Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Toxicol In Vitro ; 65: 104772, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31935485

ABSTRACT

Lapachol is a plant-derived naphthoquinone that kills several types of cancer cells. Derivatives of this molecule may therefore prove to be useful chemotherapeutic agents. In this study, we explored whether glycosylation increases the cytotoxic potency of lapachol towards HL-60 human leukemia cells. Two beta-glycosides were synthesized and characterized: LA4A (lapachol-ß-glucoside) and LA4C (lapachol-N-acetylglucosamine-ß-glucoside). The sugar moieties of both novel molecules were per-acetylated to facilitate cellular uptake. The IC50 values (in µM) for LA4A (5.7) and LA4C (5.3) were lower than those for lapachol (25). LA4A and LA4C triggered typical signs of apoptosis, such as the exposure of phosphatidylserine on the outside of cells, chromatin condensation, DNA fragmentation and a decrease of the mitochondrial transmembrane potential (ΔΨm) prior to cell lysis. Moreover, DNA fragmentation triggered by the lapachol-glycosides was reduced by pre-treatment with the caspase inhibitor, z-VAD-fmk. While LA4A and LA4C activated caspases-3, -8 and -9, lapachol failed to activate these apoptotic proteases, even when used at high concentrations. Finally, the toxicity of lapachol and its derivatives was also tested on non-tumor cells. We used human peripheral neurons (PeriTox test) to evaluate the side effect potential of these compounds. LA4C was clearly less toxic than LA4A. We conclude that LA4C had the most favorable profile as drug candidate (high tumor cell toxicity, reduced neurotoxicity). In general, this study shows that the cytotoxicity of lapachol towards HL-60 can be enhanced by glycosylation, and that the therapeutic ratio may be modified by the type of sugar added.


Subject(s)
Antineoplastic Agents/toxicity , Naphthoquinones/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Glycosylation/drug effects , HL-60 Cells , Humans
8.
Mater Sci Eng C Mater Biol Appl ; 107: 110264, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31761183

ABSTRACT

In vitro drug screening is widely used in the development of new drugs, because they constitute a cost-effective approach to select compounds with more potential for therapy. They are also an attractive alternative to in vivo testing. However, most of these assays are done in two-dimensional culture models, where cells are grown on a polystyrene or glass flat surface. In order to develop in vitro models that would more closely resemble physiological conditions, three-dimensional models have been developed. Here, we introduce two novel fully synthetic scaffolds produced using the polymer polyhydroxybutyrate (PHB): a Solvent-Casting Particle-Leaching (SCPL) membrane; and an electrospun membrane, to be used for 3D cultures of B16 F10 murine melanoma cells and 4T1 murine breast cancer cells. A 2D cell culture system in regular tissue culture plates and a classical 3D model where cells are grown on a commercially available gel derived from Engelbreth-Holm Swarm (EHS) tumor were used for comparison with the synthetic scaffolds. Cells were also collected from in vivo tumors grown as grafts in syngeneic mice. Morphology, cell viability, response to chemotherapy and gene expression analysis were used to compare all systems. In the electrospun membrane model, cells were grown on nanometer-scale fibers and in the SCPL membrane, which provides a foam-like structure for cell growth, pore sizes varied. Cells grown on all 3D models were able to form aggregates and spheroids, allowing for increased cell-cell contact when compared with the 2D system. Cell morphology was also more similar between 3D systems and cells collected from the in vivo tumors. Cells grown in 3D models showed an increase in resistance to dacarbazine, and cisplatin. Gene expression analysis also revealed similarities among all 3D platforms. The similarities between the two synthetic systems to the classic EHS gel model highlight their potential application as cost effective substitutes in drug screening, in which fully synthetic models could represent a step towards higher reproducibility. We conclude PHB synthetic membranes offer a valuable alternative for 3D cultures.


Subject(s)
Cell Culture Techniques/methods , Cell Proliferation , Gene Expression , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
9.
J Leukoc Biol ; 106(3): 531-540, 2019 09.
Article in English | MEDLINE | ID: mdl-31071239

ABSTRACT

Autophagy and inflammasome activation are cell-autonomous and cross-regulated processes involved in host resistance against infections. Our group previously described that NLRP3 inflammasome is required for the control of Trypanosoma cruzi, the causative agent of Chagas disease. However, the involvement of autophagy in this process was unclear. Here, we demonstrated that T. cruzi was able to induce an increase in LC3-II expression as well as autophagosome and autolysosome formation in peritoneal macrophages (PMs) from C57BL/6 wild-type mice. Moreover, the pharmacologic inhibition of autophagic machinery impaired the ability of PMs to control T. cruzi replication. Importantly, NLRP3 was required for the induction of a regular autophagic flux in response to T. cruzi, an effect mediated by its participation in the autolysosomes formation. Together, these results indicate autophagy as an effector mechanism mediated by NLRP3 to control T. cruzi infection.


Subject(s)
Autophagy , Chagas Disease/metabolism , Chagas Disease/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acids/metabolism , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Lysosomes/metabolism , Lysosomes/ultrastructure , Macrophages/metabolism , Macrophages/parasitology , Mice, Inbred C57BL , Trypanosoma cruzi
10.
J Leukoc Biol ; 106(1): 201-207, 2019 07.
Article in English | MEDLINE | ID: mdl-30997938

ABSTRACT

Trypanossoma cruzi (T. cruzi), the causative protozoan of Chagas disease (CD) invades many cell types, including central nervous system (CNS) cells triggering local lesions and neurological impact. Previous work from our group described NLRP3 inflammasomes as central effectors for the parasite control by macrophages. Recent evidences demonstrate that NLRP3 can be activated in CNS cells with controversial consequences to the control of infections and inflammatory pathologies. However, the relative contribution of NLRP3 in different cell types remains to be elucidated. In this article, we described an effector response mediated by NLRP3 that works on microglia but not on astrocytes to control T. cruzi infection. Despite T. cruzi ability to invade astrocytes and microglia, astrocytes were clearly more permissive to parasite replication. Moreover, the absence of NLRP3 renders microglia but not astrocytes more permissive to T. cruzi replication. In fact, microglia but not astrocytes were able to secrete NLRP3-dependent IL-1ß and NO in response to T. cruzi. Importantly, the pharmacological inhibition of iNOS with aminoguanidine resulted in a significant increase in the numbers of amastigotes found in microglia from wild-type but not from NLRP3-/- mice, indicating the importance of NLRP3-mediated NO secretion to the infection control by these cells. Taken together, our findings revealed that T. cruzi differentially activates NLRP3 inflammasomes in astrocytes and microglia and established a role for these platforms in the control of a protozoan infection by glial cells from CNS.


Subject(s)
Astrocytes/parasitology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Nitric Oxide/biosynthesis , Trypanosoma cruzi/physiology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Microglia/parasitology
11.
Front Immunol ; 9: 2379, 2018.
Article in English | MEDLINE | ID: mdl-30459758

ABSTRACT

Pattern Recognition Receptors (PRRs) are proteins capable of recognizing molecules frequently found in pathogens (the so-called Pathogen-Associated Molecular Patterns-PAMPs), or molecules released by damaged cells (the Damage-Associated Molecular Patterns-DAMPs). They emerged phylogenetically prior to the appearance of the adaptive immunity and, therefore, are considered part of the innate immune system. Signals derived from the engagement of PRRs on the immune cells activate microbicidal and pro-inflammatory responses required to eliminate or, at least, to contain infectious agents. Molecularly controlled forms of cell death are also part of a very ancestral mechanism involved in key aspects of the physiology of multicellular organism, including the elimination of unwanted, damaged or infected cells. Interestingly, each form of cell death has its particular effect on inflammation and on the development of innate and adaptive immune responses. In this review article, we discuss some aspects of the molecular interplay between the cell death machinery and signals initiated by the activation of PRRs by PAMPs and DAMPs.


Subject(s)
Pathogen-Associated Molecular Pattern Molecules/metabolism , Receptors, Pattern Recognition/metabolism , Animals , Apoptosis , Cell Death , Host-Pathogen Interactions , Humans , Inflammation/etiology , Inflammation/metabolism , Pyroptosis
12.
Cancer Cell ; 32(3): 342-359.e10, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28898696

ABSTRACT

Concomitant hepatocyte apoptosis and regeneration is a hallmark of chronic liver diseases (CLDs) predisposing to hepatocellular carcinoma (HCC). Here, we mechanistically link caspase-8-dependent apoptosis to HCC development via proliferation- and replication-associated DNA damage. Proliferation-associated replication stress, DNA damage, and genetic instability are detectable in CLDs before any neoplastic changes occur. Accumulated levels of hepatocyte apoptosis determine and predict subsequent hepatocarcinogenesis. Proliferation-associated DNA damage is sensed by a complex comprising caspase-8, FADD, c-FLIP, and a kinase-dependent function of RIPK1. This platform requires a non-apoptotic function of caspase-8, but no caspase-3 or caspase-8 cleavage. It may represent a DNA damage-sensing mechanism in hepatocytes that can act via JNK and subsequent phosphorylation of the histone variant H2AX.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Caspase 8/metabolism , DNA Damage , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Cellular Senescence , Chronic Disease , Crosses, Genetic , DNA Repair , Fas-Associated Death Domain Protein/metabolism , Female , Genomic Instability , Hepatectomy , Hepatocytes/pathology , Histones/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/metabolism , Liver/pathology , Liver Regeneration , Male , Mice , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Risk Factors
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...