Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
J Allergy Clin Immunol ; 151(1): 247-259.e7, 2023 01.
Article in English | MEDLINE | ID: mdl-35973477

ABSTRACT

BACKGROUND: Familial hemophagocytic lymphohistiocytosis is a life-threatening hyperinflammatory disease caused by genetic defects in the granule-mediated cytotoxic pathway. Success of hematopoietic cell transplantation, the only cure, is correlated with the extent of disease control before transplantation. Unfortunately, disease refractoriness and toxicities to standard chemotherapy-based regimens are fatal in a fraction of patients. Novel targeted immunotherapies, such as IFN-γ blocking antibodies or ruxolitinib, a Janus kinase (JAK) 1/2 inhibitor, are promising but only partially effective at controlling disease. OBJECTIVE: We asked whether combinations of cytokine-targeted therapies, using antibodies or JAK inhibitor, work synergistically to counteract HLH. METHODS: Genetically predisposed mice were infected and treated with distinct combinations of immunotherapies. Disease outcome was monitored and compared to monotherapies. RESULTS: We showed that inhibiting IL-6 or IL-18 signaling in combination with IFN-γ blockade or ruxolitinib did not increase disease control compared to anti-IFN-γ antibodies or ruxolitinib monotherapies. In contrast, clinically relevant doses of ruxolitinib combined with low doses of anti-IFN-γ blocking antibodies corrected cytopenias, prevented overt neutrophilia, limited cytokinemia, and resolved HLH immunopathology and symptomatology. CONCLUSIONS: Our findings demonstrate that IFN-γ blockade and ruxolitinib act synergistically to suppress HLH progression. This supports the use of combined cytokine-targeted therapies as a bridge to hematopoietic cell transplantation in severe familial hemophagocytic lymphohistiocytosis.


Subject(s)
Lymphohistiocytosis, Hemophagocytic , Animals , Mice , Lymphohistiocytosis, Hemophagocytic/drug therapy , Antibodies, Blocking/therapeutic use , Interferon-gamma/genetics , Cytokines/metabolism
2.
Aesthet Surg J ; 44(4): 363-372, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38078367

ABSTRACT

BACKGROUND: Breast implants have always been composed of a silicone elastomer envelope filled with either silicone gel or saline. Breast implant illness (BII) is a set of symptoms that has previously been linked to the leakage of silicone particles from the implants into the body. OBJECTIVES: Our research aimed to quantify the number of silicone particles present in the capsules of breast implants available in North America. METHODS: Thirty-five periprosthetic capsules were sampled and analyzed, and silicone particles were counted and measured. The capsule surface area was then measured and utilized to calculate particle density and total number of silicone particles. RESULTS: Eighty-five percent of capsules analyzed from silicone gel implants contained silicone, with an average of 62 particles per mm3 of capsular tissue. These implants had approximately 1 million silicone particles per capsule. In contrast, none of the saline implant capsules contained silicone. Capsules from macrotextured tissue expanders contained fewer and larger silicone particles. CONCLUSIONS: Silicone gel implants presented silicone particle bleeding into the periprosthetic capsule, totaling on average 1 million silicone particles per capsule. On the other hand, no silicone particle bleeding was observed from saline breast implants. These data suggest that particle bleeding comes from the inner silicone gel, and not from the smooth outer silicone shell. Previous studies have reported the presence of breast implant illness in patients with both silicone- and saline-filled implants. Therefore, our data suggest that silicone migration is not the sole cause of BII.


Subject(s)
Breast Implantation , Breast Implants , Humans , Breast Implants/adverse effects , Silicone Gels/adverse effects , Breast/surgery , Breast Implantation/adverse effects , Saline Solution , North America
3.
J Immunol ; 207(6): 1599-1615, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34408010

ABSTRACT

GFI1 is a DNA-binding transcription factor that regulates hematopoiesis by repressing target genes through its association with complexes containing histone demethylases such as KDM1A (LSD1) and histone deacetylases (HDACs). To study the consequences of the disruption of the complex between GFI1 and histone-modifying enzymes, we have used knock-in mice harboring a P2A mutation in GFI1 coding region that renders it unable to bind LSD1 and associated histone-modifying enzymes such as HDACs. GFI1P2A mice die prematurely and show increased numbers of memory effector and regulatory T cells in the spleen accompanied by a severe systemic inflammation with high serum levels of IL-6, TNF-α, and IL-1ß and overexpression of the gene encoding the cytokine oncostatin M (OSM). We identified lung alveolar macrophages, CD8 T cell from the spleen and thymic eosinophils, and monocytes as the sources of these cytokines in GFI1P2A mice. Chromatin immunoprecipitation showed that GFI1/LSD1 complexes occupy sites at the Osm promoter and an intragenic region of the Tnfα gene and that a GFI1P2A mutant still remains bound at these sites even without LSD1. Methylation and acetylation of histone H3 at these sites were enriched in cells from GFI1P2A mice, the H3K27 acetylation being the most significant. These data suggest that the histone modification facilitated by GFI1 is critical to control inflammatory pathways in different cell types, including monocytes and eosinophils, and that a disruption of GFI1-associated complexes can lead to systemic inflammation with fatal consequences.


Subject(s)
DNA-Binding Proteins/deficiency , Histone Demethylases/metabolism , Mutant Proteins/metabolism , Signal Transduction/genetics , Systemic Inflammatory Response Syndrome/blood , Transcription Factors/deficiency , Animals , Bone Marrow Transplantation/methods , CD8-Positive T-Lymphocytes/immunology , Cytokines/blood , Cytokines/genetics , DNA-Binding Proteins/genetics , Female , Gene Expression , Gene Knock-In Techniques , Histones/metabolism , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Protein Binding , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/immunology , Transcription Factors/genetics
4.
J Immunol ; 205(5): 1268-1280, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32690660

ABSTRACT

Regulation of MHC class I (MHC I) expression has been studied almost exclusively in hematolymphoid cells. We report that thymic epithelial cells (TECs), particularly the medullary TECs, constitutively express up to 100-fold more cell surface MHC I proteins than epithelial cells (ECs) from the skin, colon, and lung. Differential abundance of cell surface MHC I in primary ECs is regulated via transcription of MHC I and of genes implicated in the generation of MHC I-binding peptides. Superior MHC I expression in TECs is unaffected by deletion of Ifnar1 or Ifngr1, but is lessened by deletion of Aire, Ifnlr1, Stat1, or Nlrc5, and is driven mainly by type III IFN produced by medullary TECs. Ifnlr1 -/- mice show impaired negative selection of CD8 thymocytes and, at 9 mo of age, present autoimmune manifestations. Our study shows unanticipated variation in MHC I expression by ECs from various sites and provides compelling evidence that superior expression of MHC I in TECs is crucial for proper thymocyte education.


Subject(s)
Epithelial Cells/immunology , Histocompatibility Antigens Class I/immunology , Interferons/immunology , Receptors, Interferon/immunology , Thymus Gland/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Thymocytes/immunology , Interferon Lambda
5.
Cell Mol Biol (Noisy-le-grand) ; 68(9): 113-124, 2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36905266

ABSTRACT

Apelin-13 is an adipokine known for its growth-inducing effects on human breast cancer cells in an estrogen-containing environment. However, the response of these cells to apelin-13 in the absence of estrogen and its association with the expression of the apelin receptor (APLNR) has not yet been investigated. In the present study, we show that the breast cancer cell line MCF-7 expresses the APLNR as shown by immunofluorescence and flow cytometry, under conditions of ER starvation and that culture of these cells in the presence of apelin-13 results in an increased growth rate and a diminished autophagy flux.  Moreover, the binding of APLNR by apelin-13 resulted in an increased growth rate (assayed by AlamarBlue) and a diminished autophagy flux (monitored by Lysotracker Green). The latter observations were reversed in the presence of exogenous estrogen. Finally, apelin-13 induces the deactivation of the apoptotic kinase AMPK. Taken together, our results show that APLNR signaling in breast cancer cells is functional and prevents tumor growth under conditions of estrogen starvation. They furthermore suggest an alternative mechanism of estrogen-independent tumor growth thereby positioning the APLNR-AMPK axis as a novel pathway and a possible therapeutic target in endocrine resistance of breast cancer cells.


Subject(s)
Breast Neoplasms , Humans , Female , Apelin , Breast Neoplasms/pathology , Apelin Receptors , Estrogens , MCF-7 Cells , Autophagy , Cell Line, Tumor
6.
Am J Transplant ; 20(3): 726-738, 2020 03.
Article in English | MEDLINE | ID: mdl-31729155

ABSTRACT

Tertiary lymphoid structures (TLS) accumulate at sites of chronic injury where they function as an ectopic germinal center, fostering local autoimmune responses. Vascular injury leads to the release of endothelial-derived apoptotic exosome-like vesicles (ApoExo) that contribute to rejection in transplanted organs. The purpose of the study was to evaluate the impact of ApoExo on TLS formation in a model of vascular allograft rejection. Mice transplanted with an allogeneic aortic transplant were injected with ApoExo. The formation of TLS was significantly increased by ApoExo injection along with vascular remodeling and increased levels of antinuclear antibodies and anti-perlecan/LG3 autoantibodies. ApoExo also enhanced allograft infiltration by γδT17 cells. Recipients deficient in γδT cells showed reduced TLS formation and lower autoantibodies levels following ApoExo injection. ApoExo are characterized by proteasome activity, which can be blocked by bortezomib. Bortezomib treated ApoExo reduced the recruitment of γδT17 cells to the allograft, lowered TLS formation, and reduced autoantibody production. This study identifies vascular injury-derived extracellular vesicles (ApoExo), as initiators of TLS formation and demonstrates the pivotal role of γδT17 in coordinating TLS formation and autoantibody production. Finally, our results suggest proteasome inhibition with bortezomib as a potential option for controlling TLS formation in rejected allografts.


Subject(s)
Extracellular Vesicles , Tertiary Lymphoid Structures , Allografts , Animals , Graft Rejection/etiology , Graft Rejection/prevention & control , Mice , Mice, Inbred C57BL , Transplantation, Homologous
7.
Ann Surg Oncol ; 26(13): 4337-4345, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31605348

ABSTRACT

BACKGROUND: The sentinel node biopsy following neoadjuvant chemotherapy (SN FNAC) study has shown that in node-positive (N+) breast cancer, sentinel node biopsy (SNB) can be performed following neoadjuvant chemotherapy (NAC), with a low false negative rate (FNR = 8.4%). A secondary endpoint of the SN FNAC study was to determine whether axillary ultrasound (AxUS) could predict axillary pathological complete response (ypN0) and increase the accuracy of SNB. METHODS: The SN FNAC trial is a study of patients with biopsy-proven N+ breast cancer who underwent SNB followed by completion node dissection. All patients had AxUS following NAC and the axillary nodes were classified as either positive (AxUS+) or negative (AxUS-). AxUS was compared with the final axillary pathology results. RESULTS: There was no statistical difference in the baseline characteristics of patients with AxUS+ versus those with AxUS-. Overall, 82.5% (47/57) of AxUS+ patients had residual positive lymph nodes (ypN+) at surgery and 53.8% (42/78) of AxUS- patients had ypN+. Post NAC AxUS sensitivity was 52.8%, specificity 78.3%, and negative predictive value 46.2%. AxUS FNR was 47.2%, versus 8.4% for SNB. If post-NAC AxUS- was used to select patients for SNB, FNR would decrease from 8.4 to 2.7%. However, using post-NAC AxUS in addition to SNB as an indication for ALND would have led to unnecessary ALND in 7.8% of all patients. CONCLUSION: AxUS is not appropriate as a standalone staging procedure, and SNB itself is sufficient to assess the axilla post NAC in patients who present with N+ breast cancer.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Lymph Nodes/pathology , Neoadjuvant Therapy/methods , Ultrasonography, Mammary/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Axilla , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/diagnostic imaging , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Lobular/diagnostic imaging , Carcinoma, Lobular/drug therapy , Chemotherapy, Adjuvant , Female , Follow-Up Studies , Humans , Lymph Nodes/diagnostic imaging , Lymph Nodes/drug effects , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Sentinel Lymph Node Biopsy
8.
Br J Cancer ; 119(5): 580-590, 2018 08.
Article in English | MEDLINE | ID: mdl-30078843

ABSTRACT

BACKGROUND: Distinguishing lung adenocarcinoma (ADC) from squamous cell carcinoma (SCC) has a tremendous therapeutic implication. Sometimes, the commonly used immunohistochemistry (IHC) markers fail to discriminate between them, urging for the identification of new diagnostic biomarkers. METHODS: We performed IHC on tissue microarrays from two cohorts of lung cancer patients to analyse the expression of beta-arrestin-1, beta-arrestin-2 and clinically used diagnostic markers in ADC and SCC samples. Logistic regression models were applied for tumour subtype prediction. Parallel reaction monitoring (PRM)-based mass spectrometry was used to quantify beta-arrestin-1 in plasma from cancer patients and healthy donors. RESULTS: Beta-arrestin-1 expression was significantly higher in ADC versus SCC samples. Beta-arrestin-1 displayed high sensitivity, specificity and negative predictive value. Its usefulness in an IHC panel was also shown. Plasma beta-arrestin-1 levels were considerably higher in lung cancer patients than in healthy donors and were higher in patients who later experienced a progressive disease than in patients showing complete/partial response following EGFR inhibitor therapy. CONCLUSIONS: Our data identify beta-arrestin-1 as a diagnostic marker to differentiate ADC from SCC and indicate its potential as a plasma biomarker for non-invasive diagnosis of lung cancer. Its utility to predict response to EGFR inhibitors is yet to be confirmed.


Subject(s)
Adenocarcinoma of Lung/diagnosis , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/diagnosis , Lung Neoplasms/diagnosis , Up-Regulation , beta-Arrestin 1/metabolism , Adenocarcinoma of Lung/blood , Adenocarcinoma of Lung/metabolism , Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/metabolism , Case-Control Studies , Diagnosis, Differential , Disease Progression , Early Detection of Cancer , Gene Expression Regulation, Neoplastic , Humans , Logistic Models , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Predictive Value of Tests , Tissue Array Analysis , beta-Arrestin 1/blood
9.
Mod Pathol ; 30(5): 682-697, 2017 05.
Article in English | MEDLINE | ID: mdl-28084344

ABSTRACT

Breast cancer is a heterogeneous disease comprising a diversity of tumor subtypes that manifest themselves in a wide variety of clinical, pathological, and molecular features. One important subset, luminal breast cancers, comprises two clinically distinct subtypes luminal A and B each of them endowed with its own genetic program of differentiation and proliferation. Luminal breast cancers were operationally defined as follows: Luminal A: ER+, PR+, HER2-, Ki-67<14% and Luminal B: ER+ and/or PR+, HER2-,Ki-67≥14% or, alternatively ER+ and/or PR+, HER2+, any Ki-67. There is currently a need for a clinically robust and validated immunohistochemical assay that can help distinguish between luminal A and B breast cancer. MCM2 is a family member of the minichromosome maintenance protein complex whose role in DNA replication and cell proliferation is firmly established. As MCM2 appears to be an attractive alternative to Ki-67, we sought to study the expression of MCM2 and Ki-67 in different histological grades and molecular subtypes of breast cancer focusing primarily on ER-positive tumors. MCM2 and Ki-67 mRNA expression were studied using in silico analysis of available DNA microarray and RNA-sequencing data of human breast cancer. We next used immunohistochemistry to evaluate protein expression of MCM2 and Ki-67 on tissue microarrays of invasive breast carcinoma. We found that MCM2 and Ki-67 are highly expressed in breast tumors of high histological grades, comprising clinically aggressive tumors such as triple-negative, HER2-positive and luminal B subtypes. MCM2 expression was detected at higher levels than that of Ki-67 in normal breast tissues and in breast cancers. The bimodal distribution of MCM2 scores in ER+/HER2- breast tumors led to the identification of two distinct subgroups with different relapse-free survival rates. In conclusion, MCM2 expression can help sorting out two clinically important subsets of luminal breast cancer whose treatment and clinical outcomes are likely to diverge.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/classification , Breast Neoplasms/pathology , Minichromosome Maintenance Complex Component 2/biosynthesis , Breast Neoplasms/mortality , Cell Proliferation , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Ki-67 Antigen/analysis , Ki-67 Antigen/biosynthesis , Minichromosome Maintenance Complex Component 2/analysis , Neoplasm Grading/methods
10.
BMC Cancer ; 17(1): 356, 2017 05 22.
Article in English | MEDLINE | ID: mdl-28532474

ABSTRACT

BACKGROUND: IKKε is an oncogenic kinase that was found amplified and overexpressed in a substantial percentage of human breast cancer cell lines and primary tumors using genomic and gene expression analyses. Molecular studies have provided the rational for a key implication of IKKε in breast cancer cells proliferation and invasiveness through the phosphorylation of several substrates. METHODS: Here, we performed immunohistochemical detection of IKKε expression on tissue microarrays constituted of 154 characterized human breast cancer tumors. We further determined the association with multiple clinicopathological parameters and 5-years overall, disease-free and distant disease free survival. RESULTS: We observed expression of IKKε in 60.4% of the breast cancer tumors. IKKε expression status showed no association with a panel of markers used for molecular classification of the tumors, including ER/PR/HER2 status, or with the molecular subtypes. However, IKKε expression was inversely associated with lymph node metastasis status (p = 0.0032). Additionally, we identified a novel association between IKKε and EGFR expression (p = 0.0011). CONCLUSIONS: The unexpected observation of an inverse association between IKKε and lymph node metastasis advocates for larger scale immunohistochemical profiling of primary breast tumors to clarify the role of IKKε in metastasis. This study suggests that breast cancer tumors expressing EGFR and IKKε may be potential targets for drugs aiming at inhibiting IKKε activity or expression.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , ErbB Receptors/metabolism , I-kappa B Kinase/metabolism , Adult , Antibodies, Monoclonal/chemistry , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/secondary , Disease-Free Survival , Female , Humans , I-kappa B Kinase/immunology , Immunohistochemistry , Kaplan-Meier Estimate , Lymphatic Metastasis , MCF-7 Cells , Middle Aged , Prognosis
11.
Biol Chem ; 397(4): 323-36, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26841446

ABSTRACT

The mechanism by which kinin B1 receptor (B1R) contributes to type 1 diabetes is addressed by determining the impact of its inhibition on diabetes and on its pancreatic expression and cellular localisation on immunocompetent cells and primary sensory C-fibres. Rats were made diabetic with streptozotocin (STZ). On day 4, they were treated daily for 7 days with a B1R antagonist (SSR240612, 10 mg/kg) or its vehicle. The surviving ß-cells were measured by immunostaining. The expression of B1R, iNOS, TNF-α, macrophages, TCD4+, CGRP and TRPV1 was measured by Western blotting, qRT-PCR and immunofluorescence. Macrophages and TCD4+ lymphocytes were absent in control, but distributed abundantly in the pancreas of STZ-diabetic rats. B1R was upregulated on these immune cells infiltrating the diabetic rat pancreas while it was not expressed on primary sensory C-fibres even if the expression of TRPV1 and CGRP was enhanced. SSR240612 prevented the infiltration of macrophages and TCD4+ lymphocytes and the upregulation of B1R, iNOS, TNF-α and TRPV1. SSR240612 corrected hyperglycaemia and hypoinsulinaemia by improving the Langerhans islets survival or regeneration. It is concluded that kinin B1R antagonism exerts anti-diabetic action by preventing the infiltration of immune cells in the pancreas and by preserving the integrity of Langerhans islets ß-cells.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Dioxoles/pharmacology , Hypoglycemic Agents/pharmacology , Pancreas/drug effects , Pancreas/pathology , Receptor, Bradykinin B1/metabolism , Sulfonamides/pharmacology , Animals , Diabetes Mellitus, Experimental/pathology , Dioxoles/chemistry , Dioxoles/therapeutic use , Dose-Response Relationship, Drug , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Male , Pancreas/metabolism , Rats , Rats, Sprague-Dawley , Streptozocin , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/therapeutic use
12.
Biol Blood Marrow Transplant ; 21(10): 1721-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26151303

ABSTRACT

Graft-versus-host disease (GVHD) impairs immune reconstitution after allogeneic stem cell transplantation (allo-SCT) and effective therapies aimed at restoring T cell counts in GVHD patients have yet to be developed. During GVHD, CD4(+) T cell reconstitution is particularly affected and current models hold that GVHD insult to the peripheral lymphoid niche is responsible for this effect. Here, we show that naïve CD4(+) T cell homeostatic proliferation (HP) is lost during GVHD because of low systemic IL-7 and impaired dendritic cell (DC) regeneration. We assessed factors involved in DC differentiation and found that although fms-like tyrosine kinase 3 ligand (Flt3-L) levels were normal, stromal-derived factor-1α (SDF-1α) was diminished in the blood of GVHD mice. Unlike Flt3-L treatment, the administration of SDF-1α specifically increased CD8α(+) DC numbers and did not worsen GVHD. Importantly, CD4(+) T cell HP was enhanced only when IL-7 and SDF-1α or Flt3L were coadministered, confirming the crucial role of DCs and IL-7 in restoring CD4(+) T cell regeneration during GVHD. Altogether, our results indicate that CD8α(+) DCs are part of the peripheral niche that controls CD4(+) T cell HP and that their depletion, combined with low systemic IL-7, explains how GVHD constrains naïve CD4(+) T cell reconstitution after allo-SCT.


Subject(s)
Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , Chemokine CXCL12/therapeutic use , Graft vs Host Disease/drug therapy , Interleukin-7/therapeutic use , Membrane Proteins/therapeutic use , Adoptive Transfer , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/transplantation , Chemokine CXCL12/blood , Chemokine CXCL12/deficiency , Dendritic Cells/immunology , Drug Synergism , Drug Therapy, Combination , Female , Graft vs Host Disease/blood , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Interleukin-7/deficiency , Interleukin-7/physiology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/deficiency , Recombinant Proteins/therapeutic use , Stromal Cells/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transplantation, Homologous
13.
Radiology ; 275(3): 666-74, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25496215

ABSTRACT

PURPOSE: To develop a classification method based on the statistical backscatter properties of tissues that can be used as an ancillary tool to the usual Breast Imaging Reporting and Data System (BI-RADS) classification for solid breast lesions identified at ultrasonography (US). MATERIALS AND METHODS: This study received institutional review board approval, and all subjects provided informed consent. Eighty-nine women (mean age, 50 years; age range, 22-82 years) with 96 indeterminate solid breast lesions (BI-RADS category 4-5; mean size, 13.2 mm; range, 2.6-44.7 mm) were enrolled. Prior to biopsy, additional radiofrequency US images were obtained, and a 3-second cine sequence was used. The research data were analyzed at a later time and were not used to modify patient management decisions. The lesions were segmented manually, and parameters of the homodyned K distribution (α, k, and µn values) were extracted for three regions: the intratumoral zone, a 3-mm supratumoral zone, and a 5-mm infratumoral zone. The Mann-Whitney rank sum test was used to identify parameters with the best discriminating value, yielding intratumoral α, supratumoral k, and infratumoral µn values. RESULTS: The 96 lesions were classified as follows: 48 BI-RADS category 4A lesions, 16 BI-RADS category 4B lesions, seven BI-RADS category 4C lesions, and 25 BI-RADS category 5 lesions. There were 24 cancers (25%). The area under the receiver operating characteristic curve was 0.76 (95% confidence interval: 0.65, 0.86). Overall, 24% of biopsies (in 17 of 72 lesions) could have been spared. By limiting analysis to lesions with a lower likelihood of malignancy (BI-RADS category 4A-4B), this percentage increased to 26% (16 of 62 lesions). Among benign lesions, the model was used to correctly classify 10 of 38 fibroadenomas (26%) and three of seven stromal fibroses (43%). CONCLUSION: The statistical model performs well in the classification of solid breast lesions at US, with the potential of preventing one in four biopsies without missing any malignancy.


Subject(s)
Breast Neoplasms/classification , Breast Neoplasms/diagnostic imaging , Models, Statistical , Ultrasonography, Mammary , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Prospective Studies , Young Adult
14.
BMC Immunol ; 15: 49, 2014 Oct 26.
Article in English | MEDLINE | ID: mdl-25344377

ABSTRACT

BACKGROUND: The tissue-signaling cytokines IL-17 and IL-22 are critical to host defense against oral Candida albicans infection, by their induction of oral antimicrobial peptide expression and recruitment of neutrophils. Mucosal Th17 cells which produce these cytokines are preferentially depleted in HIV-infected patients. Here, we tested the hypothesis that defective IL-17- and IL-22-dependent host responses to C. albicans determine the phenotype of susceptibility to oropharyngeal candidiasis (OPC) in transgenic (Tg) mice expressing HIV-1. RESULTS: Naïve CD4+ T-cells and the differentiated Th1, Th2, Th17, Th1Th17 and Treg lineages were all profoundly depleted in cervical lymph nodes (CLNs) of these Tg mice. However, naive CD4+ cells from Tg mice maintained the capacity to differentiate into these lineages in response to polarizing cytokines in vitro. Expression of Il17, Il22, S100a8 and Ccl20 was enhanced in oral mucosal tissue of non-Tg, but not of Tg mice, after oral infection with C. albicans. Treatment of infected Tg mice with the combination of IL-17 and IL-22, but not IL-17 or Il-22 alone, significantly reduced oral burdens of C. albicans and abundance of Candida hyphae in the epithelium of tongues of infected Tg mice, and restored the ability of the Tg mice to up-regulate expression of S100a8 and Ccl20 in response to C. albicans infection. CONCLUSIONS: These findings demonstrate that defective IL-17- and IL-22-dependent induction of innate mucosal immunity to C. albicans is central to the phenotype of susceptibility to OPC in these HIV transgenic mice.


Subject(s)
Candida albicans/immunology , Candidiasis, Oral , HIV Infections , HIV-1 , Immunity, Mucosal , Interleukin-17/immunology , Interleukins/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Candidiasis, Oral/genetics , Candidiasis, Oral/immunology , Candidiasis, Oral/pathology , HIV Infections/genetics , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Interleukin-17/genetics , Interleukins/genetics , Mice , Mice, Transgenic , Transgenes/immunology , Interleukin-22
15.
BMC Cancer ; 14: 609, 2014 Aug 23.
Article in English | MEDLINE | ID: mdl-25151367

ABSTRACT

BACKGROUND: In 2014, breast cancer remains a major cause of mortality worldwide mostly due to tumor relapse and metastasis. There is currently a great interest in identifying cancer biomarkers and signalling pathways mechanistically related to breast cancer progression. Matrix metalloproteinase-9 (MMP-9) is a member of matrix degrading enzymes involved in cancer development, invasion and metastasis. Our objective was to investigate MMP-9 expression in normal human breast tissue and to compare it to that of breast cancer of various histological grades and molecular subtypes. We also sought to correlate MMP-9 expression with the incidence of metastasis, survival rates and relapse in breast cancer patients. METHODS: MMP-9 was first studied using in silico analysis on available DNA microarray and RNA sequencing data of human breast cancer tissues and human breast cancer cell lines. We next ascertained MMP-9 expression in both normal breast tissue and in human breast carcinoma tissue microarrays. RESULTS: Significant increase in MMP-9 expression was found in breast cancer cells where compared to normal breast tissue. A positive correlation could also be established between elevated levels of MMP-9 and breast cancer of high histological grade. Furthermore, our results indicate that not only MMP-9 is differentially expressed between each molecular subset but also, more importantly MMP-9 overexpression revealed itself as a startling feature of triple-negative and HER2-positive breast cancers. Lastly, the clinical relevance of MMP-9 overexpression is strongly supported by its significant association with a higher incidence of metastasis and relapse. CONCLUSIONS: Differential expression of MMP-9 reflects the extent of cellular differentiation in breast cancer cells and is closely related to the most aggressive subtypes of breast cancer. Hence, MMP-9 is a promising prognostic biomarker of high-grade breast cancer. In our opinion, MMP-9 expression could help segregate subsets of aggressive breast cancer into clinically meaningful subtypes.


Subject(s)
Breast Neoplasms/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , MCF-7 Cells , Prognosis , Receptor, ErbB-2/metabolism , Retrospective Studies , Survival Rate
16.
BMC Cancer ; 14: 801, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25367122

ABSTRACT

BACKGROUND: Resistance to apoptosis induced by anti-cancer drugs is a major obstacle for the treatment of aggressive forms of breast cancer. Galectin-7 (gal-7) was recently shown to be specifically expressed in basal-like but not in luminal subtypes of human breast cancer. METHODS: We generated a mutant form of gal-7 (R74S). Arginine 74 is the structural equivalent of arginine 186 found in human galectin-3. Mutation R186S was previously shown to abolish the biological function of galectin-3. RESULTS: Mutation of arginine 74 induced only limited and local changes to the gal-7 fold. Recombinant forms of R74S and wtgal-7 were also equally effective at forming dimers in solution. Analysis of the thermodynamic parameters by isothermal titration calorimetry (ITC) indicated, however, that binding of lactose to gal-7 was inhibited by the R74S mutation. Using confocal microscopy and electron microscopy, we confirmed the expression of gal-7 in the cytosolic and nuclear compartments of breast cancer cells and the ability of gal-7 to translocate to mitochondria. The mutation at position 74, however, greatly reduced the expression of gal-7 in the nuclear and mitochondrial compartments. Interestingly, cells expressing mutated gal-7 were equally if not even more resistant to drug-induced apoptosis when compared to cells expressing wtgal-7. We also found that both wtgal-7 and R74S inhibited dox-induced PARP-1 cleavage and p53 protein expression. The inhibition of p53 correlated with a decrease in p21 protein expression and CDKN1A mRNA. Furthermore, analysis of nuclear and cytoplasmic fractions showed that both wild type and R74S mutant gal-7 inhibited p53 nuclear translocation, possibly by increasing degradation of cytosolic p53. CONCLUSIONS: These findings pose a challenge to the paradigm that has guided the design of galectin-specific inhibitors for the treatment of cancer. This study suggests that targeting CRD-independent cytosolic gal-7 in breast cancer cells may be a valuable strategy for the treatment of this disease. Our study will thus complement efforts towards improving selectivity of targeted anticancer agents.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Galectins/genetics , Galectins/metabolism , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms/ultrastructure , Cell Line, Tumor , Female , Galectins/chemistry , Gene Expression Regulation, Neoplastic , Humans , Intracellular Space/metabolism , Models, Molecular , Mutation , Protein Conformation , Protein Transport , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
17.
Infect Immun ; 81(4): 1100-13, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23340313

ABSTRACT

Cryptococcus neoformans var. grubii is the most frequent cause of AIDS-associated cryptococcosis worldwide, while Cryptococcus gattii usually infects immunocompetent people. To understand the mechanisms which cause differential susceptibility to these cryptococcal species in HIV infection, we established and characterized a model of cryptococcosis in CD4C/HIV(MutA) transgenic (Tg) mice expressing gene products of HIV-1 and developing an AIDS-like disease. Tg mice infected intranasally with C. neoformans var. grubii strain H99 or C23 consistently displayed reduced survival compared to non-Tg mice at three graded inocula, while shortened survival of Tg mice infected with C. gattii strain R265 or R272 was restricted to a single high inoculum. HIV-1 transgene expression selectively augmented systemic dissemination to the liver and spleen for strains H99 and C23 but not strains R265 and R272. Histopathologic examination of lungs of Tg mice revealed large numbers of widely scattered H99 cells, with a minimal inflammatory cell response, while in the non-Tg mice H99 was almost completely embedded within extensive mixed inflammatory cell infiltrates. In contrast to H99, R265 was dispersed throughout the lung parenchyma and failed to induce a strong inflammatory response in both Tg and non-Tg mice. HIV-1 transgene expression reduced pulmonary production of CCL2 and CCL5 after infection with H99 or R265, and production of these two chemokines was lower after infection with R265. These results indicate that an altered immune response in these Tg mice markedly enhances C. neoformans but not C. gattii infection. This model therefore provides a powerful new tool to further investigate the immunopathogenesis of cryptococcosis.


Subject(s)
Cryptococcus gattii/pathogenicity , Cryptococcus neoformans/pathogenicity , Disease Susceptibility , HIV-1/pathogenicity , Animals , Colony Count, Microbial , Cryptococcus gattii/immunology , Cryptococcus neoformans/immunology , Disease Models, Animal , HIV-1/immunology , Histocytochemistry , Liver/microbiology , Lung/microbiology , Lung/pathology , Mice , Mice, Transgenic , Spleen/microbiology , Survival Analysis
18.
Blood ; 117(5): 1734-44, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21119112

ABSTRACT

Gene expression profiling of human donor T cells before allogeneic hematopoietic cell transplantation revealed that expression of selected genes correlated with the occurrence of graft-versus-host disease (GVHD) in recipients. The gene with the best GVHD predictive accuracy was SMAD3, a core component of the transforming growth factor-ß signaling pathway, whose expression levels vary more than a 6-fold range in humans. The putative role of SMAD3 in the establishment of graft-host tolerance remained elusive. We report that SMAD3-KO mice present ostensibly normal lymphoid and myeloid cell subsets. However, the lack of SMAD3 dramatically increased the frequency and severity of GVHD after allogeneic hematopoietic cell transplantation into major histocompatibility complex-identical recipients. Lethal GVHD induced by SMAD3-KO donors affected mainly the intestine and resulted from massive tissue infiltration by T-bet(+) CD4 T cells and granulocytes that caused tissue damage by in situ release of Th1 cytokines and oxidative-nitrosative mediators, respectively. Our report reveals the nonredundant roles of SMAD3 in the development of tolerance to the host. Furthermore, our data support the concept that SMAD3 levels in donor cells dictate the risk of GVHD and that SMAD3 agonists would be attractive for prevention of GVHD.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Colon/pathology , Graft vs Host Disease/prevention & control , Granulocytes/metabolism , Smad3 Protein/physiology , Th1 Cells/cytology , Animals , Blotting, Western , Bone Marrow Transplantation , Cell Proliferation , Cells, Cultured , Colon/immunology , Colon/metabolism , Graft vs Host Disease/immunology , Granulocytes/cytology , Hematopoiesis , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Transplantation, Homologous
19.
Proc Natl Acad Sci U S A ; 107(32): 14134-9, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20679199

ABSTRACT

Translational regulation plays a critical role in the control of cell growth and proliferation. A key player in translational control is eIF4E, the mRNA 5' cap-binding protein. Aberrant expression of eIF4E promotes tumorigenesis and has been implicated in cancer development and progression. The activity of eIF4E is dysregulated in cancer. Regulation of eIF4E is partly achieved through phosphorylation. However, the physiological significance of eIF4E phosphorylation in mammals is not clear. Here, we show that knock-in mice expressing a nonphosphorylatable form of eIF4E are resistant to tumorigenesis in a prostate cancer model. By using a genome-wide analysis of translated mRNAs, we show that the phosphorylation of eIF4E is required for translational up-regulation of several proteins implicated in tumorigenesis. Accordingly, increased phospho-eIF4E levels correlate with disease progression in patients with prostate cancer. Our findings establish eIF4E phosphorylation as a critical event in tumorigenesis. These findings raise the possibility that chemical compounds that prevent the phosphorylation of eIF4E could act as anticancer drugs.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Neoplasms/etiology , Neoplasms/pathology , Animals , Disease Progression , Eukaryotic Initiation Factor-4E/genetics , Gene Expression Regulation , Gene Knock-In Techniques , Humans , Male , Mice , Neoplasm Proteins/genetics , Neoplasms/genetics , Phosphorylation/physiology , Up-Regulation
20.
ACS Appl Bio Mater ; 6(3): 1173-1184, 2023 03 20.
Article in English | MEDLINE | ID: mdl-36795958

ABSTRACT

We designed, synthesized, and characterized a Raman nanoprobe made of dye-sensitized single-walled carbon nanotubes (SWCNTs) that can selectively target biomarkers of breast cancer cells. The nanoprobe is composed of Raman-active dyes encapsulated inside a SWCNT, whose surface is covalently grafted with poly(ethylene glycol) (PEG) at a density of ∼0.7% per carbon. Using α-sexithiophene- and ß-carotene-derived nanoprobes covalently bound to an antibody, either anti-E-cadherin (E-cad) or anti-keratin-19 (KRT19), we prepared two distinct nanoprobes that specifically recognize biomarkers on breast cancer cells. Immunogold experiments and transmission electron microscopy (TEM) images are first used to guide the synthesis protocol for higher PEG-antibody attachment and biomolecule loading capacity. The duplex of nanoprobes was then applied to target E-cad and KRT19 biomarkers in T47D and MDA-MB-231 breast cancer cell lines. Hyperspectral imaging of specific Raman bands allows for simultaneous detection of this nanoprobe duplex on target cells without the need for additional filters or subsequent incubation steps. Our results confirm the high reproducibility of the nanoprobe design for duplex detection and highlight the potential of Raman imaging for advanced biomedical applications in oncology.


Subject(s)
Nanotubes, Carbon , Neoplasms , Reproducibility of Results , Biomarkers , Polyethylene Glycols , Antibodies , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL