Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Cancer ; 5(4): 659-672, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38286828

ABSTRACT

The mitochondrial genome (mtDNA) encodes essential machinery for oxidative phosphorylation and metabolic homeostasis. Tumor mtDNA is among the most somatically mutated regions of the cancer genome, but whether these mutations impact tumor biology is debated. We engineered truncating mutations of the mtDNA-encoded complex I gene, Mt-Nd5, into several murine models of melanoma. These mutations promoted a Warburg-like metabolic shift that reshaped tumor microenvironments in both mice and humans, consistently eliciting an anti-tumor immune response characterized by loss of resident neutrophils. Tumors bearing mtDNA mutations were sensitized to checkpoint blockade in a neutrophil-dependent manner, with induction of redox imbalance being sufficient to induce this effect in mtDNA wild-type tumors. Patient lesions bearing >50% mtDNA mutation heteroplasmy demonstrated a response rate to checkpoint blockade that was improved by ~2.5-fold over mtDNA wild-type cancer. These data nominate mtDNA mutations as functional regulators of cancer metabolism and tumor biology, with potential for therapeutic exploitation and treatment stratification.


Subject(s)
DNA, Mitochondrial , Glycolysis , Immune Checkpoint Inhibitors , Melanoma , Mutation , DNA, Mitochondrial/genetics , Animals , Melanoma/genetics , Melanoma/drug therapy , Mice , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Glycolysis/genetics , Tumor Microenvironment , Cell Line, Tumor , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Neutrophils/metabolism , Neutrophils/immunology , Mitochondria/metabolism , Mitochondria/genetics , Oxidative Phosphorylation/drug effects
2.
Curr Biol ; 33(18): R957-R959, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37751709

ABSTRACT

A new study reports that dendritic cells actively shape the CCL19 chemokine gradient to which they respond and that the chemokine receptor CCR7 both senses CCL19 and mediates its internalisation. Generation of local changes in chemokines allows coordination of movement over longer distances than previous models could explain.


Subject(s)
Chemotaxis , Defense Mechanisms , Movement , Immunity , Dendritic Cells
3.
Sci Immunol ; 8(85): eadg8249, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37478193

ABSTRACT

T cell responses against infections and cancer are directed by conventional dendritic cells (cDCs) in lymph nodes distant from the site of challenge. Migratory cDCs, which travel from the tissue to the lymph node, not only drive initial T cell activation but also transfer antigen to lymph node-resident cDCs. These resident cells have essential roles defining the character of the resulting T cell response; however, it is unknown how they can appropriately process and present antigens to suitably direct responses given their spatial separation. Here, using a novel strain of influenza A and a modified melanoma model, we show that tissue and lymph node cDC activation is harmonized and that this is driven by cotransfer of contextual cues. In the tumor, incomplete cDC activation in the tumor microenvironment is mirrored by lymph node-resident cDCs, whereas during influenza infection, pathogen-associated molecular patterns cotransferred with antigen drive TLR signaling in resident cDCs and their subsequent robust activation. This cotransfer mechanism explains how individual antigens can be handled distinctly by resident cDCs and how signals driving poor tumoral cDC activation further impact the lymph node. Our findings clarify how tissue context dictates antigenic and, consequently, T cell fate in the lymph node.


Subject(s)
Influenza, Human , Humans , Dendritic Cells , Antigens , Lymph Nodes , T-Lymphocytes
4.
bioRxiv ; 2023 Mar 23.
Article in English | MEDLINE | ID: mdl-36993533

ABSTRACT

The mitochondrial genome encodes essential machinery for respiration and metabolic homeostasis but is paradoxically among the most common targets of somatic mutation in the cancer genome, with truncating mutations in respiratory complex I genes being most over-represented1. While mitochondrial DNA (mtDNA) mutations have been associated with both improved and worsened prognoses in several tumour lineages1-3, whether these mutations are drivers or exert any functional effect on tumour biology remains controversial. Here we discovered that complex I-encoding mtDNA mutations are sufficient to remodel the tumour immune landscape and therapeutic resistance to immune checkpoint blockade. Using mtDNA base editing technology4 we engineered recurrent truncating mutations in the mtDNA-encoded complex I gene, Mt-Nd5, into murine models of melanoma. Mechanistically, these mutations promoted utilisation of pyruvate as a terminal electron acceptor and increased glycolytic flux without major effects on oxygen consumption, driven by an over-reduced NAD pool and NADH shuttling between GAPDH and MDH1, mediating a Warburg-like metabolic shift. In turn, without modifying tumour growth, this altered cancer cell-intrinsic metabolism reshaped the tumour microenvironment in both mice and humans, promoting an anti-tumour immune response characterised by loss of resident neutrophils. This subsequently sensitised tumours bearing high mtDNA mutant heteroplasmy to immune checkpoint blockade, with phenocopy of key metabolic changes being sufficient to mediate this effect. Strikingly, patient lesions bearing >50% mtDNA mutation heteroplasmy also demonstrated a >2.5-fold improved response rate to checkpoint inhibitor blockade. Taken together these data nominate mtDNA mutations as functional regulators of cancer metabolism and tumour biology, with potential for therapeutic exploitation and treatment stratification.

5.
Int J Biochem Cell Biol ; 147: 106227, 2022 06.
Article in English | MEDLINE | ID: mdl-35605877

ABSTRACT

Despite their low abundance in tumours conventional dendritic cells play an outsized role in initiating and perpetuating anti-tumour immunity; however progressively growing tumours suppress dendritic cell function in a range of ways preventing effective anti-tumour T cell responses. While the success of immune checkpoint blockade has focused attention on T-cell directed therapies, activating tumour dendritic cells has been shown to be critical for the efficacy of several immunotherapies and other conventional therapies owing to their ability to activate and restimulate anti-tumour T-cells. As such, the importance of understanding the mechanisms by which dendritic cell function is impaired are being investigated further. Yet, while much attention has been paid to the tumour microenvironment less has been given to the macroenvironment including effects in the bone marrow and the lymph node. It is now clear that dendritic cell function can be impaired in a variety of ways at different anatomical sites and understanding these mechanisms will be critical for developing effective strategies to tune the dendritic cell response in cancer.


Subject(s)
Dendritic Cells , Neoplasms , Humans , Immunotherapy , Lymph Nodes , Neoplasms/therapy , Tumor Microenvironment
6.
Mucosal Immunol ; 15(6): 1257-1269, 2022 06.
Article in English | MEDLINE | ID: mdl-35428872

ABSTRACT

Type 2 immunity is activated in response to both allergens and helminth infection. It can be detrimental or beneficial, and there is a pressing need to better understand its regulation. The immunosuppressive cytokine IL-10 is known as a T helper 2 (Th2) effector molecule, but it is currently unclear whether IL-10 dampens or promotes Th2 differentiation during infection. Here we show that helminth infection in mice elicits IL-10 expression in both the intestinal lamina propria and the draining mesenteric lymph node, with higher expression in the infected tissue. In vitro, exogenous IL-10 enhanced Th2 differentiation in isolated CD4+ T cells, increasing expression of GATA3 and production of IL-5 and IL-13. The ability of IL-10 to amplify the Th2 response coincided with its suppression of IFNγ expression and in vivo we found that, in intestinal helminth infection, IL-10 receptor expression was higher on Th1 cells in the small intestine than on Th2 cells in the same tissue, or on any Th cell in the draining lymph node. In vivo blockade of IL-10 signalling during helminth infection resulted in an expansion of IFNγ+ and Tbet+ Th1 cells in the small intestine and a coincident decrease in IL-13, IL-5 and GATA3 expression by intestinal T cells. These changes in Th2 cytokines correlated with reduced expression of type 2 effector molecules, such as RELMα, and increased parasite egg production. Together our data indicate that IL-10 signalling promotes Th2 differentiation during helminth infection at least in part by regulating competing Th1 cells in the infected tissue.


Subject(s)
Helminths , Interleukin-13 , Mice , Animals , Interleukin-13/metabolism , Interleukin-10/metabolism , Interleukin-5/metabolism , Th2 Cells , Th1 Cells , Interferon-gamma/metabolism , Cytokines/metabolism
7.
Cancer Immunol Immunother ; 69(6): 1015-1027, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32088771

ABSTRACT

Oncolytic virus (OV) therapy is an emerging approach with the potential to redefine treatment options across a range of cancer indications and in patients who remain resistant to existing standards of care, including immuno-oncology (IO) drugs. MEDI5395, a recombinant Newcastle disease virus (NDV), engineered to express granulocyte-macrophage colony-stimulating factor (GM-CSF), exhibits potent oncolytic activity. It was hypothesized that activation of immune cells by MEDI5395, coupled with its oncolytic activity, would enhance the priming of antitumor immunity. Using MEDI5395 and recombinant NDVs encoding fluorescent reporter genes, we demonstrated preferential virus uptake and non-productive infection in myeloid cells, including monocytes, macrophages, and dendritic cells (DCs). Infection resulted in immune-cell activation, with upregulation of cell surface activation markers (e.g., CD80, PD-L1, HLA-DR) and secretion of proinflammatory cytokines (IFN-α2a, IL-6, IL-8, TNF-α). Interestingly, in vitro M2-polarized macrophages were more permissive to virus infection than were M1-polarized macrophages. In a co-culture system, infected myeloid cells were effective virus vectors and mediated the transfer of infectious NDV particles to tumor cells, resulting in cell death. Furthermore, NDV-infected DCs stimulated greater proliferation of allogeneic T cells than uninfected DCs. Antigens released after NDV-induced tumor cell lysis were cross-presented by DCs and drove activation of tumor antigen-specific autologous T cells. MEDI5395 therefore exhibited potent immunostimulatory activity and an ability to enhance antigen-specific T-cell priming. This, coupled with its tumor-selective oncolytic capacity, underscores the promise of MEDI5395 as a multimodal therapeutic, with potential to both enhance current responding patient populations and elicit de novo responses in resistant patients.


Subject(s)
Newcastle disease virus/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Cell Line, Tumor , Genetic Vectors , Humans , Immunity, Innate
8.
J Immunol Methods ; 477: 112702, 2020 02.
Article in English | MEDLINE | ID: mdl-31705860

ABSTRACT

The use of helminth infections as tools to understand the type 2 immune response is a well-established technique and important to many areas of immunological research. The phenotype and function of immune cell populations at the site of infection is a key determinant of pathogen clearance. However, infections with helminths such as the murine nematode Heligomosmoides polygryrus cause increased mucus production and thickening of the intestinal wall, which can result in extensive cell death when isolating and analysing cells from the lamina propria (LP). Populations of larger immune cells such as macrophages and dendritic cells are often trapped within mucus or dying tissues. Here we describe an optimised protocol for isolating LP leukocytes from the small intestine of H.polygyrus -infected mice, and we demonstrate phenotypic and functional identification of myeloid and CD4+ T cell subsets using cytokine staining and flow cytometry. Our protocol may provide a useful experimental method for the immunological analysis of the affected tissue site during helminth infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Separation/methods , Intestinal Diseases, Parasitic/immunology , Intestinal Mucosa/cytology , Strongylida Infections/immunology , Adaptive Immunity , Animals , CD4-Positive T-Lymphocytes/metabolism , Cytokines/chemistry , Cytokines/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Female , Flow Cytometry/methods , Intestinal Diseases, Parasitic/parasitology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestine, Small/cytology , Intestine, Small/immunology , Intestine, Small/parasitology , Macrophages/immunology , Mice , Nematospiroides dubius/immunology , Staining and Labeling/methods , Strongylida Infections/parasitology
9.
Pharmacol Res ; 145: 104258, 2019 07.
Article in English | MEDLINE | ID: mdl-31063806

ABSTRACT

Greater understanding of tumour immunobiology has led to a new era of cancer treatment in which immuno-oncology (IO) therapies are used to boost anti-cancer immune responses. Prominent among these therapies are immune checkpoint inhibitors (ICIs), antibody-based drugs that can unleash the power of tumour-specific CD8 + T-cells. ICIs targeting the Programmed cell death protein 1 (PD-1) cell surface receptor or its ligand PD-L1 are particularly effective, with clinical studies reporting powerful and durable therapeutic impact against many cancer types, including melanoma and non-small cell lung cancer. ICIs have the potential to transform the landscape of cancer treatment, and their development was recognised by the award of the 2018 Nobel Prize in Physiology or Medicine to James Allison and Tasuku Honjo. However, the proportion of patients responding to anti-PD-1/PD-L1 monotherapy can be low. The next major challenge involves understanding and overcoming the innate and acquired resistance that prevents most patients from responding to PD-1/PD-L1 blockade. In this review, we outline the physiological function of PD-1 and its exploitation by developing tumours. We give an overview of current FDA-approved drugs targeting PD-1 or PD-L1 and summarise clinical progress so far. We then discuss key mechanisms thought to underpin resistance to PD-1/PD-L1 blockade, describing biomarkers that could allow patient responses to be predicted before treatment, and tracked once treatment has started. We also present clinical and pre-clinical combination therapies that have been developed to overcome resistance and which have the potential to substantially extend the therapeutic reach of these revolutionary drugs.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , B7-H1 Antigen/immunology , Humans , Immune Evasion/drug effects , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...