Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Annu Rev Immunol ; 32: 51-82, 2014.
Article in English | MEDLINE | ID: mdl-24313777

ABSTRACT

The cytokine TGF-ß plays an integral role in regulating immune responses. TGF-ß has pleiotropic effects on adaptive immunity, especially in the regulation of effector and regulatory CD4(+) T cell responses. Many immune and nonimmune cells can produce TGF-ß, but it is always produced as an inactive complex that must be activated to exert functional effects. Thus, activation of latent TGF-ß provides a crucial layer of regulation that controls TGF-ß function. In this review, we highlight some of the important functional roles for TGF-ß in immunity, focusing on its context-specific roles in either dampening or promoting T cell responses. We also describe how activation of TGF-ß controls its function in the immune system, with a focus on the key roles for members of the integrin family in this process.


Subject(s)
Immunity/physiology , Transforming Growth Factor beta/metabolism , Adaptive Immunity , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immune System/physiology , Integrins/metabolism , Protein Binding , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transforming Growth Factor beta/chemistry
2.
Immunity ; 44(4): 860-74, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27067057

ABSTRACT

The role of dendritic cells (DCs) in intestinal immune homeostasis remains incompletely defined. Here we show that mice lacking IRF8 transcription-factor-dependent DCs had reduced numbers of T cells in the small intestine (SI), but not large intestine (LI), including an almost complete absence of SI CD8αß(+) and CD4(+)CD8αα(+) T cells; the latter requiring ß8 integrin expression by migratory IRF8 dependent CD103(+)CD11b(-) DCs. SI homing receptor induction was impaired during T cell priming in mesenteric lymph nodes (MLN), which correlated with a reduction in aldehyde dehydrogenase activity by SI-derived MLN DCs, and inefficient T cell localization to the SI. These mice also lacked intestinal T helper 1 (Th1) cells, and failed to support Th1 cell differentiation in MLN and mount Th1 cell responses to Trichuris muris infection. Collectively these results highlight multiple non-redundant roles for IRF8 dependent DCs in the maintenance of intestinal T cell homeostasis.


Subject(s)
Dendritic Cells/immunology , Homeostasis/immunology , Interferon Regulatory Factors/metabolism , Intestines/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Aldehyde Dehydrogenase/metabolism , Animals , Antigen Presentation/immunology , CD11 Antigens/genetics , CD8 Antigens/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Cells, Cultured , Integrin alpha Chains/genetics , Integrin beta Chains/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestines/cytology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/cytology , Trichuris/immunology
3.
J Immunol ; 210(5): 618-627, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36602520

ABSTRACT

Innate lymphoid cells (ILCs) are capable of rapid response to a wide variety of immune challenges, including various respiratory pathogens. Despite this, their role in the immune response against the lethal intracellular bacterium Francisella tularensis is not yet known. In this study, we demonstrate that infection of the airways with F. tularensis results in a significant reduction in lung type 2 ILCs (ILC2s) in mice. Conversely, the expansion of ILC2s via treatment with the cytokine IL-33, or by adoptive transfer of ILC2s, resulted in significantly enhanced bacterial burdens in the lung, liver, and spleen, suggesting that ILC2s may favor severe infection. Indeed, specific reduction of ILC2s in a transgenic mouse model results in a reduction in lung bacterial burden. Using an in vitro culture system, we show that IFN-γ from the live vaccine strain-infected lung reduces ILC2 numbers, suggesting that this cytokine in the lung environment is mechanistically important in reducing ILC2 numbers during infection. Finally, we show Ab-mediated blockade of IL-5, of which ILC2s are a major innate source, reduces bacterial burden postinfection, suggesting that IL-5 production by ILC2s may play a role in limiting protective immunity. Thus, overall, we highlight a negative role for ILC2s in the control of infection with F. tularensis. Our work therefore highlights the role of ILC2s in determining the severity of potentially fatal airway infections and raises the possibility of interventions targeting innate immunity during infection with F. tularensis to benefit the host.


Subject(s)
Francisella tularensis , Animals , Mice , Immunity, Innate , Lymphocytes , Interleukin-5 , Cytokines
4.
Immunity ; 42(5): 903-15, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25979421

ABSTRACT

Regulatory T (Treg) cells play a pivotal role in suppressing self-harmful T cell responses, but how Treg cells mediate suppression to maintain immune homeostasis and limit responses during inflammation is unclear. Here we show that effector Treg cells express high amounts of the integrin αvß8, which enables them to activate latent transforming growth factor-ß (TGF-ß). Treg-cell-specific deletion of integrin αvß8 did not result in a spontaneous inflammatory phenotype, suggesting that this pathway is not important in Treg-cell-mediated maintenance of immune homeostasis. However, Treg cells lacking expression of integrin αvß8 were unable to suppress pathogenic T cell responses during active inflammation. Thus, our results identify a mechanism by which Treg cells suppress exuberant immune responses, highlighting a key role for effector Treg-cell-mediated activation of latent TGF-ß in suppression of self-harmful T cell responses during active inflammation.


Subject(s)
Inflammation/immunology , Integrins/metabolism , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Cell Proliferation , Colitis/immunology , Disease Models, Animal , Gene Expression Regulation/immunology , Humans , Inflammation Mediators/immunology , Integrins/genetics , Mice , Models, Immunological , T-Lymphocytes, Regulatory/cytology
7.
PLoS Pathog ; 15(4): e1007657, 2019 04.
Article in English | MEDLINE | ID: mdl-30998782

ABSTRACT

Helminths are highly prevalent metazoan parasites that infect over a billion of the world's population. Hosts have evolved numerous mechanisms to drive the expulsion of these parasites via Th2-driven immunity, but these responses must be tightly controlled to prevent equally devastating immunopathology. However, mechanisms that regulate this balance are still unclear. Here we show that the vigorous Th2 immune response driven by the small intestinal helminth Trichinella spiralis, is associated with increased TGFß signalling responses in CD4+ T-cells. Mechanistically, enhanced TGFß signalling in CD4+ T-cells is dependent on dendritic cell-mediated TGFß activation which requires expression of the integrin αvß8. Importantly, mice lacking integrin αvß8 on DCs had a delayed ability to expel a T. spiralis infection, indicating an important functional role for integrin αvß8-mediated TGFß activation in promoting parasite expulsion. In addition to maintaining regulatory T-cell responses, the CD4+ T-cell signalling of this pleiotropic cytokine induces a Th17 response which is crucial in promoting the intestinal muscle hypercontractility that drives worm expulsion. Collectively, these results provide novel insights into intestinal helminth expulsion beyond that of classical Th2 driven immunity, and highlight the importance of IL-17 in intestinal contraction which may aid therapeutics to numerous diseases of the intestine.


Subject(s)
Dendritic Cells/immunology , Intestine, Small/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/parasitology , Intestine, Small/parasitology , Male , Mice , Mice, Inbred C57BL , Th17 Cells/parasitology , Trichinellosis/parasitology
8.
Immunology ; 160(2): 139-148, 2020 06.
Article in English | MEDLINE | ID: mdl-31792952

ABSTRACT

Transforming growth factor ß (TGF-ß) is a multifunctional cytokine that regulates cell growth, differentiation, adhesion, migration and death dependent on cell type, developmental stage, or tissue conditions. Various cell types secrete TGF-ß, but always as an inactive complex. Hence, for TGF-ß to function, this latent complex must somehow be activated. Work in recent years has highlighted a critical role for members of the αv integrin family, including αv ß1 , αv ß3 , αv ß5 , αv ß6 and αv ß8 that are involved in TGF-ß activation in various contexts, particularly at barrier sites such as the gut, lung and skin. The integrins facilitating this context- and location-specific regulation can be dysregulated in certain diseases, so are potential therapeutic targets in a number of disorders. In this review, we discuss the role of TGF-ß at these barrier sites with a focus on how integrin-mediated TGF-ß activation regulates tissue and immune homeostasis, and how this is altered in disease.


Subject(s)
Homeostasis/immunology , Integrin alphaV/metabolism , Signal Transduction/immunology , Transforming Growth Factor beta/metabolism , Animals , Disease Models, Animal , Gastrointestinal Microbiome/immunology , Humans , Intestinal Diseases/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lung/immunology , Lung/metabolism , Lung Diseases/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Skin/immunology , Skin/metabolism , Skin/microbiology , Skin Diseases/immunology
9.
J Biol Chem ; 293(22): 8543-8553, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29581231

ABSTRACT

Cross-talk between different components of the intestinal barrier and the immune system may be important in maintaining gut homeostasis. A crucial part of the gut barrier is the mucus layer, a cross-linked gel on top of the intestinal epithelium that consists predominantly of the mucin glycoprotein MUC2. However, whether the mucin layer actively regulates intestinal immune cell responses is not clear. Because recent evidence suggests that intestinal dendritic cells (DCs) may be regulated by the mucus layer, we purified intestinal mucin, incubated it with human DCs, and determined the functional effects. Here we show that expression of the chemokine IL-8 and co-stimulatory DC markers CD86 and CD83 are significantly up-regulated on human DCs in the presence of intestinal mucins. Additionally, mucin-exposed DCs promoted neutrophil migration in an IL-8-dependent manner. The stimulatory effects of mucins on DCs were not due to mucin sample contaminants such as lipopolysaccharide, DNA, or contaminant proteins. Instead, mucin glycans are important for the pro-inflammatory effects on DCs. Thus, intestinal mucins are capable of inducing important pro-inflammatory functions in DCs, which could be important in driving inflammatory responses upon intestinal barrier damage.


Subject(s)
Dendritic Cells/metabolism , Interleukin-8/metabolism , Intestinal Mucosa/metabolism , Mucins/pharmacology , Polysaccharides/chemistry , Animals , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Glycosylation , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Polysaccharides/metabolism
10.
J Immunol ; 199(8): 2652-2667, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28904128

ABSTRACT

TCR signaling pathways cooperate to activate the inducible transcription factors NF-κB, NFAT, and AP-1. In this study, using the calcium ionophore ionomycin and/or PMA on Jurkat T cells, we show that the gene expression program associated with activation of TCR signaling is closely related to specific chromatin landscapes. We find that calcium and kinase signaling cooperate to induce chromatin remodeling at ∼2100 chromatin regions, which demonstrate enriched binding motifs for inducible factors and correlate with target gene expression. We found that these regions typically function as inducible enhancers. Many of these elements contain composite NFAT/AP-1 sites, which typically support cooperative binding, thus further reinforcing the need for cooperation between calcium and kinase signaling in the activation of genes in T cells. In contrast, treatment with PMA or ionomycin alone induces chromatin remodeling at far fewer regions (∼600 and ∼350, respectively), which mostly represent a subset of those induced by costimulation. This suggests that the integration of TCR signaling largely occurs at the level of chromatin, which we propose plays a crucial role in regulating T cell activation.


Subject(s)
Calcium/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Calcium Ionophores/immunology , Humans , Jurkat Cells , Lymphocyte Activation , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Phosphotransferases/metabolism , Receptor Cross-Talk , Signal Transduction , Transcription Factor AP-1/metabolism
11.
Trends Biochem Sci ; 36(1): 47-54, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20870411

ABSTRACT

Transforming growth factor beta (TGFß) controls numerous cellular responses, including proliferation, differentiation, apoptosis and migration. This cytokine is produced by many different cell types and has been implicated in the pathogenesis of many diseases, ranging from autoimmune disorders and infectious diseases to fibrosis and cancer. However, TGFß is always produced as an inactive complex that must be activated to enable binding to its receptor and subsequent function. Recent evidence highlights a crucial role for members of the integrin receptor family in controlling the activation of TGFß. These pathways are important in human health and disease, and new insights into the biochemical mechanisms that allow integrins to control TGFß activation could prove useful in the design of therapeutics.


Subject(s)
Integrins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Humans , Integrins/immunology , Protein Binding , Signal Transduction , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/immunology
12.
PLoS Pathog ; 9(10): e1003675, 2013.
Article in English | MEDLINE | ID: mdl-24098124

ABSTRACT

Chronic intestinal parasite infection is a major global health problem, but mechanisms that promote chronicity are poorly understood. Here we describe a novel cellular and molecular pathway involved in the development of chronic intestinal parasite infection. We show that, early during development of chronic infection with the murine intestinal parasite Trichuris muris, TGFß signalling in CD4+ T-cells is induced and that antibody-mediated inhibition of TGFß function results in protection from infection. Mechanistically, we find that enhanced TGFß signalling in CD4+ T-cells during infection involves expression of the TGFß-activating integrin αvß8 by dendritic cells (DCs), which we have previously shown is highly expressed by a subset of DCs in the intestine. Importantly, mice lacking integrin αvß8 on DCs were completely resistant to chronic infection with T. muris, indicating an important functional role for integrin αvß8-mediated TGFß activation in promoting chronic infection. Protection from infection was dependent on CD4+ T-cells, but appeared independent of Foxp3+ Tregs. Instead, mice lacking integrin αvß8 expression on DCs displayed an early increase in production of the protective type 2 cytokine IL-13 by CD4+ T-cells, and inhibition of this increase by crossing mice to IL-4 knockout mice restored parasite infection. Our results therefore provide novel insights into how type 2 immunity is controlled in the intestine, and may help contribute to development of new therapies aimed at promoting expulsion of gut helminths.


Subject(s)
Dendritic Cells/immunology , Integrins/immunology , Intestinal Diseases, Parasitic/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/immunology , Trichuriasis/immunology , Trichuris/immunology , Animals , Chronic Disease , Dendritic Cells/metabolism , Dendritic Cells/pathology , Integrins/genetics , Integrins/metabolism , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Intestinal Diseases, Parasitic/genetics , Intestinal Diseases, Parasitic/pathology , Mice , Mice, Knockout , Th2 Cells/metabolism , Th2 Cells/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Trichuriasis/genetics , Trichuriasis/metabolism , Trichuriasis/pathology , Trichuris/genetics , Trichuris/metabolism
13.
Exp Cell Res ; 318(6): 716-22, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22309779

ABSTRACT

Transforming Growth Factor Beta (TGF-ß) is involved in regulating many biological processes and disease states. Cells secrete cytokine as a latent complex that must be activated for it to exert its biological functions. We previously discovered that the epithelial-restricted integrin α(v)ß(6) activates TGF-ß and that this process is important in a number of in vivo models of disease. Here, we show that agonists of G-protein coupled receptors (Sphingosine-1-Phosphate and Lysophosphatidic Acid) which are ligated under conditions of epithelial injury directly stimulate primary airway epithelial cells to activate latent TGF-ß through a pathway that involves Rho Kinase, non-muscle myosin, the α(v)ß(6) integrin, and the generation of mechanical tension. Interestingly, lung epithelial cells appear to exert force on latent TGF-ß using sub-cortical actin/myosin rather than the stress fibers utilized by fibroblasts and other traditionally "contractile" cells. These findings extend recent evidence suggesting TGF-ß can be activated by integrin-mediated mechanical force and suggest that this mechanism is important for an integrin (α(v)ß(6)) and a cell type (epithelial cells) that have important roles in biologically relevant TGF-ß activation in vivo.


Subject(s)
Actins/metabolism , Antigens, Neoplasm/metabolism , Epithelial Cells/metabolism , Integrins/metabolism , Myosins/metabolism , Transforming Growth Factor beta/metabolism , Cells, Cultured , Epithelial Cells/cytology , Humans , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism
14.
Nature ; 449(7160): 361-5, 2007 Sep 20.
Article in English | MEDLINE | ID: mdl-17694047

ABSTRACT

The cytokine transforming growth factor-beta (TGF-beta) is an important negative regulator of adaptive immunity. TGF-beta is secreted by cells as an inactive precursor that must be activated to exert biological effects, but the mechanisms that regulate TGF-beta activation and function in the immune system are poorly understood. Here we show that conditional loss of the TGF-beta-activating integrin alpha(v)beta8 on leukocytes causes severe inflammatory bowel disease and age-related autoimmunity in mice. This autoimmune phenotype is largely due to lack of alpha(v)beta8 on dendritic cells, as mice lacking alpha(v)beta8 principally on dendritic cells develop identical immunological abnormalities as mice lacking alpha(v)beta8 on all leukocytes, whereas mice lacking alpha(v)beta8 on T cells alone are phenotypically normal. We further show that dendritic cells lacking alpha(v)beta8 fail to induce regulatory T cells (T(R) cells) in vitro, an effect that depends on TGF-beta activity. Furthermore, mice lacking alpha(v)beta8 on dendritic cells have reduced proportions of T(R) cells in colonic tissue. These results suggest that alpha(v)beta8-mediated TGF-beta activation by dendritic cells is essential for preventing immune dysfunction that results in inflammatory bowel disease and autoimmunity, effects that are due, at least in part, to the ability of alpha(v)beta8 on dendritic cells to induce and/or maintain tissue T(R) cells.


Subject(s)
Autoimmunity/immunology , Colitis/metabolism , Dendritic Cells/metabolism , Integrins/deficiency , Integrins/metabolism , Leukocytes/metabolism , Aging/immunology , Animals , Colitis/immunology , Colon/cytology , Colon/immunology , Immunoglobulins/blood , Immunologic Memory , Integrins/genetics , Interferon-gamma/metabolism , Interleukin-4/metabolism , Leukocytes/immunology , Leukocytes/pathology , Lymphocyte Activation , Lymphocyte Count , Mice , Phenotype , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/metabolism
15.
Gastroenterology ; 141(5): 1802-12, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21723222

ABSTRACT

BACKGROUND & AIMS: The intestinal immune system is tightly regulated to prevent responses against the many nonpathogenic antigens in the gut. Transforming growth factor (TGF)-ß is a cytokine that maintains intestinal homeostasis, in part by inducing Foxp3(+) regulatory T cells (Tregs) that suppress immune responses. TGF-ß is expressed at high levels in the gastrointestinal tract as a latent complex that must be activated. However, the pathways that control TGF-ß activation in the intestine are poorly defined. We investigated the cellular and molecular pathways that control activation of TGF-ß and induction of Foxp3(+) Tregs in the intestines of mice to maintain immune homeostasis. METHODS: Subsets of intestinal dendritic cells (DCs) were examined for their capacity to activate TGF-ß and induce Foxp3(+) Tregs in vitro. Mice were fed oral antigen, and induction of Foxp3(+) Tregs was measured. RESULTS: A tolerogenic subset of intestinal DCs that express CD103 were specialized to activate latent TGF-ß, and induced Foxp3(+) Tregs independently of the vitamin A metabolite retinoic acid. The integrin αvß8, which activates TGF-ß, was significantly up-regulated on CD103(+) intestinal DCs. DCs that lack expression of integrin αvß8 had reduced ability to activate latent TGF-ß and induce Foxp3(+) Tregs in vitro and in vivo. CONCLUSIONS: CD103(+) intestinal DCs promote a tolerogenic environment in the intestines of mice via integrin αvß8-mediated activation of TGF-ß.


Subject(s)
Dendritic Cells/physiology , Forkhead Transcription Factors/metabolism , Integrins/metabolism , Intestines/cytology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/metabolism , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Homeostasis/physiology , Immune System/physiology , In Vitro Techniques , Integrin alpha Chains/metabolism , Integrins/genetics , Mice , Mice, Knockout , Models, Animal , Ovalbumin/pharmacology , T-Lymphocytes, Regulatory/cytology , Tretinoin/metabolism
16.
Contact Dermatitis ; 67(4): 179-83, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22804346

ABSTRACT

It is well established that, in common with other adaptive immune responses, the acquisition of skin sensitization is carefully orchestrated and finely controlled. This is achieved in a number of ways. However, in recent years, there has been an increasing interest in the roles that regulatory T cells (Tregs) play in allergic contact dermatitis. Here, we review briefly the phenotype and function of Tregs, and consider how they may impact on various aspects of skin sensitization, including: inter-individual differences in susceptibility; variations in the potency of chemical allergens; and the prevention of excessive, and potentially damaging, levels of sensitization.


Subject(s)
Dermatitis, Allergic Contact/immunology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Immunity, Cellular , Mice , T-Lymphocytes, Helper-Inducer/immunology
17.
Nat Rev Immunol ; 22(2): 124-138, 2022 02.
Article in English | MEDLINE | ID: mdl-34211187

ABSTRACT

Radiotherapy (RT) is a highly effective anticancer treatment that is delivered to more than half of all patients with cancer. In addition to the well-documented direct cytotoxic effects, RT can have immunomodulatory effects on the tumour and surrounding tissues. These effects are thought to underlie the so-called abscopal responses, whereby RT generates systemic antitumour immunity outside the irradiated tumour. The full scope of these immune changes remains unclear but is likely to involve multiple components, such as immune cells, the extracellular matrix, endothelial and epithelial cells and a myriad of chemokines and cytokines, including transforming growth factor-ß (TGFß). In normal tissues exposed to RT during cancer therapy, acute immune changes may ultimately lead to chronic inflammation and RT-induced toxicity and organ dysfunction, which limits the quality of life of survivors of cancer. Here we discuss the emerging understanding of RT-induced immune effects with particular focus on the lungs and gut and the potential immune crosstalk that occurs between these tissues.


Subject(s)
Neoplasms , Quality of Life , Humans , Immunity , Immunomodulation , Immunotherapy
18.
Sci Rep ; 11(1): 7667, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33828158

ABSTRACT

SapM is a secreted virulence factor from Mycobacterium tuberculosis critical for pathogen survival and persistence inside the host. Its full potential as a target for tuberculosis treatment has not yet been exploited because of the lack of potent inhibitors available. By screening over 1500 small molecules, we have identified new potent and selective inhibitors of SapM with an uncompetitive mechanism of inhibition. The best inhibitors share a trihydroxy-benzene moiety essential for activity. Importantly, the inhibitors significantly reduce mycobacterial burden in infected human macrophages at 1 µM, and they are selective with respect to other mycobacterial and human phosphatases. The best inhibitor also reduces intracellular burden of Francisella tularensis, which secretes the virulence factor AcpA, a homologue of SapM, with the same mechanism of catalysis and inhibition. Our findings demonstrate that inhibition of SapM with small molecule inhibitors is efficient in reducing intracellular mycobacterial survival in host macrophages and confirm SapM as a potential therapeutic target. These initial compounds have favourable physico-chemical properties and provide a basis for exploration towards the development of new tuberculosis treatments. The efficacy of a SapM inhibitor in reducing Francisella tularensis intracellular burden suggests the potential for developing broad-spectrum antivirulence agents to treat microbial infections.


Subject(s)
Mycobacterium tuberculosis/drug effects , Virulence Factors/antagonists & inhibitors , Alkaline Phosphatase/antagonists & inhibitors , Francisella tularensis/enzymology , Humans , Molecular Targeted Therapy , Mycobacterium tuberculosis/pathogenicity , Small Molecule Libraries , Structure-Activity Relationship , Tuberculosis/drug therapy
19.
Nat Commun ; 12(1): 6228, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34711823

ABSTRACT

Presence of TGFß in the tumor microenvironment is one of the most relevant cancer immune-escape mechanisms. TGFß is secreted in an inactive form, and its activation within the tumor may depend on different cell types and mechanisms than its production. Here we show in mouse melanoma and breast cancer models that regulatory T (Treg) cells expressing the ß8 chain of αvß8 integrin (Itgß8) are the main cell type in the tumors that activates TGFß, produced by the cancer cells and stored in the tumor micro-environment. Itgß8 ablation in Treg cells impairs TGFß signalling in intra-tumoral T lymphocytes but not in the tumor draining lymph nodes. Successively, the effector function of tumor infiltrating CD8+ T lymphocytes strengthens, leading to efficient control of tumor growth. In cancer patients, anti-Itgß8 antibody treatment elicits similar improved cytotoxic T cell activation. Thus, this study reveals that Treg cells work in concert with cancer cells to produce bioactive-TGFß and to create an immunosuppressive micro-environment.


Subject(s)
Integrins/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Female , Humans , Integrins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Tumor Microenvironment
20.
Pharmacol Ther ; 225: 107839, 2021 09.
Article in English | MEDLINE | ID: mdl-33774068

ABSTRACT

Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-ß induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.


Subject(s)
Lung Diseases/drug therapy , Lung Diseases/physiopathology , Airway Remodeling/physiology , Asthma/drug therapy , Asthma/physiopathology , Calcium-Binding Proteins/metabolism , Extracellular Matrix/metabolism , Fibroblasts , Fibrosis/physiopathology , Glycoproteins/metabolism , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/physiopathology , Matrix Metalloproteinases/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/physiopathology , Transforming Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL