Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Annu Rev Immunol ; 31: 635-674, 2013.
Article in English | MEDLINE | ID: mdl-23330956

ABSTRACT

To directly study complex human hemato-lymphoid system physiology and respective system-associated diseases in vivo, human-to-mouse xenotransplantation models for human blood and blood-forming cells and organs have been developed over the past three decades. We here review the fundamental requirements and the remarkable progress made over the past few years in improving these systems, the current major achievements reached by use of these models, and the future challenges to more closely model and study human health and disease and to achieve predictive preclinical testing of both prevention measures and potential new therapies.


Subject(s)
Hematopoiesis/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/transplantation , Models, Animal , Animals , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/trends , Humans , Immunophenotyping , Lymphoid Tissue/pathology , Mice , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends , Transplantation, Heterologous
2.
Cell ; 159(7): 1563-77, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525875

ABSTRACT

The mechanism by which cells undergo death determines whether dying cells trigger inflammatory responses or remain immunologically silent. Mitochondria play a central role in the induction of cell death, as well as in immune signaling pathways. Here, we identify a mechanism by which mitochondria and downstream proapoptotic caspases regulate the activation of antiviral immunity. In the absence of active caspases, mitochondrial outer membrane permeabilization by Bax and Bak results in the expression of type I interferons (IFNs). This induction is mediated by mitochondrial DNA-dependent activation of the cGAS/STING pathway and results in the establishment of a potent state of viral resistance. Our results show that mitochondria have the capacity to simultaneously expose a cell-intrinsic inducer of the IFN response and to inactivate this response in a caspase-dependent manner. This mechanism provides a dual control, which determines whether mitochondria initiate an immunologically silent or a proinflammatory type of cell death.


Subject(s)
Apoptosis , Caspases/metabolism , Interferon Type I/metabolism , Signal Transduction , Animals , DNA, Mitochondrial/metabolism , Inflammation/immunology , Inflammation/metabolism , Interferon Type I/immunology , Mice , Mice, Knockout , Virus Diseases/immunology
3.
Nat Immunol ; 17(11): 1282-1290, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27618552

ABSTRACT

Glioma cells recruit and exploit microglia (the resident immune cells of the brain) for their proliferation and invasion ability. The underlying molecular mechanism used by glioma cells to transform microglia into a tumor-supporting phenotype has remained elusive. We found that glioma-induced microglia conversion was coupled to a reduction in the basal activity of microglial caspase-3 and increased S-nitrosylation of mitochondria-associated caspase-3 through inhibition of thioredoxin-2 activity, and that inhibition of caspase-3 regulated microglial tumor-supporting function. Furthermore, we identified the activity of nitric oxide synthase 2 (NOS2, also known as iNOS) originating from the glioma cells as a driving stimulus in the control of microglial caspase-3 activity. Repression of glioma NOS2 expression in vivo led to a reduction in both microglia recruitment and tumor expansion, whereas depletion of microglial caspase-3 gene promoted tumor growth. Our results provide evidence that inhibition of the denitrosylation of S-nitrosylated procaspase-3 mediated by the redox protein Trx2 is a part of the microglial pro-tumoral activation pathway initiated by glioma cancer cells.


Subject(s)
Caspase 3/metabolism , Glioma/metabolism , Glioma/pathology , Microglia/metabolism , Phenotype , Animals , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Enzyme Activation , Gene Knockdown Techniques , Glioma/immunology , Heterografts , Humans , Male , Mice , Microglia/immunology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Thioredoxins/metabolism , Tumor Burden
4.
Nature ; 605(7911): 728-735, 2022 05.
Article in English | MEDLINE | ID: mdl-35545675

ABSTRACT

Immunotherapies have achieved remarkable successes in the treatment of cancer, but major challenges remain1,2. An inherent weakness of current treatment approaches is that therapeutically targeted pathways are not restricted to tumours, but are also found in other tissue microenvironments, complicating treatment3,4. Despite great efforts to define inflammatory processes in the tumour microenvironment, the understanding of tumour-unique immune alterations is limited by a knowledge gap regarding the immune cell populations in inflamed human tissues. Here, in an effort to identify such tumour-enriched immune alterations, we used complementary single-cell analysis approaches to interrogate the immune infiltrate in human head and neck squamous cell carcinomas and site-matched non-malignant, inflamed tissues. Our analysis revealed a large overlap in the composition and phenotype of immune cells in tumour and inflamed tissues. Computational analysis identified tumour-enriched immune cell interactions, one of which yields a large population of regulatory T (Treg) cells that is highly enriched in the tumour and uniquely identified among all haematopoietically-derived cells in blood and tissue by co-expression of ICOS and IL-1 receptor type 1 (IL1R1). We provide evidence that these intratumoural IL1R1+ Treg cells had responded to antigen recently and demonstrate that they are clonally expanded with superior suppressive function compared with IL1R1- Treg cells. In addition to identifying extensive immunological congruence between inflamed tissues and tumours as well as tumour-specific changes with direct disease relevance, our work also provides a blueprint for extricating disease-specific changes from general inflammation-associated patterns.


Subject(s)
Neoplasms , Humans , Immunotherapy , Inflammation , Neoplasms/pathology , T-Lymphocytes, Regulatory , Tumor Microenvironment
5.
J Immunol ; 209(3): 606-620, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35817516

ABSTRACT

Despite recent therapeutic progress, advanced melanoma remains lethal for many patients. The composition of the immune tumor microenvironment (TME) has decisive impacts on therapy response and disease outcome, and high-dimensional analyses of patient samples reveal the heterogeneity of the immune TME. Macrophages infiltrate TMEs and generally associate with tumor progression, but the underlying mechanisms are incompletely understood. Because experimental systems are needed to elucidate the functional properties of these cells, we developed a humanized mouse model reconstituted with human immune cells and human melanoma. We used two strains of recipient mice, supporting or not supporting the development of human myeloid cells. We found that human myeloid cells favored metastatic spread of the primary tumor, thereby recapitulating the cancer-supportive role of macrophages. We next analyzed the transcriptome of human immune cells infiltrating tumors versus other tissues. This analysis identified a cluster of myeloid cells present in the TME, but not in other tissues, which do not correspond to canonical M2 cells. The transcriptome of these cells is characterized by high expression of glycolytic enzymes and multiple chemokines and by low expression of gene sets associated with inflammation and adaptive immunity. Compared with humanized mouse results, we found transcriptionally similar myeloid cells in patient-derived samples of melanoma and other cancer types. The humanized mouse model described here thus complements patient sample analyses, enabling further elucidation of fundamental principles in melanoma biology beyond M1/M2 macrophage polarization. The model can also support the development and evaluation of candidate antitumor therapies.


Subject(s)
Macrophages , Melanoma , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Macrophage Activation , Melanoma/pathology , Mice , Tumor Microenvironment
6.
Nature ; 517(7534): 381-5, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25561180

ABSTRACT

Despite antiretroviral therapy (ART), human immunodeficiency virus (HIV)-1 persists in a stable latent reservoir, primarily in resting memory CD4(+) T cells. This reservoir presents a major barrier to the cure of HIV-1 infection. To purge the reservoir, pharmacological reactivation of latent HIV-1 has been proposed and tested both in vitro and in vivo. A key remaining question is whether virus-specific immune mechanisms, including cytotoxic T lymphocytes (CTLs), can clear infected cells in ART-treated patients after latency is reversed. Here we show that there is a striking all or none pattern for CTL escape mutations in HIV-1 Gag epitopes. Unless ART is started early, the vast majority (>98%) of latent viruses carry CTL escape mutations that render infected cells insensitive to CTLs directed at common epitopes. To solve this problem, we identified CTLs that could recognize epitopes from latent HIV-1 that were unmutated in every chronically infected patient tested. Upon stimulation, these CTLs eliminated target cells infected with autologous virus derived from the latent reservoir, both in vitro and in patient-derived humanized mice. The predominance of CTL-resistant viruses in the latent reservoir poses a major challenge to viral eradication. Our results demonstrate that chronically infected patients retain a broad-spectrum viral-specific CTL response and that appropriate boosting of this response may be required for the elimination of the latent reservoir.


Subject(s)
Genes, Dominant/genetics , Genes, Viral/genetics , HIV-1/genetics , HIV-1/immunology , Mutation/genetics , T-Lymphocytes, Cytotoxic/immunology , Virus Latency/immunology , Acute Disease/therapy , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Chronic Disease/drug therapy , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , HIV Infections/blood , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/growth & development , Humans , Male , Mice , RNA, Viral/blood , Viral Load/drug effects , Virus Latency/genetics , Virus Replication/immunology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
7.
Proc Natl Acad Sci U S A ; 115(46): E10888-E10897, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30381458

ABSTRACT

Cell death and inflammation are intimately linked during Yersinia infection. Pathogenic Yersinia inhibits the MAP kinase TGFß-activated kinase 1 (TAK1) via the effector YopJ, thereby silencing cytokine expression while activating caspase-8-mediated cell death. Here, using Yersinia pseudotuberculosis in corroboration with costimulation of lipopolysaccharide and (5Z)-7-Oxozeaenol, a small-molecule inhibitor of TAK1, we show that caspase-8 activation during TAK1 inhibition results in cleavage of both gasdermin D (GSDMD) and gasdermin E (GSDME) in murine macrophages, resulting in pyroptosis. Loss of GsdmD delays membrane rupture, reverting the cell-death morphology to apoptosis. We found that the Yersinia-driven IL-1 response arises from asynchrony of macrophage death during bulk infections in which two cellular populations are required to provide signal 1 and signal 2 for IL-1α/ß release. Furthermore, we found that human macrophages are resistant to YopJ-mediated pyroptosis, with dampened IL-1ß production. Our results uncover a form of caspase-8-mediated pyroptosis and suggest a hypothesis for the increased sensitivity of humans to Yersinia infection compared with the rodent reservoir.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspase 8/metabolism , Yersinia Infections/metabolism , Animals , Apoptosis/physiology , Bacterial Proteins/metabolism , Humans , Interleukin-1/metabolism , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phosphate-Binding Proteins , Pyroptosis/physiology , Yersinia Infections/pathology , Yersinia pseudotuberculosis/metabolism
8.
PLoS Pathog ; 12(10): e1005910, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27737018

ABSTRACT

Caspases regulate cell death programs in response to environmental stresses, including infection and inflammation, and are therefore critical for the proper operation of the mammalian immune system. Caspase-8 is necessary for optimal production of inflammatory cytokines and host defense against infection by multiple pathogens including Yersinia, but whether this is due to death of infected cells or an intrinsic role of caspase-8 in TLR-induced gene expression is unknown. Caspase-8 activation at death signaling complexes results in its autoprocessing and subsequent cleavage and activation of its downstream apoptotic targets. Whether caspase-8 activity is also important for inflammatory gene expression during bacterial infection has not been investigated. Here, we report that caspase-8 plays an essential cell-intrinsic role in innate inflammatory cytokine production in vivo during Yersinia infection. Unexpectedly, we found that caspase-8 enzymatic activity regulates gene expression in response to bacterial infection as well as TLR signaling independently of apoptosis. Using newly-generated mice in which caspase-8 autoprocessing is ablated (Casp8DA/DA), we now demonstrate that caspase-8 enzymatic activity, but not autoprocessing, mediates induction of inflammatory cytokines by bacterial infection and a wide variety of TLR stimuli. Because unprocessed caspase-8 functions in an enzymatic complex with its homolog cFLIP, our findings implicate the caspase-8/cFLIP heterodimer in control of inflammatory cytokines during microbial infection, and provide new insight into regulation of antibacterial immune defense.


Subject(s)
Caspase 8/immunology , Cytokines/biosynthesis , Immunity, Innate/immunology , Signal Transduction/immunology , Yersinia Infections/immunology , Animals , Apoptosis , Caspase 8/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation/immunology , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Toll-Like Receptors/immunology
9.
Blood ; 128(14): 1829-1833, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27543436

ABSTRACT

Human CD34+ hematopoietic stem and progenitor cells (HSPCs) can reconstitute a human hemato-lymphoid system when transplanted into immunocompromised mice. Although fetal liver-derived and cord blood-derived CD34+ cells lead to high engraftment levels, engraftment of mobilized, adult donor-derived CD34+ cells has remained poor. We generated so-called MSTRG and MISTRG humanized mice on a Rag2-/-Il2rg-/- background carrying a transgene for human signal regulatory protein α (SIRPα) and human homologs of the cytokine macrophage colony-stimulating factor, thrombopoietin, with or without interleukin-3 and granulocyte-macrophage colony-stimulating factor under murine promoters. Here we transplanted mobilized peripheral blood (PB) CD34+ cells in sublethally irradiated newborn and adult recipients. Human hematopoietic engraftment levels were significantly higher in bone marrow (BM), spleen, and PB in newborn transplanted MSTRG/MISTRG as compared with nonobese diabetic/severe combined immunodeficient Il2rg-/- or human SIRPα-transgenic Rag2-/-Il2rg-/- recipients. Furthermore, newborn transplanted MSTRG/MISTRG mice supported higher engraftment levels of human phenotypically defined HSPCs in BM, T cells in the thymus, and myeloid cells in nonhematopoietic organs such as liver, lung, colon, and skin, approximating the levels in the human system. Similar results were obtained in adult recipient mice. Thus, human cytokine knock-in mice might open new avenues for personalized studies of human pathophysiology of the hematopoietic and immune system in vivo.


Subject(s)
Antigens, CD34/metabolism , Cytokines/metabolism , Gene Knock-In Techniques , Animals , Animals, Newborn , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Lymphoid Tissue/metabolism , Mice, Transgenic
10.
Nature ; 484(7395): 510-3, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-22538615

ABSTRACT

NLRs (nucleotide-binding domain leucine-rich-repeat-containing receptors; NOD-like receptors) are a class of pattern recognition receptor (PRR) that respond to host perturbation from either infectious agents or cellular stress. The function of most NLR family members has not been characterized and their role in instructing adaptive immune responses remains unclear. NLRP10 (also known as PYNOD, NALP10, PAN5 and NOD8) is the only NLR lacking the putative ligand-binding leucine-rich-repeat domain, and has been postulated to be a negative regulator of other NLR members, including NLRP3 (refs 4-6). We did not find evidence that NLRP10 functions through an inflammasome to regulate caspase-1 activity nor that it regulates other inflammasomes. Instead, Nlrp10(-/-) mice had a profound defect in helper T-cell-driven immune responses to a diverse array of adjuvants, including lipopolysaccharide, aluminium hydroxide and complete Freund's adjuvant. Adaptive immunity was impaired in the absence of NLRP10 because of a dendritic cell (DC) intrinsic defect in emigration from inflamed tissues, whereas upregulation of DC costimulatory molecules and chemotaxis to CCR7-dependent and -independent ligands remained intact. The loss of antigen transport to the draining lymph nodes by a subset of migratory DCs resulted in an almost absolute loss in naive CD4(+) T-cell priming, highlighting the critical link between diverse innate immune stimulation, NLRP10 activity and the immune function of mature DCs.


Subject(s)
Adaptive Immunity/immunology , Apoptosis Regulatory Proteins/metabolism , Dendritic Cells/immunology , Adaptor Proteins, Signal Transducing , Adjuvants, Immunologic , Animals , Antigens/immunology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Caspase 1 , Cell Movement , Chemokines/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Deletion , Inflammasomes , Ligands , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines/immunology
11.
Nature ; 475(7357): 514-8, 2011 Jul 17.
Article in English | MEDLINE | ID: mdl-21765430

ABSTRACT

Interleukin (IL)-17-producing T helper cells (T(H)17) are a recently identified CD4(+) T cell subset distinct from T helper type 1 (T(H)1) and T helper type 2 (T(H)2) cells. T(H)17 cells can drive antigen-specific autoimmune diseases and are considered the main population of pathogenic T cells driving experimental autoimmune encephalomyelitis (EAE), the mouse model for multiple sclerosis. The factors that are needed for the generation of T(H)17 cells have been well characterized. However, where and how the immune system controls T(H)17 cells in vivo remains unclear. Here, by using a model of tolerance induced by CD3-specific antibody, a model of sepsis and influenza A viral infection (H1N1), we show that pro-inflammatory T(H)17 cells can be redirected to and controlled in the small intestine. T(H)17-specific IL-17A secretion induced expression of the chemokine CCL20 in the small intestine, facilitating the migration of these cells specifically to the small intestine via the CCR6/CCL20 axis. Moreover, we found that T(H)17 cells are controlled by two different mechanisms in the small intestine: first, they are eliminated via the intestinal lumen; second, pro-inflammatory T(H)17 cells simultaneously acquire a regulatory phenotype with in vitro and in vivo immune-suppressive properties (rT(H)17). These results identify mechanisms limiting T(H)17 cell pathogenicity and implicate the gastrointestinal tract as a site for control of T(H)17 cells.


Subject(s)
Intestine, Small/immunology , Th17 Cells/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cell Movement/drug effects , Chemokine CCL20/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Expression Profiling , Gene Expression Regulation/immunology , Influenza A virus/immunology , Interleukin-17/immunology , Intestine, Small/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Receptors, CCR6/immunology , Sepsis/immunology , Staphylococcal Infections/immunology
13.
J Cell Sci ; 127(Pt 10): 2217-26, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24610949

ABSTRACT

Caspase-3 is an effector caspase that is activated downstream of mitochondrial outer-membrane permeabilization (MOMP) during apoptosis. However, previous work has demonstrated that caspase-3-deficient mouse embryonic fibroblasts (MEFs) are resistant to mitochondrially mediated cell death and display a delay in the mitochondrial events of apoptosis, including Bax activation, MOMP and release of cytochrome c. Here, we show that caspase-3 regulates fibronectin secretion and impacts on cell morphology, adhesion and migration. Surprisingly, the catalytic activity of caspase-3 is not required for these non-apoptotic functions. Moreover, we found that caspase-3-deficient MEFs are not resistant to death by anoikis and that exogenous fibronectin protects wild-type MEFs from cell death induced by serum withdrawal. Taken together, our data indicate that procaspase-3 has a non-apoptotic function; it regulates the secretion of fibronectin and influences morphology, adhesion and migration. Furthermore, this novel procaspase-3 function might alter the apoptotic threshold of the cell.


Subject(s)
Caspase 3/metabolism , Cell Adhesion/physiology , Cell Movement/physiology , Fibronectins/metabolism , Animals , Apoptosis , Catalysis , Cell Survival/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Mice
14.
Haematologica ; 101(1): 5-19, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26721800

ABSTRACT

Over the last decades, incrementally improved xenograft mouse models, supporting the engraftment and development of a human hemato-lymphoid system, have been developed and now represent an important research tool in the field. The most significant contributions made by means of humanized mice are the identification of normal and leukemic hematopoietic stem cells, the characterization of the human hematopoietic hierarchy, and their use as preclinical therapy models for malignant hematopoietic disorders. Successful xenotransplantation depends on three major factors: tolerance by the mouse host, correct spatial location, and appropriately cross-reactive support and interaction factors such as cytokines and major histocompatibility complex molecules. Each of these can be modified. Experimental approaches include the genetic modification of mice to faithfully express human support factors as non-cross-reactive cytokines, to create free niche space, the co-transplantation of human mesenchymal stem cells, the implantation of humanized ossicles or other stroma, and the implantation of human thymic tissue. Besides the source of hematopoietic cells, the conditioning regimen and the route of transplantation also significantly affect human hematopoietic development in vivo. We review here the achievements, most recent developments, and the remaining challenges in the generation of pre-clinically-predictive systems for human hematology and immunology, closely resembling the human situation in a xenogeneic mouse environment.


Subject(s)
Hematopoietic System/immunology , Lymphatic System/immunology , Mesenchymal Stem Cell Transplantation , Thymus Gland/transplantation , Animals , Heterografts , Humans , Mice , Mice, SCID , Thymus Gland/immunology
15.
J Biol Chem ; 289(38): 26642-26657, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25086041

ABSTRACT

Transcriptome analyses have recently identified PARP12, a member of a large family of ADP-ribosyl transferases, as an interferon-induced gene (ISG), whose function remains incompletely characterized. We demonstrate herein that PARP12 is a genuine ISG, whose expressed protein displays at least two distinct subcellular locations and related functions. Upon ectopic expression or exposure to oxidative stress, PARP12 is recruited to stress-granules (SGs), known sites of mRNA translational arrest. Accordingly, PARP12 was found to block mRNA translation, possibly upon association to the translational machinery. Both the N-terminal domain (containing an RNA-binding domain characterized by the presence of five CCCH-type Zn-fingers) and integrity of the catalytic domain are required for this suppressive function. In contrast, stimulation with LPS leads to the localization of PARP12 to p62/SQSTM1 (an adaptor protein involved in innate signaling and autophagy) containing structures, unrelated to SGs. Deletion of the N-terminal domain promotes the association of the protein to p62/SQSTM1, suggesting that the RNA-binding domain is responsible for the subcellular localization of PARP12. Association to p62/SQSTM1 was found to correlate with increased NF-κB signaling, suggesting a role for PARP12 in inflammation. Collectively, these observations suggest that PARP12 can alternate between two distinct subcellular compartments associated to two distinct cellular functions. The present work therefore identifies PARP12 as an ISG with a potential role in cellular defenses against viral infections.


Subject(s)
Poly(ADP-ribose) Polymerases/genetics , Protein Biosynthesis , Amino Acid Sequence , Animals , Cytoplasmic Granules/metabolism , Enzyme Induction , HEK293 Cells , HeLa Cells , Humans , Inflammation/enzymology , Interferon-beta/physiology , Mice , Molecular Sequence Data , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Transport , Transcriptional Activation , Ubiquitin/metabolism , Zinc Fingers
16.
J Cell Sci ; 126(Pt 7): 1682-91, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23424201

ABSTRACT

Polypyrimidine tract binding protein (PTB) regulates pre-mRNA splicing, having special relevance for determining gene expression in the differentiating muscle. We have previously shown that PTB protein abundance is progressively reduced during heart development without reduction of its own transcript. Simultaneous reduction of histone deacetylase (HDAC) expression prompted us to investigate the potential link between these events. HDAC5-deficient mice have reduced cardiac PTB protein abundance, and HDAC inhibition in myocytes causes a reduction in endogenous expression of cellular FLICE-like inhibitory protein (cFLIP) and caspase-dependent cleavage of PTB. In agreement with this, cardiac PTB expression is abnormally high in mice with cardiac-specific executioner caspase deficiency, and cFLIP overexpression prevents PTB cleavage in vitro. Caspase-dependent cleavage triggers further fragmentation of PTB, and these fragments accumulate in the presence of proteasome inhibitors. Experimental modification of the above processes in vivo and in vitro results in coherent changes in the alternative splicing of genes encoding tropomyosin-1 (TPM1), tropomyosin-2 (TPM2) and myocyte enhancer factor-2 (MEF2). Thus, we report a pathway connecting HDAC, cFLIP and caspases regulating the progressive disappearance of PTB, which enables the expression of the adult variants of proteins involved in the regulation of contraction and transcription during cardiac muscle development.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspases/metabolism , Histone Deacetylases/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Animals , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Caspases/genetics , Electrophoresis, Polyacrylamide Gel , Histone Deacetylases/genetics , Mice , Mice, Knockout , Myocardium/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tropomyosin/genetics , Tropomyosin/metabolism
17.
Trends Immunol ; 32(7): 321-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21697012

ABSTRACT

Human hemato-lymphoid-system mice hold great promise for modeling and studying important human diseases in vivo, and to enable vaccine testing. Until now, several major limitations have restricted the utility of human hemato-lymphoid-system mice in translational research. Recently, however, significant advances have been made in improving these mice, based on the delivery of human cytokines to create a better environment for human cells in the mouse host. In this review, we discuss the various approaches with a particular focus on improving human hemato-lymphoid-system mice by human cytokine knock-in gene replacement.


Subject(s)
Cytokines/immunology , Hematopoietic System/immunology , Lymphoid Tissue/immunology , Animals , Cell Lineage , Cytokines/genetics , Gene Knock-In Techniques , Hematopoietic System/cytology , Humans , Lymphoid Tissue/cytology , Mice , Translational Research, Biomedical
18.
Proc Natl Acad Sci U S A ; 108(32): 13218-23, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788509

ABSTRACT

Transplantation of human hematopoietic stem cells into severely immunocompromised newborn mice allows the development of a human hematopoietic and immune system in vivo. NOD/scid/γ(c)(-/-) (NSG) and BALB/c Rag2(-/-)γ(c)(-/-) mice are the most commonly used mouse strains for this purpose and a number of studies have demonstrated the high value of these model systems in areas spanning from basic to translational research. However, limited cross-reactivity of many murine cytokines on human cells and residual host immune function against the xenogeneic grafts results in defective development and maintenance of human cells in vivo. Whereas NSG mice have higher levels of absolute human engraftment than similar mice on a BALB/c background, they have a shorter lifespan and NOD ES cells are unsuitable for the complex genetic engineering that is required to improve human hematopoiesis and immune responses by transgenesis or knockin of human genes. We have generated mice that faithfully express a transgene of human signal regulatory protein alpha (SIRPa), a receptor that negatively regulates phagocytosis, in Rag2(-/-)γ(c)(-/-) mice on a mixed 129/BALB/c background, which can easily be genetically engineered. These mice allow significantly increased engraftment and maintenance of human hematopoietic cells reaching levels comparable to NSG mice. Furthermore, we found improved functionality of the human immune system in these mice. In summary, hSIRPa-transgenic Rag2(-/-)γ(c)(-/-) mice represent a unique mouse strain supporting high levels of human cell engraftment, which can easily be genetically manipulated.


Subject(s)
Antigens, Differentiation/metabolism , DNA-Binding Proteins/deficiency , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Interleukin Receptor Common gamma Subunit/deficiency , Receptors, Immunologic/metabolism , Transgenes/genetics , Animals , Antigens, Differentiation/genetics , Bone Marrow Cells/pathology , Cell Lineage , DNA-Binding Proteins/metabolism , Epitopes/immunology , Humans , Immunity, Humoral/immunology , Interleukin Receptor Common gamma Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptors, Immunologic/genetics
19.
Proc Natl Acad Sci U S A ; 108(6): 2390-5, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21262803

ABSTRACT

Mice with a functional human immune system have the potential to allow in vivo studies of human infectious diseases and to enable vaccine testing. To this end, mice need to fully support the development of human immune cells, allow infection with human pathogens, and be capable of mounting effective human immune responses. A major limitation of humanized mice is the poor development and function of human myeloid cells and the absence of human immune responses at mucosal surfaces, such as the lung. To overcome this, we generated human IL-3/GM-CSF knock-in (hIL-3/GM-CSF KI) mice. These mice faithfully expressed human GM-CSF and IL-3 and developed pulmonary alveolar proteinosis because of elimination of mouse GM-CSF. We demonstrate that hIL-3/GM-CSF KI mice engrafted with human CD34(+) hematopoietic cells had improved human myeloid cell reconstitution in the lung. In particular, hIL-3/GM-CSF KI mice supported the development of human alveolar macrophages that partially rescued the pulmonary alveolar proteinosis syndrome. Moreover, human alveolar macrophages mounted correlates of a human innate immune response against influenza virus. The hIL-3/GM-CSF KI mice represent a unique mouse model that permits the study of human mucosal immune responses to lung pathogens.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Interleukin-3/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/immunology , Animals , Cord Blood Stem Cell Transplantation , Gene Knock-In Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Immunity, Mucosal/genetics , Interleukin-3/genetics , Lung/virology , Macrophages, Alveolar/virology , Mice , Mice, Transgenic , Models, Immunological , Orthomyxoviridae Infections/genetics , Transplantation Chimera/genetics , Transplantation Chimera/immunology , Transplantation Chimera/virology , Transplantation, Heterologous
20.
Proc Natl Acad Sci U S A ; 108(6): 2378-83, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21262827

ABSTRACT

Hematopoietic stem cells (HSCs) both self-renew and give rise to all blood cells for the lifetime of an individual. Xenogeneic mouse models are broadly used to study human hematopoietic stem and progenitor cell biology in vivo. However, maintenance, differentiation, and function of human hematopoietic cells are suboptimal in these hosts. Thrombopoietin (TPO) has been demonstrated as a crucial cytokine supporting maintenance and self-renewal of HSCs. We generated RAG2(-/-)γ(c)(-/-) mice in which we replaced the gene encoding mouse TPO by its human homolog. Homozygous humanization of TPO led to increased levels of human engraftment in the bone marrow of the hosts, and multilineage differentiation of hematopoietic cells was improved, with an increased ratio of myelomonocytic verus lymphoid lineages. Moreover, maintenance of human stem and progenitor cells was improved, as demonstrated by serial transplantation. Therefore, RAG2(-/-)γ(c)(-/-) TPO-humanized mice represent a useful model to study human hematopoiesis in vivo.


Subject(s)
Hematopoiesis , Thrombopoietin/metabolism , Animals , Gene Knock-In Techniques , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Transgenic , Thrombopoietin/genetics , Transplantation Chimera/genetics , Transplantation Chimera/metabolism , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL