Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Histochem Cytochem ; 69(9): 611-615, 2021 09.
Article in English | MEDLINE | ID: mdl-34353148

ABSTRACT

With the advent of checkpoint inhibitors, there is increasing need to study the dynamics of CD8+ T-cells in the tumor microenviroment. In this article, we describe a semi-automated method to quantify and interrogate spatial relationships between T-cells and collagenous stroma in human and mouse tissue samples. The assay combines CD8 immunohistochemistry with modified Masson's trichrome. Slides are scanned and digital images are analyzed using an adjustable MATLAB algorithm, allowing for high-throughput quantification of cytotoxic T-cells and collagen. This method provides a flexible tool for unbiased quantification of T-cells and their interactions with tumor cells and tumor microenvironment in tissue samples.


Subject(s)
CD8 Antigens/analysis , High-Throughput Screening Assays , Algorithms , Animals , Humans , Immunohistochemistry , Mice , Tumor Microenvironment
2.
Cancer Res ; 80(8): 1656-1668, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31988076

ABSTRACT

The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk for mesothelioma and melanocytic tumors. Here, we show that pancreatic intraepithelial neoplasia driven by oncogenic mutant KrasG12D progressed to pancreatic adenocarcinoma in the absence of BAP1. The Hippo pathway was deregulated in BAP1-deficient pancreatic tumors, with the tumor suppressor LATS exhibiting enhanced ubiquitin-dependent proteasomal degradation. Therefore, BAP1 may limit tumor progression by stabilizing LATS and thereby promoting activity of the Hippo tumor suppressor pathway. SIGNIFICANCE: BAP1 is mutated in a broad spectrum of tumors. Pancreatic Bap1 deficiency causes acinar atrophy but combines with oncogenic Ras to produce pancreatic tumors. BAP1-deficient tumors exhibit deregulation of the Hippo pathway.See related commentary by Brekken, p. 1624.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Hippo Signaling Pathway , Humans , Protein Serine-Threonine Kinases , Signal Transduction , Tumor Suppressor Proteins , Ubiquitin Thiolesterase
3.
Cell Death Differ ; 26(11): 2416-2429, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30824837

ABSTRACT

Emerging research suggests that multiple tumor compartments can influence treatment responsiveness and relapse, yet the search for therapeutic resistance mechanisms remains largely focused on acquired genomic alterations in cancer cells. Here we show how treatment-induced changes occur in multiple tumor compartments during tumor relapse and can reduce benefit of follow-on therapies. By using serial biopsies, next-generation sequencing, and single-cell transcriptomics, we tracked the evolution of multiple cellular compartments within individual lesions during first-line treatment response, relapse, and second-line therapeutic interventions in an autochthonous model of melanoma. We discovered that although treatment-relapsed tumors remained genetically stable, they converged on a shared resistance phenotype characterized by dramatic changes in tumor cell differentiation state, immune infiltration, and extracellular matrix (ECM) composition. Similar alterations in tumor cell differentiation were also observed in more than half of our treatment-relapsed patient tumors. Tumor cell-state changes were coincident with ECM remodeling and increased tumor stiffness, which alone was sufficient to alter tumor cell fate and reduce treatment responses in melanoma cell lines in vitro. Despite the absence of acquired mutations in the targeted pathway, resistant tumors showed significantly decreased responsiveness to second-line therapy intervention within the same pathway. The ability to preclinically model relapse and refractory settings-while capturing dynamics within and crosstalk between all relevant tumor compartments-provides a unique opportunity to better design and sequence appropriate clinical interventions.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Extracellular Matrix/pathology , Melanoma/drug therapy , Melanoma/pathology , Animals , Azetidines/pharmacology , Biomarkers, Tumor/genetics , Cell Line, Tumor , DNA Copy Number Variations/genetics , Drug Resistance, Neoplasm/physiology , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Melanoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Piperidines/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib/pharmacology , Exome Sequencing
4.
Am J Physiol Renal Physiol ; 316(1): F76-F89, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30256127

ABSTRACT

Glomeruli number and size are important for determining the pathogenesis of glomerular disease, chronic kidney disease, and hypertension. Moreover, renal injury can occur in specific cortical layers and alter glomerular spatial distribution. In this study, we present a comprehensive structural analysis of glomeruli in a model of Adriamycin (doxorubicin) nephropathy. Glomeruli are imaged (micro-CT at 10 × 10 × 10 µm3) in kidney specimens from C57Bl/6 mouse cohorts: control treated with saline ( n = 9) and Adriamycin treated with 20 mg/kg Adriamycin ( n = 7). Several indices were examined, including glomerular number, glomerular volume, glomerular volume heterogeneity, and spatial density at each glomerulus and in each cortical layer (superficial, midcortical, and juxtamedullary). In the Adriamycin-treated animals, glomerular number decreased significantly in the left kidney [control: 8,298 ± 221, Adriamycin: 6,781 ± 630 (mean ± SE)] and right kidney (control: 7,317 ± 367, Adriamycin: 5,522 ± 508), and glomerular volume heterogeneity increased significantly in the left kidney (control: 0.642 ± 0.015, Adriamycin: 0.786 ± 0.018) and right kidney (control: 0.739 ± 0.016, Adriamycin: 0.937 ± 0.023). Glomerular spatial density was not affected. Glomerular volume heterogeneity increased significantly in the superficial and midcortical layers of the Adriamycin cohort. Adriamycin did not affect glomerular volume or density metrics in the juxtamedullary region, suggesting a compensatory mechanism of juxtamedullary glomeruli to injury in the outer cortical layers. Left/right asymmetry was observed in kidney size and various glomeruli metrics. The methods presented here can be used to evaluate renal disease models with subtle changes in glomerular endowment locally or across the entire kidney, and they provide an imaging tool to investigate diverse interventions and therapeutic drugs.


Subject(s)
Doxorubicin , Glomerulosclerosis, Focal Segmental/diagnostic imaging , Kidney Glomerulus/diagnostic imaging , X-Ray Microtomography , Algorithms , Animals , Barium Sulfate/administration & dosage , Contrast Media/administration & dosage , Disease Models, Animal , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/pathology , Image Interpretation, Computer-Assisted , Kidney Glomerulus/pathology , Male , Mice, Inbred C57BL , Predictive Value of Tests
5.
J Crohns Colitis ; 12(10): 1191-1199, 2018 Nov 09.
Article in English | MEDLINE | ID: mdl-29912405

ABSTRACT

BACKGROUND: Recent findings suggest that αE expression is enriched on effector T cells and that intestinal αE+ T cells have increased expression of inflammatory cytokines. αE integrin expression is a potential predictive biomarker for response to etrolizumab, a monoclonal antibody against ß7 integrin that targets both α4ß7 and αEß7. We evaluated the prevalence and localization of αE+ cells as well as total αE gene expression in healthy and inflammatory bowel disease patients. METHODS: αE+ cells were identified in ileal and colonic biopsies by immunohistochemistry and counted using an automated algorithm. Gene expression was assessed by quantitative reverse-transcriptase polymerase chain reaction. RESULTS: In both healthy and inflammatory bowel disease patients, significantly more αE+ cells were present in the epithelium and lamina propria of ileal compared with colonic biopsies. αE gene expression levels were also significantly higher in ileal compared with colonic biopsies. Paired biopsies from the same patient showed moderate correlation of αE expression between the ileum and colon. Inflammation did not affect αE expression, and neither endoscopy nor histology scores correlated with αE gene expression. αE expression was not different between patients based on concomitant medication use except 5-aminosalicylic acid. CONCLUSION: αE+ cells, which have been shown to have inflammatory potential, are increased in the ileum in comparison with the colon in both Crohn's disease and ulcerative colitis, as well as in healthy subjects. In inflammatory bowel disease patients, αE levels are stable, regardless of inflammatory status or most concomitant medications, which could support its use as a biomarker for etrolizumab.


Subject(s)
Colon , Ileum , Inflammatory Bowel Diseases , Adult , Antigens, CD , Biopsy/methods , Colon/immunology , Colon/pathology , Correlation of Data , Endoscopy, Digestive System/methods , Female , Gene Expression Profiling , Humans , Ileum/immunology , Ileum/pathology , Immunohistochemistry , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Integrin alpha Chains , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Middle Aged
6.
J Crohns Colitis ; 11(5): 610-620, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28453768

ABSTRACT

BACKGROUND AND AIMS: The αEß7 integrin is crucial for retention of T lymphocytes at mucosal surfaces through its interaction with E-cadherin. Pathogenic or protective functions of these cells during human intestinal inflammation, such as ulcerative colitis [UC], have not previously been defined, with understanding largely derived from animal model data. Defining this phenotype in human samples is important for understanding UC pathogenesis and is of translational importance for therapeutic targeting of αEß7-E-cadherin interactions. METHODS: αEß7+ and αEß7- colonic T cell localization, inflammatory cytokine production and expression of regulatory T cell-associated markers were evaluated in cohorts of control subjects and patients with active UC by immunohistochemistry, flow cytometry and real-time PCR of FACS-purified cell populations. RESULTS: CD4+αEß7+ T lymphocytes from both healthy controls and UC patients had lower expression of regulatory T cell-associated genes, including FOXP3, IL-10, CTLA-4 and ICOS in comparison with CD4+αEß7- T lymphocytes. In UC, CD4+αEß7+ lymphocytes expressed higher levels of IFNγ and TNFα in comparison with CD4+αEß7- lymphocytes. Additionally the CD4+αEß7+ subset was enriched for Th17 cells and the recently described Th17/Th1 subset co-expressing both IL-17A and IFNγ, both of which were found at higher frequencies in UC compared to control. CONCLUSION: αEß7 integrin expression on human colonic CD4+ T cells was associated with increased production of pro-inflammatory Th1, Th17 and Th17/Th1 cytokines, with reduced expression of regulatory T cell-associated markers. These data suggest colonic CD4+αEß7+ T cells are pro-inflammatory and may play a role in UC pathobiology.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis, Ulcerative/immunology , Colon/cytology , Integrins/immunology , Adult , Aged , Case-Control Studies , Colitis, Ulcerative/metabolism , Colon/immunology , Cytokines/metabolism , Female , Flow Cytometry , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Young Adult
7.
Gastroenterology ; 150(2): 477-87.e9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26522261

ABSTRACT

BACKGROUND & AIMS: Etrolizumab is a humanized monoclonal antibody against the ß7 integrin subunit that has shown efficacy vs placebo in patients with moderate to severely active ulcerative colitis (UC). Patients with colon tissues that expressed high levels of the integrin αE gene (ITGAE) appeared to have the best response. We compared differences in colonic expression of ITGAE and other genes between patients who achieved clinical remission with etrolizumab vs those who did. METHODS: We performed a retrospective analysis of data collected from 110 patients with UC who participated in a phase 2 placebo-controlled trial of etrolizumab, as well as from 21 patients with UC or without inflammatory bowel disease (controls) enrolled in an observational study at a separate site. Colon biopsies were collected from patients in both studies and analyzed by immunohistochemistry and gene expression profiling. Mononuclear cells were isolated and analyzed by flow cytometry. We identified biomarkers associated with response to etrolizumab. In the placebo-controlled trial, clinical remission was defined as total Mayo Clinic Score ≤2, with no individual subscore >1, and mucosal healing was defined as endoscopic score ≤1. RESULTS: Colon tissues collected at baseline from patients who had a clinical response to etrolizumab expressed higher levels of T-cell-associated genes than patients who did not respond (P < .05). Colonic CD4(+) integrin αE(+) cells from patients with UC expressed higher levels of granzyme A messenger RNA (GZMA mRNA) than CD4(+) αE(-) cells (P < .0001); granzyme A and integrin αE protein were detected in the same cells. Of patients receiving 100 mg etrolizumab, a higher proportion of those with high levels of GZMA mRNA (41%) or ITGAE mRNA (38%) than those with low levels of GZMA (6%) or ITGAE mRNA (13%) achieved clinical remission (P < .05) and mucosal healing (41% GZMA(high) vs 19% GZMA(low) and 44% ITGAE(high) vs 19% ITGAE(low)). Compared with ITGAE(low) and GZMA(low) patients, patients with ITGAE(high) and GZMA(high) had higher baseline numbers of epithelial crypt-associated integrin αE(+) cells (P < .01 for both), but a smaller number of crypt-associated integrin αE(+) cells after etrolizumab treatment (P < .05 for both). After 10 weeks of etrolizumab treatment, expression of genes associated with T-cell activation and genes encoding inflammatory cytokines decreased by 40%-80% from baseline (P < .05) in patients with colon tissues expressing high levels of GZMA at baseline. CONCLUSIONS: Levels of GZMA and ITGAE mRNAs in colon tissues can identify patients with UC who are most likely to benefit from etrolizumab; expression levels decrease with etrolizumab administration in biomarker(high) patients. Larger, prospective studies of markers are needed to assess their clinical value.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, CD/metabolism , Colitis, Ulcerative/drug therapy , Colon/drug effects , Gastrointestinal Agents/therapeutic use , Granzymes/metabolism , Integrin alpha Chains/metabolism , Antigens, CD/genetics , Biopsy , Clinical Trials, Phase II as Topic , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Colon/enzymology , Colon/pathology , Gene Expression Profiling/methods , Granzymes/genetics , Humans , Immunohistochemistry , Integrin alpha Chains/genetics , Predictive Value of Tests , RNA, Messenger/metabolism , Randomized Controlled Trials as Topic , Remission Induction , Retrospective Studies , Time Factors , Treatment Outcome , Wound Healing/drug effects
8.
Sci Signal ; 8(405): ra122, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26628680

ABSTRACT

Interleukin-2 (IL-2)-inducible T cell kinase (ITK) mediates T cell receptor (TCR) signaling primarily to stimulate the production of cytokines, such as IL-4, IL-5, and IL-13, from T helper 2 (TH2) cells. Compared to wild-type mice, ITK knockout mice are resistant to asthma and exhibit reduced lung inflammation and decreased amounts of TH2-type cytokines in the bronchoalveolar lavage fluid. We found that a small-molecule selective inhibitor of ITK blocked TCR-mediated signaling in cultured TH2 cells, including the tyrosine phosphorylation of phospholipase C-γ1 (PLC-γ1) and the secretion of IL-2 and TH2-type cytokines. Unexpectedly, inhibition of the kinase activity of ITK during or after antigen rechallenge in an ovalbumin-induced mouse model of asthma failed to reduce airway hyperresponsiveness and inflammation. Rather, in mice, pharmacological inhibition of ITK resulted in T cell hyperplasia and the increased production of TH2-type cytokines. Thus, our studies predict that inhibition of the kinase activity of ITK may not be therapeutic in patients with asthma.


Subject(s)
Asthma/immunology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Th2 Cells/immunology , Animals , Asthma/genetics , Asthma/pathology , Cell Death/drug effects , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Female , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Th2 Cells/pathology
9.
Cell Rep ; 12(3): 495-510, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26166562

ABSTRACT

Understanding the regulation of islet cell mass has important implications for the discovery of regenerative therapies for diabetes. The liver plays a central role in metabolism and the regulation of endocrine cell number, but liver-derived factors that regulate α-cell and ß-cell mass remain unidentified. We propose a nutrient-sensing circuit between liver and pancreas in which glucagon-dependent control of hepatic amino acid metabolism regulates α-cell mass. We found that glucagon receptor inhibition reduced hepatic amino acid catabolism, increased serum amino acids, and induced α-cell proliferation in an mTOR-dependent manner. In addition, mTOR inhibition blocked amino-acid-dependent α-cell replication ex vivo and enabled conversion of α-cells into ß-like cells in vivo. Serum amino acids and α-cell proliferation were increased in neonatal mice but fell throughout postnatal development in a glucagon-dependent manner. These data reveal that amino acids act as sensors of glucagon signaling and can function as growth factors that increase α-cell proliferation.


Subject(s)
Amino Acids/metabolism , Glucagon/metabolism , Liver/cytology , Liver/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Proliferation , Metabolism , Mice , Signal Transduction
10.
Stem Cell Reports ; 4(5): 768-79, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25937372

ABSTRACT

The adult prostate possesses a significant regenerative capacity that is of great interest for understanding adult stem cell biology. We demonstrate that leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5) is expressed in a rare population of prostate epithelial progenitor cells, and a castration-resistant Lgr5(+) population exists in regressed prostate tissue. Genetic lineage tracing revealed that Lgr5(+) cells and their progeny are primarily luminal. Lgr5(+) castration-resistant cells are long lived and upon regeneration, both luminal Lgr5(+) cells and basal Lgr5(+) cells expand. Moreover, single Lgr5(+) cells can generate multilineage prostatic structures in renal transplantation assays. Additionally, Lgr5(+) cell depletion revealed that the regenerative potential of the castrated adult prostate depends on Lgr5(+) cells. Together, these data reveal insights into the cellular hierarchy of castration-resistant Lgr5+ cells, indicate a requirement for Lgr5(+) cells during prostatic regeneration, and identify an Lgr5(+) adult stem cell population in the prostate.


Subject(s)
Prostate/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Androgens/pharmacology , Animals , Cell Lineage , Cells, Cultured , Immunohistochemistry , Male , Mice , Mice, Nude , Orchiectomy , Prostate/cytology , Prostate/pathology , Rats , Rats, Sprague-Dawley , Regeneration/drug effects , Stem Cells/cytology , Testosterone/pharmacology
11.
Proc Natl Acad Sci U S A ; 108(23): 9589-94, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21597001

ABSTRACT

Hedgehog (Hh) signaling is critical to the patterning and development of a variety of organ systems, and both ligand-dependent and ligand-independent Hh pathway activation are known to promote tumorigenesis. Recent studies have shown that in tumors promoted by Hh ligands, activation occurs within the stromal microenvironment. Testing whether ligand-driven Hh signaling promotes tumor angiogenesis, we found that Hh antagonism reduced the vascular density of Hh-producing LS180 and SW480 xenografts. In addition, ectopic expression of sonic hedgehog in low-Hh-expressing DLD-1 xenografts increased tumor vascular density, augmented angiogenesis, and was associated with canonical Hh signaling within perivascular tumor stromal cells. To better understand the molecular mechanisms underlying Hh-mediated tumor angiogenesis, we established an Hh-sensitive angiogenesis coculture assay and found that fibroblast cell lines derived from a variety of human tissues were Hh responsive and promoted angiogenesis in vitro through a secreted paracrine signal(s). Affymetrix array analyses of cultured fibroblasts identified VEGF-A, hepatocyte growth factor, and PDGF-C as candidate secreted proangiogenic factors induced by Hh stimulation. Expression studies of xenografts and angiogenesis assays using combinations of Hh and VEGF-A inhibitors showed that it is primarily Hh-induced VEGF-A that promotes angiogenesis in vitro and augments tumor-derived VEGF to promote angiogenesis in vivo.


Subject(s)
Hedgehog Proteins/genetics , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Line , Cell Line, Tumor , Culture , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Mice , Mice, Nude , Myofibroblasts/cytology , Myofibroblasts/metabolism , Neoplasms/blood supply , Neoplasms/pathology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neovascularization, Physiologic/genetics , Oligonucleotide Array Sequence Analysis , Patched Receptors , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Stromal Cells/pathology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...