Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
1.
Mol Cancer Res ; 19(10): 1699-1711, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34131071

RÉSUMÉ

HER2-positive breast cancers are among the most heterogeneous breast cancer subtypes. The early amplification of HER2 and its known oncogenic isoforms provide a plausible mechanism in which distinct programs of tumor heterogeneity could be traced to the initial oncogenic event. Here a Cancer rainbow mouse simultaneously expressing fluorescently barcoded wildtype (WTHER2), exon-16 null (d16HER2), and N-terminally truncated (p95HER2) HER2 isoforms is used to trace tumorigenesis from initiation to invasion. Tumorigenesis was visualized using whole-gland fluorescent lineage tracing and single-cell molecular pathology. We demonstrate that within weeks of expression, morphologic aberrations were already present and unique to each HER2 isoform. Although WTHER2 cells were abundant throughout the mammary ducts, detectable lesions were exceptionally rare. In contrast, d16HER2 and p95HER2 induced rapid tumor development. d16HER2 incited homogenous and proliferative luminal-like lesions which infrequently progressed to invasive phenotypes whereas p95HER2 lesions were heterogenous and invasive at the smallest detectable stage. Distinct cancer trajectories were observed for d16HER2 and p95HER2 tumors as evidenced by oncogene-dependent changes in epithelial specification and the tumor microenvironment. These data provide direct experimental evidence that intratumor heterogeneity programs begin very early and well in advance of screen or clinically detectable breast cancer. IMPLICATIONS: Although all HER2 breast cancers are treated equally, we show a mechanism by which clinically undetected HER2 isoforms program heterogenous cancer phenotypes through biased epithelial specification and adaptations within the tumor microenvironment.


Sujet(s)
Tumeurs du sein/génétique , Carcinogenèse/génétique , Isoformes de protéines/génétique , Récepteur ErbB-2/génétique , Animaux , Femelle , Régulation de l'expression des gènes tumoraux/génétique , Souris , Souris knockout , Microenvironnement tumoral/génétique
2.
Breast Cancer Res ; 19(1): 102, 2017 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-28865492

RÉSUMÉ

BACKGROUND: Stat1 gene-targeted knockout mice (129S6/SvEvTac-Stat1 tm1Rds) develop estrogen receptor-positive (ER+), luminal-type mammary carcinomas at an advanced age. There is evidence for both host environment as well as tumor cell-intrinsic mechanisms to initiate tumorigenesis in this model. In this report, we summarize details of the systemic and mammary pathology at preneoplastic and tumor-bearing time points. In addition, we investigate tumor progression in the 129:Stat1 -/- host compared with wild-type 129/SvEv, and we describe the immune cell reaction to the tumors. METHODS: Mice housed and treated according to National Institutes of Health guidelines and Institutional Animal Care and Use Committee-approved methods were evaluated by histopathology, and their tissues were subjected to immunohistochemistry with computer-assisted quantitative image analysis. Tumor cell culture and conditioned media from cell culture were used to perform macrophage (RAW264.7) cell migration assays, including the 129:Stat1 -/--derived SSM2 cells as well as control Met1 and NDL tumor cells and EpH4 normal cells. RESULTS: Tumorigenesis in 129:Stat1 -/- originates from a population of FoxA1+ large oval pale cells that initially appear and accumulate along the mammary ducts in segments or regions of the gland prior to giving rise to mammary intraepithelial neoplasias. Progression to invasive carcinoma is accompanied by a marked local stromal and immune cell response composed predominantly of T cells and macrophages. In conditioned media experiments, cells derived from 129:Stat1 -/- tumors secrete both chemoattractant and chemoinhibitory factors, with greater attraction in the extracellular vesicular fraction and inhibition in the soluble fraction. The result appears to be recruitment of the immune reaction to the periphery of the tumor, with exclusion of immune cell infiltration into the tumor. CONCLUSIONS: 129:Stat1 -/- is a unique model for studying the critical origins and risk reduction strategies in age-related ER+ breast cancer. In addition, it can be used in preclinical trials of hormonal and targeted therapies as well as immunotherapies.


Sujet(s)
Tumeurs du sein/étiologie , Tumeurs du sein/métabolisme , Phénotype , Récepteurs des oestrogènes/métabolisme , Facteur de transcription STAT-1/déficit , Facteurs âges , Animaux , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Chimiotaxie des leucocytes/immunologie , Modèles animaux de maladie humaine , Femelle , Humains , Immunohistochimie , Incidence , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Tumeurs expérimentales de la mamelle , Souris , Souris de souche-129 , Souris knockout
3.
J Immunother Cancer ; 5: 33, 2017.
Article de Anglais | MEDLINE | ID: mdl-28428882

RÉSUMÉ

BACKGROUND: Studies assessing immune parameters typically utilize human PBMCs or murine splenocytes to generate data that is interpreted as representative of immune status. Using splenocytes, we have shown memory CD4-T cells that expand following systemic immunostimulatory therapies undergo rapid IFNg-mediated activation induced cell death (AICD) resulting in a net loss of total CD4-T cells which correlates with elevated PD-1 expression. This is in contrast to CD8-T cells which expand with minimal PD-1 upregulation and apoptosis. In this study we expand upon our previous work by evaluating CD4 and CD8-T cell phenotype and distribution in peripheral organs which are more representative of immune responses occurring at metastatic sites following immunotherapy. METHODS: Phenotypic assessment of T cells in both lymphoid (spleen and LN) as well as peripheral organs (liver and lungs) in control and immunotherapy treated mice was performed to survey the impact of location on memory phenotype and activation marker status. Peripheral blood from patients undergoing systemic high dose IL-2 was also assessed for expression of PD-1 and memory phenotype. RESULTS: Here we reveal that, similar to what occurs in the spleen and lymph nodes, CD4-T cell numbers decreased while CD8-T cells expanded at these peripheral sites. In contrast to having differential expression of PD-1 as occurs in the spleen, both CD4 and CD8-T cells had significantly elevated levels of PD-1 in both the liver and lungs. Further analysis correlated PD-1 expression to CD62Llow (T effector/effector memory,TE/EM) expression which are more prevalent in CD4-T cells in general as well as CD8-T cells in peripheral organs. Similar elevated PD-1 expression on TE/EM cells was observed in patients undergoing systemic high-dose IL-2 therapy. CONCLUSIONS: These data highlight PD-1 expressing and/or TE/EM subsets of T cells in circulation as more representative of cells at immune sites and underscore the importance of valuation both in lymphoid as well as target organs when making determinations about immune status. TRIAL REGISTRATION: ClinicalTrials.gov NCT01416831. Registered August 12, 2011.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Immunothérapie/méthodes , Rate/immunologie , Animaux , Femelle , Humains , Souris , Souris de lignée BALB C , Phénotype
4.
PLoS One ; 10(6): e0129895, 2015.
Article de Anglais | MEDLINE | ID: mdl-26075897

RÉSUMÉ

Female 129:Stat1-null mice (129S6/SvEvTac-Stat1(tm1Rds) homozygous) uniquely develop estrogen-receptor (ER)-positive mammary tumors. Herein we report that the mammary glands (MG) of these mice have altered growth and development with abnormal terminal end buds alongside defective branching morphogenesis and ductal elongation. We also find that the 129:Stat1-null mammary fat pad (MFP) fails to sustain the growth of 129S6/SvEv wild-type and Stat1-null epithelium. These abnormalities are partially reversed by elevated serum progesterone and prolactin whereas transplantation of wild-type bone marrow into 129:Stat1-null mice does not reverse the MG developmental defects. Medium conditioned by 129:Stat1-null epithelium-cleared MFP does not stimulate epithelial proliferation, whereas it is stimulated by medium conditioned by epithelium-cleared MFP from either wild-type or 129:Stat1-null females having elevated progesterone and prolactin. Microarrays and multiplexed cytokine assays reveal that the MG of 129:Stat1-null mice has lower levels of growth factors that have been implicated in normal MG growth and development. Transplanted 129:Stat1-null tumors and their isolated cells also grow slower in 129:Stat1-null MG compared to wild-type recipient MG. These studies demonstrate that growth of normal and neoplastic 129:Stat1-null epithelium is dependent on the hormonal milieu and on factors from the mammary stroma such as cytokines. While the individual or combined effects of these factors remains to be resolved, our data supports the role of STAT1 in maintaining a tumor-suppressive MG microenvironment.


Sujet(s)
Glandes mammaires animales/croissance et développement , Glandes mammaires animales/métabolisme , Facteur de transcription STAT-1/génétique , Animaux , Tumeurs du sein/étiologie , Tumeurs du sein/anatomopathologie , Analyse de regroupements , Cytokines/métabolisme , Modèles animaux de maladie humaine , Épithélium/effets des médicaments et des substances chimiques , Épithélium/métabolisme , Épithélium/anatomopathologie , Femelle , Analyse de profil d'expression de gènes , Hormones/métabolisme , Hormones/pharmacologie , Isogreffes , Glandes mammaires animales/anatomopathologie , Souris , Souris knockout , Facteur de transcription STAT-1/déficit , Cellules stromales/effets des médicaments et des substances chimiques , Cellules stromales/métabolisme
5.
J Biol Chem ; 288(27): 19593-603, 2013 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-23696648

RÉSUMÉ

Insulin-like growth factor-1 (IGF1) is a major therapeutic target for cancer. We recently reported that IGF1 directly binds to integrins (αvß3 and α6ß4) and induces ternary complex formation (integrin-IGF1-IGF1 receptor (IGF1R)) and that the integrin binding-defective mutant of IGF1 (R36E/R37E) is defective in signaling and ternary complex formation. These findings predict that R36E/R37E competes with WT IGF1 for binding to IGF1R and inhibits IGF signaling. Here, we described that excess R36E/R37E suppressed cell viability increased by WT IGF1 in vitro in non-transformed cells. We studied the effect of R36E/R37E on viability and tumorigenesis in cancer cell lines. We did not detect an effect of WT IGF1 or R36E/R37E in cancer cells under anchorage-dependent conditions. However, under anchorage-independent conditions, WT IGF1 enhanced cell viability and induced signals, whereas R36E/R37E did not. Notably, excess R36E/R37E suppressed cell viability and signaling induced by WT IGF1 under anchorage-independent conditions. Using cancer cells stably expressing WT IGF1 or R36E/R37E, we determined that R36E/R37E suppressed tumorigenesis in vivo, whereas WT IGF1 markedly enhanced it. R36E/R37E suppressed the binding of WT IGF1 to the cell surface and the subsequent ternary complex formation induced by WT IGF1. R36E/R37E suppressed activation of IGF1R by insulin. WT IGF1, but not R36E/R37E, induced ternary complex formation with the IGF1R/insulin receptor hybrid. These findings suggest that 1) IGF1 induces signals under anchorage-independent conditions and that 2) R36E/R37E acts as a dominant-negative inhibitor of IGF1R (IGF1 decoy). Our results are consistent with a model in which ternary complex formation is critical for IGF signaling.


Sujet(s)
Substitution d'acide aminé , Transformation cellulaire néoplasique/effets des médicaments et des substances chimiques , Facteur de croissance IGF-I/pharmacologie , Mutation faux-sens , Récepteur IGF de type 1/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire tumorale , Survie cellulaire , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Humains , Insuline/génétique , Insuline/métabolisme , Facteur de croissance IGF-I/génétique , Facteur de croissance IGF-I/métabolisme , Intégrines , Souris , Modèles biologiques , Cellules NIH 3T3 , Liaison aux protéines , Structure quaternaire des protéines , Récepteur IGF de type 1/génétique , Transduction du signal/génétique
6.
Lab Invest ; 93(4): 480-97, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23399853

RÉSUMÉ

Quantitative Image Analysis (QIA) of digitized whole slide images for morphometric parameters and immunohistochemistry of breast cancer antigens was used to evaluate the technical reproducibility, biological variability, and intratumoral heterogeneity in three transplantable mouse mammary tumor models of human breast cancer. The relative preservation of structure and immunogenicity of the three mouse models and three human breast cancers was also compared when fixed with representatives of four distinct classes of fixatives. The three mouse mammary tumor cell models were an ER+/PR+ model (SSM2), a Her2+ model (NDL), and a triple negative model (MET1). The four breast cancer antigens were ER, PR, Her2, and Ki67. The fixatives included examples of (1) strong cross-linkers, (2) weak cross-linkers, (3) coagulants, and (4) combination fixatives. Each parameter was quantitatively analyzed using modified Aperio Technologies ImageScope algorithms. Careful pre-analytical adjustments to the algorithms were required to provide accurate results. The QIA permitted rigorous statistical analysis of results and grading by rank order. The analyses suggested excellent technical reproducibility and confirmed biological heterogeneity within each tumor. The strong cross-linker fixatives, such as formalin, consistently ranked higher than weak cross-linker, coagulant and combination fixatives in both the morphometric and immunohistochemical parameters.


Sujet(s)
Variation des antigènes/effets des médicaments et des substances chimiques , Marqueurs biologiques tumoraux/analyse , Fixateurs/pharmacologie , Tumeurs expérimentales de la mamelle/anatomopathologie , Manipulation d'échantillons/normes , Animaux , Lignée cellulaire tumorale , Femelle , Humains , Immunohistochimie , Foie/effets des médicaments et des substances chimiques , Souris , Souris de souche-129 , Reproductibilité des résultats
7.
Dis Model Mech ; 5(6): 914-20, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-22563073

RÉSUMÉ

Tp53 mutations are common in human prostate cancer (CaP), occurring with a frequency of ∼30% and ∼70% in localized and metastatic disease, respectively. In vitro studies have determined several common mutations of Tp53 that have specific gain-of-function properties in addition to loss of function, including the ability to promote castration-resistant (CR) growth of CaP cells in some contexts. To date, a lack of suitable mouse models has prohibited investigation of the role played by Tp53 mutations in mediating CaP progression in vivo. Here, we describe the effects of conditional expression of a mutant Tp53 (Tp53(R270H); equivalent to the human hotspot mutant R273H) in the prostate epithelium of mice. Heterozygous "Tp53(LSL-R270H/+)" [129S4(Trp53(tm3Tyj))] and "Nkx3.1-Cre" [129S(Nkx3-1(tm3(cre)Mms))] mice with prostate-specific expression of the Tp53(R270H) mutation (p53(R270H/+) Nkx3.1-Cre mice) were bred onto an FVB/N background via speed congenesis to produce strain FVB.129S4(Trp53(tm3Tyj/wt)); FVB.129S(Nkx3-1(tm3(cre)Mms/wt)) and littermate genotype negative control mice. These mutant mice had significantly increased incidences of prostatic intraepithelial neoplasia (PIN) lesions, and these appeared earlier, compared with the Nkx3.1 haploinsufficient (Nkx3.1-Cre het) littermate mice, which did not express the Tp53 mutation. PIN lesions in these mice showed consistent progression and some developed into invasive adenocarcinoma with a high grade, sarcomatoid or epithelial-mesenchymal transition (EMT) phenotype. PIN lesions were similar to those seen in PTEN conditional knockout mice, with evidence of AKT activation concomitant with neoplastic proliferation. However, the invasive tumor phenotype is rarely seen in previously described mouse models of prostatic neoplasia. These data indicate that the Tp53(R270H) mutation plays a role in CaP initiation. This finding has not previously been reported. Further characterization of this model, particularly in a setting of androgen deprivation, should allow further insight into the mechanisms by which the Tp53(R270H) mutation mediates CaP progression.


Sujet(s)
Substitution d'acide aminé/génétique , Évolution de la maladie , États précancéreux/génétique , États précancéreux/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Protéine p53 suppresseur de tumeur/génétique , Animaux , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Hétérozygote , Protéines à homéodomaine/métabolisme , Humains , Immunophénotypage , Integrases/métabolisme , Mâle , Souris , Souches mutantes de souris , Spécificité d'organe , Tumeur intraépithéliale prostate/enzymologie , Tumeur intraépithéliale prostate/anatomopathologie , Stabilité protéique , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs aux androgènes/métabolisme , Reproductibilité des résultats , Facteurs de transcription/métabolisme
8.
Clin Cancer Res ; 17(3): 550-9, 2011 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-21106723

RÉSUMÉ

PURPOSE: We apply positron emission tomography (PET) to elucidate changes in nanocarrier extravasation during the transition from premalignant to malignant cancer, providing insight into the use of imaging to characterize early cancerous lesions and the utility of nanoparticles in early disease. EXPERIMENTAL DESIGN: Albumin and liposomes were labeled with (64)Cu (half-life 12.7 hours), and longitudinal PET and CT imaging studies were conducted in a mouse model of ductal carcinoma in situ. A pharmacokinetic model was applied to estimate the tumor vascular volume and permeability. RESULTS: From early time points characterized by disseminated hyperproliferation, the enhanced vascular permeability facilitated lesion detection. During disease progression, the vascular volume fraction increased 1.6-fold and the apparent vascular permeability to albumin and liposomes increased ∼2.5-fold to 6.6 × 10(-8) and 1.3 × 10(-8) cm/s, respectively, with the accumulation of albumin increasing earlier in the disease process. In the malignant tumor, both tracers reached similar mean intratumoral concentrations of ∼6% ID/cc but the distribution of liposomes was more heterogeneous, ranging from 1% to 18% ID/cc compared with 1% to 9% ID/cc for albumin. The tumor-to-muscle ratio was 17.9 ± 8.1 and 7.1 ± 0.5 for liposomes and albumin, respectively, indicating a more specific delivery of liposomes than with albumin. CONCLUSIONS: PET imaging of radiolabeled particles, validated by confocal imaging and histology, detected the transition from premalignant to malignant lesions and effectively quantified the associated changes in vascular permeability.


Sujet(s)
Perméabilité capillaire , Épithélioma in situ/vascularisation , Transformation cellulaire néoplasique/métabolisme , Liposomes/métabolisme , Tumeurs expérimentales de la mamelle/vascularisation , Tomographie par émission de positons/méthodes , Animaux , Radio-isotopes du cuivre , Évolution de la maladie , Souris , Nanoparticules/analyse , Sérumalbumine/métabolisme
9.
J Am Assoc Lab Anim Sci ; 49(5): 610-6, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20858363

RÉSUMÉ

The purpose of this study was to determine the level of pain elicited by mammary fat pad removal surgery and the effects of postoperative analgesics on recovery. Female FVB mice were anesthetized, and mammary fat pad removal was performed. After surgery, mice received carprofen, buprenorphine, a combination of carprofen and buprenorphine, or saline treatment. Additional mice received anesthesia but no surgery or treatment. Food and water intake, body weight, wheel running activity, and a visual assessment score were recorded daily for 4 d after surgery and compared with presurgical findings. Corticosterone metabolites in fecal samples were analyzed at 12 and 24 h postsurgically and compared with baseline values. All surgical groups had significantly decreased food intake at 24 h, with a return to baseline by 48 h. The combination treatment resulted in a significantly decreased water intake and body weight at 24 h. All surgical groups had significantly decreased wheel running activity at 24 h only. The visual assessment scores indicated mild pain for all surgical groups, with the buprenorphine treated mice showing the highest pain index scores, as compared with nonsurgical controls. Fecal corticosterone metabolite levels did not differ significantly between any of the groups or across time. The parameters used in this study did not indicate that administration of these analgesic regimens improved recovery as compared with that of saline-treated mice. Care should be taken when using visual assessment scores to evaluate pain in mice, given that analgesics may have side effects that inadvertently elevate the score.


Sujet(s)
Analgésiques morphiniques/usage thérapeutique , Anti-inflammatoires non stéroïdiens/usage thérapeutique , Buprénorphine/usage thérapeutique , Carbazoles/usage thérapeutique , Association de médicaments/médecine vétérinaire , Glandes mammaires animales/chirurgie , Douleur postopératoire/médecine vétérinaire , Tissu adipeux/chirurgie , Animaux , Femelle , Souris/chirurgie , Lignées consanguines de souris , Mesure de la douleur/méthodes , Mesure de la douleur/médecine vétérinaire , Douleur postopératoire/traitement médicamenteux , Douleur postopératoire/prévention et contrôle , Répartition aléatoire , Résultat thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE