Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 305
Filter
1.
Cell ; 187(2): 235-256, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38242081

ABSTRACT

Cell death supports morphogenesis during development and homeostasis after birth by removing damaged or obsolete cells. It also curtails the spread of pathogens by eliminating infected cells. Cell death can be induced by the genetically programmed suicide mechanisms of apoptosis, necroptosis, and pyroptosis, or it can be a consequence of dysregulated metabolism, as in ferroptosis. Here, we review the signaling mechanisms underlying each cell-death pathway, discuss how impaired or excessive activation of the distinct cell-death processes can promote disease, and highlight existing and potential therapies for redressing imbalances in cell death in cancer and other diseases.


Subject(s)
Cell Death , Signal Transduction , Humans , Apoptosis , Ferroptosis , Homeostasis , Pyroptosis
2.
Cell ; 187(2): 219-224, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38242078

ABSTRACT

50 years ago, cell biology was a nascent field. Today, it is a vast discipline whose principles and tools are also applied to other disciplines; vice versa, cell biologists are inspired by other fields. So, the question begs: what is cell biology? The answers are as diverse as the people who define it.

3.
Cell ; 173(5): 1217-1230.e17, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29775594

ABSTRACT

Intrinsic apoptosis, reliant on BAX and BAK, has been postulated to be fundamental for morphogenesis, but its precise contribution to this process has not been fully explored in mammals. Our structural analysis of BOK suggests close resemblance to BAX and BAK structures. Notably, Bok-/-Bax-/-Bak-/- animals exhibited more severe defects and died earlier than Bax-/-Bak-/- mice, implying that BOK has overlapping roles with BAX and BAK during developmental cell death. By analyzing Bok-/-Bax-/-Bak-/- triple-knockout mice whose cells are incapable of undergoing intrinsic apoptosis, we identified tissues that formed well without this process. We provide evidence that necroptosis, pyroptosis, or autophagy does not substantially substitute for the loss of apoptosis. Albeit very rare, unexpected attainment of adult Bok-/-Bax-/-Bak-/- mice suggests that morphogenesis can proceed entirely without apoptosis mediated by these proteins and possibly without cell death in general.


Subject(s)
Apoptosis , Embryo, Mammalian/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics , Abnormalities, Multiple/pathology , Abnormalities, Multiple/veterinary , Animals , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/pathology , Embryonic Development/genetics , Fetus/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
4.
Nat Rev Mol Cell Biol ; 21(11): 678-695, 2020 11.
Article in English | MEDLINE | ID: mdl-32873928

ABSTRACT

The removal of functionally dispensable, infected or potentially neoplastic cells is driven by programmed cell death (PCD) pathways, highlighting their important roles in homeostasis, host defence against pathogens, cancer and a range of other pathologies. Several types of PCD pathways have been described, including apoptosis, necroptosis and pyroptosis; they employ distinct molecular and cellular processes and differ in their outcomes, such as the capacity to trigger inflammatory responses. Recent genetic and biochemical studies have revealed remarkable flexibility in the use of these PCD pathways and indicate a considerable degree of plasticity in their molecular regulation; for example, despite having a primary role in inducing pyroptosis, inflammatory caspases can also induce apoptosis, and conversely, apoptotic stimuli can trigger pyroptosis. Intriguingly, this flexibility is most pronounced in cellular responses to infection, while apoptosis is the dominant cell death process through which organisms prevent the development of cancer. In this Review, we summarize the mechanisms of the different types of PCD and describe the physiological and pathological processes that engage crosstalk between these pathways, focusing on infections and cancer. We discuss the intriguing notion that the different types of PCD could be seen as a single, coordinated cell death system, in which the individual pathways are highly interconnected and can flexibly compensate for one another.


Subject(s)
Apoptosis/physiology , Signal Transduction/physiology , Animals , Humans
5.
Immunity ; 55(3): 423-441.e9, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35139355

ABSTRACT

Cell death plays an important role during pathogen infections. Here, we report that interferon-γ (IFNγ) sensitizes macrophages to Toll-like receptor (TLR)-induced death that requires macrophage-intrinsic death ligands and caspase-8 enzymatic activity, which trigger the mitochondrial apoptotic effectors, BAX and BAK. The pro-apoptotic caspase-8 substrate BID was dispensable for BAX and BAK activation. Instead, caspase-8 reduced pro-survival BCL-2 transcription and increased inducible nitric oxide synthase (iNOS), thus facilitating BAX and BAK signaling. IFNγ-primed, TLR-induced macrophage killing required iNOS, which licensed apoptotic caspase-8 activity and reduced the BAX and BAK inhibitors, A1 and MCL-1. The deletion of iNOS or caspase-8 limited SARS-CoV-2-induced disease in mice, while caspase-8 caused lethality independent of iNOS in a model of hemophagocytic lymphohistiocytosis. These findings reveal that iNOS selectively licenses programmed cell death, which may explain how nitric oxide impacts disease severity in SARS-CoV-2 infection and other iNOS-associated inflammatory conditions.


Subject(s)
COVID-19/immunology , Caspase 8/metabolism , Interferon-gamma/metabolism , Lymphohistiocytosis, Hemophagocytic/immunology , Macrophages/immunology , Mitochondria/metabolism , SARS-CoV-2/physiology , Animals , Caspase 8/genetics , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Interferon-gamma/genetics , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Signal Transduction , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
6.
Immunity ; 54(8): 1758-1771.e7, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34256013

ABSTRACT

Apoptosis can potently defend against intracellular pathogens by directly killing microbes and eliminating their replicative niche. However, the reported ability of Mycobacterium tuberculosis to restrict apoptotic pathways in macrophages in vitro has led to apoptosis being dismissed as a host-protective process in tuberculosis despite a lack of in vivo evidence. Here we define crucial in vivo functions of the death receptor-mediated and BCL-2-regulated apoptosis pathways in mediating protection against tuberculosis by eliminating distinct populations of infected macrophages and neutrophils and priming T cell responses. We further show that apoptotic pathways can be targeted therapeutically with clinical-stage compounds that antagonize inhibitor of apoptosis (IAP) proteins to promote clearance of M. tuberculosis in mice. These findings reveal that any inhibition of apoptosis by M. tuberculosis is incomplete in vivo, advancing our understanding of host-protective responses to tuberculosis (TB) and revealing host pathways that may be targetable for treatment of disease.


Subject(s)
Apoptosis/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Neutrophils/immunology , Tuberculosis, Pulmonary/immunology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Line , Dipeptides/therapeutic use , Humans , Indoles/therapeutic use , Lymphocyte Activation/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/microbiology , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocytes/immunology , Thiazoles/therapeutic use , Tuberculosis, Pulmonary/drug therapy
7.
Immunity ; 53(3): 533-547.e7, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32735843

ABSTRACT

Programmed cell death contributes to host defense against pathogens. To investigate the relative importance of pyroptosis, necroptosis, and apoptosis during Salmonella infection, we infected mice and macrophages deficient for diverse combinations of caspases-1, -11, -12, and -8 and receptor interacting serine/threonine kinase 3 (RIPK3). Loss of pyroptosis, caspase-8-driven apoptosis, or necroptosis had minor impact on Salmonella control. However, combined deficiency of these cell death pathways caused loss of bacterial control in mice and their macrophages, demonstrating that host defense can employ varying components of several cell death pathways to limit intracellular infections. This flexible use of distinct cell death pathways involved extensive cross-talk between initiators and effectors of pyroptosis and apoptosis, where initiator caspases-1 and -8 also functioned as executioners when all known effectors of cell death were absent. These findings uncover a highly coordinated and flexible cell death system with in-built fail-safe processes that protect the host from intracellular infections.


Subject(s)
Apoptosis/immunology , Macrophages/immunology , Necroptosis/immunology , Pyroptosis/immunology , Salmonella Infections/immunology , Salmonella/immunology , Animals , Caspase 1/deficiency , Caspase 1/genetics , Caspase 12/deficiency , Caspase 12/genetics , Caspase 8/genetics , Caspases, Initiator/deficiency , Caspases, Initiator/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
8.
EMBO J ; 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160273

ABSTRACT

Investigating how transcription factors control complex cellular processes requires tools that enable responses to be visualised at the single-cell level and their cell fate to be followed over time. For example, the tumour suppressor p53 (also called TP53 in humans and TRP53 in mice) can initiate diverse cellular responses by transcriptional activation of its target genes: Puma to induce apoptotic cell death and p21 to induce cell cycle arrest/cell senescence. However, it is not known how these processes are regulated and initiated in different cell types. Also, the context-dependent interaction partners and binding loci of p53 remain largely elusive. To be able to examine these questions, we here developed knock-in mice expressing triple-FLAG-tagged p53 to facilitate p53 pull-down and two p53 response reporter mice, knocking tdTomato and GFP into the Puma/Bbc3 and p21 gene loci, respectively. By crossing these reporter mice into a p53-deficient background, we show that the new reporters reliably inform on p53-dependent and p53-independent initiation of both apoptotic or cell cycle arrest/senescence programs, respectively, in vitro and in vivo.

9.
Mol Cell ; 78(6): 1045-1054, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32516599

ABSTRACT

Cell death, or, more specifically, cell suicide, is a process of fundamental importance to human health. Throughout our lives, over a million cells are produced every second. When organismal growth has stopped, to balance cell division, a similar number of cells must be removed. This is achieved by activation of molecular mechanisms that have evolved so that cells can destroy themselves. The first clues regarding the nature of one of these mechanisms came from studying genes associated with cancer, in particular the gene for BCL-2. Subsequent studies revealed that mutations or other defects that inhibit cell death allow cells to accumulate, prevent removal of cells with damaged DNA, and increase the resistance of malignant cells to chemotherapy. Knowledge of this mechanism has allowed development of drugs that kill cancer cells by directly activating the cell death machinery and by synergizing with conventional chemotherapy as well as targeted agents to achieve improved outcomes for cancer patients.


Subject(s)
Cell Death/physiology , Neoplasms/genetics , Neoplasms/therapy , Apoptosis/genetics , Cell Death/genetics , DNA Damage/genetics , Humans , Neoplasms/physiopathology
10.
Immunity ; 48(3): 570-583.e8, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29562203

ABSTRACT

Polymorphisms in NFKB1 that diminish its expression have been linked to human inflammatory diseases and increased risk for epithelial cancers. The underlying mechanisms are unknown, and the link is perplexing given that NF-κB signaling reportedly typically exerts pro-tumorigenic activity. Here we have shown that NF-κB1 deficiency, even loss of a single allele, resulted in spontaneous invasive gastric cancer (GC) in mice that mirrored the histopathological progression of human intestinal-type gastric adenocarcinoma. Bone marrow chimeras revealed that NF-κB1 exerted tumor suppressive functions in both epithelial and hematopoietic cells. RNA-seq analysis showed that NF-κB1 deficiency resulted in aberrant JAK-STAT signaling, which dysregulated expression of effectors of inflammation, antigen presentation, and immune checkpoints. Concomitant loss of STAT1 prevented these immune abnormalities and GC development. These findings provide mechanistic insight into how polymorphisms that attenuate NFKB1 expression predispose humans to epithelial cancers, highlighting the pro-tumorigenic activity of STAT1 and identifying targetable vulnerabilities in GC.


Subject(s)
Gene Expression Regulation, Neoplastic , Inflammation/genetics , Inflammation/metabolism , NF-kappa B/deficiency , STAT1 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Animals , Antigen Presentation/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Regulatory Networks , Humans , Inflammation/pathology , Mice , Mice, Knockout , STAT1 Transcription Factor/deficiency , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology
11.
EMBO J ; 41(15): e110300, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35758142

ABSTRACT

The intrinsic apoptosis pathway, regulated by the BCL-2 protein family, is essential for embryonic development. Using mice lacking all known apoptosis effectors, BAX, BAK and BOK, we have previously defined the processes during development that require apoptosis. Rare Bok-/- Bax-/- Bak-/- triple knockout (TKO) mice developed to adulthood and several tissues that were thought to require apoptosis during development appeared normal. This raises the question if all apoptosis had been abolished in the TKO mice or if other BCL-2 family members could act as effectors of apoptosis. Here, we investigated the role of BID, generally considered to link the extrinsic and intrinsic apoptosis pathways, acting as a BH3-only protein initiating apoptosis upstream of BAX and BAK. We found that Bok-/- Bax-/- Bak-/- Bid-/- quadruple knockout (QKO) mice have additional developmental anomalies compared to TKO mice, consistent with a role of BID, not only upstream but also in parallel to BAX, BAK and BOK. Mitochondrial experiments identified a small cytochrome c-releasing activity of full-length BID. Collectively, these findings suggest a new effector role for BID in the intrinsic apoptosis pathway.


Subject(s)
BH3 Interacting Domain Death Agonist Protein , Proto-Oncogene Proteins c-bcl-2 , bcl-2 Homologous Antagonist-Killer Protein , Animals , Mice , Apoptosis , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Embryonic Development/genetics , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
12.
Nat Rev Mol Cell Biol ; 15(1): 49-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24355989

ABSTRACT

The BCL-2 protein family determines the commitment of cells to apoptosis, an ancient cell suicide programme that is essential for development, tissue homeostasis and immunity. Too little apoptosis can promote cancer and autoimmune diseases; too much apoptosis can augment ischaemic conditions and drive neurodegeneration. We discuss the biochemical, structural and genetic studies that have clarified how the interplay between members of the BCL-2 family on mitochondria sets the apoptotic threshold. These mechanistic insights into the functions of the BCL-2 family are illuminating the physiological control of apoptosis, the pathological consequences of its dysregulation and the promising search for novel cancer therapies that target the BCL-2 family.


Subject(s)
Apoptosis , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/physiology , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/physiology , Humans , Models, Molecular , Molecular Sequence Data , Molecular Targeted Therapy , Protein Multimerization , Protein Structure, Secondary , Proto-Oncogene Proteins c-bcl-2/chemistry , Signal Transduction
13.
Genes Dev ; 32(21-22): 1420-1429, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30366906

ABSTRACT

Mutations in Trp53, prevalent in human cancer, are reported to drive tumorigenesis through dominant-negative effects (DNEs) over wild-type TRP53 function as well as neomorphic gain-of-function (GOF) activity. We show that five TRP53 mutants do not accelerate lymphomagenesis on a TRP53-deficient background but strongly synergize with c-MYC overexpression in a manner that distinguishes the hot spot Trp53 mutations. RNA sequencing revealed that the mutant TRP53 DNE does not globally repress wild-type TRP53 function but disproportionately impacts a subset of wild-type TRP53 target genes. Accordingly, TRP53 mutant proteins impair pathways for DNA repair, proliferation, and metabolism in premalignant cells. This reveals that, in our studies of lymphomagenesis, mutant TRP53 drives tumorigenesis primarily through the DNE, which modulates wild-type TRP53 function in a manner advantageous for neoplastic transformation.


Subject(s)
Carcinogenesis/genetics , Mutation , Tumor Suppressor Protein p53/genetics , Animals , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Suppressor Protein p53/metabolism
14.
Nat Immunol ; 14(3): 290-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23377201

ABSTRACT

The long-term survival of plasma cells is entirely dependent on signals derived from their environment. These extrinsic factors presumably induce and sustain the expression of antiapoptotic proteins of the Bcl-2 family. It is uncertain whether there is specificity among Bcl-2 family members in the survival of plasma cells and whether their expression is linked to specific extrinsic factors. We found here that deletion of the gene encoding the antiapoptotic protein Mcl-1 in plasma cells resulted in rapid depletion of this population in vivo. Furthermore, we found that the receptor BCMA was needed to establish high expression of Mcl-1 in bone marrow but not spleen plasma cells and that establishing this survival pathway preceded the component of plasma cell differentiation that depends on the transcriptional repressor Blimp-1. Our results identify a critical role for Mcl-1 in the maintenance of plasma cells.


Subject(s)
B-Cell Maturation Antigen/metabolism , Plasma Cells/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factors/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Survival , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Plasma Cells/cytology , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-2/genetics , Spleen/immunology
15.
Nat Immunol ; 14(9): 959-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23852275

ABSTRACT

Foxp3⁺ regulatory T (Treg) cells are a crucial immunosuppressive population of CD4⁺ T cells, yet the homeostatic processes and survival programs that maintain the Treg cell pool are poorly understood. Here we report that peripheral Treg cells markedly alter their proliferative and apoptotic rates to rapidly restore numerical deficit through an interleukin 2-dependent and costimulation-dependent process. By contrast, excess Treg cells are removed by attrition, dependent on the Bim-initiated Bak- and Bax-dependent intrinsic apoptotic pathway. The antiapoptotic proteins Bcl-xL and Bcl-2 were dispensable for survival of Treg cells, whereas Mcl-1 was critical for survival of Treg cells, and the loss of this antiapoptotic protein caused fatal autoimmunity. Together, these data define the active processes by which Treg cells maintain homeostasis via critical survival pathways.


Subject(s)
Apoptosis/immunology , Forkhead Transcription Factors/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Apoptosis/genetics , Cell Survival/genetics , Cell Survival/immunology , Female , Forkhead Transcription Factors/genetics , Gene Deletion , Homeostasis/immunology , Interleukin-2/metabolism , Lymphocyte Count , Male , Mice , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction
16.
Immunity ; 45(3): 513-526, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27523270

ABSTRACT

The kinases RIPK1 and RIPK3 and the pseudo-kinase MLKL have been identified as key regulators of the necroptotic cell death pathway, although a role for MLKL within the whole animal has not yet been established. Here, we have shown that MLKL deficiency rescued the embryonic lethality caused by loss of Caspase-8 or FADD. Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice were viable and fertile but rapidly developed severe lymphadenopathy, systemic autoimmune disease, and thrombocytopenia. These morbidities occurred more rapidly and with increased severity in Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice compared to Casp8(-/-)Ripk3(-/-) or Fadd(-/-)Ripk3(-/-) mice, respectively. These results demonstrate that MLKL is an essential effector of aberrant necroptosis in embryos caused by loss of Caspase-8 or FADD. Furthermore, they suggest that RIPK3 and/or MLKL may exert functions independently of necroptosis. It appears that non-necroptotic functions of RIPK3 contribute to the lymphadenopathy, autoimmunity, and excess cytokine production that occur when FADD or Caspase-8-mediated apoptosis is abrogated.


Subject(s)
Apoptosis/physiology , Autoimmune Diseases/metabolism , Cell Death/physiology , Fas-Associated Death Domain Protein/metabolism , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Caspase 8/metabolism , Mice , Mice, Inbred C57BL , Necrosis/metabolism
17.
Proc Natl Acad Sci U S A ; 119(15): e2120149119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35394861

ABSTRACT

Immunological tolerance is established and maintained by a diverse array of safeguards that work together to protect against autoimmunity. Despite the identification of numerous tolerogenic processes, the basis for cooperation among them remains poorly understood. We sought to identify synergy among several well-defined tolerance mediators that alone provide protection only from mild autoimmune symptoms in C57BL/6 mice: BIM, AIRE, CBL-B, and PD-1. Survey of a range of compound mutant mice revealed that the combined loss of the autoimmune regulator, AIRE, with PD-1 unleashed a spontaneous, lethal autoimmune disease. Pdcd1−/−Aire−/− mice succumbed to cachexia before adulthood, with near-complete destruction of the exocrine pancreas. Such fatal autoimmunity was not observed in Pdcd1−/−Bim−/−, Bim−/−Aire−/−, or Cblb−/−Bim−/− mice, suggesting that the cooperation between AIRE-mediated and PD-1­mediated tolerance was particularly potent. Immune profiling revealed largely normal development of FOXP3+ regulatory T (Treg) cells in Pdcd1−/−Aire−/− mice, yet excessive, early activation of effector T cells. Adoptive transfer experiments demonstrated that autoimmune exocrine pancreatitis was driven by conventional CD4+ T cells and could not be prevented by the cotransfer of Treg cells from wild-type mice. The development of autoimmunity in mixed bone marrow chimeras supported these observations, indicating that failure of recessive tolerance was responsible for disease. These findings reveal a potent tolerogenic axis between AIRE and PD-1 that has implications for our understanding of how immune checkpoint blockade might synergize with subclinical defects in central tolerance to elicit autoimmune disease.


Subject(s)
Autoimmune Pancreatitis , Immune Tolerance , Peripheral Tolerance , Programmed Cell Death 1 Receptor , Transcription Factors , Animals , Autoimmune Pancreatitis/genetics , Autoimmune Pancreatitis/immunology , Autoimmunity/genetics , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Peripheral Tolerance/genetics , Peripheral Tolerance/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/physiology , Thymus Gland/immunology , Transcription Factors/genetics , Transcription Factors/physiology , AIRE Protein
18.
EMBO J ; 39(24): e105561, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33236795

ABSTRACT

Studies of gene-targeted mice identified the roles of the different pro-survival BCL-2 proteins during embryogenesis. However, little is known about the role(s) of these proteins in adults in response to cytotoxic stresses, such as treatment with anti-cancer agents. We investigated the role of BCL-XL in adult mice using a strategy where prior bone marrow transplantation allowed for loss of BCL-XL exclusively in non-hematopoietic tissues to prevent anemia caused by BCL-XL deficiency in erythroid cells. Unexpectedly, the combination of total body γ-irradiation (TBI) and genetic loss of Bcl-x caused secondary anemia resulting from chronic renal failure due to apoptosis of renal tubular epithelium with secondary obstructive nephropathy. These findings identify a critical protective role of BCL-XL in the adult kidney and inform on the use of BCL-XL inhibitors in combination with DNA damage-inducing drugs for cancer therapy. Encouragingly, the combination of DNA damage-inducing anti-cancer therapy plus a BCL-XL inhibitor could be tolerated in mice, at least when applied sequentially.


Subject(s)
Anemia/prevention & control , Kidney/radiation effects , bcl-X Protein/metabolism , bcl-X Protein/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11/genetics , DNA Damage , Female , Gamma Rays , Hematologic Neoplasms/pathology , Inflammation , Kidney/metabolism , Kidney/pathology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcriptome , Tumor Suppressor Proteins/genetics , bcl-X Protein/deficiency , bcl-X Protein/genetics
19.
Immunity ; 42(5): 890-902, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25979420

ABSTRACT

The mechanistic links between genetic variation and autoantibody production in autoimmune disease remain obscure. Autoimmune lymphoproliferative syndrome (ALPS) is caused by inactivating mutations in FAS or FASL, with autoantibodies thought to arise through failure of FAS-mediated removal of self-reactive germinal center (GC) B cells. Here we show that FAS is in fact not required for this process. Instead, FAS inactivation led to accumulation of a population of unconventional GC B cells that underwent somatic hypermutation, survived despite losing antigen reactivity, and differentiated into a large population of plasma cells that included autoantibody-secreting clones. IgE(+) plasma cell numbers, in particular, increased after FAS inactivation and a major cohort of ALPS-affected patients were found to have hyper-IgE. We propose that these previously unidentified cells, designated "rogue GC B cells," are a major driver of autoantibody production and provide a mechanistic explanation for the linked production of IgE and autoantibodies in autoimmune disease.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/cytology , Germinal Center/cytology , Germinal Center/immunology , Immunoglobulin E/immunology , fas Receptor/immunology , Animals , Autoantibodies/biosynthesis , B-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoglobulin E/biosynthesis , Mice , Polymerase Chain Reaction , fas Receptor/deficiency , fas Receptor/metabolism
20.
Nature ; 557(7703): 112-117, 2018 05.
Article in English | MEDLINE | ID: mdl-29695863

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC) is required for optimal gene activation and prevention of cell death upon activation of immune receptors, including TNFR1 1 . Deficiency in the LUBAC components SHARPIN or HOIP in mice results in severe inflammation in adulthood or embryonic lethality, respectively, owing to deregulation of TNFR1-mediated cell death2-8. In humans, deficiency in the third LUBAC component HOIL-1 causes autoimmunity and inflammatory disease, similar to HOIP deficiency, whereas HOIL-1 deficiency in mice was reported to cause no overt phenotype9-11. Here we show, by creating HOIL-1-deficient mice, that HOIL-1 is as essential for LUBAC function as HOIP, albeit for different reasons: whereas HOIP is the catalytically active component of LUBAC, HOIL-1 is required for LUBAC assembly, stability and optimal retention in the TNFR1 signalling complex, thereby preventing aberrant cell death. Both HOIL-1 and HOIP prevent embryonic lethality at mid-gestation by interfering with aberrant TNFR1-mediated endothelial cell death, which only partially depends on RIPK1 kinase activity. Co-deletion of caspase-8 with RIPK3 or MLKL prevents cell death in Hoil-1-/- (also known as Rbck1-/-) embryos, yet only the combined loss of caspase-8 with MLKL results in viable HOIL-1-deficient mice. Notably, triple-knockout Ripk3-/-Casp8-/-Hoil-1-/- embryos die at late gestation owing to haematopoietic defects that are rescued by co-deletion of RIPK1 but not MLKL. Collectively, these results demonstrate that both HOIP and HOIL-1 are essential LUBAC components and are required for embryogenesis by preventing aberrant cell death. Furthermore, they reveal that when LUBAC and caspase-8 are absent, RIPK3 prevents RIPK1 from inducing embryonic lethality by causing defects in fetal haematopoiesis.


Subject(s)
Carrier Proteins/metabolism , Cell Death , Embryonic Development , Hematopoiesis , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Caspase 8/genetics , Caspase 8/metabolism , Cell Death/genetics , Embryo Loss/genetics , Embryonic Development/genetics , Endothelial Cells/cytology , Female , Hematopoiesis/genetics , Mice , Mice, Inbred C57BL , Protein Domains , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL