Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Blood ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38820590

ABSTRACT

Deficiency of X-linked Inhibitor of Apoptosis Protein (XIAP) is a rare genetic condition that can present with recurrent episodes of hemophagocytic lymphohistiocytosis (HLH), though the exact mechanisms leading to this hyperinflammatory disorder are unclear. Understanding its biology is critical to developing targeted therapies for this potentially fatal disease. Here we report on a novel multi-exonic intragenic duplication leading to XIAP deficiency with recurrent HLH that demonstrated complete response to interleukin (IL)-1b blockade. We further demonstrate using both primary patient cells and genetically modified THP-1 monocyte cell lines that, contrary to what has previously been shown in mouse cells, XIAP-deficient human macrophages do not produce excess IL-1b when stimulated under standard conditions. Instead, NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated hyperproduction of IL-1b is observed only when the XIAP-deficient cells are stimulated under autophagy-promoting conditions and this correlates with defective autophagic flux as measured by decreased accumulation of the early autophagy marker LC3-II. This work therefore highlights IL-1b blockade as a therapeutic option for patients with XIAP deficiency experiencing recurrent HLH and identifies a critical role for XIAP in promoting autophagy as a means of limiting IL-1b-mediated hyperinflammation during periods of cellular stress.

2.
Nature ; 577(7788): 109-114, 2020 01.
Article in English | MEDLINE | ID: mdl-31827280

ABSTRACT

Activation of RIPK1 controls TNF-mediated apoptosis, necroptosis and inflammatory pathways1. Cleavage of human and mouse RIPK1 after residues D324 and D325, respectively, by caspase-8 separates the RIPK1 kinase domain from the intermediate and death domains. The D325A mutation in mouse RIPK1 leads to embryonic lethality during mouse development2,3. However, the functional importance of blocking caspase-8-mediated cleavage of RIPK1 on RIPK1 activation in humans is unknown. Here we identify two families with variants in RIPK1 (D324V and D324H) that lead to distinct symptoms of recurrent fevers and lymphadenopathy in an autosomal-dominant manner. Impaired cleavage of RIPK1 D324 variants by caspase-8 sensitized patients' peripheral blood mononuclear cells to RIPK1 activation, apoptosis and necroptosis induced by TNF. The patients showed strong RIPK1-dependent activation of inflammatory signalling pathways and overproduction of inflammatory cytokines and chemokines compared with unaffected controls. Furthermore, we show that expression of the RIPK1 mutants D325V or D325H in mouse embryonic fibroblasts confers not only increased sensitivity to RIPK1 activation-mediated apoptosis and necroptosis, but also induction of pro-inflammatory cytokines such as IL-6 and TNF. By contrast, patient-derived fibroblasts showed reduced expression of RIPK1 and downregulated production of reactive oxygen species, resulting in resistance to necroptosis and ferroptosis. Together, these data suggest that human non-cleavable RIPK1 variants promote activation of RIPK1, and lead to an autoinflammatory disease characterized by hypersensitivity to apoptosis and necroptosis and increased inflammatory response in peripheral blood mononuclear cells, as well as a compensatory mechanism to protect against several pro-death stimuli in fibroblasts.


Subject(s)
Caspase 8/metabolism , Hereditary Autoinflammatory Diseases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , Child , Child, Preschool , Female , HEK293 Cells , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/pathology , Humans , Male , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequence Alignment , Sequence Homology, Amino Acid
3.
Nat Immunol ; 14(1): 27-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23179078

ABSTRACT

The E3 ligase ARIH2 has an unusual structure and mechanism of elongating ubiquitin chains. To understand its physiological role, we generated gene-targeted mice deficient in ARIH2. ARIH2 deficiency resulted in the embryonic death of C57BL/6 mice. On a mixed genetic background, the lethality was attenuated, with some mice surviving beyond weaning and then succumbing to an aggressive multiorgan inflammatory response. We found that in dendritic cells (DCs), ARIH2 caused degradation of the inhibitor IκBß in the nucleus, which abrogated its ability to sequester, protect and transcriptionally coactivate the transcription factor subunit p65 in the nucleus. Loss of ARIH2 caused dysregulated activation of the transcription factor NF-κB in DCs, which led to lethal activation of the immune system in ARIH2-sufficent mice reconstituted with ARIH2-deficient hematopoietic stem cells. Our data have therapeutic implications for targeting ARIH2 function.


Subject(s)
Dendritic Cells/immunology , Embryonic Development/immunology , Multiple Organ Failure/immunology , Ubiquitin-Protein Ligases/physiology , Animals , Cells, Cultured , Disease Models, Animal , Embryonic Development/genetics , Hematopoiesis/genetics , Humans , Immune System/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Multiple Organ Failure/genetics , NF-kappa B/metabolism , Transcriptional Activation/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/immunology
4.
Support Care Cancer ; 26(8): 2851-2859, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29525841

ABSTRACT

PURPOSE: One in three breast cancer survivors experiences persistent cognitive changes that can negatively impact daily functioning and quality of life. In our cancer center, the largest tertiary cancer center in Canada, patients with self-reported cancer-related cognitive dysfunction (CRCD) are offered psychoeducation intended to reduce distress about CRCD symptoms and improve everyday cognitive performance, but evidence regarding this intervention's impact is lacking. Here, we assess whether a 1-hour (h), individual psychoeducational intervention designed to promote self-management of CRCD symptoms can improve attitudes and coping with memory-related difficulties in women with breast cancer. METHODS: Breast cancer survivors with self-reported CRCD (N = 100) were assessed immediately before, immediately after, and 6 weeks following the intervention. Participants' memory contentment, knowledge of CRCD, symptom distress, and self-efficacy to cope with symptoms were measured. RESULTS: Participants showed improvements in memory contentment immediately after the intervention (Cohen's d effect size and 95% CI = 0.87 [0.58, 1.16]) and 6 weeks later (d = 0.77 [0.48, 1.05]). Significant improvements in secondary study outcomes, including knowledge of CRCD (d = 1.32 [1.01, 1.63]), symptom distress (d = - 0.82 [- 1.11, - 0.53]), and self-efficacy to cope with cognitive symptoms (d = 1.45 [1.14, 1.76]), were also observed. CONCLUSIONS: A single, 1-hour psychoeducational intervention can achieve lasting and improved adjustment to memory symptoms in breast cancer survivors with self-reported CRCD. Further investigation using a randomized controlled study design is warranted. Comparisons with previously reported psychoeducational interventions for CRCD are made, and next steps for this research are discussed.


Subject(s)
Breast Neoplasms/psychology , Cancer Survivors/psychology , Memory/physiology , Quality of Life/psychology , Adaptation, Psychological , Adult , Aged , Female , Humans , Longitudinal Studies , Middle Aged , Prospective Studies
5.
J Immunol ; 195(10): 4632-40, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26447227

ABSTRACT

TLR-induced maturation of dendritic cells (DCs) leads to the production of proinflammatory cytokines as well as the upregulation of various molecules involved in T cell activation. These are believed to be the critical events that account for the induction of the adaptive immune response. In this study, we have examined the role of miR-155 in DC function and the induction of immunity. Using a model in which the transfer of self-Ag-pulsed, TLR-matured DCs can induce a functional CD8 T cell response and autoimmunity, we find that DCs lacking miR-155 have an impaired ability to break immune tolerance. Importantly, transfer of self- Ag-pulsed DCs overexpressing miR-155 was sufficient to break tolerance in the absence of TLR stimuli. Although these unstimulated DCs induced T cell function in vivo, there was no evidence for the upregulation of costimulatory ligands or cytokine secretion. Further analysis showed that miR-155 influenced the level of the phosphatase SHIP1 in DCs and that the lack of SHIP1 in DCs was sufficient to break T cell tolerance in vivo, again in the absence of TLR-induced DC maturation. Our study demonstrates that the overexpression of miR-155 in DCs is a critical event that is alone sufficient to break self-tolerance and promote a CD8-mediated autoimmune response in vivo. This process is independent of the induction of conventional DC maturation markers, indicating that miR-155 regulation of SHIP represents a unique axis that regulates DC function in vivo.


Subject(s)
Dendritic Cells/metabolism , Immune Tolerance/immunology , MicroRNAs/biosynthesis , Phosphoric Monoester Hydrolases/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Immune Tolerance/genetics , Inositol Polyphosphate 5-Phosphatases , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Up-Regulation
7.
Proc Natl Acad Sci U S A ; 109(4): 1210-5, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22167808

ABSTRACT

Infections with HIV, hepatitis B virus, and hepatitis C virus can turn into chronic infections, which currently affect more than 500 million patients worldwide. It is generally thought that virus-mediated T-cell exhaustion limits T-cell function, thus promoting chronic disease. Here we demonstrate that natural killer (NK) cells have a negative impact on the development of T-cell immunity by using the murine lymphocytic choriomeningitis virus. NK cell-deficient (Nfil3(-/-), E4BP4(-/-)) mice exhibited a higher virus-specific T-cell response. In addition, NK cell depletion caused enhanced T-cell immunity in WT mice, which led to rapid virus control and prevented chronic infection in lymphocytic choriomeningitis virus clone 13- and reduced viral load in DOCILE-infected animals. Further experiments showed that NKG2D triggered regulatory NK cell functions, which were mediated by perforin, and limited T-cell responses. Therefore, we identified an important role of regulatory NK cells in limiting T-cell immunity during virus infection.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Communicable Diseases/immunology , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus , Analysis of Variance , Animals , Cytotoxicity Tests, Immunologic , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Histological Techniques , Interferon-alpha/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily K/immunology , Real-Time Polymerase Chain Reaction
8.
Immunol Rev ; 241(1): 164-79, 2011 May.
Article in English | MEDLINE | ID: mdl-21488897

ABSTRACT

Self-reactive T cells in the body are controlled by mechanisms of peripheral tolerance that limit their activation and induction of immune pathology. Our understanding of these mechanisms has been advanced by the use of tissue-specific promoters to express neo-self-antigens. Here, we present findings using the RIP-gp (rat insulin promoter-glycoprotein) transgenic mouse, which expresses the lymphocytic choriomeningitis virus glycoprotein (LCMV-gp) specifically in the pancreatic ß islet cells. T cells responsive to this antigen remain ignorant of the LCMV-gp expressed by the islets, and breaking tolerance is dependent upon the maturation status of antigen-presenting cells, the avidity of the T-cell receptor ligation, and the level of major histocompatibility complex expression in the pancreas. Furthermore, decreased activity of Casitas B-lineage lymphoma b, a negative regulator of T-cell receptor signaling, can allow recognition and destruction of the pancreatic islets. This review discusses the roles of these factors in the context of anti-tissue responses, both in the setting of autoimmunity and in anti-tumor immunity.


Subject(s)
Autoantigens/metabolism , Autoimmune Diseases/immunology , Glycoproteins/metabolism , Insulin-Secreting Cells/metabolism , Neoplasms/immunology , Viral Envelope Proteins/metabolism , Animals , Antigens, Neoplasm/immunology , Autoantigens/genetics , Autoantigens/immunology , Glycoproteins/genetics , Humans , Insulin/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Transgenic , Organ Specificity , Promoter Regions, Genetic/genetics , Self Tolerance , Transgenes/genetics , Viral Envelope Proteins/genetics
9.
J Immunol ; 186(4): 1923-33, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21220694

ABSTRACT

GM-CSF is an important cytokine involved in myeloid differentiation and inflammatory processes. Signaling through the GM-CSFR also plays a critical role in the generation of monocyte-derived dendritic cells (DC). In this article, we report that the Src-like adaptor protein (SLAP) functions as a negative regulator of the GM-CSFR. In bone marrow-derived DC (BM-DC) lacking SLAP and the closely related SLAP2, downregulation of GM-CSFRß is impaired, leading to enhanced phosphorylation of Jak2 and prolonged activation of Akt and Erk1/2 in response to GM-CSF stimulation. Compared with wild-type bone marrow, SLAP/SLAP2(-/-) bone marrow gave rise to similar numbers of CD11c(+) and CD11b(+) DC, but SLAP/SLAP2(-/-) BM-DC failed to acquire high levels of MHC class II, CD80, and CD86, indicating an impairment in maturation. Furthermore, MHC class II expression in SLAP/SLAP2(-/-) BM-DC was rescued by decreasing GM-CSF concentration, suggesting that enhanced GM-CSF signaling mediates the block in maturation. In addition, SLAP/SLAP2(-/-) BM-DC produced less IL-12 and TNF-α in response to LPS compared with controls and failed to stimulate T cells in an MLR. Ag-specific T cell activation assays showed that SLAP/SLAP2(-/-) BM-DC were less robust at inducing IFN-γ secretion by DO11.10 T cells. These results indicated that SLAP-mediated GM-CSFR regulation is important for the generation of functionally mature monocytic DC.


Subject(s)
Cell Differentiation/immunology , Cytokine Receptor Common beta Subunit/physiology , Dendritic Cells/immunology , Growth Inhibitors/physiology , Monocytes/immunology , Proto-Oncogene Proteins pp60(c-src)/physiology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Differentiation/genetics , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Cytokine Receptor Common beta Subunit/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Down-Regulation/genetics , Down-Regulation/immunology , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Mice , Mice, Knockout , Mice, Transgenic , Monocytes/metabolism , Monocytes/pathology , Proto-Oncogene Proteins pp60(c-src)/deficiency , Proto-Oncogene Proteins pp60(c-src)/genetics , Signal Transduction/genetics
10.
Front Immunol ; 14: 1127085, 2023.
Article in English | MEDLINE | ID: mdl-36926348

ABSTRACT

RELA haploinsufficiency is a recently described autoinflammatory condition presenting with intermittent fevers and mucocutaneous ulcerations. The RELA gene encodes the p65 protein, one of five NF-κB family transcription factors. As RELA is an essential regulator of mucosal homeostasis, haploinsufficiency leads to decreased NF-κB signaling which promotes TNF-driven mucosal apoptosis with impaired epithelial recovery. Thus far, only eight cases have been reported in the literature. Here, we report four families with three novel and one previously described pathogenic variant in RELA. These four families included 23 affected individuals for which genetic testing was available in 16. Almost half of these patients had been previously diagnosed with more common rheumatologic entities (such as Behcet's Disease; BD) prior to the discovery of their pathogenic RELA variants. The most common clinical features were orogenital ulcers, rash, joint inflammation, and fever. The least common were conjunctivitis and recurrent infections. Clinical variability was remarkable even among familial cases, and incomplete penetrance was observed. Patients in our series were treated with a variety of medications, and benefit was observed with glucocorticoids, colchicine, and TNF inhibitors. Altogether, our work adds to the current literature and doubles the number of reported cases with RELA-Associated Inflammatory Disease (RAID). It reaffirms the central importance of the NF-κB pathway in immunity and inflammation, as well as the important regulatory role of RELA in mucosal homeostasis. RELA associated inflammatory disease should be considered in all patients with BD, particularly those with early onset and/or with a strong family history.


Subject(s)
Behcet Syndrome , NF-kappa B , Humans , NF-kappa B/metabolism , Behcet Syndrome/drug therapy , Behcet Syndrome/genetics , Genetic Testing , Inflammation/genetics , Signal Transduction/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
11.
Pediatr Rheumatol Online J ; 19(1): 54, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33892719

ABSTRACT

BACKGROUND: Biallelic loss-of-function variants in NCF1 lead to reactive oxygen species deficiency and chronic granulomatous disease (CGD). Heterozygosity for the p.Arg90His variant in NCF1 has been associated with susceptibility to systemic lupus erythematosus, rheumatoid arthritis, and Sjögren's syndrome in adult patients. This study demonstrates the association of the homozygous p.Arg90His variant with interferonopathy with features of autoinflammation and autoimmunity in a pediatric patient. CASE PRESENTATION: A 5-year old female of Indian ancestry with early-onset recurrent fever and headache, and persistently elevated antinuclear, anti-Ro, and anti-La antibodies was found to carry the homozygous p.Arg90His variant in NCF1 through exome sequencing. Her unaffected parents and three other siblings were carriers for the mutant allele. Because the presence of two NCF1 pseudogenes, this variant was confirmed by independent genotyping methods. Her intracellular neutrophil oxidative burst and NCF1 expression levels were normal, and no clinical features of CGD were apparent. Gene expression analysis in peripheral blood detected an interferon gene expression signature, which was further supported by cytokine analyses of supernatants of cultured patient's cells. These findings suggested that her inflammatory disease is at least in part mediated by type I interferons. While her fever episodes responded well to systemic steroids, treatment with the JAK inhibitor tofacitinib resulted in decreased serum ferritin levels and reduced frequency of fevers. CONCLUSION: Homozygosity for p.Arg90His in NCF1 should be considered contributory in young patients with an atypical systemic inflammatory antecedent phenotype that may evolve into autoimmunity later in life. The complex genomic organization of NCF1 poses a difficulty for high-throughput genotyping techniques and variants in this gene should be carefully evaluated when using the next generation and Sanger sequencing technologies. The p.Arg90His variant is found at a variable allele frequency in different populations, and is higher in people of South East Asian ancestry. In complex genetic diseases such as SLE, other rare and common susceptibility alleles might be necessary for the full disease expressivity.


Subject(s)
Autoimmune Diseases/genetics , Interferons , NADPH Oxidases/genetics , Child, Preschool , Female , Homozygote , Humans , Pedigree
12.
Nat Genet ; 53(4): 500-510, 2021 04.
Article in English | MEDLINE | ID: mdl-33782605

ABSTRACT

Spleen tyrosine kinase (SYK) is a critical immune signaling molecule and therapeutic target. We identified damaging monoallelic SYK variants in six patients with immune deficiency, multi-organ inflammatory disease such as colitis, arthritis and dermatitis, and diffuse large B cell lymphomas. The SYK variants increased phosphorylation and enhanced downstream signaling, indicating gain of function. A knock-in (SYK-Ser544Tyr) mouse model of a patient variant (p.Ser550Tyr) recapitulated aspects of the human disease that could be partially treated with a SYK inhibitor or transplantation of bone marrow from wild-type mice. Our studies demonstrate that SYK gain-of-function variants result in a potentially treatable form of inflammatory disease.


Subject(s)
Arthritis/genetics , Colitis/genetics , Dermatitis/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Syk Kinase/genetics , Adult , Animals , Arthritis/immunology , Arthritis/pathology , Arthritis/therapy , Base Sequence , Bone Marrow Transplantation , Colitis/immunology , Colitis/pathology , Colitis/therapy , Dermatitis/immunology , Dermatitis/pathology , Dermatitis/therapy , Family , Female , Gene Expression , Gene Knock-In Techniques , Humans , Infant , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Male , Mice , Mice, Knockout , Middle Aged , Mutation , Pedigree , Protein Kinase Inhibitors/pharmacology , Syk Kinase/antagonists & inhibitors , Syk Kinase/deficiency
13.
Cell Immunol ; 250(1-2): 55-67, 2007.
Article in English | MEDLINE | ID: mdl-18313652

ABSTRACT

Virus-based recombinant vaccines have proven highly effective at generating protective CD8+ T cell responses. Multiple vector platforms are available, however, little is known about the relative influence of the different vectors on the transgene-specific CD8+ T cell population. To address this question, we compared several characteristics of the CD8+ T cell response elicited by recombinant adenovirus (rAd) and vaccinia virus (rVV). We found that following rAd immunization the transgene-specific CD8+ T cell response peaked around day 12 and was larger and more sustained than the response produced by rVV. In addition, the CD8+ T cell response generated by rAd was directed primarily against the transgene, whereas the CD8+ T cell response produced by rVV principally targeted the vector backbone. In addition, we also observed that transgene selection also impacted on the magnitude of the CD8+ T cell response elicited by both vectors. Despite differences in the magnitude of the anti-transgene CD8+ T cell response, both vectors elicited CD8+ T cell populations with similar cytokine production, functional avidity and cytolytic activity. In addition, plasmid priming prior to immunization with either rAd or rVV only impacted the magnitude of the transgene gene specific CD8+ T cell response. Our study demonstrates that both vector and transgene selection can influence the magnitude of the CD8+ T cell response, but they do not influence functionality.


Subject(s)
Adenoviridae/genetics , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/genetics , Recombinant Proteins/genetics , Vaccinia virus/immunology , Animals , Antigens/genetics , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Recombinant Proteins/biosynthesis , Transgenes/genetics , Vaccinia virus/genetics , Viral Proteins/genetics , Viral Proteins/immunology
15.
PLoS One ; 9(3): e92380, 2014.
Article in English | MEDLINE | ID: mdl-24647761

ABSTRACT

Vaccines for cancer immunotherapy are of interest but in general have not yet achieved the desired therapeutic efficacy in clinical trials. We present here a novel model to evaluate vaccine strategies by following tissue destruction in a transgenic model, where a defined antigen is expressed on pancreatic islets. We found that the transfer of syngeneic antigen-pulsed dendritic cells (DCs) resulted in autoimmune cytotoxic T-lymphocyte activation that was not observed following vaccinations that were based on peptides and adjuvants. Importantly, the induction of diabetes by DC transfer is dependent upon the maturation of DCs prior to transfer. Furthermore, diabetes induction only occurred if DCs were pulsed with the immunodominant epitope in addition to at least one other peptide, suggesting greater cytolytic activity upon engagement of multiple T-cell specificities. While the tumor environment undoubtedly will be more complex than healthy tissue, the insights gained through this model provide useful information on variables that can affect CD8-mediated tissue cytolysis in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Peptides/pharmacology , Animals , Cells, Cultured , Humans , Mice , T-Lymphocytes, Cytotoxic/immunology
16.
J Clin Invest ; 123(1): 315-28, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23202731

ABSTRACT

Despite efforts to understand and treat acute myeloid leukemia (AML), there remains a need for more comprehensive therapies to prevent AML-associated relapses. To identify new therapeutic strategies for AML, we screened a library of on- and off-patent drugs and identified the antimalarial agent mefloquine as a compound that selectively kills AML cells and AML stem cells in a panel of leukemia cell lines and in mice. Using a yeast genome-wide functional screen for mefloquine sensitizers, we identified genes associated with the yeast vacuole, the homolog of the mammalian lysosome. Consistent with this, we determined that mefloquine disrupts lysosomes, directly permeabilizes the lysosome membrane, and releases cathepsins into the cytosol. Knockdown of the lysosomal membrane proteins LAMP1 and LAMP2 resulted in decreased cell viability, as did treatment of AML cells with known lysosome disrupters. Highlighting a potential therapeutic rationale for this strategy, leukemic cells had significantly larger lysosomes compared with normal cells, and leukemia-initiating cells overexpressed lysosomal biogenesis genes. These results demonstrate that lysosomal disruption preferentially targets AML cells and AML progenitor cells, providing a rationale for testing lysosomal disruption as a novel therapeutic strategy for AML.


Subject(s)
Intracellular Membranes/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Lysosomes/metabolism , Neoplastic Stem Cells/metabolism , Animals , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Cell Survival/drug effects , Female , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Intracellular Membranes/pathology , K562 Cells , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomes/genetics , Lysosomes/physiology , Male , Mefloquine/pharmacokinetics , Mefloquine/pharmacology , Mice , Neoplastic Stem Cells/pathology , Permeability/drug effects , Saccharomyces cerevisiae/genetics
17.
PLoS One ; 7(4): e34343, 2012.
Article in English | MEDLINE | ID: mdl-22529912

ABSTRACT

Changes in expression and localization of proteins that regulate cell and tissue polarity are frequently observed in carcinoma. However, the mechanisms by which changes in cell polarity proteins regulate carcinoma progression are not well understood. Here, we report that loss of polarity protein expression in epithelial cells primes them for cooperation with oncogenes or changes in tissue microenvironment to promote invasive behavior. Activation of ErbB2 in cells lacking the polarity regulators Scribble, Dlg1 or AF-6, induced invasive properties. This cooperation required the ability of ErbB2 to regulate the Par6/aPKC polarity complex. Inhibition of the ErbB2-Par6 pathway was sufficient to block ErbB2-induced invasion suggesting that two polarity hits may be needed for ErbB2 to promote invasion. Interestingly, in the absence of ErbB2 activation, either a combined loss of two polarity proteins, or exposure of cells lacking one polarity protein to cytokines IL-6 or TNFα induced invasive behavior in epithelial cells. We observed the invasive behavior only when cells were plated on a stiff matrix (Matrigel/Collagen-1) and not when plated on a soft matrix (Matrigel alone). Cells lacking two polarity proteins upregulated expression of EGFR and activated Akt. Inhibition of Akt activity blocked the invasive behavior identifying a mechanism by which loss of polarity promotes invasion of epithelial cells. Thus, we demonstrate that loss of polarity proteins confers phenotypic plasticity to epithelial cells such that they display normal behavior under normal culture conditions but display aggressive behavior in response to activation of oncogenes or exposure to cytokines.


Subject(s)
Cell Movement , Cell Polarity , Cellular Microenvironment , Epithelial Cells/metabolism , Oncogenes , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Movement/genetics , Cell Polarity/genetics , Cytokines/metabolism , Discs Large Homolog 1 Protein , Epithelial Cells/cytology , Epithelial Cells/physiology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression , Gene Expression Regulation , Humans , Inflammation Mediators/metabolism , Kinesins/genetics , Kinesins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Myosins/genetics , Myosins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
18.
PLoS One ; 6(9): e23940, 2011.
Article in English | MEDLINE | ID: mdl-21931625

ABSTRACT

Although toll-like receptor (TLR) signals are critical for promoting antigen presenting cell maturation, it remains unclear how stimulation via different TLRs influence dendritic cell (DC) function and the subsequent adaptive response in vivo. Furthermore, the relationship between TLR-induced cytokine production by DCs and the consequences on the induction of a functional immune response is not clear. We have established a murine model to examine whether TLR3 or TLR4 mediated DC maturation has an impact on the cytokines required to break tolerance and induce T-cell-mediated autoimmunity. Our study demonstrates that IL-12 is not absolutely required for the induction of a CD8 T-cell-mediated tissue specific immune response, but rather the requirement for IL-12 is determined by the stimuli used to mature the DCs. Furthermore, we found that IFNα is a critical pathogenic component of the cytokine milieu that circumvents the requirement for IL-12 in the induction of autoimmunity. These studies illustrate how different TLR stimuli have an impact on DC function and the induction of immunity.


Subject(s)
Autoimmunity/immunology , Cytokines/metabolism , Immune Tolerance/immunology , Toll-Like Receptors/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glycoproteins/genetics , Interferon-alpha/biosynthesis , Interferon-alpha/metabolism , Interleukin-12/biosynthesis , Interleukin-12/deficiency , Interleukin-12/metabolism , Lipopolysaccharides/immunology , Lymphocytic choriomeningitis virus/genetics , Mice , Toll-Like Receptors/immunology
19.
Nat Med ; 17(12): 1663-7, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22081022

ABSTRACT

Mature dendritic cells (DCs) are crucial for the induction of adaptive immune responses and perturbed DC homeostasis can result in autoimmune disease. Either uncontrolled expansion or enhanced survival of DCs can result in a variety of autoimmune diseases in mouse models. In addition, increased maturation signals, through overexpression of surface Toll-like receptors (TLRs) or stimulation by type I interferon (IFN), has been associated with systemic autoimmunity. Whereas recent studies have focused on identifying factors required for initiating the maturation process, the possibility that resting DCs also express molecules that 'hold' them in an immature state has generally not been considered. Here we show that nuclear factor-κB1 (NF-κB1) is crucial for maintaining the resting state of DCs. Self-antigen-pulsed unstimulated DCs that do not express NF-κB1 were able to activate CD8(+) T lymphocytes and induce autoimmunity. We further show that NF-κB1 negatively regulates the spontaneous production of tumor necrosis factor-α (TNF-α), which is associated with increased granzyme B expression in cytotoxic T lymphocytes (CTLs). These findings provide a new perspective on functional DC maturation and a potential mechanism that may account for pathologic T cell activation.


Subject(s)
Adaptive Immunity , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , NF-kappa B/metabolism , Animals , Autoantigens/immunology , Autoantigens/metabolism , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Flow Cytometry , Gene Expression Regulation , Granzymes/metabolism , Immunohistochemistry , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
Nat Med ; 15(5): 528-36, 2009 May.
Article in English | MEDLINE | ID: mdl-19396174

ABSTRACT

Identifying key factors that enhance immune responses is crucial for manipulating immunity to tumors. We show that after a vaccine-induced immune response, adjuvant interleukin-7 (IL-7) improves antitumor responses and survival in an animal model. The improved immune response is associated with increased IL-6 production and augmented T helper type 17 cell differentiation. Furthermore, IL-7 modulates the expression of two ubiquitin ligases: Casitas B-lineage lymphoma b (Cbl-b), a negative regulator of T cell activation, is repressed, and SMAD-specific E3 ubiquitin protein ligase-2 (Smurf2) is enhanced, which antagonizes transforming growth factor-beta signaling. Notably, we show that although short term IL-7 therapy potently enhances vaccine-mediated immunity, in the absence of vaccination it is inefficient in promoting antitumor immune responses, despite inducing homeostatic proliferation of T cells. The ability of adjuvant IL-7 to antagonize inhibitory networks at the cellular and molecular level has major implications for immunotherapy in the treatment of tumors.


Subject(s)
Arenaviridae Infections/immunology , Cancer Vaccines/therapeutic use , Immunotherapy/methods , Interleukin-7/therapeutic use , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Neoplasms/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Count , Mice , Neoplasms, Experimental/immunology , Survivors , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL