Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Science ; 384(6694): 428-437, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38662827

ABSTRACT

A role for vitamin D in immune modulation and in cancer has been suggested. In this work, we report that mice with increased availability of vitamin D display greater immune-dependent resistance to transplantable cancers and augmented responses to checkpoint blockade immunotherapies. Similarly, in humans, vitamin D-induced genes correlate with improved responses to immune checkpoint inhibitor treatment as well as with immunity to cancer and increased overall survival. In mice, resistance is attributable to the activity of vitamin D on intestinal epithelial cells, which alters microbiome composition in favor of Bacteroides fragilis, which positively regulates cancer immunity. Our findings indicate a previously unappreciated connection between vitamin D, microbial commensal communities, and immune responses to cancer. Collectively, they highlight vitamin D levels as a potential determinant of cancer immunity and immunotherapy success.


Subject(s)
Bacteroides fragilis , Gastrointestinal Microbiome , Immune Checkpoint Inhibitors , Neoplasms , Vitamin D , Animals , Female , Humans , Male , Mice , Bacteroides fragilis/metabolism , Gastrointestinal Microbiome/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/microbiology , Neoplasms/therapy , Vitamin D/administration & dosage , Vitamin D/metabolism , Diet , Cell Line, Tumor , Calcifediol/administration & dosage , Calcifediol/metabolism , Vitamin D-Binding Protein/genetics , Vitamin D-Binding Protein/metabolism
2.
Cell Rep ; 42(12): 113506, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38019655

ABSTRACT

Cross-presentation of dead cell-associated antigens by conventional dendritic cells type 1 (cDC1s) is critical for CD8+ T cells response against many tumors and viral infections. It is facilitated by DNGR-1 (CLEC9A), an SYK-coupled cDC1 receptor that detects dead cell debris. Here, we report that DNGR-1 engagement leads to rapid activation of CBL and CBL-B E3 ligases to cause K63-linked ubiquitination of SYK and terminate signaling. Genetic deletion of CBL E3 ligases or charge-conserved mutation of target lysines within SYK abolishes SYK ubiquitination and results in enhanced DNGR-1-dependent antigen cross-presentation. We also find that cDC1 deficient in CBL E3 ligases are more efficient at cross-priming CD8+ T cells to dead cell-associated antigens and promoting host resistance to tumors. Our findings reveal a role for CBL-dependent ubiquitination in limiting cross-presentation of dead cell-associated antigens and highlight an axis of negative regulation of cDC1 activity that could be exploited to increase anti-tumor immunity.


Subject(s)
Cross-Priming , Ubiquitin-Protein Ligases , CD8-Positive T-Lymphocytes , Proto-Oncogene Proteins c-cbl , Ubiquitination , Dendritic Cells , Syk Kinase
3.
Cell Rep ; 42(8): 112881, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37523265

ABSTRACT

Conventional dendritic cells (cDCs) are found in most tissues and play a key role in initiation of immunity. cDCs require constant replenishment from progenitors called pre-cDCs that develop in the bone marrow (BM) and enter the blood circulation to seed all tissues. This process can be markedly accelerated in response to inflammation (emergency cDCpoiesis). Here, we identify two populations of BM pre-cDC marked by differential expression of CXCR4. We show that CXCR4lo cells constitute the migratory pool of BM pre-cDCs, which exits the BM and can be rapidly mobilized during challenge. We further show that exit of CXCR4lo pre-cDCs from BM at steady state is partially dependent on CCR2 and that CCR2 upregulation in response to type I IFN receptor signaling markedly increases efflux during infection with influenza A virus. Our results highlight a fine balance between retention and efflux chemokine cues that regulates steady-state and emergency cDCpoiesis.


Subject(s)
Bone Marrow , Dendritic Cells , Receptors, CCR2 , Receptors, CXCR4 , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Dendritic Cells/metabolism , Inflammation/metabolism , Receptors, CCR2/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Animals
4.
J Immunother Cancer ; 10(9)2022 09.
Article in English | MEDLINE | ID: mdl-36162919

ABSTRACT

Type 1 conventional dendritic cells (cDC1) play a critical role in priming anticancer cytotoxic CD8+ T cells. DNGR-1 (a.k.a. CLEC9A) is a cDC1 receptor that binds to F-actin exposed on necrotic cancer and normal cells. DNGR-1 signaling enhances cross-presentation of dead-cell associated antigens, including tumor antigens. We have recently shown that secreted gelsolin (sGSN), a plasma protein, competes with DNGR-1 for binding to dead cell-exposed F-actin and dampens anticancer immunity. Here, we investigated the effects of loss of sGSN on various anticancer therapies that are thought to induce cell death and provoke an immune response to cancer. We compared WT (wildtype) with Rag1-/- , Batf3-/- , Clec9agfp/gfp , sGsn-/- or sGsn-/- Clec9agfp/gfp mice implanted with transplantable tumor cell lines, including MCA-205 fibrosarcoma, 5555 BrafV600E melanoma and B16-F10 LifeAct (LA)-ovalbumin (OVA)-mCherry melanoma. Tumor-bearing mice were treated with (1) doxorubicin (intratumoral) chemotherapy for MCA-205, (2) BRAF-inhibitor PLX4720 (oral gavage) targeted therapy for 5555 BrafV600E, and (3) X-ray radiotherapy for B16 LA-OVA-mCherry. We confirmed that efficient tumor control following each therapy requires an immunocompetent host as efficacy was markedly reduced in Rag1-/- compared with WT mice. Notably, across all the therapeutic modalities, loss of sGSN significantly enhanced tumor control compared with treated WT controls. This was an on-target effect as mice deficient in both sGSN and DNGR-1 behaved no differently from WT mice following therapy. In sum, we find that mice deficient in sGsn display enhanced DNGR-1-dependent responsiveness to chemotherapy, targeted therapy and radiotherapy. Our findings are consistent with the notion some cancer therapies induce immunogenic cell death (ICD), which mobilizes anticancer T cells. Our results point to cDC1 and DNGR-1 as decoders of ICD and to sGSN as a negative regulator of such decoding, highlighting sGSN as a possible target in cancer treatment. Further prospective studies are warranted to identify patients who may benefit most from inhibition of sGSN function.


Subject(s)
Gelsolin , Melanoma, Experimental , Actins/metabolism , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes , Doxorubicin/metabolism , Gelsolin/genetics , Gelsolin/metabolism , Homeodomain Proteins , Lectins, C-Type , Mice , Ovalbumin , Proto-Oncogene Proteins B-raf/metabolism , Receptors, Immunologic/metabolism
5.
Cell ; 184(15): 4016-4031.e22, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34081922

ABSTRACT

Cross-presentation of antigens from dead tumor cells by type 1 conventional dendritic cells (cDC1s) is thought to underlie priming of anti-cancer CD8+ T cells. cDC1 express high levels of DNGR-1 (a.k.a. CLEC9A), a receptor that binds to F-actin exposed by dead cell debris and promotes cross-presentation of associated antigens. Here, we show that secreted gelsolin (sGSN), an extracellular protein, decreases DNGR-1 binding to F-actin and cross-presentation of dead cell-associated antigens by cDC1s. Mice deficient in sGsn display increased DNGR-1-dependent resistance to transplantable tumors, especially ones expressing neoantigens associated with the actin cytoskeleton, and exhibit greater responsiveness to cancer immunotherapy. In human cancers, lower levels of intratumoral sGSN transcripts, as well as presence of mutations in proteins associated with the actin cytoskeleton, are associated with signatures of anti-cancer immunity and increased patient survival. Our results reveal a natural barrier to cross-presentation of cancer antigens that dampens anti-tumor CD8+ T cell responses.


Subject(s)
Cross-Priming/immunology , Gelsolin/metabolism , Immunity , Lectins, C-Type/metabolism , Neoplasms/immunology , Receptors, Immunologic/metabolism , Receptors, Mitogen/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cross-Priming/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gelsolin/chemistry , Gelsolin/deficiency , Gene Expression Regulation, Neoplastic/drug effects , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunity/drug effects , Mice, Inbred C57BL , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Protein Binding/drug effects , Survival Analysis
6.
Nat Commun ; 11(1): 5387, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33144593

ABSTRACT

The Human Silencing Hub (HUSH) complex is necessary for epigenetic repression of LINE-1 elements. We show that HUSH-depletion in human cell lines and primary fibroblasts leads to induction of interferon-stimulated genes (ISGs) through JAK/STAT signaling. This effect is mainly attributed to MDA5 and RIG-I sensing of double-stranded RNAs (dsRNAs). This coincides with upregulation of primate-conserved LINE-1s, as well as increased expression of full-length hominid-specific LINE-1s that produce bidirectional RNAs, which may form dsRNA. Notably, LTRs nearby ISGs are derepressed likely rendering these genes more responsive to interferon. LINE-1 shRNAs can abrogate the HUSH-dependent response, while overexpression of an engineered LINE-1 construct activates interferon signaling. Finally, we show that the HUSH component, MPP8 is frequently downregulated in diverse cancers and that its depletion leads to DNA damage. These results suggest that LINE-1s may drive physiological or autoinflammatory responses through dsRNA sensing and gene-regulatory roles and are controlled by the HUSH complex.


Subject(s)
Epigenesis, Genetic/physiology , Gene Expression Regulation, Neoplastic , Gene Silencing/physiology , Interferon Type I/metabolism , Long Interspersed Nucleotide Elements/physiology , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , DNA Damage , Down-Regulation , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Inflammation , Interferon-Induced Helicase, IFIH1/metabolism , Long Interspersed Nucleotide Elements/genetics , Phosphoproteins/metabolism , RNA, Double-Stranded , Receptors, Immunologic , Sequence Analysis, RNA , Signal Transduction
7.
Immunity ; 50(3): 645-654.e6, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30770250

ABSTRACT

The epidermal growth factor receptor ligand Amphiregulin has a well-documented role in the restoration of tissue homeostasis after injury; however, the mechanism by which Amphiregulin contributes to wound repair remains unknown. Here we show that Amphiregulin functioned by releasing bioactive transforming growth factor beta (TGF-ß) from latent complexes via integrin-αV activation. Using acute injury models in two different tissues, we found that by inducing TGF-ß activation on mesenchymal stromal cells (pericytes), Amphiregulin induced their differentiation into myofibroblasts, thereby selectively contributing to the restoration of vascular barrier function within injured tissue. Furthermore, we identified macrophages as a critical source of Amphiregulin, revealing a direct effector mechanism by which these cells contribute to tissue restoration after acute injury. Combined, these observations expose a so far under-appreciated mechanism of how cells of the immune system selectively control the differentiation of tissue progenitor cells during tissue repair and inflammation.


Subject(s)
Amphiregulin/metabolism , Macrophages/metabolism , Pericytes/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/physiology , Female , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism
9.
Nat Cell Biol ; 19(9): 1116-1129, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28846096

ABSTRACT

Apoptosis represents a key anti-cancer therapeutic effector mechanism. During apoptosis, mitochondrial outer membrane permeabilization (MOMP) typically kills cells even in the absence of caspase activity. Caspase activity can also have a variety of unwanted consequences that include DNA damage. We therefore investigated whether MOMP-induced caspase-independent cell death (CICD) might be a better way to kill cancer cells. We find that cells undergoing CICD display potent pro-inflammatory effects relative to apoptosis. Underlying this, MOMP was found to stimulate NF-κB activity through the downregulation of inhibitor of apoptosis proteins. Strikingly, engagement of CICD displays potent anti-tumorigenic effects, often promoting complete tumour regression in a manner dependent on intact immunity. Our data demonstrate that by activating NF-κB, MOMP can exert additional signalling functions besides triggering cell death. Moreover, they support a rationale for engaging caspase-independent cell death in cell-killing anti-cancer therapies.


Subject(s)
Caspases/metabolism , Colonic Neoplasms/enzymology , Inflammation Mediators/metabolism , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , NF-kappa B/metabolism , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Genotype , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , Macrophage Activation , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/pathology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/immunology , Mitochondrial Membranes/pathology , NF-kappa B/deficiency , Necrosis , Permeability , Phenotype , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Sulfonamides/pharmacology , Time Factors , Transfection , Tumor Necrosis Factor-alpha/metabolism , NF-kappaB-Inducing Kinase
10.
Curr Biol ; 27(5): 624-637, 2017 Mar 06.
Article in English | MEDLINE | ID: mdl-28238662

ABSTRACT

The individual molecular pathways downstream of Cdc42, Rac, and Rho GTPases are well documented, but we know surprisingly little about how these pathways are coordinated when cells move in a complex environment in vivo. In the developing embryo, melanoblasts originating from the neural crest must traverse the dermis to reach the epidermis of the skin and hair follicles. We previously established that Rac1 signals via Scar/WAVE and Arp2/3 to effect pseudopod extension and migration of melanoblasts in skin. Here we show that RhoA is redundant in the melanocyte lineage but that Cdc42 coordinates multiple motility systems independent of Rac1. Similar to Rac1 knockouts, Cdc42 null mice displayed a severe loss of pigmentation, and melanoblasts showed cell-cycle progression, migration, and cytokinesis defects. However, unlike Rac1 knockouts, Cdc42 null melanoblasts were elongated and displayed large, bulky pseudopods with dynamic actin bursts. Despite assuming an elongated shape usually associated with fast mesenchymal motility, Cdc42 knockout melanoblasts migrated slowly and inefficiently in the epidermis, with nearly static pseudopods. Although much of the basic actin machinery was intact, Cdc42 null cells lacked the ability to polarize their Golgi and coordinate motility systems for efficient movement. Loss of Cdc42 de-coupled three main systems: actin assembly via the formin FMNL2 and Arp2/3, active myosin-II localization, and integrin-based adhesion dynamics.


Subject(s)
Actins/metabolism , Cell Adhesion , Cell Movement , Melanocytes/metabolism , cdc42 GTP-Binding Protein/genetics , Animals , Cell Lineage , Mice/embryology , Neuropeptides/genetics , Neuropeptides/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
11.
Nat Commun ; 7: 10538, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26833356

ABSTRACT

Most apoptotic stimuli require mitochondrial outer membrane permeabilization (MOMP) in order to execute cell death. As such, MOMP is subject to tight control by Bcl-2 family proteins. We have developed a powerful new technique to investigate Bcl-2-mediated regulation of MOMP. This method, called mito-priming, uses co-expression of pro- and anti-apoptotic Bcl-2 proteins to engineer Bcl-2 addiction. On addition of Bcl-2 targeting BH3 mimetics, mito-primed cells undergo apoptosis in a rapid and synchronous manner. Using this method we have comprehensively surveyed the efficacy of BH3 mimetic compounds, identifying potent and specific MCL-1 inhibitors. Furthermore, by combining different pro- and anti-apoptotic Bcl-2 pairings together with CRISPR/Cas9-based genome editing, we find that tBID and PUMA can preferentially kill in a BAK-dependent manner. In summary, mito-priming represents a facile and robust means to trigger mitochondrial apoptosis.


Subject(s)
Mitochondrial Membranes/physiology , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CRISPR-Cas Systems , Cell Line , Gene Expression Regulation , Genetic Engineering , Humans , Mice , Proto-Oncogene Proteins c-bcl-2/genetics
12.
FEBS J ; 283(5): 803-14, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26607558

ABSTRACT

Mitochondria have traditionally been viewed as the powerhouse of the cell, where they serve, amongst other functions, as a major source of ATP generation. More recently, mitochondria have also been shown to have active roles in a variety of other processes, including apoptotic cell death and inflammation. Here we review the various ways in which mitochondrial functions affect cancer. Although there are many diverse types of cancer, hallmarks have been defined that are applicable to most cancer types. We provide an overview of how mitochondrial functions affect some of these hallmarks, which include evasion of cell death, de-regulated bioenergetics, genome instability, tumour-promoting inflammation and metastasis. In addition to discussing the underlying mitochondrial roles in each of these processes, we also highlight the considerable potential of targeting mitochondrial functions to improve cancer treatment.


Subject(s)
Gene Expression Regulation, Neoplastic , Mitochondria/metabolism , Neoplasms/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Cell Death , Energy Metabolism , Genes, Tumor Suppressor , Genome , Humans , Hypoxia , Inflammation , Mice , Mutation , Neoplasm Metastasis , Neoplasms/pathology , Oxygen/metabolism , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism
13.
Mol Cell ; 57(5): 860-872, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25702873

ABSTRACT

During apoptosis, the mitochondrial outer membrane is permeabilized, leading to the release of cytochrome c that activates downstream caspases. Mitochondrial outer membrane permeabilization (MOMP) has historically been thought to occur synchronously and completely throughout a cell, leading to rapid caspase activation and apoptosis. Using a new imaging approach, we demonstrate that MOMP is not an all-or-nothing event. Rather, we find that a minority of mitochondria can undergo MOMP in a stress-regulated manner, a phenomenon we term "minority MOMP." Crucially, minority MOMP leads to limited caspase activation, which is insufficient to trigger cell death. Instead, this caspase activity leads to DNA damage that, in turn, promotes genomic instability, cellular transformation, and tumorigenesis. Our data demonstrate that, in contrast to its well-established tumor suppressor function, apoptosis also has oncogenic potential that is regulated by the extent of MOMP. These findings have important implications for oncogenesis following either physiological or therapeutic engagement of apoptosis.


Subject(s)
Apoptosis/physiology , DNA Damage , Genomic Instability , Mitochondrial Membranes/physiology , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p19/deficiency , Cyclin-Dependent Kinase Inhibitor p19/genetics , Dose-Response Relationship, Drug , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , HCT116 Cells , HeLa Cells , Histones/metabolism , Humans , MCF-7 Cells , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Nitrophenols/pharmacology , Permeability , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Staurosporine/pharmacology , Sulfonamides/pharmacology , Time Factors
14.
Autophagy ; 10(12): 2269-78, 2014.
Article in English | MEDLINE | ID: mdl-25629932

ABSTRACT

During macroautophagy, conjugation of ATG12 to ATG5 is essential for LC3 lipidation and autophagosome formation. Additionally, ATG12 has ATG5-independent functions in diverse processes including mitochondrial fusion and mitochondrial-dependent apoptosis. In this study, we investigated the regulation of free ATG12. In stark contrast to the stable ATG12-ATG5 conjugate, we find that free ATG12 is highly unstable and rapidly degraded in a proteasome-dependent manner. Surprisingly, ATG12, itself a ubiquitin-like protein, is directly ubiquitinated and this promotes its proteasomal degradation. As a functional consequence of its turnover, accumulation of free ATG12 contributes to proteasome inhibitor-mediated apoptosis, a finding that may be clinically important given the use of proteasome inhibitors as anticancer agents. Collectively, our results reveal a novel interconnection between autophagy, proteasome activity, and cell death mediated by the ubiquitin-like properties of ATG12.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Mitochondria/metabolism , Proteasome Endopeptidase Complex/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitination/physiology , Animals , Autophagy-Related Protein 12 , Cell Death/physiology , Humans , Mice , Microtubule-Associated Proteins/metabolism , Protein Processing, Post-Translational/physiology , Ubiquitin-Conjugating Enzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...