Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
J Allergy Clin Immunol ; 147(6): 2134-2145.e20, 2021 06.
Article in English | MEDLINE | ID: mdl-33378691

ABSTRACT

BACKGROUND: NLRP1 is an innate immune sensor that can form cytoplasmic inflammasome complexes. Polymorphisms in NLRP1 are linked to asthma; however, there is currently no functional or mechanistic explanation for this. OBJECTIVE: We sought to clarify the role of NLRP1 in asthma pathogenesis. METHODS: Results from the GALA II cohort study were used to identify a link between NLRP1 and asthma in Mexican Americans. In vitro and in vivo models for NLRP1 activation were applied to investigate the role of this inflammasome in asthma at the molecular level. RESULTS: We document the association of an NLRP1 haplotype with asthma for which the single nucleotide polymorphism rs11651270 (M1184V) individually is the most significant. Surprisingly, M1184V increases NLRP1 activation in the context of N-terminal destabilization, but decreases NLRP1 activation on dipeptidyl peptidase 9 inhibition. In vitro studies demonstrate that M1184V increases binding to dipeptidyl peptidase 9, which can account for its inhibitory role in this context. In addition, in vivo data from a mouse model of airway inflammation reveal a protective role for NLRP1 inflammasome activation reducing eosinophilia in this setting. CONCLUSIONS: Linking our in vitro and in vivo results, we found that the NLRP1 variant M1184V reduces inflammasome activation in the context of dipeptidyl peptidase 9 inhibition and could thereby increase asthma severity. Our studies may have implications for the treatment of asthma in patients carrying this variant of NLRP1.


Subject(s)
Alleles , Asthma/etiology , Asthma/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Inflammasomes/metabolism , Mutation , NLR Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Asthma/diagnosis , Cell Line , Disease Models, Animal , Disease Susceptibility , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Genetic Predisposition to Disease , Humans , Mice , Mice, Knockout , NLR Proteins/chemistry , NLR Proteins/metabolism , Polymorphism, Single Nucleotide , Structure-Activity Relationship , Trauma Severity Indices
3.
Cell Death Discov ; 6(1): 114, 2020 Oct 31.
Article in English | MEDLINE | ID: mdl-33298868

ABSTRACT

Despite having one of the lowest survival rates of all cancers, there have been no new approved treatments for malignant pleural mesothelioma (MPM) in over a decade. Standard-of-care treatment relies on Cisplatin plus Pemetrexed chemotherapy. Here, we tested a suite of BH3-mimetic drugs targeting BCL-2 pro-survival proteins of the intrinsic apoptotic pathway. We found BCL-XL is the dominant pro-survival protein in a panel of cell lines in vitro, though potent, synergistic cell killing occurred with MCL-1 co-targeting. This correlates with high-level expression of BCL-XL and MCL-1 in cell lines and a large cohort of patient tumour samples. BCL-XL inhibition combined with Cisplatin also enhanced cell killing. In vivo BCL-XL inhibition was as effective as Cisplatin, and the combination enhanced tumour growth control and survival. Genetic ablation of MCL-1 also enhanced the effects of BCL-XL inhibitors, in vivo. Combined, these data provide a compelling rationale for the clinical investigation of BH3-mimetics targeting BCL-XL in MPM.

4.
Cancer Immunol Res ; 8(4): 428-435, 2020 04.
Article in English | MEDLINE | ID: mdl-31992566

ABSTRACT

Persistent activation of the latent transcription factor STAT3 is observed in gastric tumor epithelial and immune cells and is associated with a poor patient prognosis. Although targeting STAT3-activating upstream kinases offers therapeutically viable targets with limited specificity, direct inhibition of STAT3 remains challenging. Here we provide functional evidence that myeloid-specific hematopoietic cell kinase (HCK) activity can drive STAT3-dependent epithelial tumor growth in mice and is associated with alternative macrophage activation alongside matrix remodeling and tumor cell invasion. Accordingly, genetic reduction of HCK expression in bone marrow-derived cells or systemic pharmacologic inhibition of HCK activity suppresses alternative macrophage polarization and epithelial STAT3 activation, and impairs tumor growth. These data validate HCK as a molecular target for the treatment of human solid tumors harboring excessive STAT3 activity.


Subject(s)
Proto-Oncogene Proteins c-hck/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/drug therapy , Animals , Female , Humans , Macrophage Activation/drug effects , Male , Mice , Mice, Transgenic , Phosphorylation/drug effects , Proto-Oncogene Proteins c-hck/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Survival Rate
5.
Nat Commun ; 10(1): 2735, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227713

ABSTRACT

The contribution of mast cells in the microenvironment of solid malignancies remains controversial. Here we functionally assess the impact of tumor-adjacent, submucosal mast cell accumulation in murine and human intestinal-type gastric cancer. We find that genetic ablation or therapeutic inactivation of mast cells suppresses accumulation of tumor-associated macrophages, reduces tumor cell proliferation and angiogenesis, and diminishes tumor burden. Mast cells are activated by interleukin (IL)-33, an alarmin produced by the tumor epithelium in response to the inflammatory cytokine IL-11, which is required for the growth of gastric cancers in mice. Accordingly, ablation of the cognate IL-33 receptor St2 limits tumor growth, and reduces mast cell-dependent production and release of the macrophage-attracting factors Csf2, Ccl3, and Il6. Conversely, genetic or therapeutic macrophage depletion reduces tumor burden without affecting mast cell abundance. Therefore, tumor-derived IL-33 sustains a mast cell and macrophage-dependent signaling cascade that is amenable for the treatment of gastric cancer.


Subject(s)
Interleukin-33/immunology , Macrophages/immunology , Mast Cells/immunology , Stomach Neoplasms/immunology , Aminopyridines/administration & dosage , Animals , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cromolyn Sodium/administration & dosage , Disease Models, Animal , Epithelium/immunology , Epithelium/pathology , Female , Gastric Mucosa/cytology , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Humans , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Male , Mice , Mice, Transgenic , Pyrroles/administration & dosage , Signal Transduction/drug effects , Signal Transduction/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tissue Array Analysis , Tumor Microenvironment/immunology
6.
Cancer Cell ; 31(4): 563-575.e5, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28399411

ABSTRACT

Aberrant activation of the SRC family kinase hematopoietic cell kinase (HCK) triggers hematological malignancies as a tumor cell-intrinsic oncogene. Here we find that high HCK levels correlate with reduced survival of colorectal cancer patients. Likewise, increased Hck activity in mice promotes the growth of endogenous colonic malignancies and of human colorectal cancer cell xenografts. Furthermore, tumor-associated macrophages of the corresponding tumors show a pronounced alternatively activated endotype, which occurs independently of mature lymphocytes or of Stat6-dependent Th2 cytokine signaling. Accordingly, pharmacological inhibition or genetic reduction of Hck activity suppresses alternative activation of tumor-associated macrophages and the growth of colon cancer xenografts. Thus, Hck may serve as a promising therapeutic target for solid malignancies.


Subject(s)
Colonic Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Proto-Oncogene Proteins c-hck/metabolism , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Macrophage Activation/genetics , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-hck/antagonists & inhibitors , Proto-Oncogene Proteins c-hck/genetics , Pyrimidines/pharmacology , Pyrroles/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction , Stromal Cells/metabolism , Xenograft Model Antitumor Assays
7.
Oncoscience ; 4(11-12): 150-151, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29344543
8.
J Gastroenterol Hepatol ; 31(7): 1257-72, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26809278

ABSTRACT

Gastric cancer is the third leading cause of cancer-related mortality worldwide. This is in part due to the asymptomatic nature of the disease, which often results in late-stage diagnosis, at which point there are limited treatment options. Even when treated successfully, gastric cancer patients have a high risk of tumor recurrence and acquired drug resistance. It is vital to gain a better understanding of the molecular mechanisms underlying gastric cancer pathogenesis to facilitate the design of new-targeted therapies that may improve patient survival. A number of chemically and genetically engineered mouse models of gastric cancer have provided significant insight into the contribution of genetic and environmental factors to disease onset and progression. This review outlines the strengths and limitations of current mouse models of gastric cancer and their relevance to the pre-clinical development of new therapeutics.


Subject(s)
Disease Models, Animal , Mice, Inbred Strains , Mice, Transgenic , Stomach Neoplasms/etiology , Animals , Gastrins , Helicobacter Infections , Helicobacter felis , Helicobacter pylori , Methylnitrosourea , Molecular Targeted Therapy , Stomach Neoplasms/classification , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy
9.
Oncotarget ; 6(18): 15752-71, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26087188

ABSTRACT

The hematopoietic cell kinase (HCK) is a member of the SRC family of cytoplasmic tyrosine kinases (SFKs), and is expressed in cells of the myeloid and B-lymphocyte cell lineages. Excessive HCK activation is associated with several types of leukemia and enhances cell proliferation and survival by physical association with oncogenic fusion proteins, and with functional interactions with receptor tyrosine kinases. Elevated HCK activity is also observed in many solid malignancies, including breast and colon cancer, and correlates with decreased patient survival rates. HCK enhances the secretion of growth factors and pro-inflammatory cytokines from myeloid cells, and promotes macrophage polarization towards a wound healing and tumor-promoting alternatively activated phenotype. Within tumor associated macrophages, HCK stimulates the formation of podosomes that facilitate extracellular matrix degradation, which enhance immune and epithelial cell invasion. By virtue of functional cooperation between HCK and bona fide oncogenic tyrosine kinases, excessive HCK activation can also reduce drug efficacy and contribute to chemo-resistance, while genetic ablation of HCK results in minimal physiological consequences in healthy mice. Given its known crystal structure, HCK therefore provides an attractive therapeutic target to both, directly inhibit the growth of cancer cells, and indirectly curb the source of tumor-promoting changes in the tumor microenvironment.


Subject(s)
Neoplasms/immunology , Neoplasms/therapy , Proto-Oncogene Proteins c-hck/immunology , Animals , Cell Proliferation/physiology , Humans , Signal Transduction
10.
Stem Cell Res ; 11(3): 1222-33, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24029687

ABSTRACT

Tissue resident mesenchymal stromal cells (MSCs) contribute to tissue regeneration through various mechanisms, including the secretion of trophic factors that act directly on epithelial stem cells to promote epithelialization. However, MSCs in tissues constitute a heterogeneous population of stromal cells and different subtypes may have different functions. In this study we show that CD166(neg) and CD166(pos) lung stromal cells have different proliferative and differentiative potential. CD166(neg) lung stromal cells exhibit high proliferative potential with the capacity to differentiate along the lipofibroblastic and myofibroblastic lineages, whereas CD166(pos) lung stromal cells have limited proliferative potential and are committed to the myofibroblastic lineage. Moreover, we show that CD166(pos) lung stromal cells do not share the same epithelial-supportive capacity as their CD166(neg) counterparts, which support the growth of lung epithelial stem cell (EpiSPC) colonies in vitro. In addition, ex vivo expansion of lung stromal cells also resulted in the loss of epithelial-supportive capacity, which could be reinstated by inhibition of the TGF-ß signaling pathway. We show that epithelial-supportive capacity correlated with the level of FGF-10 expression and the reactivation of several lung development-associated genes. In summary, these studies suggest that TGF-ß signaling in stromal cells acts upstream of FGF-10 to regulate epithelial stem cell growth in the adult lung.


Subject(s)
Epithelial Cells/cytology , Fibroblast Growth Factor 10/metabolism , Lung/physiology , Signal Transduction , Stem Cells/metabolism , Stromal Cells/cytology , Transforming Growth Factor beta/metabolism , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Animals , Benzamides/pharmacology , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Dioxoles/pharmacology , Epithelial Cells/metabolism , Female , Lung/cytology , Mice , Mice, Inbred C57BL , Myofibroblasts/cytology , Myofibroblasts/metabolism , Signal Transduction/drug effects , Stem Cells/cytology , Stromal Cells/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Up-Regulation
11.
Proc Am Thorac Soc ; 9(3): 177-82, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22802294

ABSTRACT

Pulmonary fibrosis is a devastating, relentlessly progressive, and lethal disease. There is a significant unmet need for effective treatment since currently no FDA-approved therapies exist. Current thinking suggests that idiopathic pulmonary fibrosis (IPF) is initiated by pathways similar to normal wound healing, but relentless fibrosis occurs secondary to absent or defective inhibitory mechanisms that normally terminate wound healing. The heterogeneous pathological presentation of fibrosis suggests that the anatomic location and origin of fibroblasts and other cells might be critical for their phenotype and function and will impact on strategies to prevent or treat fibrotic lung diseases. This review summarizes our current understanding of the pathobiology of IPF, with a specific focus on the role of STAT3 in regulating cellular responses that may contribute to or inhibit pro-fibrotic processes. An improved understanding of the complex cell-type specific roles that this transcription factor plays in normal lung and in fibrosis is required to determine its suitability as an effective therapeutic target.


Subject(s)
Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Pulmonary Alveoli/metabolism , STAT3 Transcription Factor/metabolism , Apoptosis , Cellular Senescence , Epithelial Cells/metabolism , Humans , Inflammation/metabolism , Pulmonary Alveoli/cytology
12.
EMBO Mol Med ; 4(9): 939-51, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684844

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal disease that is unresponsive to current therapies and characterized by excessive collagen deposition and subsequent fibrosis. While inflammatory cytokines, including interleukin (IL)-6, are elevated in IPF, the molecular mechanisms that underlie this disease are incompletely understood, although the development of fibrosis is believed to depend on canonical transforming growth factor (TGF)-ß signalling. We examined bleomycin-induced inflammation and fibrosis in mice carrying a mutation in the shared IL-6 family receptor gp130. Using genetic complementation, we directly correlate the extent of IL-6-mediated, excessive Stat3 activity with inflammatory infiltrates in the lung and the severity of fibrosis in corresponding gp130(757F) mice. The extent of fibrosis was attenuated in B lymphocyte-deficient gp130(757F);µMT(-/-) compound mutant mice, but fibrosis still occurred in their Smad3(-/-) counterparts consistent with the capacity of excessive Stat3 activity to induce collagen 1α1 gene transcription independently of canonical TGF-ß/Smad3 signalling. These findings are of therapeutic relevance, since we confirmed abundant STAT3 activation in fibrotic lungs from IPF patients and showed that genetic reduction of Stat3 protected mice from bleomycin-induced lung fibrosis.


Subject(s)
Interleukin-6/metabolism , Pulmonary Fibrosis/genetics , STAT3 Transcription Factor/biosynthesis , Animals , Bleomycin/toxicity , Cytokine Receptor gp130/deficiency , Genetic Complementation Test , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Smad3 Protein/deficiency
13.
J Immunol ; 182(12): 7990-6, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494324

ABSTRACT

Lung neutrophilia is common to a variety of lung diseases. The production of reactive oxygen and nitrogen species during neutrophil oxidative burst has been associated with protein and DNA damage. Myeloperoxidase (MPO) is an enzyme stored in the azurophilic granula of neutrophils. It is important in host defense because it generates the reactive oxidant hypochlorous acid and has been described to play a role in the activation of neutrophils during extravasation. We hypothesized that MPO contributes directly to the development of acute lung neutrophilia via stimulation of neutrophil extravasation and indirectly to the subsequent production of cytokines and chemokines in the lung. To test this hypothesis, wild-type (WT) and Mpo(-/-) mice were given a single LPS instillation, after which the development of neutrophil-dominated lung inflammation, oxidative stress, and cytokine and chemokine levels were examined. Mpo(-/-) mice demonstrated a decreased lung neutrophilia that peaked earlier than neutrophilia in WT mice, which can be explained by decreased neutrophil chemoattractant levels in LPS-exposed Mpo(-/-) compared with WT mice. However, oxidative stress levels were not different in LPS-exposed WT and Mpo(-/-) mice. Furthermore, in vivo findings were confirmed by in vitro studies, using isolated neutrophils. These results indicate that MPO promotes the development of lung neutrophilia and indirectly influences subsequent chemokine and cytokine production by other cell types in the lung.


Subject(s)
Cytokines/biosynthesis , Cytokines/immunology , Lipopolysaccharides/pharmacology , Peroxidase/metabolism , Pneumonia/enzymology , Pneumonia/immunology , Acute Disease , Animals , Cell Movement , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/enzymology , Peroxidase/deficiency , Peroxidase/genetics , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...