of the intrinsic mitochondrial death pathway. Monocytic suppressors tolerate the absence of MCL-1 provided cytokines increase expression of the MCL-1-related protein A1. Monocytic suppressors mediate T cell suppression, whereas their granulocytic counterparts lack suppressive function. The loss of the granulocytic subset via conditional MCL-1 deletion did not alter tumor incidence implicating the monocytic compartment as the functionally immunosuppressive subset in vivo. Thus, death pathway modulation defines the development, survival, and function of myeloid suppressor cells.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Granulocytes/physiology , Monocytes/physiology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cells/physiology , Neoplasms, Experimental/immunology , Animals , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/genetics , Caspase 8/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Lineage/genetics , Coculture Techniques , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Signal Transduction/genetics
5.
Mol Cell ; 56(4): 469-80, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25459879

ABSTRACT

Receptor Interacting Protein Kinase-1 (RIPK1), a key player in inflammation and cell death, assumes opposite functions depending on the cellular context and its posttranslational modifications. Genetic evidence supported by biochemical and cellular biology approaches sheds light on the circumstances in which RIPK1 promotes or inhibits these processes.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis , Humans , Inflammation/enzymology , Signal Transduction , Ubiquitination
6.
Eur Respir J ; 57(6)2021 06.
Article in English | MEDLINE | ID: mdl-33303545

ABSTRACT

Respiratory syncytial virus (RSV) is the major cause of acute bronchiolitis in infants under 2 years old. Necroptosis has been implicated in the outcomes of respiratory virus infections. We report that RSV infection triggers necroptosis in primary mouse macrophages and human monocytes in a RIPK1-, RIPK3- and MLKL-dependent manner. Moreover, necroptosis pathways are harmful to RSV clearance from alveolar macrophages. Additionally, Ripk3-/- mice were protected from RSV-induced weight loss and presented with reduced viral loads in the lungs.Alveolar macrophage depletion also protected mice from weight loss and decreased lung RSV virus load. Importantly, alveolar macrophage depletion abolished the upregulation of Ripk3 and Mlkl gene expression induced by RSV infection in the lung tissue.Autocrine tumor necrosis factor (TNF)-mediated RSV-triggered macrophage necroptosis and necroptosis pathways were also involved in TNF secretion even when macrophages were committed to cell death, which can worsen lung injury during RSV infection. In line, Tnfr1-/- mice had a marked decrease in Ripk3 and Mlkl gene expression and a sharp reduction in the numbers of necrotic alveolar macrophages in the lungs. Finally, we provide evidence that elevated nasal levels of TNF are associated with disease severity in infants with RSV bronchiolitis.We propose that targeting TNF and/or the necroptotic machinery may be valuable therapeutic approaches to reduce the respiratory morbidity caused by RSV infection in young children.


Subject(s)
Bronchiolitis , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Animals , Macrophages, Alveolar , Mice , Necroptosis
7.
Adv Exp Med Biol ; 1301: 123-138, 2021.
Article in English | MEDLINE | ID: mdl-34370290

ABSTRACT

The past decades witnessed the discovery of novel modes of cell death, such as ferroptosis, pyroptosis and necroptosis, all of them presenting common necrotic traits. In this chapter, we revisit the early discoveries that unveiled necroptosis as a distinct cell death mechanism. We describe necroptosis, its main regulators and their role in maintaining cellular homeostasis and in the disease state. We conclude by discussing its phenotypic similarities with ferroptosis and the possible crosstalk between these pathways.


Subject(s)
Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Apoptosis , Caspases/genetics , Cell Death , Humans , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
8.
J Neurooncol ; 147(3): 587-594, 2020 May.
Article in English | MEDLINE | ID: mdl-32222932

ABSTRACT

PURPOSE: Necroptosis is a necrotic-like cell death pathway in which Receptor-interacting serine/threonine-protein kinase 3 (RIPK3) plays a central role and may induce inflammation and immunity. Lower RIPK3 levels have been correlated with a poor prognosis in breast and colorectal cancer patients. Instead, in gliomas, the most prevalent among central nervous system cancers, necrosis concurs with a more aggressive and lethal outcome, suggesting that, in these cases, necrotic-like pathways may be linked to worse prognoses. Lower-grade gliomas (LGG) exhibit highly diverse clinical behaviors, ranging from slow-paced growth to fast progression to glioblastoma yet patient outcomes cannot be fully predicted through the available markers. To date, IDH mutational status is the most broadly used prognostic marker, albeit several candidates have been proposed to refine LGG subgrouping. Here, we aimed to assess RIPK3 role as a prognostic marker for LGG patients, independently of or in combination with IDH. METHODS: Using publicly available discovery (513 patients) and validation (134 patients) cohorts, we performed Kaplan Meier survival analysis and uni- and multivariate Cox regression models. RESULTS: RIPK3 is an independent prognostic marker in LGG patients, even when controlled by age and molecular or histological diagnostic criteria. Contrary to what was previously reported for other cancers, high RIPK3 expression levels correlates with an increased risk of death. Importantly, RIPK3 expression levels further split both the mutant and wild-type IDH patients into distinct risk groups. CONCLUSION: RIPK3 expression levels can be used in combination with IDH mutational status to better subgroup LGG patients regarding overall survival.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Glioma/diagnosis , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Adult , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Prognosis
9.
Nature ; 506(7489): 451-5, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24553141

ABSTRACT

Members of the nuclear factor-κB (NF-κB) family of transcriptional regulators are central mediators of the cellular inflammatory response. Although constitutive NF-κB signalling is present in most human tumours, mutations in pathway members are rare, complicating efforts to understand and block aberrant NF-κB activity in cancer. Here we show that more than two-thirds of supratentorial ependymomas contain oncogenic fusions between RELA, the principal effector of canonical NF-κB signalling, and an uncharacterized gene, C11orf95. In each case, C11orf95-RELA fusions resulted from chromothripsis involving chromosome 11q13.1. C11orf95-RELA fusion proteins translocated spontaneously to the nucleus to activate NF-κB target genes, and rapidly transformed neural stem cells--the cell of origin of ependymoma--to form these tumours in mice. Our data identify a highly recurrent genetic alteration of RELA in human cancer, and the C11orf95-RELA fusion protein as a potential therapeutic target in supratentorial ependymoma.


Subject(s)
Cell Transformation, Neoplastic , Ependymoma/genetics , Ependymoma/metabolism , NF-kappa B/metabolism , Proteins/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Base Sequence , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 11/genetics , Ependymoma/pathology , Female , Humans , Mice , Models, Genetic , Molecular Sequence Data , NF-kappa B/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteins/genetics , Transcription Factor RelA/genetics , Transcription Factors , Translocation, Genetic/genetics , YAP-Signaling Proteins
10.
Mol Cell ; 44(1): 9-16, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21981915

ABSTRACT

Caspase-8, FADD, and FLIP orchestrate apoptosis in response to death receptor ligation. Mysteriously however, these proteins are also required for normal embryonic development and immune cell proliferation, an observation that has led to their implication in several nonapoptotic processes. While many scenarios have been proposed, recent genetic and biochemical evidence points to unregulated signaling by the receptor-interacting protein kinases-1 (RIPK1) and RIPK3 as the lethal defect in caspase-8-, FADD-, and FLIP-deficient animals and tissues. The RIPKs are known killers, being responsible for a nonapoptotic form of cell death with features similar to necrosis. However, the mechanism by which caspase-8, FADD, and FLIP prevent runaway RIPK activation is unknown, and the signals that trigger these events during development and immune cell activation remain at large. In this review, we will lay out the evidence as it now stands, reinterpreting earlier observations in light of new clues and considering where the investigation might lead.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Fas-Associated Death Domain Protein/metabolism , Gene Expression Regulation, Enzymologic , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Humans , Mice , Mice, Knockout
11.
Nature ; 471(7338): 363-7, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21368763

ABSTRACT

Caspase-8 has two opposing biological functions--it promotes cell death by triggering the extrinsic pathway of apoptosis, but also has a survival activity, as it is required for embryonic development, T-lymphocyte activation, and resistance to necrosis induced by tumour necrosis factor-α (TNF-α) and related family ligands. Here we show that development of caspase-8-deficient mice is completely rescued by ablation of receptor interacting protein kinase-3 (RIPK3). Adult animals lacking both caspase-8 and RIPK3 display a progressive lymphoaccumulative disease resembling that seen with defects in CD95 or CD95-ligand (also known as FAS and FASLG, respectively), and resist the lethal effects of CD95 ligation in vivo. We have found that caspase-8 prevents RIPK3-dependent necrosis without inducing apoptosis by functioning in a proteolytically active complex with FLICE-like inhibitory protein long (FLIP(L), also known as CFLAR), and this complex is required for the protective function.


Subject(s)
Biocatalysis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Caspase 8/genetics , Caspase Inhibitors , Cell Line , Female , Male , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Phenotype , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Serpins/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Viral Proteins/pharmacology , fas Receptor/deficiency , fas Receptor/metabolism
12.
Proc Natl Acad Sci U S A ; 111(47): 16836-41, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385600

ABSTRACT

Receptor-interacting protein kinase 3 (RIPK3)-mediated necroptosis is thought to be the pathophysiologically predominant pathway that leads to regulated necrosis of parenchymal cells in ischemia-reperfusion injury (IRI), and loss of either Fas-associated protein with death domain (FADD) or caspase-8 is known to sensitize tissues to undergo spontaneous necroptosis. Here, we demonstrate that renal tubules do not undergo sensitization to necroptosis upon genetic ablation of either FADD or caspase-8 and that the RIPK1 inhibitor necrostatin-1 (Nec-1) does not protect freshly isolated tubules from hypoxic injury. In contrast, iron-dependent ferroptosis directly causes synchronized necrosis of renal tubules, as demonstrated by intravital microscopy in models of IRI and oxalate crystal-induced acute kidney injury. To suppress ferroptosis in vivo, we generated a novel third-generation ferrostatin (termed 16-86), which we demonstrate to be more stable, to metabolism and plasma, and more potent, compared with the first-in-class compound ferrostatin-1 (Fer-1). Even in conditions with extraordinarily severe IRI, 16-86 exerts strong protection to an extent which has not previously allowed survival in any murine setting. In addition, 16-86 further potentiates the strong protective effect on IRI mediated by combination therapy with necrostatins and compounds that inhibit mitochondrial permeability transition. Renal tubules thus represent a tissue that is not sensitized to necroptosis by loss of FADD or caspase-8. Finally, ferroptosis mediates postischemic and toxic renal necrosis, which may be therapeutically targeted by ferrostatins and by combination therapy.


Subject(s)
Apoptosis , Kidney Tubules/cytology , Animals , Body Weight , Caspase 8/genetics , Caspase 8/physiology , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/physiology , Mice , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Reperfusion Injury/prevention & control
13.
J Immunol ; 192(4): 1835-46, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24453255

ABSTRACT

The Nlrp3 inflammasome is critical for host immunity, but the mechanisms controlling its activation are enigmatic. In this study, we show that loss of FADD or caspase-8 in a RIP3-deficient background, but not RIP3 deficiency alone, hampered transcriptional priming and posttranslational activation of the canonical and noncanonical Nlrp3 inflammasome. Deletion of caspase-8 in the presence or absence of RIP3 inhibited caspase-1 and caspase-11 activation by Nlrp3 stimuli but not the Nlrc4 inflammasome. In addition, FADD deletion prevented caspase-8 maturation, positioning FADD upstream of caspase-8. Consequently, FADD- and caspase-8-deficient mice had impaired IL-1ß production when challenged with LPS or infected with the enteropathogen Citrobacter rodentium. Thus, our results reveal FADD and caspase-8 as apical mediators of canonical and noncanonical Nlrp3 inflammasome priming and activation.


Subject(s)
Carrier Proteins/immunology , Caspase 8/metabolism , Fas-Associated Death Domain Protein/metabolism , Inflammasomes/immunology , Animals , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Caspase 1/metabolism , Caspase 8/genetics , Caspase 8/immunology , Caspases/metabolism , Caspases, Initiator , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Enzyme Activation , Fas-Associated Death Domain Protein/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides , Macrophages/immunology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Transcription, Genetic
14.
Proc Natl Acad Sci U S A ; 110(29): 12024-9, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818611

ABSTRACT

Regulated necrosis (RN) may result from cyclophilin (Cyp)D-mediated mitochondrial permeability transition (MPT) and receptor-interacting protein kinase (RIPK)1-mediated necroptosis, but it is currently unclear whether there is one common pathway in which CypD and RIPK1 act in or whether separate RN pathways exist. Here, we demonstrate that necroptosis in ischemia-reperfusion injury (IRI) in mice occurs as primary organ damage, independent of the immune system, and that mice deficient for RIPK3, the essential downstream partner of RIPK1 in necroptosis, are protected from IRI. Protection of RIPK3-knockout mice was significantly stronger than of CypD-deficient mice. Mechanistically, in vivo analysis of cisplatin-induced acute kidney injury and hyperacute TNF-shock models in mice suggested the distinctness of CypD-mediated MPT from RIPK1/RIPK3-mediated necroptosis. We, therefore, generated CypD-RIPK3 double-deficient mice that are viable and fertile without an overt phenotype and that survived prolonged IRI, which was lethal to each single knockout. Combined application of the RIPK1 inhibitor necrostatin-1 and the MPT inhibitor sanglifehrin A confirmed the results with mutant mice. The data demonstrate the pathophysiological coexistence and corelevance of two separate pathways of RN in IRI and suggest that combination therapy targeting distinct RN pathways can be beneficial in the treatment of ischemic injury.


Subject(s)
Apoptosis/physiology , Cyclophilins/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Necrosis/physiopathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Reperfusion Injury/complications , Animals , Cell Line , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , DNA Primers/genetics , Genotype , Kaplan-Meier Estimate , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Permeability Transition Pore , Necrosis/etiology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
15.
J Virol ; 88(1): 503-15, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24173220

ABSTRACT

Enhancement of cell death is a distinguishing feature of H1N1 influenza virus A/Puerto Rico/8/34 protein PB1-F2. Comparing the sequences (amino acids [aa] 61 to 87 using PB1-F2 amino acid numbering) of the PB1-F2-derived C-terminal peptides from influenza A viruses inducing high or low levels of cell death, we identified a unique I68, L69, and V70 motif in A/Puerto Rico/8/34 PB1-F2 responsible for promotion of the peptide's cytotoxicity and permeabilization of the mitochondrial membrane. When administered to mice, a 27-mer PB1-F2-derived C-terminal peptide with this amino acid motif caused significantly greater weight loss and pulmonary inflammation than the peptide without it (due to I68T, L69Q, and V70G mutations). Similar to the wild-type peptide, A/Puerto Rico/8/34 elicited significantly higher levels of macrophages, neutrophils, and cytokines in the bronchoalveolar lavage fluid of mice than its mutant counterpart 7 days after infection. Additionally, infection of mice with A/Puerto Rico/8/34 significantly enhanced the levels of morphologically transformed epithelial and immune mononuclear cells recruited in the airways compared with the mutant virus. In the mouse bacterial superinfection model, both peptide and virus with the I68, L69, and V70 sequence accelerated development of pneumococcal pneumonia, as reflected by increased levels of viral and bacterial lung titers and by greater mortality. Here we provide evidence suggesting that the newly identified cytotoxic sequence I68, L69, and V70 of A/Puerto Rico/8/34 PB1-F2 contributes to the pathogenesis of both primary viral and secondary bacterial infections.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/immunology , Pneumonia, Bacterial/complications , Viral Proteins/physiology , Amino Acid Sequence , Animals , Bronchoalveolar Lavage Fluid , Dogs , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/virology , Sequence Homology, Amino Acid , Viral Proteins/chemistry
16.
Cell Rep ; 43(6): 114335, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38850531

ABSTRACT

Perturbation of the apoptosis and necroptosis pathways critically influences embryogenesis. Receptor-associated protein kinase-1 (RIPK1) interacts with Fas-associated via death domain (FADD)-caspase-8-cellular Flice-like inhibitory protein long (cFLIPL) to regulate both extrinsic apoptosis and necroptosis. Here, we describe Ripk1-mutant animals (Ripk1R588E [RE]) in which the interaction between FADD and RIPK1 is disrupted, leading to embryonic lethality. This lethality is not prevented by further removal of the kinase activity of Ripk1 (Ripk1R588E K45A [REKA]). Both Ripk1RE and Ripk1REKA animals survive to adulthood upon ablation of Ripk3. While embryonic lethality of Ripk1RE mice is prevented by ablation of the necroptosis effector mixed lineage kinase-like (MLKL), animals succumb to inflammation after birth. In contrast, Mlkl ablation does not prevent the death of Ripk1REKA embryos, but animals reach adulthood when both MLKL and caspase-8 are removed. Ablation of the nucleic acid sensor Zbp1 largely prevents lethality in both Ripk1RE and Ripk1REKA embryos. Thus, the RIPK1-FADD interaction prevents Z-DNA binding protein-1 (ZBP1)-induced, RIPK3-caspase-8-mediated embryonic lethality, affected by the kinase activity of RIPK1.


Subject(s)
Caspase 8 , Fas-Associated Death Domain Protein , Inflammation , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Fas-Associated Death Domain Protein/metabolism , Inflammation/metabolism , Inflammation/pathology , Mice , Caspase 8/metabolism , Protein Kinases/metabolism , Apoptosis , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Necroptosis , Protein Binding , Mice, Inbred C57BL
17.
Transplant Cell Ther ; 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38986739

ABSTRACT

Natural killer (NK) cells play a crucial role in the immune system's response against cancer. However, the challenge of obtaining the required quantity of NK cells for effective therapeutic response necessitates the development of strategies for their ex vivo expansion. This study aimed to develop a novel feeder cell line, K562.Clone1, capable of promoting the ex vivo expansion of NK cells while preserving their cytotoxic potential. he K562 leukemic cell line was transduced with mbIL-21 and 4-1BBL proteins to generate K562.Clone1 cells. NK cells were then co-cultured with these feeder cells, and their expansion rate was monitored over 14 days. The cytotoxic potential of the expanded NK cells was evaluated against acute myeloid leukemia blasts and tumor cell lines of leukemia and glial origin. Statistical analysis was performed to determine the significance of the results. The K562.Clone1 co-cultured with peripheral NK showed a significant increase in cell count, with an approximate 94-fold expansion over 14 days. Expanded NK cells demonstrated cytotoxicity against the tested tumor cell lines, indicating preservation of their cytotoxic characteristics. Additionally, the CD56, CD16, inhibitory KIRs, and activation receptors were conserved and present in a well-balanced manner. The study successfully developed a feeder cell line, K562.Clone1, that effectively promotes the expansion of NK cells ex vivo while maintaining their cytotoxic potential. This development could significantly contribute to the advancement of NK cell therapy, especially in Brazil.

18.
PLoS One ; 18(8): e0291019, 2023.
Article in English | MEDLINE | ID: mdl-37651429

ABSTRACT

INTRODUCTION: Recently, the search for novel molecular markers in adult-type diffuse gliomas has grown substantially, yet with few novel breakthroughs. As the presence of a necrotic center is a differential diagnosis for more aggressive entities, we hypothesized that genes involved in necroptosis may play a role in tumor progression. AIM: Given that MLKL is the executioner of the necroptotic pathway, we evaluated whether this gene would help to predict prognosis of adult gliomas patients. METHODS: We analyzed a publicly available retrospective cohort (n = 530) with Kaplan Meier survival analysis (p<0.0001) and both uni- and multivariate Cox regression models. RESULTS: We determined that MLKL is an independent predictive prognostic marker for overall survival in these patients (HR: 2.56, p<0.001), even when controlled by the CNS5 gold-standard markers, namely IDH mutation and 1p/19q Codeletion (HR: 1.68, p = 0.013). These findings were confirmed in a validation cohort (n = 325), using the same cutoff value. Interestingly, higher expression of MLKL is associated with worse clinical outcome for adult-type diffuse glioma patients, which is opposite to what was found in other cell cancer types, suggesting that necroptosis undertakes an atypical detrimental role in glioma progression.


Subject(s)
Genes, Regulator , Glioma , Humans , Adult , Retrospective Studies , Transcription Factors , Glioma/genetics , Aggression , Protein Kinases
19.
Expert Opin Biol Ther ; 23(4): 353-364, 2023 04.
Article in English | MEDLINE | ID: mdl-36920351

ABSTRACT

INTRODUCTION: Lysosomal storage disorders (LSD) are a group of monogenic rare diseases caused by pathogenic variants in genes that encode proteins related to lysosomal function. These disorders are good candidates for gene therapy for different reasons: they are monogenic, most of lysosomal proteins are enzymes that can be secreted and cross-correct neighboring cells, and small quantities of these proteins are able to produce clinical benefits in many cases. Ex vivo gene therapy allows for autologous transplant of modified cells from different sources, including stem cells and hematopoietic precursors. AREAS COVERED: Here, we summarize the main gene therapy and genome editing strategies that are currently being used as ex vivo gene therapy approaches for lysosomal disorders, highlighting important characteristics, such as vectors used, strategies, types of cells that are modified and main results in different disorders. EXPERT OPINION: Clinical trials are already ongoing, and soon approved therapies for LSD based on ex vivo gene therapy approaches should reach the market.


Subject(s)
Lysosomal Storage Diseases , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Genetic Vectors , Genetic Therapy/methods , Lysosomes
20.
Mol Med ; 18: 577-86, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22371307

ABSTRACT

Tumor necrosis factor receptor (TNFR) signaling may result in survival, apoptosis or programmed necrosis. The latter is called necroptosis if the receptor-interacting protein 1 (RIP1) inhibitor necrostatin-1 (Nec-1) or genetic knockout of RIP3 prevents it. In the lethal mouse model of TNFα-mediated shock, addition of the pan-caspase inhibitor zVAD-fmk (zVAD) accelerates time to death. Here, we demonstrate that RIP3-deficient mice are protected markedly from TNFα-mediated shock in the presence and absence of caspase inhibition. We further show that the fusion protein TAT-crmA, previously demonstrated to inhibit apoptosis, also prevents necroptosis in L929, HT29 and FADD-deficient Jurkat cells. In contrast to RIP3-deficient mice, blocking necroptosis by Nec-1 or TAT-crmA did not protect from TNFα/zVAD-mediated shock, but further accelerated time to death. Even in the absence of caspase inhibition, Nec-1 application led to similar kinetics. Depletion of macrophages, natural killer (NK) cells, granulocytes or genetic deficiency for T lymphocytes did not influence this model. Because RIP3-deficient mice are known to be protected from cerulein-induced pancreatitis (CIP), we applied Nec-1 and TAT-crmA in this model and demonstrated the deterioration of pancreatic damage upon addition of these substances. These data highlight the importance of separating genetic RIP3 deficiency from RIP1 inhibition by Nec-1 application in vivo and challenge the current definition of necroptosis.


Subject(s)
Apoptosis , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Shock/genetics , Shock/pathology , Tumor Necrosis Factor-alpha/toxicity , Animals , Apoptosis/drug effects , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor , Female , Gene Products, tat/genetics , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Necrosis , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology , Recombinant Fusion Proteins/pharmacology , Serpins/genetics , Shock/chemically induced , Shock/mortality , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL