Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cancers (Basel) ; 16(3)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38339231

ABSTRACT

Predicting which patients will progress to metastatic disease after surgery for non-metastatic clear cell renal cell carcinoma (ccRCC) is difficult; however, recent data suggest that tumor immune cell infiltration could be used as a biomarker. We evaluated the quantity and type of immune cells infiltrating ccRCC tumors for associations with metastatic progression following attempted curative surgery. We quantified immune cell densities in the tumor microenvironment and validated our findings in two independent patient cohorts with multi-region sampling to investigate the impact of heterogeneity on prognostic accuracy. For non-metastatic ccRCC, increased CD8+ T cell infiltration was associated with a reduced likelihood of progression to metastatic disease. Interestingly, patients who progressed to metastatic disease also had increased percentages of exhausted CD8+ T cells. Finally, we evaluated the spatial heterogeneity of the immune infiltration and demonstrated that patients without metastatic progression had CD8+ T cells in closer proximity to ccRCC cells. These data strengthen the evidence for CD8+ T cell infiltration as a prognostic biomarker in non-metastatic ccRCC and demonstrate that multi-region sampling may be necessary to fully characterize immune infiltration within heterogeneous tumors. Tumor CD8+ T cell infiltration should be investigated as a biomarker in adjuvant systemic therapy clinical trials for high-risk non-metastatic RCC.

2.
Cell Rep ; 40(7): 111201, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977482

ABSTRACT

Stimulatory type 1 conventional dendritic cells (cDC1s) engage in productive interactions with CD8+ effectors along tumor-stroma boundaries. The paradoxical accumulation of "poised" cDC1s within stromal sheets is unlikely to simply reflect passive exclusion from tumor cores. Drawing parallels with embryonic morphogenesis, we hypothesized that invasive margin stromal remodeling generates developmentally conserved cell fate cues that regulate cDC1 behavior. We find that, in human T cell-inflamed tumors, CD8+ T cells penetrate tumor nests, whereas cDC1s are confined within adjacent stroma that recurrently displays site-specific proteolysis of the matrix proteoglycan versican (VCAN), an essential organ-sculpting modification in development. VCAN is necessary, and its proteolytic fragment (matrikine) versikine is sufficient for cDC1 accumulation. Versikine does not influence tumor-seeding pre-DC differentiation; rather, it orchestrates a distinctive cDC1 activation program conferring exquisite sensitivity to DNA sensing, supported by atypical innate lymphoid cells. Thus, peritumoral stroma mimicking embryonic provisional matrix remodeling regulates cDC1 abundance and activity to elicit T cell-inflamed tumor microenvironments.


Subject(s)
Neoplasms , Tumor Microenvironment , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Humans , Immunity, Innate , Lymphocytes/metabolism , Neoplasms/pathology , Versicans/metabolism
3.
Sci Transl Med ; 13(610): eabd4811, 2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34516829

ABSTRACT

Paclitaxel (Taxol) is a cornerstone of cancer treatment. However, its mechanism of cytotoxicity is incompletely understood and not all patients benefit from treatment. We show that patients with breast cancer did not accumulate sufficient intratumoral paclitaxel to induce mitotic arrest in tumor cells. Instead, clinically relevant concentrations induced multipolar mitotic spindle formation. However, the extent of early multipolarity did not predict patient response. Whereas multipolar divisions frequently led to cell death, multipolar spindles focused into bipolar spindles before division at variable frequency, and maintaining multipolarity throughout mitosis was critical to induce the high rates of chromosomal instability necessary for paclitaxel to elicit cell death. Increasing multipolar divisions in paclitaxel resulted in improved cytotoxicity. Conversely, decreasing paclitaxel-induced multipolar divisions reduced paclitaxel efficacy. Moreover, we found that preexisting chromosomal instability sensitized breast cancer cells to paclitaxel. Both genetic and pharmacological methods of inducing chromosomal instability were sufficient to increase paclitaxel efficacy. In patients, the amount of pretreatment chromosomal instability directly correlated with taxane response in metastatic breast cancer such that patients with a higher rate of preexisting chromosomal instability showed improved response to taxanes. Together, these results support the use of baseline rates of chromosomal instability as a predictive biomarker for paclitaxel response. Furthermore, they suggest that agents that increase chromosomal instability or maintain multipolar spindles throughout mitosis will improve the clinical utility of paclitaxel.


Subject(s)
Breast Neoplasms , Paclitaxel , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Chromosomal Instability , Female , Humans
4.
PLoS One ; 16(8): e0255798, 2021.
Article in English | MEDLINE | ID: mdl-34383787

ABSTRACT

RATIONALE: Murine syngeneic tumor models have revealed efficacious systemic antitumor responses following primary tumor in situ vaccination combined with targeted radionuclide therapy to secondary or metastatic tumors. Here we present studies on the safety and feasibility of this approach in a relevant translational companion dog model (n = 17 dogs) with advanced cancer. METHODS: The three component of the combination immuno-radiotherapy approach were employed either separately or in combination in companion dogs with advanced stage cancer. In situ vaccination was achieved through the administration of hypofractionated external beam radiotherapy and intratumoral hu14.18-IL2 fusion immunocytokine injections to the index tumor. In situ vaccination was subsequently combined with targeted radionuclide therapy using a theranostic pairing of IV 86Y-NM600 (for PET imaging and subject-specific dosimetry) and IV 90Y-NM600 (therapeutic radionuclide) prescribed to deliver an immunomodulatory 2 Gy dose to all metastatic sites in companion dogs with metastatic melanoma or osteosarcoma. In a subset of dogs, immunologic parameters preliminarily assessed. RESULTS: The components of the immuno-radiotherapy combination were well tolerated either alone or in combination, resulting in only transient low grade (1 or 2) adverse events with no dose-limiting events observed. In subject-specific dosimetry analyses, we observed 86Y-NM600 tumor:bone marrow absorbed-dose differential uptakes ≥2 in 4 of 5 dogs receiving the combination, which allowed subsequent safe delivery of at least 2 Gy 90Y-NM600 TRT to tumors. NanoString gene expression profiling and immunohistochemistry from pre- and post-treatment biopsy specimens provide evidence of tumor microenvironment immunomodulation by 90Y-NM600 TRT. CONCLUSIONS: The combination of external beam radiotherapy, intratumoral immunocytokine, and targeted radionuclide immuno-radiotherapy known to have activity against syngeneic melanoma in murine models is feasible and well tolerated in companion dogs with advanced stage, spontaneously arising melanoma or osteosarcoma and has immunomodulatory potential. Further studies evaluating the dose-dependent immunomodulatory effects of this immuno-radiotherapy combination are currently ongoing.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Interleukin-2/therapeutic use , Melanoma/therapy , Osteosarcoma/therapy , Radiopharmaceuticals/therapeutic use , Animals , Antibodies, Monoclonal/adverse effects , Bone Marrow/chemistry , Bone Marrow/metabolism , Bone Marrow/pathology , Combined Modality Therapy , Dogs , Feasibility Studies , Female , Gene Expression , Interleukin-2/adverse effects , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Melanoma/immunology , Melanoma/pathology , Melanoma/veterinary , Osteosarcoma/immunology , Osteosarcoma/veterinary , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/chemistry , Vaccination , Yttrium Radioisotopes/chemistry
5.
Ann Diagn Pathol ; 48: 151586, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32836178

ABSTRACT

Quantification of Ki67 and mitosis is time consuming and subject to inter-observer variabilities. Limited studies explored the impact of those variables on the results and the correlation between mitotic count and Ki67 index in endocrine/neuroendocrine tumors, particularly so since the advent of PHH3 antibody and digital pathology. Using Ki67 and mitosis as examples, this study is intended to reveal variables affecting accurate quantification of biomarkers, and to explore the relationship of Ki67 index and mitotic count/index in endocrine/neuroendocrine tumors. Using both manual and pathologist supervised digital image analysis (PSDIA) methods, we examined the impact of post-analytical variables on the quantification of mitosis and Ki67 index and studied the correlation between them in 41 cases of endocrine/neuroendocrine tumors of variable histological grades/proliferating rates. We found that the selection of hotspots, field size and especially threshold affected the outcome of quantification of mitosis and Ki67 index; that mitotic count/index strongly (p < 0.05) correlated with Ki67 index only in the tumors with peak Ki67 index less than 30% and the correlation was more monotonic (positive, non-linear) than linear. In the hotspots of these tumors, the ratio of mitotic count to proliferating cells defined by Ki67 detection averaged 0.04. We also found that the PHH3 antibody could markedly increase the efficiency and accuracy of mitotic quantification. A consensus among pathologists is needed for the selection of hotspots, field size and threshold for quantification of mitosis and Ki67 index.


Subject(s)
Biomarkers/metabolism , Ki-67 Antigen/metabolism , Mitotic Index/methods , Neuroendocrine Tumors/pathology , Animals , Cell Proliferation , Evaluation Studies as Topic , Histones/metabolism , Humans , Image Processing, Computer-Assisted/methods , Ki-67 Antigen/immunology , Mice , Mitotic Index/statistics & numerical data , Neuroendocrine Tumors/immunology , Observer Variation , Pathologists
6.
Front Oncol ; 10: 553, 2020.
Article in English | MEDLINE | ID: mdl-32500020

ABSTRACT

New tools are needed to match cancer patients with effective treatments. Patient-derived organoids offer a high-throughput platform to personalize treatments and discover novel therapies. Currently, methods to evaluate drug response in organoids are limited because they overlook cellular heterogeneity. In this study, non-invasive optical metabolic imaging (OMI) of cellular heterogeneity was characterized in breast cancer (BC) and pancreatic cancer (PC) patient-derived organoids. Baseline heterogeneity was analyzed for each patient, demonstrating that single-cell techniques, such as OMI, are required to capture the complete picture of heterogeneity present in a sample. Treatment-induced changes in heterogeneity were also analyzed, further demonstrating that these measurements greatly complement current techniques that only gauge average cellular response. Finally, OMI of cellular heterogeneity in organoids was evaluated as a predictor of clinical treatment response for the first time. Organoids were treated with the same drugs as the patient's prescribed regimen, and OMI measurements of heterogeneity were compared to patient outcome. OMI distinguished subpopulations of cells with divergent and dynamic responses to treatment in living organoids without the use of labels or dyes. OMI of organoids agreed with long-term therapeutic response in patients. With these capabilities, OMI could serve as a sensitive high-throughput tool to identify optimal therapies for individual patients, and to develop new effective therapies that address cellular heterogeneity in cancer.

7.
Mol Oncol ; 14(8): 1898-1909, 2020 08.
Article in English | MEDLINE | ID: mdl-32255253

ABSTRACT

Centrosome amplification (CA) is a common phenomenon in cancer, promotes genomic stability and cancer evolution, and has been reported to promote metastasis. CA promotes a stochastic gain/loss of chromosomes during cell division, known as chromosomal instability (CIN). However, it is unclear whether CA is present in circulating tumor cells (CTCs), the seeds for metastasis. Here, we surveyed CA in CTCs from human subjects with metastatic breast cancer. CTCs were captured by CD45 exclusion and selection of EpCAM-positive cells using an exclusion-based sample preparation technology platform known as VERSA (versatile exclusion-based rare sample analysis). Centriole amplification (centrin foci> 4) is the definitive assay for CA. However, determination of centrin foci is technically challenging and incompatible with automated analysis. To test if the more technically accessible centrosome marker pericentrin could serve as a surrogate for centriole amplification in CTCs, cells were stained with pericentrin and centrin antibodies to evaluate CA. This assay was first validated using breast cancer cell lines and a nontransformed epithelial cell line model of inducible CA, then translated to CTCs. Pericentrin area and pericentrin area x intensity correlate well with centrin foci, validating pericentrin as a surrogate marker of CA. CA is found in CTCs from 75% of subjects, with variability in the percentage and extent of CA in individual circulating cells in a given subject, similar to the variability previously seen in primary tumors and cell lines. In summary, we created, validated, and implemented a novel method to assess CA in CTCs from subjects with metastatic breast cancer. Such an assay will be useful for longitudinal monitoring of CA in cancer patients and in prospective clinical trials for assessing the impact of CA on response to therapy.


Subject(s)
Antigens/metabolism , Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Centrosome/metabolism , Neoplastic Cells, Circulating/metabolism , Aged , Antigens/blood , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Centrioles/metabolism , Centrosome/pathology , Epithelial Cell Adhesion Molecule/metabolism , Female , Humans , Leukocyte Common Antigens/metabolism , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Up-Regulation
8.
Endocr Pathol ; 30(4): 262-269, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31468286

ABSTRACT

Long non-coding RNAs (lncRNAs) participate in transcription and in epigenetic or post-transcriptional regulation of gene expression. They also have roles in epithelial to mesenchymal transition and in carcinogenesis. Because lncRNAs may also have a role in thyroid cancer progression, we examined a group of thyroid tumors which included papillary thyroid carcinomas and anaplastic thyroid carcinomas to determine the specific lncRNAs that were upregulated during thyroid tumor progression. An RT2 Profiler PCR Array Human Cancer Pathway Finder consisting of 84 lncRNAs (Qiagen) and fresh tissues of normal thyroid, PTCs, and ATCs with gene expression profiling was used to determine genes upregulated and downregulated in ATCs. Two of the most highly upregulated genes, prostate cancer antigen 3 (PCA3) and HOX antisense intergenic RNA myeloid 1 (HOTAIRM1 or HAM-1), were selected for further studies using a thyroid tissue microarray(TMA) with formalin-fixed paraffin-embedded tissues of normal thyroid (NT, n = 10), nodular goiters (NG, n = 10), follicular adenoma (FA, n = 32), follicular carcinoma (FCA, n = 28), papillary thyroid carcinoma (PTC, n = 28), follicular variant of papillary thyroid carcinoma (FVPTC, n = 28), and anaplastic thyroid carcinoma (ATC, n = 10). TMA sections were analyzed by in situ hybridization (ISH) using RNAscope technology. The results of ISH analyses were imaged with Vectra imaging technology and quantified with Nuance® and inForm® software. The TMA analysis was validated by qRT-PCR using FFPE tissues for RNA preparation. Cultured thyroid carcinoma cell lines (n = 7) were also used to analyze for lncRNAs by qRT-PCR. The results showed 11 lncRNAs upregulated and 7 downregulated lncRNAs more than twofold in the ATCS compared with PTCs. Two of the upregulated lncRNAs, PCA3 and HAM-1, were analyzed on a thyroid carcinoma TMA. There was increased expression of both lncRNAs in ATCs and PTCs compared with NT after TMA analysis. qRT-PCR analyses showed increased expression of both lncRNAs in ATCs compared with NT and PTCs. Analyses of these lncRNAs from cultured thyroid carcinoma cell lines by qRT-PCR showed the highest levels of lncRNA expression in ATCs. TGF-ß treatment of cultured PTC and ATC cells for 21 days led to increased expression of PCA3 lncRNA in both cell lines by day 14. These results show that the lncRNAs PCA3 and HAM-1 are upregulated during thyroid tumor development and progression and may function as oncogenes during tumor progression.


Subject(s)
Antigens, Neoplasm/biosynthesis , MicroRNAs/biosynthesis , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Neoplasms/genetics , Adult , Aged , Female , Humans , Male , Middle Aged , Oncogenes/genetics , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Up-Regulation
9.
Neoplasia ; 21(6): 615-626, 2019 06.
Article in English | MEDLINE | ID: mdl-31078067

ABSTRACT

Heterogeneous populations within a tumor have varying metabolic profiles, which can muddle the interpretation of bulk tumor imaging studies of treatment response. Although methods to study tumor metabolism at the cellular level are emerging, these methods provide a single time point "snapshot" of tumor metabolism and require a significant time and animal burden while failing to capture the longitudinal metabolic response of a single tumor to treatment. Here, we investigated a novel method for longitudinal, single-cell tracking of metabolism across heterogeneous tumor cell populations using optical metabolic imaging (OMI), which measures autofluorescence of metabolic coenzymes as a report of metabolic activity. We also investigated whether in vivo cellular metabolic heterogeneity can be accurately captured using tumor-derived three-dimensional organoids in a genetically engineered mouse model of breast cancer. OMI measurements of response to paclitaxel and the phosphatidylinositol-3-kinase inhibitor XL147 in tumors and organoids taken at single cell resolution revealed parallel shifts in metaboltruic heterogeneity. Interestingly, these previously unappreciated heterogeneous metabolic responses in tumors and organoids could not be attributed to tumor cell fate or varying leukocyte content within the microenvironment, suggesting that heightened metabolic heterogeneity upon treatment is largely due to heterogeneous metabolic shifts within tumor cells. Together, these studies show that OMI revealed remarkable heterogeneity in response to treatment, which could provide a novel approach to predict the presence of potentially unresponsive tumor cell subpopulations lurking within a largely responsive bulk tumor population, which might otherwise be overlooked by traditional measurements.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Organoids/diagnostic imaging , Single-Cell Analysis , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Female , Humans , Mice , Optical Imaging , Organoids/metabolism , Quinoxalines/pharmacology , Sulfonamides/pharmacology , Tumor Microenvironment/genetics
10.
Nat Commun ; 10(1): 1540, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948704

ABSTRACT

Mitotic arrest deficient 1 (Mad1) plays a well-characterized role in the mitotic checkpoint. However, interphase roles of Mad1 that do not impact mitotic checkpoint function remain largely uncharacterized. Here we show that upregulation of Mad1, which is common in human breast cancer, prevents stress-induced stabilization of the tumor suppressor p53 in multiple cell types. Upregulated Mad1 localizes to ProMyelocytic Leukemia (PML) nuclear bodies in breast cancer and cultured cells. The C-terminus of Mad1 directly interacts with PML, and this interaction is enhanced by sumoylation. PML stabilizes p53 by sequestering MDM2, an E3 ubiquitin ligase that targets p53 for degradation, to the nucleolus. Upregulated Mad1 displaces MDM2 from PML, freeing it to ubiquitinate p53. Upregulation of Mad1 accelerates growth of orthotopic mammary tumors, which show decreased levels of p53 and its downstream effector p21. These results demonstrate an unexpected interphase role for Mad1 in tumor promotion via p53 destabilization.


Subject(s)
Cell Cycle Proteins/genetics , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Breast Neoplasms/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , DNA Damage , Escherichia coli/genetics , Female , HEK293 Cells , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints/genetics , Mice, Nude , Nuclear Proteins/metabolism , Protein Domains , Sumoylation , Up-Regulation
11.
Am J Pathol ; 188(3): 559-573, 2018 03.
Article in English | MEDLINE | ID: mdl-29429545

ABSTRACT

Inflammation, and the organization of collagen in the breast tumor microenvironment, is an important mediator of breast tumor progression. However, a direct link between markers of inflammation, collagen organization, and patient outcome has yet to be established. A tumor microarray of 371 invasive breast carcinoma biopsy specimens was analyzed for expression of inflammatory markers, including cyclooxygenase 2 (COX-2), macrophages, and several collagen features in the tumor nest (TN) or the tumor-associated stroma (TS). The tumor microarray cohort included females, aged 18 to 80 years, with a median follow-up of 8.4 years. High expression of COX-2 (TN), CD68 (TS), and CD163 (TN and TS) predicted worse patient overall survival (OS). This notion was strengthened by the finding from the multivariate analysis that high numbers of CD163+ macrophages in the TS is an independent prognostic factor. Overall collagen deposition was associated with high stromal expression of COX-2 and CD163; however, total collagen deposition was not a predictor for OS. Conversely, local collagen density, alignment and perpendicular alignment to the tumor boundary (tumor-associated collagen signature-3) were predictors of OS. These results suggest that in invasive carcinoma, the localization of inflammatory cells and aligned collagen orientation predict poor patient survival. Additional clinical studies may help validate whether therapy with selective COX-2 inhibitors alters expression of CD68 and CD163 inflammatory markers.


Subject(s)
Breast Neoplasms/metabolism , Collagen/metabolism , Cyclooxygenase 2/metabolism , Macrophages/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Female , Humans , Macrophages/pathology , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Young Adult
12.
Hum Pathol ; 70: 77-83, 2017 12.
Article in English | MEDLINE | ID: mdl-29079172

ABSTRACT

Cystic pancreatic tumors account for 10% of cystic lesions in the pancreas. Evaluation focuses on identifying lesions that require surgical resection due to actual or potential malignancy. Cystic tumors with malignant potential include mucinous cystic neoplasms (MCNs), intraductal papillary mucinous neoplasms (IPMNs), and cystic neuroendocrine tumors (NETs). The sensitivity of endoscopic fine needle aspiration (FNA) to diagnose such lesions is low, and a more accurate marker of malignant potential is needed. Aldo-keto reductase 1B10 (AKR1B10) was originally found in human hepatocellular carcinoma. Since then, it has been identified in pancreatic adenocarcinoma and pancreatic intraepithelial neoplasia. Because there is difficulty in determining the malignant potential of cystic pancreatic tumors, we set out to examine the expression of AKR1B10 in these lesions as a potential biomarker of malignancy. AKR1B10 expression was analyzed in cell blocks from FNAs and surgical resection specimens using immunohistochemistry. We examined MCN (n=28), IPMN (n=18), and cystic NET (n=20) as well as nonmucinous cysts including pseudocysts (n=13) and serous cystadenomas (n=16). AKR1B10 expression was seen in 45 of 46 (98%) mucinous lesions evaluated. Strong staining (2+-3+/60%-100% staining) was seen in 16 of 18 (89%) IPMNs and 25 of 28 (90%) MCNs. No staining was seen in the nonmucinous lesions (n=49). In conclusion, AKR1B10 is upregulated in mucinous cystic pancreatic tumors, and this staining can be accomplished in cytology FNA material, making AKR1B10 a promising biomarker of malignant potential. Most importantly, this application could impact the clinical management of these patients by determining the best candidates for surgical resection.


Subject(s)
Aldehyde Reductase/analysis , Biomarkers, Tumor/analysis , Biopsy, Fine-Needle , Immunohistochemistry , Neoplasms, Cystic, Mucinous, and Serous/enzymology , Pancreatic Neoplasms/enzymology , Aldo-Keto Reductases , Clinical Decision-Making , Humans , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/surgery , Pancreatectomy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Patient Selection , Predictive Value of Tests
13.
Breast Cancer Res ; 18(1): 35, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-27000374

ABSTRACT

BACKGROUND: High breast density is linked to an increased risk of breast cancer, and correlates with changes in collagen. In a mouse model of mammary carcinoma in the context of increased collagen deposition, the MMTV-PyMT/Col1a1 (tm1jae) , there is accelerated mammary tumor formation and progression. Previous gene expression analysis suggests that increased collagen density elevates expression of PTGS2 (prostaglandin-endoperoxide synthase 2), the gene for cyclooxygenase-2 (COX-2). METHODS: To understand the role of COX-2 in tumor progression within a collagen-dense microenvironment, we treated MMTV-PyMT or MMTV-PyMT/Col1a1 (tm1jae) tumors prior to and after tumor formation. Animals received treatment with celecoxib, a specific COX-2 inhibitor, or placebo. Mammary tumors were examined for COX-2, inflammatory and stromal cell components, and collagen deposition through immunohistochemical analysis, immunofluorescence, multiplex cytokine ELISA and tissue imaging techniques. RESULTS: PyMT/Col1a1 (tm1jae) tumors were larger, more proliferative, and expressed higher levels of COX-2 and PGE2 than PyMT tumors in wild type (WT) mice. Treatment with celecoxib significantly decreased the induced tumor size and metastasis of the PyMT/Col1a1 tumors, such that their size was not different from the smaller PyMT tumors. Celecoxib had minimal effect on the PyMT tumors. Celecoxib decreased expression levels of COX-2, PGE2, and Ki-67. Several cytokines were over-expressed in PyMT/Col1a1 compared to PyMT, and celecoxib treatment prevented their over-expression. Furthermore, macrophage and neutrophil recruitment were enhanced in PyMT/Col1a1 tumors, and this effect was inhibited by celecoxib. Notably, COX-2 inhibition reduced overall collagen deposition. Finally, when celecoxib was used prior to tumor formation, PyMT/Col1a1 tumors were fewer and smaller than in untreated animals. CONCLUSION: These findings suggest that COX-2 has a direct role in modulating tumor progression in tumors arising within collagen-dense microenvironments, and suggest that COX-2 may be an effective therapeutic target for women with dense breast tissue and early-stage breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cyclooxygenase 2/biosynthesis , Mammary Neoplasms, Animal/genetics , Tumor Microenvironment/genetics , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Celecoxib/administration & dosage , Cell Line, Tumor , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Dinoprostone/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Macrophages/pathology , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , Mice , Neutrophils/pathology , Tumor Microenvironment/drug effects
14.
Ann Surg Oncol ; 19(10): 3236-45, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22847119

ABSTRACT

BACKGROUND: Achieving negative surgical margins is critical to minimizing the risk of tumor recurrence in patients undergoing breast conservation surgery (BCS) for a breast malignancy. Our objective was to perform a systematic review comparing reexcision rates, sensitivity and specificity of the intraoperative use of the margin assessment techniques of imprint cytology (IC) and frozen section analysis (FSA), against permanent histopathologic section (PS). METHODS: The databases PubMed, Web of Knowledge, Cochrane Library and CINAHL Plus were searched for literature published from 1997 to 2011. Original investigations of patients who underwent BCS for breast cancer that evaluated margin assessment with PS and/or IC or FSA were included. Of 182 titles identified, 41 patient cohorts from 37 articles met inclusion criteria: PS (n = 19), IC (n = 7) and FSA (n = 15). Studies were summarized qualitatively using the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) checklist for cohort studies and the Strength of Recommendation Taxonomy (SORT) numerical scale for diagnostic studies. RESULTS: The final reexcision rates after primary BCS were 35 % for PS, 11 % for IC (p = 0.001 vs. PS) and 10 % for FSA (p < 0.0001 vs. PS). For IC, reexcision rates decreased from 26 to 4 % (p = 0.18) and for FSA, reexcision rates decreased from 27 to 6 % (p < 0.0001). The pooled sensitivity of IC and FSA were 72 and 83 %. The pooled specificity of IC and FSA were 97 and 95 %. The average length of each technique was 13 min for IC and 27 min for FSA. CONCLUSIONS: Patients who underwent BCS with intraoperative IC or FSA to assess negative surgical margins had significantly fewer secondary surgical procedures for excision of their breast malignancies.


Subject(s)
Breast Neoplasms/pathology , Cytodiagnosis , Frozen Sections , Mastectomy, Segmental , Monitoring, Intraoperative , Breast Neoplasms/surgery , Female , Humans , Prognosis
15.
J Biomed Biotechnol ; 2010: 753675, 2010.
Article in English | MEDLINE | ID: mdl-20339475

ABSTRACT

Titin is an extremely large protein found in highest concentrations in heart and skeletal muscle. The single mammalian gene is expressed in multiple isoforms as a result of alternative splicing. Although titin isoform expression is controlled developmentally and in a tissue specific manner, the vast number of potential splicing pathways far exceeds those described in any other alternatively spliced gene. Over 1 million human splice pathways for a single individual can be potentially derived from the PEVK region alone. A new splicing pattern for the human cardiac N2BA isoform type has been found in which the PEVK region includes only the N2B type exons. The alterations in splicing and titin isoform expression in human heart disease provide impetus for future detailed study of the splicing mechanisms for this giant protein.


Subject(s)
Muscle Proteins/genetics , Muscle Proteins/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Alternative Splicing , Animals , Connectin , Humans , Muscle Proteins/chemistry , Protein Kinases/chemistry , Sarcomeres/genetics , Sarcomeres/metabolism
16.
Am J Physiol Heart Circ Physiol ; 297(6): H2253-61, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19837952

ABSTRACT

Apoptotic death of vascular smooth muscle cells (SMCs) is a prominent feature of blood vessel remodeling and various vascular diseases. We have previously shown that protein kinase C-delta (PKC-delta) plays a critical role in SMC apoptosis. In this study, we tested the importance of PKC-delta proteolytic cleavage and tyrosine phosphorylation within the apoptosis pathway. Using hydrogen peroxide as a paradigm for oxidative stress, we showed that proteolytic cleavage of PKC-delta occurred in SMCs that underwent apoptosis, while tyrosine phosphorylation was detected only in necrotic cells. Furthermore, using a peptide (z-DIPD-fmk) that mimics the caspase-3 binding motif within the linker region of PKC-delta, we were able to prevent the cleavage of PKC-delta, as well as apoptosis. Inhibition of PKC-delta with rottlerin or small-interfering RNA diminished caspase-3 cleavage, caspase-3 activity, cleavage of poly (ADP-ribose) polymerase, cleavage of PKC-delta, and DNA fragmentation, confirming the previously reported role of PKC-delta in initiation of apoptosis. In contrast, z-DIPD-fmk markedly diminished caspase-3 activity, cleavage of PKC-delta, and DNA fragmentation without affecting cleavage of caspase-3 and poly (ADP-ribose) polymerase. Taken together, our data suggest that caspase-3-mediated PKC-delta cleavage underlies SMC apoptosis induced by oxidative stress, and that PKC-delta acts both upstream and downstream of caspase-3.


Subject(s)
Apoptosis , Caspase 3/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Protein Kinase C-delta/metabolism , Protein Processing, Post-Translational , Acetophenones/pharmacology , Animals , Apoptosis/drug effects , Benzopyrans/pharmacology , Cell Line , DNA Fragmentation , Dose-Response Relationship, Drug , Enzyme Activation , Hydrogen Peroxide/toxicity , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Oligopeptides/pharmacology , Oxidants/toxicity , Oxidative Stress , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , RNA Interference , Rats , Time Factors , Tyrosine
17.
J Mol Cell Cardiol ; 44(6): 983-991, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18387630

ABSTRACT

Titin is a very large alternatively spliced protein that performs multiple functions in heart and skeletal muscles. A rat strain is described with an autosomal dominant mutation that alters the isoform expression of titin. While wild type animals go through a developmental program where the 3.0 MDa N2B becomes the major isoform expressed by two to three weeks after birth (approximately 85%), the appearance of the N2B is markedly delayed in heterozygotes and never reaches more than 50% of the titin in the adult. Homozygote mutants express a giant titin of the N2BA isoform type (3.9 MDa) that persists as the primary titin species through ages of more than one and a half years. The mutation does not affect the isoform switching of troponin T, a protein that is also alternatively spliced with developmental changes. The basis for the apparently greater size of the giant titin in homozygous mutants was not determined, but the additional length was not due to inclusion of sequence from larger numbers of PEVK exons or the Novex III exon. Passive tension measurements using isolated cardiomyocytes from homozygous mutants showed that cells could be stretched to sarcomere lengths greater than 4 mum without breakage. This novel rat model should be useful for exploring the potential role of titin in the Frank-Starling relationship and mechano-sensing/signaling mechanisms.


Subject(s)
Alternative Splicing/genetics , Exons/genetics , Muscle Proteins/biosynthesis , Mutation , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Connectin , Heart/growth & development , Homozygote , Mechanotransduction, Cellular/genetics , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Rats , Rats, Inbred F344 , Rats, Mutant Strains , Rats, Sprague-Dawley , Sarcomeres/genetics , Sarcomeres/metabolism , Troponin T/biosynthesis , Troponin T/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...