Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS Med ; 17(11): e1003323, 2020 11.
Article in English | MEDLINE | ID: mdl-33147277

ABSTRACT

BACKGROUND: The tumor microenvironment (TME) is increasingly appreciated as an important determinant of cancer outcome, including in multiple myeloma (MM). However, most myeloma microenvironment studies have been based on bone marrow (BM) aspirates, which often do not fully reflect the cellular content of BM tissue itself. To address this limitation in myeloma research, we systematically characterized the whole bone marrow (WBM) microenvironment during premalignant, baseline, on treatment, and post-treatment phases. METHODS AND FINDINGS: Between 2004 and 2019, 998 BM samples were taken from 436 patients with newly diagnosed MM (NDMM) at the University of Arkansas for Medical Sciences in Little Rock, Arkansas, United States of America. These patients were 61% male and 39% female, 89% White, 8% Black, and 3% other/refused, with a mean age of 58 years. Using WBM and matched cluster of differentiation (CD)138-selected tumor gene expression to control for tumor burden, we identified a subgroup of patients with an adverse TME associated with 17 fewer months of progression-free survival (PFS) (95% confidence interval [CI] 5-29, 49-69 versus 70-82 months, χ2 p = 0.001) and 15 fewer months of overall survival (OS; 95% CI -1 to 31, 92-120 versus 113-129 months, χ2 p = 0.036). Using immunohistochemistry-validated computational tools that identify distinct cell types from bulk gene expression, we showed that the adverse outcome was correlated with elevated CD8+ T cell and reduced granulocytic cell proportions. This microenvironment develops during the progression of premalignant to malignant disease and becomes less prevalent after therapy, in which it is associated with improved outcomes. In patients with quantified International Staging System (ISS) stage and 70-gene Prognostic Risk Score (GEP-70) scores, taking the microenvironment into consideration would have identified an additional 40 out of 290 patients (14%, premutation p = 0.001) with significantly worse outcomes (PFS, 95% CI 6-36, 49-73 versus 74-90 months) who were not identified by existing clinical (ISS stage III) and tumor (GEP-70) criteria as high risk. The main limitations of this study are that it relies on computationally identified cell types and that patients were treated with thalidomide rather than current therapies. CONCLUSIONS: In this study, we observe that granulocyte signatures in the MM TME contribute to a more accurate prognosis. This implies that future researchers and clinicians treating patients should quantify TME components, in particular monocytes and granulocytes, which are often ignored in microenvironment studies.


Subject(s)
Bone Marrow/pathology , Multiple Myeloma/diagnosis , Multiple Myeloma/pathology , Tumor Microenvironment , Adult , Cohort Studies , Female , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Prognosis , Tumor Burden
2.
J Immunol Methods ; 478: 112714, 2020 03.
Article in English | MEDLINE | ID: mdl-31783023

ABSTRACT

With the explosion of immuno-oncology and the approval of many immune checkpoint therapies by regulatory agencies in the last few years, understanding the tumor microenvironment (TME) in the context of patients' immune status has become essential. Among available immune profiling techniques, multiplex immunofluorescence (mIF) assays offer the unique advantage of preserving the architectural features of the tumor and revealing the spatial relationships between tumor cells and immune cells. A number of mIF and image analysis assays have been described for solid tumors but most are not sufficiently suitable in lymphoma, where the lack of clear tumor-stromal boundaries and high tumor density present significant challenges. Here we describe the development and optimization of a reliable workflow using Akoya Opal staining kits to label and analyze 6 markers per slide in diffuse large B-cell lymphoma (DLBCL) tissue sections. Five panels totaling 30 markers were developed to characterize infiltrating immune cells and relevant check-point proteins such as PD1, PD-L1, ICOS, SIRP-alpha and Lag3 on 70 DLBCL sections. Multiplexed sections were scanned using an Akoya multispectral scanner. An image analysis workflow using InForm and Matlab was developed to overcome challenges inherent to the DLBCL environment. Using the assays and workflows detailed here, we were able to quantify cell densities of subsets of infiltrating immune cells and observe their spatial patterns within the tumors. We highlight heterogeneous distribution of cytotoxic T cells across tumors with similar T cell density to underscores the importance of considering spatial context when studying the effects of immunological therapies in DLBCL.


Subject(s)
Biomarkers, Tumor/analysis , Fluorescent Antibody Technique/methods , High-Throughput Screening Assays/methods , Lymphoma, Large B-Cell, Diffuse/immunology , Tumor Microenvironment/immunology , Algorithms , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Feasibility Studies , Fluorescent Antibody Technique/instrumentation , Fluorescent Dyes/chemistry , High-Throughput Screening Assays/instrumentation , Humans , Image Processing, Computer-Assisted , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Reproducibility of Results , Software , Spatial Analysis , Staining and Labeling , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Workflow
3.
J Pathol ; 227(4): 417-30, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22611036

ABSTRACT

Resistance to anti-angiogenic therapy can occur via several potential mechanisms. Unexpectedly, recent studies showed that short-term inhibition of either VEGF or VEGFR enhanced tumour invasiveness and metastatic spread in preclinical models. In an effort to evaluate the translational relevance of these findings, we examined the consequences of long-term anti-VEGF monoclonal antibody therapy in several well-validated genetically engineered mouse tumour models of either neuroendocrine or epithelial origin. Anti-VEGF therapy decreased tumour burden and increased overall survival, either as a single agent or in combination with chemotherapy, in all four models examined. Importantly, neither short- nor long-term exposure to anti-VEGF therapy altered the incidence of metastasis in any of these autochthonous models, consistent with retrospective analyses of clinical trials. In contrast, we observed that sunitinib treatment recapitulated previously reported effects on tumour invasiveness and metastasis in a pancreatic neuroendocrine tumour (PNET) model. Consistent with these results, sunitinib treatment resulted in an up-regulation of the hypoxia marker GLUT1 in PNETs, whereas anti-VEGF did not. These results indicate that anti-VEGF mediates anti-tumour effects and therapeutic benefits without a paradoxical increase in metastasis. Moreover, these data underscore the concept that drugs targeting VEGF ligands and receptors may affect tumour metastasis in a context-dependent manner and are mechanistically distinct from one another.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Anti-Idiotypic/therapeutic use , Lung Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Vascular Endothelial Growth Factor A/immunology , Adenocarcinoma/genetics , Angiogenesis Inhibitors/therapeutic use , Animals , Disease Models, Animal , Drug Therapy, Combination , Genetic Engineering , Indoles/therapeutic use , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Mice , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrroles/therapeutic use , Small Cell Lung Carcinoma/genetics , Sunitinib , Vascular Endothelial Growth Factor A/antagonists & inhibitors
4.
Nature ; 474(7351): 403-6, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21572435

ABSTRACT

The proto-oncogenes ETV1, ETV4 and ETV5 encode transcription factors in the E26 transformation-specific (ETS) family, which includes the most frequently rearranged and overexpressed genes in prostate cancer. Despite being critical regulators of development, little is known about their post-translational regulation. Here we identify the ubiquitin ligase COP1 (also known as RFWD2) as a tumour suppressor that negatively regulates ETV1, ETV4 and ETV5. ETV1, which is mutated in prostate cancer more often, was degraded after being ubiquitinated by COP1. Truncated ETV1 encoded by prostate cancer translocation TMPRSS2:ETV1 lacks the critical COP1 binding motifs and was 50-fold more stable than wild-type ETV1. Almost all patient translocations render ETV1 insensitive to COP1, implying that this confers a selective advantage to prostate epithelial cells. Indeed, COP1 deficiency in mouse prostate elevated ETV1 and produced increased cell proliferation, hyperplasia, and early prostate intraepithelial neoplasia. Combined loss of COP1 and PTEN enhanced the invasiveness of mouse prostate adenocarcinomas. Finally, rare human prostate cancer samples showed hemizygous loss of the COP1 gene, loss of COP1 protein, and elevated ETV1 protein while lacking a translocation event. These findings identify COP1 as a tumour suppressor whose downregulation promotes prostatic epithelial cell proliferation and tumorigenesis.


Subject(s)
Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Animals , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Male , Mice , Nuclear Proteins/deficiency , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination
5.
PLoS One ; 6(3): e17449, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21394210

ABSTRACT

MYC and phosphoinositide 3-kinase (PI3K)-pathway deregulation are common in human prostate cancer. Through examination of 194 human prostate tumors, we observed statistically significant co-occurrence of MYC amplification and PI3K-pathway alteration, raising the possibility that these two lesions cooperate in prostate cancer progression. To investigate this, we generated bigenic mice in which both activated human AKT1 and human MYC are expressed in the prostate (MPAKT/Hi-MYC model). In contrast to mice expressing AKT1 alone (MPAKT model) or MYC alone (Hi-MYC model), the bigenic phenotype demonstrates accelerated progression of mouse prostate intraepithelial neoplasia (mPIN) to microinvasive disease with disruption of basement membrane, significant stromal remodeling and infiltration of macrophages, B- and T-lymphocytes, similar to inflammation observed in human prostate tumors. In contrast to the reversibility of mPIN lesions in young MPAKT mice after treatment with mTOR inhibitors, Hi-MYC and bigenic MPAKT/Hi-MYC mice were resistant. Additionally, older MPAKT mice showed reduced sensitivity to mTOR inhibition, suggesting that additional genetic events may dampen mTOR dependence. Since increased MYC expression is an early feature of many human prostate cancers, these data have implications for treatment of human prostate cancers with PI3K-pathway alterations using mTOR inhibitors.


Subject(s)
Precancerous Conditions/enzymology , Prostatic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Disease Models, Animal , Disease Progression , Enzyme Activation/drug effects , Humans , Male , Mice , Mice, Transgenic , Neoplasm Invasiveness , Phenotype , Precancerous Conditions/pathology , Prostate/drug effects , Prostate/pathology , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
6.
J Am Assoc Lab Anim Sci ; 49(3): 316-22, 2010 May.
Article in English | MEDLINE | ID: mdl-20587163

ABSTRACT

We and others frequently have noted serum potassium levels of 8.0 +/- 0.85 mEq/L or greater in laboratory mice; this concentration has even been published as the upper limit of a 'normal' reference range. However, if bone fide, this potassium concentration would be incompatible with life in all species. We investigated conditions frequently encountered in the research setting to distinguish artifactual from true hyperkalemia. Variables evaluated included site of collection, time allowed for clot formation before serum separation, time elapsed between collection and analysis of samples collected in a serum separator tube, precollection method of anesthesia, and euthanasia technique. Serum potassium was measured from 75 C57BL/6NTac 10-wk-old female mice and divided into at least 5 mice per variable. Animals were euthanized by exsanguination immediately after terminal CO2 or ketamine-xylazine (KX) administration. Mice euthanized with CO2 had higher mean serum potassium (7.0 +/- 0.5 mEq/L) and range serum potassium (6.0 to 8.1 mEq/L) than did KX-treated mice. CO2 inhalation resulted in significantly lower blood pH (6.9 +/- 0.1), higher pCO2 (153.3 +/- 38.8 mm Hg), and higher lactate levels (3.9 +/- 0.9 mmol/L) than did KX anesthesia followed by exsanguination. These results suggest that antemortem respiratory acidosis from CO2 administration causes artifactual hyperkalemia in mice. Therefore, blood collection under KX anesthesia is preferable over CO2 inhalation to obtain accurate potassium values from mice.


Subject(s)
Carbon Dioxide/toxicity , Euthanasia, Animal/methods , Mice/blood , Potassium/blood , Anesthesia/methods , Anesthesia/veterinary , Animals , Carbon Dioxide/administration & dosage , Female , Hematologic Tests/methods , Hematologic Tests/veterinary , Hyperkalemia/etiology , Hyperkalemia/veterinary , Ketamine/pharmacology , Mice, Inbred C57BL , Rodent Diseases/etiology , Xylazine/pharmacology
7.
Nat Genet ; 41(5): 524-6, 2009 May.
Article in English | MEDLINE | ID: mdl-19396167

ABSTRACT

The TMPRSS2-ERG fusion, present in approximately 50% of prostate cancers, is less common in prostatic intraepithelial neoplasia (PIN), raising questions about whether TMPRSS2-ERG contributes to disease initiation. We identified the translational start site of a common TMPRSS2-ERG fusion and showed that transgenic TMPRSS2-ERG mice develop PIN, but only in the context of PI3-kinase pathway activation. TMPRSS2-ERG-positive human tumors are also enriched for PTEN loss, suggesting cooperation in prostate tumorigenesis.


Subject(s)
Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction , Animals , Humans , Male , Mice , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Prostate/pathology , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology
8.
Differentiation ; 77(1): 103-11, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19281769

ABSTRACT

Tumor suppressor gene PTEN is important in the initiation and progression of human prostate carcinoma, whereas the role of TP53 remains controversial. Since Pten/Trp53 double conditional knockout mice show earlier onset and fast progression of prostate cancer when compared to Pten knockout mice, we asked whether heterozygosity of these two tumor suppressor genes was sufficient to accelerate prostatic tumorigenesis. To answer this question we examined prostatic lesion progression of Pten/Trp53 double heterozygous mice and a series of controls such as Pten heterozygous, Pten conditional knockout, Trp53 heterozygous and Trp53 knockout mice. Tissue recombination of adult prostatic epithelium coupled with embryonic rat seminal vesicle mesenchyme was used as a tool to stimulate prostatic epithelial proliferation. In our study, high-grade prostatic intraepithelial neoplasia (PIN) was found with high frequency at 8 weeks post-tissue recombination transplantation. PIN lesions in Pten/Trp53 double heterozygous mice were more severe than those seen in Pten heterozygous alone. Furthermore, morphologic features attributable to Pten or Trp53 loss appeared to be enhanced in double heterozygous tissues. LOH analysis of Pten and Trp53 in genomic DNA collected from high-grade PIN lesions in Pten heterozygous and Pten/Trp53 double heterozygous mice showed an intact wild-type allele for both genes in all samples examined. In conclusion, simultaneous heterozygosity of Pten and Trp53 accelerates prostatic tumorigenesis in this mouse model of prostate cancer independently of loss of heterozygosity of either gene.


Subject(s)
PTEN Phosphohydrolase/metabolism , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Disease Models, Animal , Heterozygote , Loss of Heterozygosity , Male , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , Rats , Tumor Suppressor Protein p53/genetics
9.
Endocr Pathol ; 19(3): 139-47, 2008.
Article in English | MEDLINE | ID: mdl-18758692

ABSTRACT

The genome has been sequenced. However, the functions of each gene remain to be elucidated through phenotypic analysis. This analysis has been called phenogenomics. That part of phenogenomics related to disease can be called pathogenomics or Genomic Pathology. The initial phases of disease analysis will use genetically modified mice. The proliferation of ambitious programs designed to use mice for phenogenomics has been met with alarm by comparative pathologists who note the lack of qualified genomic pathologists and of training programs in genomic pathology. While endocrine pathology offers a number of excellent examples of the contributions made by pathologists to the scientific literature, it also contains examples of the hazards of working with untrained, unwary personnel.


Subject(s)
Endocrine System Diseases/genetics , Endocrinology/trends , Genomics/trends , Animals , Humans
10.
Mol Cell ; 31(1): 21-32, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18614044

ABSTRACT

The Mre11 complex (Mre11, Rad50, and Nbs1) and Chk2 have been implicated in the DNA-damage response, an inducible process required for the suppression of malignancy. The Mre11 complex is predominantly required for repair and checkpoint activation in S phase, whereas Chk2 governs apoptosis. We examined the relationship between the Mre11 complex and Chk2 in the DNA-damage response via the establishment of Nbs1(DeltaB/DeltaB) Chk2(-/-) and Mre11(ATLD1/ATLD1) Chk2(-/-) mice. Chk2 deficiency did not modify the checkpoint defects or chromosomal instability of Mre11 complex mutants; however, the double-mutant mice exhibited synergistic defects in DNA-damage-induced p53 regulation and apoptosis. Nbs1(DeltaB/DeltaB) Chk2(-/-) and Mre11(ATLD1/ATLD1) Chk2(-/-) mice were also predisposed to tumors. In contrast, DNA-PKcs-deficient mice, in which G1-specific chromosome breaks are present, did not exhibit synergy with Chk2(-/-) mutants. These data suggest that Chk2 suppresses the oncogenic potential of DNA damage arising during S and G2 phases of the cell cycle.


Subject(s)
DNA Damage , DNA Replication , Precancerous Conditions/enzymology , Protein Serine-Threonine Kinases/metabolism , Alleles , Animals , Apoptosis , Cell Cycle , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2 , Chromosomal Instability , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Repair , DNA Repair Enzymes/metabolism , DNA, Complementary/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Exons/genetics , Genome/genetics , MRE11 Homologue Protein , Mice , Mutation/genetics , Nuclear Proteins/metabolism , Precancerous Conditions/pathology , Protein Serine-Threonine Kinases/deficiency , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
11.
Genes Dev ; 22(13): 1773-85, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18550783

ABSTRACT

Dyskeratosis congenita (DC) is an inherited bone marrow failure syndrome characterized by cutaneous symptoms, including hyperpigmentation and nail dystrophy. Some forms of DC are caused by mutations in telomerase, the enzyme that counteracts telomere shortening, suggesting a telomere-based disease mechanism. However, mice with extensively shortened telomeres due to telomerase deficiency do not develop the characteristics of DC, raising questions about the etiology of DC and/or mouse models for human telomere dysfunction. Here we describe mice engineered to undergo telomere degradation due to the absence of the shelterin component POT1b. When combined with reduced telomerase activity, POT1b deficiency elicits several characteristics of DC, including hyperpigmentation and fatal bone marrow failure at 4-5 mo of age. These results provide experimental support for the notion that DC is caused by telomere dysfunction, and demonstrate that key aspects of a human telomere-based disease can be modeled in the mouse.


Subject(s)
Dyskeratosis Congenita , Telomerase/metabolism , Telomere/ultrastructure , Animals , Bone Marrow/abnormalities , Bone Marrow/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Disease Models, Animal , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/pathology , Longevity , Mice , Phenotype , Skin Pigmentation , Telomerase/genetics
12.
Am J Pathol ; 172(1): 236-46, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18156212

ABSTRACT

The transgenic adenocarcinoma of mouse prostate (TRAMP) model is widely used in prostate cancer research because of rapid tumor onset and progression. The transgenic mouse is on a C57BL/6 (B6) background and expresses SV40 T-antigen under the probasin promoter. The strong genetic component of susceptibility to prostate cancer in humans prompted us to investigate the effect of mouse strain background (FVB and B6) on incidence, progression, and pathology of prostate cancer in this model. Because TRAMP lesions are unique but differ from conventional prostatic intraepithelial neoplasia because the epithelium and stroma are affected diffusely, we designated them as "atypical hyperplasia of Tag." Although the incidence and severity of atypical hyperplasia of Tag is similar, FVB-TRAMP mice live significantly shorter lives than B6-TRAMP mice because of the rapid development and progression of neuroendocrine carcinomas. This is associated with an increased frequency of neuroendocrine precursor lesions in young TRAMP mice, detectable at 4 weeks after birth. These lesions show properties of bipotential stem cells and co-express markers of epithelial (E-cadherin) and neuroendocrine (synaptophysin) lineages, as well as the transcription factors Foxa1 and Foxa2. Transplantation studies using TRAMP prostatic ducts suggested that neuroendocrine carcinomas arise independently from atypical hyperplasias or other epithelial lesions. Adenocarcinomas were not seen in our cohort. Thus, neuroendocrine carcinomas are the principal malignancy in this model and may develop from bipotential progenitor cells at an early stage of prostate tumorigenesis.


Subject(s)
Adenocarcinoma/genetics , Prostatic Neoplasms/genetics , Adenocarcinoma/pathology , Animals , Cell Lineage , Disease Models, Animal , Humans , Kidney/metabolism , Male , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis , Neoplasms, Glandular and Epithelial/pathology , Neuroendocrine Tumors/pathology , Prostatic Neoplasms/pathology , Transgenes
13.
Nature ; 447(7141): 218-21, 2007 May 10.
Article in English | MEDLINE | ID: mdl-17429352

ABSTRACT

The MRE11 complex (MRE11, RAD50 and NBS1) and the ataxia-telangiectasia mutated (ATM) kinase function in the same DNA damage response pathway to effect cell cycle checkpoint activation and apoptosis. The functional interaction between the MRE11 complex and ATM has been proposed to require a conserved C-terminal domain of NBS1 for recruitment of ATM to sites of DNA damage. Human Nijmegen breakage syndrome (NBS) cells and those derived from multiple mouse models of NBS express a hypomorphic NBS1 allele that exhibits impaired ATM activity despite having an intact C-terminal domain. This indicates that the NBS1 C terminus is not sufficient for ATM function. We derived Nbs1(DeltaC/DeltaC) mice in which the C-terminal ATM interaction domain is deleted. Nbs1(DeltaC/DeltaC) cells exhibit intra-S-phase checkpoint defects, but are otherwise indistinguishable from wild-type cells with respect to other checkpoint functions, ionizing radiation sensitivity and chromosome stability. However, multiple tissues of Nbs1(DeltaC/DeltaC) mice showed a severe apoptotic defect, comparable to that of ATM- or CHK2-deficient animals. Analysis of p53 transcriptional targets and ATM substrates showed that, in contrast to the phenotype of Chk2(-/-) mice, NBS1(DeltaC) does not impair the induction of proapoptotic genes. Instead, the defects observed in Nbs1(DeltaC/DeltaC) result from impaired phosphorylation of ATM targets including SMC1 and the proapoptotic factor, BID.


Subject(s)
Apoptosis , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , ATP-Binding Cassette Transporters/metabolism , Acid Anhydride Hydrolases , Alleles , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , BH3 Interacting Domain Death Agonist Protein/deficiency , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Cycle Proteins/genetics , Cell Line , Checkpoint Kinase 2 , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Humans , MRE11 Homologue Protein , Mice , Molecular Sequence Data , Multiprotein Complexes/metabolism , Nuclear Proteins/genetics , Phenotype , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Sequence Deletion/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
Theriogenology ; 65(3): 517-27, 2006 Feb.
Article in English | MEDLINE | ID: mdl-15993483

ABSTRACT

Uterine smooth muscle specimens were collected from euthanatized mares in estrus and diestrus. Longitudinal and circular specimens were mounted in organ baths and the signals transcribed to a Grass polygraph. After equilibration time and 2 g preload, their physiologic isometric contractility was recorded for a continuous 2.0 h. Area under the curve, frequency and time occupied by contractions were studied. Differences between cycle phases, between muscle layers, and over the recorded time periods were statistically evaluated using linear mixed-effect models. In the mare, physiologic contractility of the uterus decreased significantly over time for all variables evaluated (time as covariate on a continuous scale). For area under the curve, there was a significant effect of muscle layer (longitudinal > circular). For frequency, higher values were recorded in estrus for circular smooth muscle layer, whereas higher values were seen in longitudinal smooth muscle layers during diestrus. In longitudinal layer and in diestrus, more time was occupied by contractions than in circular layer, and in estrus. This study is describing physiologic myometrial motility in the organ bath depending on cycle phase.


Subject(s)
Diestrus/physiology , Estrus/physiology , Horses/physiology , Myometrium/physiology , Uterine Contraction/physiology , Animals , Area Under Curve , Female , In Vitro Techniques , Linear Models , Muscle, Smooth/physiology
SELECTION OF CITATIONS
SEARCH DETAIL