Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Toxicol Pathol ; 51(6): 375-389, 2023 08.
Article in English | MEDLINE | ID: mdl-38179962

ABSTRACT

Direct delivery of therapeutics to the central nervous system (CNS) greatly expands opportunities to treat neurological diseases but is technically challenging. This opinion outlines principal technical aspects of direct CNS delivery via intracerebroventricular (ICV) or intrathecal (IT) injection to common nonclinical test species (rodents, dogs, and nonhuman primates) and describes procedure-related clinical and histopathological effects that confound interpretation of test article-related effects. Direct dosing is by ICV injection in mice due to their small body size, while other species are dosed IT in the lumbar cistern. The most frequent procedure-related functional effects are transient absence of lower spinal reflexes after IT injection or death soon after ICV dosing. Common procedure-related microscopic findings in all species include leukocyte infiltrates in CNS meninges or perivascular (Virchow-Robin) spaces; nerve fiber degeneration in the spinal cord white matter (especially dorsal and lateral tracts compressed by dosing needles or indwelling catheters), spinal nerve roots, and sciatic nerve; meningeal fibrosis at or near IT injection sites; hemorrhage; and gliosis. Findings typically are minimal to occasionally mild. Findings tend to be more severe and/or have a higher incidence in the spinal cord segments and spinal nerve roots at or close to the site of administration.


Subject(s)
Oligonucleotides , Rodentia , Dogs , Mice , Animals , Central Nervous System/pathology , Spinal Cord/pathology , Nerve Degeneration/pathology , Primates
2.
Cancer Res ; 80(3): 430-443, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31740444

ABSTRACT

Emerging evidence indicates the deubiquitinase USP22 regulates transcriptional activation and modification of target substrates to promote pro-oncogenic phenotypes. Here, in vivo characterization of tumor-associated USP22 upregulation and unbiased interrogation of USP22-regulated functions in vitro demonstrated critical roles for USP22 in prostate cancer. Specifically, clinical datasets validated that USP22 expression is elevated in prostate cancer, and a novel murine model demonstrated a hyperproliferative phenotype with prostate-specific USP22 overexpression. Accordingly, upon overexpression or depletion of USP22, enrichment of cell-cycle and DNA repair pathways was observed in the USP22-sensitive transcriptome and ubiquitylome using prostate cancer models of clinical relevance. Depletion of USP22 sensitized cells to genotoxic insult, and the role of USP22 in response to genotoxic insult was further confirmed using mouse adult fibroblasts from the novel murine model of USP22 expression. As it was hypothesized that USP22 deubiquitylates target substrates to promote protumorigenic phenotypes, analysis of the USP22-sensitive ubiquitylome identified the nucleotide excision repair protein, XPC, as a critical mediator of the USP22-mediated response to genotoxic insult. Thus, XPC undergoes deubiquitylation as a result of USP22 function and promotes USP22-mediated survival to DNA damage. Combined, these findings reveal unexpected functions of USP22 as a driver of protumorigenic phenotypes and have significant implications for the role of USP22 in therapeutic outcomes. SIGNIFICANCE: The studies herein present a novel mouse model of tumor-associated USP22 overexpression and implicate USP22 in modulation of cellular survival and DNA repair, in part through regulation of XPC.


Subject(s)
Carcinogenesis/pathology , Cell Proliferation , DNA Repair Enzymes/metabolism , DNA Repair , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , Ubiquitin Thiolesterase/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , DNA Damage , DNA Repair Enzymes/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured , Ubiquitin Thiolesterase/genetics , Xenograft Model Antitumor Assays
3.
Vet Pathol ; 55(2): 331-340, 2018 03.
Article in English | MEDLINE | ID: mdl-29338616

ABSTRACT

C-terminal Src kinase (Csk) is one of the critical negative regulators of the Src family of kinases. The Src family of kinases are nonreceptor tyrosine kinases that regulate inflammation, cell proliferation, motility, and adhesion. To investigate potential histologic lesions associated with systemic loss of Csk gene activity in adult mice, conditional Csk-knockout mice were examined. Cre-mediated systemic excision of Csk induced by tamoxifen treatment resulted in multiorgan inflammation. Specifically, induction of Csk gene excision with three days of tamoxifen treatment resulted in greater than 90% gene excision. Strikingly, these mice developed enteritis that ranged from minimal and suppurative to severe, fibrinonecrosuppurative and hemorrhagic. Other inflammatory lesions included suppurative pneumonia, gastritis, and myocarditis, and increased numbers of inflammatory cells within the hepatic parenchyma. When tamoxifen treatment was reduced from three days to one day in an effort to lower the level of Csk gene excision and limit lesion development, the mice developed severe suppurative to pyogranulomatous pneumonia and minimal to mild suppurative enteritis. Lesions observed secondary to Csk gene excision suggest important roles for Csk in downregulating the proinflammatory activity of the Src family of kinases and limiting neutrophil-mediated inflammation.


Subject(s)
Inflammation/veterinary , Mice, Knockout/metabolism , Suppuration/veterinary , src-Family Kinases/metabolism , Animals , Blotting, Southern , CSK Tyrosine-Protein Kinase , Female , Gene Expression , Inflammation/metabolism , Inflammation/pathology , Male , Suppuration/metabolism , Suppuration/pathology
4.
Cell Rep ; 13(10): 2312-23, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26673324

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is a neurodegenerative disease caused by a polyglutamine expansion in the androgen receptor (AR) and is associated with misfolding and aggregation of the mutant AR. We investigated the role of an interdomain interaction between the amino (N)-terminal FxxLF motif and carboxyl (C)-terminal AF-2 domain in a mouse model of SBMA. Male transgenic mice expressing polyQ-expanded AR with a mutation in the FxxLF motif (F23A) to prevent the N/C interaction displayed substantially improved motor function compared with N/C-intact AR-expressing mice and showed reduced pathological features of SBMA. Serine 16 phosphorylation was substantially enhanced by the F23A mutation; moreover, the protective effect of AR F23A was dependent on this phosphorylation. These results reveal an important role for the N/C interaction on disease onset in mice and suggest that targeting AR conformation could be a therapeutic strategy for patients with SBMA.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/physiopathology , Receptors, Androgen/chemistry , Animals , Disease Models, Animal , Immunoprecipitation , Male , Mice , Mice, Transgenic , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , Receptors, Androgen/metabolism
5.
Cancer Res ; 75(17): 3554-67, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26206558

ABSTRACT

The treatment options remain limited for patients with melanoma who are wild-type for both BRAF and NRAS (WT/WT). We demonstrate that a subgroup of WT/WT melanomas display high basal phosphorylation of ErbB3 that is associated with autocrine production of the ErbB3 ligand neuregulin-1 (NRG1). In WT/WT melanoma cells displaying high levels of phospho-ErbB3, knockdown of NRG1 reduced cell viability and was associated with decreased phosphorylation of ErbB3, its coreceptor ErbB2, and its downstream target, AKT. Similar effects were observed by targeting ErbB3 with either siRNAs or the neutralizing ErbB3 monoclonal antibodies huHER3-8 and NG33. In addition, pertuzumab-mediated inhibition of ErbB2 heterodimerization decreased AKT phosphorylation, cell growth in vitro, and xenograft growth in vivo. Pertuzumab also potentiated the effects of MEK inhibitor on WT/WT melanoma growth in vitro and in vivo. These findings demonstrate that targeting ErbB3-ErbB2 signaling in a cohort of WT/WT melanomas leads to tumor growth reduction. Together, these studies support the rationale to target the NRG1-ErbB3-ErbB2 axis as a novel treatment strategy in a subset of cutaneous melanomas.


Subject(s)
Melanoma/genetics , Neuregulin-1/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Antibodies, Monoclonal, Humanized/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/genetics , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , Melanoma/pathology , Membrane Proteins/genetics , Molecular Targeted Therapy , Neuregulin-1/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Skin Neoplasms , Melanoma, Cutaneous Malignant
6.
EMBO Mol Med ; 7(5): 628-47, 2015 May.
Article in English | MEDLINE | ID: mdl-25787974

ABSTRACT

Clinical evidence suggests that cyclin D1b, a variant of cyclin D1, is associated with tumor progression and poor outcome. However, the underlying molecular basis was unknown. Here, novel models were created to generate a genetic switch from cyclin D1 to cyclin D1b. Extensive analyses uncovered overlapping but non-redundant functions of cyclin D1b compared to cyclin D1 on developmental phenotypes, and illustrated the importance of the transcriptional regulatory functions of cyclin D1b in vivo. Data obtained identify cyclin D1b as an oncogene, wherein cyclin D1b expression under the endogenous promoter induced cellular transformation and further cooperated with known oncogenes to promote tumor growth in vivo. Further molecular interrogation uncovered unexpected links between cyclin D1b and the DNA damage/PARP1 regulatory networks, which could be exploited to suppress cyclin D1b-driven tumors. Collectively, these data are the first to define the consequence of cyclin D1b expression on normal cellular function, present evidence for cyclin D1b as an oncogene, and provide pre-clinical evidence of effective methods to thwart growth of cells dependent upon this oncogenic variant.


Subject(s)
Cell Transformation, Neoplastic , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Gene Regulatory Networks , Mice, Inbred C57BL , Mice, Transgenic
7.
Clin Cancer Res ; 21(4): 795-807, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25691773

ABSTRACT

PURPOSE: To improve the outcomes of patients with castration-resistant prostate cancer (CRPC), there is an urgent need for more effective therapies and approaches that individualize specific treatments for patients with CRPC. These studies compared the novel taxane cabazitaxel with the previous generation docetaxel, and aimed to determine which tumors are most likely to respond. EXPERIMENTAL DESIGN: Cabazitaxel and docetaxel were compared via in vitro modeling to determine the molecular mechanism, biochemical and cell biologic impact, and cell proliferation, which was further assessed ex vivo in human tumor explants. Isogenic pairs of RB knockdown and control cells were interrogated in vitro and in xenograft tumors for cabazitaxel response. RESULTS: The data herein show that (i) cabazitaxel exerts stronger cytostatic and cytotoxic response compared with docetaxel, especially in CRPC; (ii) cabazitaxel induces aberrant mitosis, leading to pyknotic and multinucleated cells; (iii) taxanes do not act through the androgen receptor (AR); (iv) gene-expression profiling reveals distinct molecular actions for cabazitaxel; and (v) tumors that have progressed to castration resistance via loss of RB show enhanced sensitivity to cabazitaxel. CONCLUSIONS: Cabazitaxel not only induces improved cytostatic and cytotoxic effects, but also affects distinct molecular pathways, compared with docetaxel, which could underlie its efficacy after docetaxel treatment has failed in patients with CRPC. Finally, RB is identified as the first potential biomarker that could define the therapeutic response to taxanes in metastatic CRPC. This would suggest that loss of RB function induces sensitization to taxanes, which could benefit up to 50% of CRPC cases.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Taxoids/pharmacology , Animals , Cell Line, Tumor , Docetaxel , Flow Cytometry , Humans , Immunoblotting , Male , Mice , Microscopy, Fluorescence , Models, Molecular , Oligonucleotide Array Sequence Analysis , Transcriptome , Xenograft Model Antitumor Assays
8.
Endocr Relat Cancer ; 22(1): R33-49, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25349195

ABSTRACT

Prostate cancer remains the second leading cause of cancer death in men in the USA and most western countries. Prostatic acinar adenocarcinoma is the most commonly diagnosed form of prostate cancer. Small-cell neuroendocrine carcinoma is less frequently identified at the time of initial diagnosis, but this highly aggressive form of prostate cancer is increasingly observed in patients who have failed first- and second-line hormone therapy. Thus, developing and exploring models of neuroendocrine prostate cancer (NePC) are of increasing importance. This review examines the relevant xenograft tumor and genetically engineered mouse models of NePC, with the aim of addressing salient features and clinical relevance.


Subject(s)
Carcinoma, Neuroendocrine/pathology , Disease Models, Animal , Prostatic Neoplasms/pathology , Animals , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Heterografts , Humans , Male , Mice , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
9.
Toxicol Pathol ; 43(2): 186-97, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24742627

ABSTRACT

Male transgenic adenocarcinoma of the mouse prostate (TRAMP) mice are frequently used in prostate cancer research because their prostates consistently develop a series of preneoplastic and neoplastic lesions. Disease progression in TRAMP mouse prostates culminates in metastatic, poorly differentiated carcinomas with neuroendocrine features. The androgen dependence of the rat probasin promoter largely limits transgene expression to the prostatic epithelium. However, extra-prostatic transgene-positive lesions have been described in TRAMP mice, including renal tubuloacinar carcinomas, neuroendocrine carcinomas of the urethra, and phyllodes-like tumors of the seminal vesicle. Here, we describe the histologic and immunohistochemical features of 2 novel extra-prostatic lesions in TRAMP mice: primary anaplastic tumors of uncertain cell origin in the midbrain and poorly differentiated adenocarcinomas of the submandibular salivary gland. These newly characterized tumors apparently result from transgene expression in extra-prostatic locations rather than representing metastatic prostate neoplasms because lesions were identified in both male and female mice and in male TRAMP mice without histologically apparent prostate tumors. In this article, we also calculate the incidences of the urethral carcinomas and renal tubuloacinar carcinomas, further elucidate the biological behavior of the urethral carcinomas, and demonstrate the critical importance of complete necropsies even when evaluating presumably well characterized phenotypes in genetically engineered mice.


Subject(s)
Adenocarcinoma/genetics , Prostatic Neoplasms/genetics , Transgenes/genetics , Adenocarcinoma/pathology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Female , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/secondary , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Necrosis , Prostatic Neoplasms/pathology , Submandibular Gland Neoplasms/genetics , Submandibular Gland Neoplasms/pathology , Submandibular Gland Neoplasms/secondary , Urethral Neoplasms/genetics , Urethral Neoplasms/pathology , Urethral Neoplasms/secondary
10.
Oncotarget ; 5(19): 9460-71, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25294806

ABSTRACT

The emergence of targeted cancer therapy has been limited by the paucity of determinants which are tumor-specific and generally associated with disease, and have cell dynamics which effectively deploy cytotoxic payloads. Guanylyl cyclase C (GUCY2C) may be ideal for targeting because it is normally expressed only in insulated barrier compartments, including intestine and brain, but over-expressed by systemic metastatic colorectal tumors. Here, we reveal that GUCY2C rapidly internalizes from the cell surface to lysosomes in intestinal and colorectal cancer cells. Endocytosis is independent of ligand binding and receptor activation, and is mediated by clathrin. This mechanism suggests a design for immunotoxins comprising a GUCY2C-directed monoclonal antibody conjugated through a reducible disulfide linkage to ricin A chain, which is activated to a potent cytotoxin in lysosomes. Indeed, this immunotoxin specifically killed GUCY2C-expressing colorectal cancer cells in a lysosomal- and clathrin-dependent fashion. Moreover, this immunotoxin reduced pulmonary tumors>80% (p<0.001), and improved survival 25% (p<0.001), in mice with established colorectal cancer metastases. Further, therapeutic efficacy was achieved without histologic evidence of toxicity in normal tissues. These observations support GUCY2C-targeted immunotoxins as novel therapeutics for metastatic tumors originating in the GI tract, including colorectum, stomach, esophagus, and pancreas.


Subject(s)
Antibodies, Monoclonal/immunology , Gastrointestinal Neoplasms/drug therapy , Immunotoxins/pharmacology , Receptors, Guanylate Cyclase-Coupled/metabolism , Receptors, Peptide/metabolism , Ricin/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Caveolins/metabolism , Cell Line, Tumor , Clathrin/metabolism , Drug Carriers , Endocytosis , Gastrointestinal Neoplasms/pathology , Immunotherapy , Immunotoxins/administration & dosage , Lysosomes , Mice , Molecular Targeted Therapy , Protein Transport , RNA Interference , RNA, Small Interfering , Receptors, Enterotoxin , Ricin/administration & dosage , Ricin/genetics
11.
J Med Food ; 17(11): 1177-82, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24983303

ABSTRACT

Broccoli is rich in bioactive components, such as sulforaphane and indole-3-carbinol, which may impact cancer risk. The glucosinolate profile of broccoli can be manipulated through treatment with the plant stress hormone methyl jasmonate (MeJA). Our objective was to produce broccoli with enhanced levels of indole glucosinolates and determine its impact on prostate carcinogenesis. Brassica oleracea var. Green Magic was treated with a 250 µM MeJA solution 4 days prior to harvest. MeJA-treated broccoli had significantly increased levels of glucobrassicin, neoglucobrassicin, and gluconasturtiin (P < .05). Male transgenic adenocarcinoma of mouse prostate (TRAMP) mice (n = 99) were randomized into three diet groups at 5-7 weeks of age: AIN-93G control, 10% standard broccoli powder, or 10% MeJA broccoli powder. Diets were fed throughout the study until termination at 20 weeks of age. Hepatic CYP1A was induced with MeJA broccoli powder feeding, indicating biological activity of the indole glucosinolates. Following ∼ 15 weeks on diets, neither of the broccoli treatments significantly altered genitourinary tract weight, pathologic score, or metastasis incidence, indicating that broccoli powder at 10% of the diet was ineffective at reducing prostate carcinogenesis in the TRAMP model. Whereas broccoli powder feeding had no effect in this model of prostate cancer, our work demonstrates the feasibility of employing plant stress hormones exogenously to stimulate changes in phytochemical profiles, an approach that may be useful for optimizing bioactive component patterns in foods for chronic-disease-prevention studies.


Subject(s)
Acetates/pharmacology , Brassica/metabolism , Carcinogenesis/drug effects , Cyclopentanes/pharmacology , Glucosinolates/pharmacology , Indoles/pharmacology , Oxylipins/pharmacology , Plant Extracts/pharmacology , Prostatic Neoplasms , Animals , Brassica/drug effects , Glucosinolates/metabolism , Glucosinolates/therapeutic use , Indoles/metabolism , Indoles/therapeutic use , Male , Mice, Inbred C57BL , Plant Extracts/therapeutic use , Plant Growth Regulators/pharmacology , Prostatic Neoplasms/drug therapy
12.
Mol Nutr Food Res ; 58(6): 1226-38, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24668769

ABSTRACT

SCOPE: Ulcerative colitis (UC) is a chronic inflammatory disease of the colon. α-Mangostin (α-MG), the most abundant xanthone in mangosteen fruit, exerts anti-inflammatory and antibacterial activities in vitro. We evaluated the impact of dietary α-MG on murine experimental colitis and on the gut microbiota of healthy mice. METHODS AND RESULTS: Colitis was induced in C57BL/6J mice by administration of dextran sulfate sodium (DSS). Mice were fed control diet or diet with α-MG (0.1%). α-MG exacerbated the pathology of DSS-induced colitis. Mice fed diet with α-MG had greater colonic inflammation and injury, as well as greater infiltration of CD3(+) and F4/80(+) cells, and colonic myeloperoxidase, than controls. Serum levels of granulocyte colony-stimulating factor, IL-6, and serum amyloid A were also greater in α-MG-fed animals than in controls. The colonic and cecal microbiota of healthy mice fed α-MG but no DSS shifted to an increased abundance of Proteobacteria and decreased abundance of Firmicutes and Bacteroidetes, a profile similar to that found in human UC. CONCLUSION: α-MG exacerbated colonic pathology during DSS-induced colitis. These effects may be associated with an induction of intestinal dysbiosis by α-MG. Our results suggest that the use of α-MG-containing supplements by patients with UC may have unintentional risk.


Subject(s)
Colitis, Ulcerative/pathology , Dysbiosis/pathology , Garcinia mangostana/chemistry , Xanthones/adverse effects , Amyloid/blood , Animals , Colitis, Ulcerative/chemically induced , Colon/drug effects , Colon/enzymology , Colon/microbiology , Dextran Sulfate , Diet , Dietary Supplements , Disease Models, Animal , Dysbiosis/chemically induced , Female , Fruit/chemistry , Interleukin-6/blood , Mice , Mice, Inbred C57BL , Peroxidase/metabolism , Proteobacteria/isolation & purification
13.
Cancer Prev Res (Phila) ; 6(3): 232-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23275006

ABSTRACT

Cells undergoing malignant transformation often exhibit a shift in cellular metabolism from oxidative phosphorylation to glycolysis. This glycolytic shift, called the Warburg effect, provides a mechanistic basis for targeting glycolysis to suppress carcinogenesis through the use of dietary caloric restriction and energy restriction-mimetic agents (ERMA). We recently reported the development of a novel class of ERMAs that exhibits high potency in eliciting starvation-associated cellular responses and epigenetic changes in cancer cells though glucose uptake inhibition. The lead ERMA in this class, OSU-CG5, decreases the production of ATP and NADH in LNCaP prostate cancer cells. In this study, we examined the effect of OSU-CG5 on the severity of preneoplastic lesions in male transgenic adenocarcinoma of the mouse prostate (TRAMP) mice. Daily oral treatment with OSU-CG5 at 100 mg/kg from 6 to 10 weeks of age resulted in a statistically significant decrease in the weight of urogenital tract and microdissected dorsal, lateral, and anterior prostatic lobes relative to vehicle controls. The suppressive effect of OSU-CG5 was evidenced by marked decreases in Ki67 immunostaining and proliferating cell nuclear antigen (PCNA) expression in the prostate. OSU-CG5 treatment was not associated with evidence of systemic toxicity. Microarray analysis indicated a central role for Akt, and Western blot analysis showed reduced phosphorylation and/or expression levels of Akt, Src, androgen receptor, and insulin-like growth factor-1 receptor in prostate lobes. These findings support further investigation of OSU-CG5 as a potential chemopreventive agent.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects , Thiazolidinediones/pharmacology , Adenocarcinoma/pathology , Animals , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Glycolysis/drug effects , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Prostatic Neoplasms/pathology
14.
Toxicol Pathol ; 40(1): 5-17, 2012.
Article in English | MEDLINE | ID: mdl-22021166

ABSTRACT

The transgenic adenocarcinoma of the mouse prostate (TRAMP) model is well established and offers several advantages for the study of chemopreventive agents, including its well-defined course of disease progression and high incidence of poorly differentiated carcinomas within a relatively short length of time. However, there is no consensus on the grading of prostatic lesions in these mice. In particular, agreement is lacking on the criteria for differentiating prostatic intraepithelial neoplasia (PIN) from well-differentiated adenocarcinoma, specifically as it relates to evidence of invasion. This differentiation is critical for evaluating the effects of putative chemopreventive agents on progression to neoplasia. Moreover, only one of the published grading schemes assigns numerical grades to prostatic lesions, which facilitate statistical analysis. Here, we review five currently available grading schemes and propose a refined scheme that provides a useful definition of invasion for the differentiation of PIN from well-differentiated adenocarcinoma and includes a numerical scoring system that accounts for both the most severe and most common histopathological lesions in each of the lobes of the prostate and their distributions. We expect that researchers will find this refined grading scheme to be useful for chemoprevention studies in TRAMP mice.


Subject(s)
Adenocarcinoma/pathology , Prostatic Neoplasms/pathology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Grading/methods
15.
Hepatology ; 53(6): 1943-58, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21391227

ABSTRACT

UNLABELLED: Accumulating evidence suggests the therapeutic potential of the immunosuppressive agent FTY720 (fingolimod) in hepatocellular carcinoma (HCC). Based on our previous finding that FTY720 mediates apoptosis in HCC cells by activating reactive oxygen species (ROS)-protein kinase Cδ (PKCδ) signaling independent of effects on sphingosine-1-phosphate (S1P) receptors, we embarked on the pharmacological exploitation of FTY720 to develop a nonimmunosuppressive analogue with antitumor activity. This effort led to the development of OSU-2S, which exhibits higher potency than FTY720 in suppressing HCC cell growth through PKCδ activation. In contrast to FTY720, OSU-2S was not phosphorylated by sphingosine kinase 2 (SphK2) in vitro, and did not cause S1P1 receptor internalization in HCC cells or T lymphocyte homing in immunocompetent mice. Although devoid of S1P1 receptor activity, OSU-2S exhibited higher in vitro antiproliferative efficacy relative to FTY720 against HCC cells without cytotoxicity in normal hepatocytes. Several lines of pharmacological and molecular genetic evidence indicate that ROS-PKCδ-caspase-3 signaling underlies OSU-2S-mediated antitumor effects, and that differences in the antitumor activity between FTY720 and OSU-2S were attributable to SphK2-mediated phosphorylation of FTY720, which represents a metabolic inactivation of its antitumor activity. Finally, OSU-2S exhibited high in vivo potency in suppressing xenograft tumor growth in both ectopic and orthotopic models without overt toxicity. CONCLUSION: Using the molecular platform of FTY720, we developed OSU-2S, a novel PKCδ-targeted antitumor agent, which is devoid of S1P1 receptor activity and is highly effective in suppressing HCC tumor growth in vivo. These findings suggest that OSU-2S has clinical value in therapeutic strategies for HCC and warrants continued investigation in this regard.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Liver Neoplasms/pathology , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/physiopathology , Caspase 3/metabolism , Cell Line, Tumor , Cells, Cultured , Fingolimod Hydrochloride , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Mice , Mice, Nude , Protein Kinase C-delta/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/pharmacology , Xenograft Model Antitumor Assays
17.
Nature ; 446(7135): 552-6, 2007 Mar 29.
Article in English | MEDLINE | ID: mdl-17322906

ABSTRACT

Intestinal epithelial cells (IECs) provide a primary physical barrier against commensal and pathogenic microorganisms in the gastrointestinal (GI) tract, but the influence of IECs on the development and regulation of immunity to infection is unknown. Here we show that IEC-intrinsic IkappaB kinase (IKK)-beta-dependent gene expression is a critical regulator of responses of dendritic cells and CD4+ T cells in the GI tract. Mice with an IEC-specific deletion of IKK-beta show a reduced expression of the epithelial-cell-restricted cytokine thymic stromal lymphopoietin in the intestine and, after infection with the gut-dwelling parasite Trichuris, fail to develop a pathogen-specific CD4+ T helper type 2 (T(H)2) response and are unable to eradicate infection. Further, these animals show exacerbated production of dendritic-cell-derived interleukin-12/23p40 and tumour necrosis factor-alpha, increased levels of CD4+ T-cell-derived interferon-gamma and interleukin-17, and develop severe intestinal inflammation. Blockade of proinflammatory cytokines during Trichuris infection ablates the requirement for IKK-beta in IECs to promote CD4+ T(H)2 cell-dependent immunity, identifying an essential function for IECs in tissue-specific conditioning of dendritic cells and limiting type 1 cytokine production in the GI tract. These results indicate that the balance of IKK-beta-dependent gene expression in the intestinal epithelium is crucial in intestinal immune homeostasis by promoting mucosal immunity and limiting chronic inflammation.


Subject(s)
Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic , Homeostasis , I-kappa B Kinase/metabolism , Intestines/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cytokines/deficiency , Cytokines/immunology , Dendritic Cells/immunology , Epithelial Cells/metabolism , I-kappa B Kinase/genetics , Immunity, Mucosal/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Intestines/cytology , Intestines/parasitology , Mice , NF-kappa B/metabolism , Trichuris/immunology , Trichuris/physiology , Thymic Stromal Lymphopoietin
SELECTION OF CITATIONS
SEARCH DETAIL
...