Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Immunol ; 209(3): 606-620, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35817516

ABSTRACT

Despite recent therapeutic progress, advanced melanoma remains lethal for many patients. The composition of the immune tumor microenvironment (TME) has decisive impacts on therapy response and disease outcome, and high-dimensional analyses of patient samples reveal the heterogeneity of the immune TME. Macrophages infiltrate TMEs and generally associate with tumor progression, but the underlying mechanisms are incompletely understood. Because experimental systems are needed to elucidate the functional properties of these cells, we developed a humanized mouse model reconstituted with human immune cells and human melanoma. We used two strains of recipient mice, supporting or not supporting the development of human myeloid cells. We found that human myeloid cells favored metastatic spread of the primary tumor, thereby recapitulating the cancer-supportive role of macrophages. We next analyzed the transcriptome of human immune cells infiltrating tumors versus other tissues. This analysis identified a cluster of myeloid cells present in the TME, but not in other tissues, which do not correspond to canonical M2 cells. The transcriptome of these cells is characterized by high expression of glycolytic enzymes and multiple chemokines and by low expression of gene sets associated with inflammation and adaptive immunity. Compared with humanized mouse results, we found transcriptionally similar myeloid cells in patient-derived samples of melanoma and other cancer types. The humanized mouse model described here thus complements patient sample analyses, enabling further elucidation of fundamental principles in melanoma biology beyond M1/M2 macrophage polarization. The model can also support the development and evaluation of candidate antitumor therapies.


Subject(s)
Macrophages , Melanoma , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Macrophage Activation , Melanoma/pathology , Mice , Tumor Microenvironment
2.
Gene Ther ; 26(1-2): 1-15, 2019 02.
Article in English | MEDLINE | ID: mdl-30323352

ABSTRACT

Intratumoral electroporation-mediated IL-12 gene therapy (IT-pIL12/EP) has been shown to be safe and effective in clinical trials, demonstrating systemic antitumor effects with local delivery of this potent cytokine. We recently optimized our IL-12 gene delivery platform to increase transgene expression and efficacy in preclinical models. Here we analyze the immunological changes induced with the new IT-pIL12/EP platform in both electroporated and distant, non-electroporated lesions. IT-pIL12/EP-treated tumors demonstrated rapid induction of IL-12-regulated pathways, as well as other cytokines and chemokines pathways, and upregulation of antigen presentation machinery. The distant tumors showed an increase in infiltrating lymphocytes and gene expression changes indicative of a de novo immune response in these untreated lesions. Flow cytometric analyses revealed a KLRG1hi CD8+ effector T-cell population uniquely present in mice treated with IT-pIL12/EP. Despite being highly activated, this population expressed diminished levels of PD-1 when re-exposed to antigen in the PD-L1-rich tumor. Other T-cell exhaustion markers appeared to be downregulated in concert, suggesting an orchestrated "armoring" of these effector T cells against T-cell checkpoints when primed in the presence of IL-12 in situ. These cells may represent an important mechanism by which local IL-12 gene therapy can induce a systemic antitumor immune response without the associated toxicity of systemic IL-12 exposure.


Subject(s)
Electroporation/methods , Genetic Therapy/methods , Interleukin-12/genetics , Neoplasms, Experimental/therapy , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Interleukin-12/metabolism , Lectins, C-Type , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
3.
Cancer Immunol Immunother ; 68(8): 1273-1286, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31243491

ABSTRACT

Therapeutic cancer vaccines have met limited clinical success. In the setting of cancer, the immune system is either tolerized and/or has a limited tumor-specific T cell repertoire. In this study, we explore whether intratumoral (IT) vaccination with an HPV vaccine can elicit quantitative and qualitative differences in immune response as compared to intramuscular (IM) vaccination to overcome immune resistance in established tumors. We report that IT administration of an HPV-16 E7 peptide vaccine formulated with polyinosinic-polycytidylic acid [poly(I:C)] generated an enhanced antitumor effect relative to IM delivery. The elicited anti-tumor effect with IT vaccination was consistent among the vaccinated groups and across various C57BL/6 substrains. IT vaccination resulted in an increased frequency of PD-1hi TILs, which represented both vaccine-targeted and non-vaccine-targeted tumor-specific CD8+ T cells. Overall, the CD8+/Treg ratio was increased within the tumor microenvironment using IT vaccination. We also assessed transcriptional changes in several immune-related genes in the tumor microenvironment of the various treated groups, and our data suggest that IT vaccination leads to upregulation of a broad complement of immunomodulatory genes, including upregulation of interferon gamma (IFNγ) and antigen presentation and processing machine (APM) components. IT vaccine delivery is superior to traditional IM vaccination routes with the potential to improve tumor immunogenicity, which has potential clinical application in the setting of accessible lesions such as head and neck squamous cell carcinomas (HNSCCs).


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation/genetics , Cancer Vaccines/immunology , Carcinoma, Squamous Cell/immunology , Cells, Cultured , Cytotoxicity, Immunologic , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/immunology , Humans , Immunity, Cellular/genetics , Injections, Intramuscular , Interferon-gamma/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Poly I-C/immunology , Programmed Cell Death 1 Receptor/metabolism , Vaccination
4.
Stem Cells ; 36(1): 103-113, 2018 01.
Article in English | MEDLINE | ID: mdl-28960647

ABSTRACT

Liver progenitor cells have the potential to repair and regenerate a diseased liver. The success of any translational efforts, however, hinges on thorough understanding of the fate of these cells after transplant, especially in terms of long-term safety and efficacy. Here, we report transplantation of a liver progenitor population isolated from human fetal livers into immune-permissive mice with follow-up up to 36 weeks after transplant. We found that human progenitor cells engraft and differentiate into functional human hepatocytes in the mouse, producing albumin, alpha-1-antitrypsin, and glycogen. They create tight junctions with mouse hepatocytes, with no evidence of cell fusion. Interestingly, they also differentiate into functional endothelial cell and bile duct cells. Transplantation of progenitor cells abrogated carbon tetrachloride-induced fibrosis in recipient mice, with downregulation of procollagen and anti-smooth muscle actin. Paradoxically, the degree of engraftment of human hepatocytes correlated negatively with the anti-fibrotic effect. Progenitor cell expansion was most prominent in cirrhotic animals, and correlated with transcript levels of pro-fibrotic genes. Animals that had resolution of fibrosis had quiescent native progenitor cells in their livers. No evidence of neoplasia was observed, even up to 9 months after transplantation. Human fetal liver progenitor cells successfully attenuate liver fibrosis in mice. They are activated in the setting of liver injury, but become quiescent when injury resolves, mimicking the behavior of de novo progenitor cells. Our data suggest that liver progenitor cells transplanted into injured livers maintain a functional role in the repair and regeneration of the liver. Stem Cells 2018;36:103-113.


Subject(s)
Liver/pathology , Stem Cell Transplantation/methods , Animals , Cell Differentiation , Disease Models, Animal , Fetal Stem Cells , Humans , Mice
5.
Am J Physiol Cell Physiol ; 310(6): C436-45, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26632601

ABSTRACT

Chronic liver injury leads to fibrosis and cirrhosis. Cirrhosis, the end stage of chronic liver disease, is a leading cause of death worldwide and increases the risk of developing hepatocellular carcinoma. Currently, there is a lack of effective antifibrotic therapies to treat fibrosis and cirrhosis. Development of antifibrotic therapies requires an in-depth understanding of the cellular and molecular mechanisms involved in inflammation and fibrosis after hepatic injury. Two growth factor signaling pathways that regulate liver fibrosis are transforming growth factor-ß (TGFß) and platelet-derived growth factor (PDGF). However, their specific contributions to fibrogenesis are not well understood. Using a genetic model of liver fibrosis, we investigated whether the canonical TGFß signaling pathway was necessary for fibrogenesis. PDGF-C transgenic (PDGF-C Tg) mice were intercrossed with mice that lack Smad3, and molecular and histological fibrosis was analyzed. PDGF-C Tg mice that also lacked Smad3 had less fibrosis and improved liver lobule architecture. Loss of Smad3 also reduced expression of collagen genes, which were induced by PDGF-C, but not the expression of genes frequently associated with hepatic stellate cell (HSC) activation. In vitro HSCs isolated from Smad3-null mice proliferated more slowly than cells from wild-type mice. Taken together, these findings indicate that PDGF-C activates TGFß/Smad3 signaling pathways to regulate HSC proliferation, collagen production and ultimately fibrosis. In summary, these results suggest that inhibition of both PDGF and TGFß signaling pathways may be required to effectively attenuate fibrogenesis in patients with chronic liver disease.


Subject(s)
Liver Cirrhosis/metabolism , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Smad3 Protein/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/physiology , Cells, Cultured , Female , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Liver/physiology , Liver Neoplasms/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Rats , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
6.
Biol Blood Marrow Transplant ; 22(7): 1173-1181, 2016 07.
Article in English | MEDLINE | ID: mdl-27060441

ABSTRACT

Iron overload is common in patients undergoing hematopoietic cell transplantation (HCT). Peritransplant events, such as total body irradiation (TBI), and the effects of donor cell infusion may contribute to iron overload, in addition to disease-associated anemia and RBC transfusions. Using murine models we show complex time- and dose-dependent interactions of TBI and transplanted donor cells with expression patterns of iron regulatory genes in the liver. Infusion of allogeneic or syngeneic donor T lymphocytes increased serum iron, transiently up-regulated interleukin-6 (IL-6) and hepcidin (Hamp), and down-regulated ferroportin1 (Fpn1). After 7 to 14 days, however, changes were significant only with allogeneic cells. TBI (200 to 400 Gy) also induced IL-6 and Hamp expression but had little effect on Fpn1. TBI combined with allogeneic donor cell infusion resulted in modest early up-regulation of IL-6, followed by a decline in IL-6 levels and Hamp as well as Fpn1, and was accompanied by increased liver iron content. Injection of Fas ligand-deficient T lymphocytes from gld mice resulted in substantially lower alterations of gene expression than infusion of wild-type T cells. The agonistic anti-Fas antibody, JO2, triggered early up-regulation of Stat3 and IL-6, followed by an increase in Hamp and decreased expression of Fpn1 by 7 to 14 days, implicating Fas as a key modulator of gene expression in HCT. Minimal histologic changes were observed in mouse liver and duodenum. These data show profound and interacting effects of TBI and cell transplantation on the expression of iron regulatory genes in murine recipients. Alterations are largely related to induction of cytokines and Fas-dependent signals.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Iron Overload/etiology , Iron/metabolism , Whole-Body Irradiation/adverse effects , Animals , Cytokines/metabolism , Gene Expression Regulation , Iron-Regulatory Proteins/genetics , Liver/metabolism , Mice , T-Lymphocytes/metabolism , Transplantation, Homologous , fas Receptor/physiology
7.
Biochim Biophys Acta ; 1842(2): 318-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24269585

ABSTRACT

Platelet-derived growth factors (PDGFs) are critical for development; their over-expression is associated with fibrogenesis. Full-length PDGF-C is secreted as an inactive dimer, requiring cleavage to allow receptor binding. Previous studies indicate that tissue-type plasminogen activator (tPA) is the specific protease that performs this cleavage; in vivo confirmation is lacking. We demonstrate that primary hepatocytes from tpa KO mice produce less cleaved active PDGF-CC than do wild type hepatocytes, suggesting that tPA is critical for in vitro activation of this growth factor. We developed mice that over-express full-length human PDGF-C in the liver; these mice develop progressive liver fibrosis. To test whether tPA is important for cleavage and activation of PDGF-C in vivo, we intercrossed PDGF-C transgenic (Tg) and tpa knock-out (KO) mice, anticipating that lack of tPA would result in decreased fibrosis due to lack of hPDGF-C cleavage. To measure levels of cleaved, dimerized PDGF-CC in sera, we developed an ELISA that specifically detects cleaved PDGF-CC. We report that the absence of tpa does not affect the phenotype of `PDGF-C Tg mice. PDGF-C Tg mice lacking tPA have high serum levels of cleaved growth factor, significant liver fibrosis, and gene expression alterations similar to those of PDGF-C Tg mice with intact tPA. Furthermore, urokinase plasminogen activator and plasminogen activator inhibitor-1 expression are increased in PDGF-C Tg; tpa KO mice. Our ELISA data suggest a difference between in vitro and in vivo activation of this growth factor, and our mouse model confirms that multiple proteases cleave and activate PDGF-C in vivo.


Subject(s)
Hepatocytes/metabolism , Liver Cirrhosis/genetics , Lymphokines/genetics , Platelet-Derived Growth Factor/genetics , Tissue Plasminogen Activator/genetics , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Hepatocytes/cytology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Lymphokines/blood , Lymphokines/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Platelet-Derived Growth Factor/metabolism , Proteolysis , Reverse Transcriptase Polymerase Chain Reaction , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
8.
Cancer Sci ; 106(9): 1143-52, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26122702

ABSTRACT

Differentially regulated microRNA (miRNA) are associated with hepatic fibrosis; however, their potential usefulness for blocking hepatic fibrosis has not been exploited fully. We examined the expression of miRNA in the liver of a transgenic mouse model in which platelet-derived growth factor C (PDGF-C) is overexpressed (Pdgf-c Tg), resulting in hepatic fibrosis and steatosis and the eventual development of hepatocellular carcinoma (HCC). Robust induction of miR-214 correlated with fibrogenesis in the liver of Pdgf-c Tg mice, atherogenic high-fat diet-induced NASH mice, and patients with chronic hepatitis B or C. Pdgf-c Tg mice were injected with locked nucleic acid (LNA)-antimiR-214 via the tail vein using Invivofectamine 2.0 and the degree of hepatic fibrosis and tumor incidence were evaluated. Pdgf-c Tg mice treated with LNA-antimiR-214 showed a marked reduction in fibrosis and tumor incidence compared with saline or LNA-miR-control-injected control mice. In vitro, LNA-antimiR-214 significantly ameliorated TGF-ß1-induced pro-fibrotic gene expression in Lx-2 cells. MiR-214 targets a negative regulator of EGFR signaling, Mig-6. Mimic-miR-214 decreased the expression of Mig-6 and increased the levels of EGF-mediated p-EGFR (Y1173 and Y845) and p-Met (Tyr1234/1235) in Huh-7 cells. Conversely, LNA-antimiR-214 repressed the expression of these genes. In conclusion, miR-214 appears to participate in the development of hepatic fibrosis by modulating the EGFR and TGF-ß signaling pathways. LNA-antimiR-214 is a potential therapy for the prevention of hepatic fibrosis.


Subject(s)
Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Lymphokines/genetics , Mice, Transgenic/genetics , MicroRNAs/genetics , Platelet-Derived Growth Factor/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Epidermal Growth Factor/genetics , ErbB Receptors/genetics , Gene Expression/genetics , HEK293 Cells , Hep G2 Cells , Humans , Incidence , Liver/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Oligonucleotides/genetics , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/genetics , Transforming Growth Factor beta1/genetics
9.
Int J Cancer ; 134(4): 778-88, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-23929039

ABSTRACT

Cirrhosis is the primary risk factor for the development of hepatocellular carcinoma (HCC), yet the mechanisms by which cirrhosis predisposes to carcinogenesis are poorly understood. Using a mouse model that recapitulates many aspects of the pathophysiology of human liver disease, we explored the mechanisms by which changes in the liver microenvironment induce dysplasia and HCC. Hepatic expression of platelet-derived growth factor C (PDGF-C) induces progressive fibrosis, chronic inflammation, neoangiogenesis and sinusoidal congestion, as well as global changes in gene expression. Using reporter mice, immunofluorescence, immunohistochemistry and liver cell isolation, we demonstrate that receptors for PDGF-CC are localized on hepatic stellate cells (HSCs), which proliferate, and transform into myofibroblast-like cells that deposit extracellular matrix and lead to production of growth factors and cytokines. We demonstrate induction of cytokine genes at 2 months, and stromal cell-derived hepatocyte growth factors that coincide with the onset of dysplasia at 4 months. Our results support a paracrine signaling model wherein hepatocyte-derived PDGF-C stimulates widespread HSC activation throughout the liver leading to chronic inflammation, liver injury and architectural changes. These complex changes to the liver microenvironment precede the development of HCC. Further, increased PDGF-CC levels were observed in livers of patients with nonalcoholic fatty steatohepatitis and correlate with the stage of disease, suggesting a role for this growth factor in chronic liver disease in humans. PDGF-C transgenic mice provide a unique model for the in vivo study of tumor-stromal interactions in the liver.


Subject(s)
Carcinoma, Hepatocellular/pathology , Fatty Liver/pathology , Hepatic Stellate Cells/pathology , Liver Neoplasms/pathology , Lymphokines/metabolism , Paracrine Communication , Platelet-Derived Growth Factor/metabolism , Stromal Cells/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cohort Studies , Cytokines/genetics , Cytokines/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Immunoenzyme Techniques , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lymphokines/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Non-alcoholic Fatty Liver Disease , Oligonucleotide Array Sequence Analysis , Platelet-Derived Growth Factor/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism
10.
Gastroenterology ; 144(5): 1055-65, 2013 May.
Article in English | MEDLINE | ID: mdl-23376645

ABSTRACT

BACKGROUND & AIMS: Phosphatidylinositide 3-kinase (PI3K) is deregulated in many human tumor types, including primary liver malignancies. The kinase v-akt murine thymoma viral oncogene homolog 1 (Akt) and mammalian target of rapamycin complex (mTORC1) are effectors of PI3K that promote cell growth and survival, but their individual roles in tumorigenesis are not well defined. METHODS: In livers of albumin (Alb)-Cre mice, we selectively deleted tuberous sclerosis (Tsc)1, a negative regulator of Ras homolog enriched in brain and mTORC1, along with Phosphatase and tensin homolog (Pten), a negative regulator of PI3K. Tumor tissues were characterized by histologic and biochemical analyses. RESULTS: The Tsc1fl/fl;AlbCre, Ptenfl/fl;AlbCre, and Tsc1fl/fl;Ptenfl/fl;AlbCre mice developed liver tumors that differed in size, number, and histologic features. Livers of Tsc1fl/fl;AlbCre mice did not develop steatosis; tumors arose later than in the other strains of mice and were predominantly hepatocellular carcinomas. Livers of the Ptenfl/fl;AlbCre mice developed steatosis and most of the tumors that formed were intrahepatic cholangiocarcinomas. Livers of Tsc1fl/fl;Ptenfl/fl;AlbCre formed large numbers of tumors, of mixed histologies, with the earliest onset of any strain, indicating that loss of Tsc1 and Pten have synergistic effects on tumorigenesis. In these mice, the combination of rapamycin and MK2206 was more effective in reducing liver cell proliferation and inducing cell death than either reagent alone. Tumor differentiation correlated with Akt and mTORC1 activities; the ratio of Akt:mTORC1 activity was high throughout the course of intrahepatic cholangiocarcinomas development and low during hepatocellular carcinoma development. Compared with surrounding nontumor liver tissue, tumors from all 3 strains had increased activities of Akt, mTORC1, and mitogen-activated protein kinase and overexpressed fibroblast growth factor receptor 1. Inhibition of fibroblast growth factor receptor 1 in Tsc1-null mice suppressed Akt and mitogen-activated protein kinase activities in tumor cells. CONCLUSIONS: Based on analyses of knockout mice, mTORC1 and Akt have different yet synergistic effects during the development of liver tumors in mice.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms, Experimental/genetics , Multiprotein Complexes/genetics , Mutation , Proto-Oncogene Proteins c-akt/genetics , RNA, Neoplasm/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
11.
PLoS Genet ; 7(6): e1002147, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21731504

ABSTRACT

We are reporting qualitative and quantitative changes of the extracellular matrix (ECM) and associated receptor proteomes, occurring during the transition from liver fibrosis and steatohepatitis to hepatocellular carcinoma (HCC). We compared two mouse models relevant to human HCC: PDGFC transgenic (Tg) and Pten null mice, models of disease progression from fibrosis and steatohepatitis to HCC. Using mass spectrometry, we identified in the liver of both models proteins for 26 collagen-encoding genes, providing the first evidence of expression at the protein level for 16 collagens. We also identified post-transcriptional protein variants for six collagens and lysine hydroxylation modifications for 14 collagens. Tumor-associated collagen proteomes were similar in both models with increased expression of collagens type IV, VI, VII, X, XIV, XV, XVI, and XVIII. Splice variants for Col4a2, Col6a2, Col6a3 were co-upregulated while only the short form of Col18a1 increased in the tumors. We also identified tumor specific increases of nidogen 1, decorin, perlecan, and of six laminin subunits. The changes in these non-collagenous ECM proteins were similar in both models with the exception of laminin ß3, detected specifically in the Pten null tumors. Pdgfa and Pdgfc mRNA expression was increased in the Pten null liver, a possible mechanism for the similarity in ECM composition observed in the tumors of both models. In contrast and besides the strong up-regulation of integrin α5 protein observed in the liver tumors of both models, the expression of the six other integrins identified was specific to each model, with integrins α2b, α3, α6, and ß1 up-regulated in Pten null tumors and integrins α8 and ß5 up-regulated in the PDGFC Tg tumors. In conclusion, HCC-associated ECM proteins and ECM-integrin networks, common or specific to HCC subtypes, were identified, providing a unique foundation to using ECM composition for HCC classification, diagnosis, prevention, or treatment.


Subject(s)
Collagen/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Proteomics , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Hydroxylation , Integrins/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver Neoplasms, Experimental/metabolism , Lymphokines/metabolism , Lysine/metabolism , Mass Spectrometry/methods , Mice , Mice, Knockout , PTEN Phosphohydrolase/metabolism , Platelet-Derived Growth Factor/metabolism , Protein Isoforms/metabolism , Up-Regulation
12.
Biol Blood Marrow Transplant ; 19(8): 1210-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23707854

ABSTRACT

Hepatic iron overload is common in patients undergoing hematopoietic cell transplantation. We showed previously in a murine model that transplantation of allogeneic T cells induced iron deposition and down-regulation of hepcidin (Hamp) in hepatocytes. We hypothesized that hepatic injury was related to disrupted iron homeostasis triggered by the interaction of Fas-ligand, expressed on activated T cells, with Fas on hepatocytes. In the current study, we determined the effects of modified expression of the Flice inhibitory protein (FLIP long [FLIPL]), which interferes with Fas signaling, on the impact of Fas-initiated signals on the expression of IL-6 and Stat3 and their downstream target, Hamp. To exclude a possible contribution by other pathways, we used agonistic anti-Fas antibodies (rather than allogeneic T cells) to trigger Fas signaling. Inhibition of FLIPL by RNA interference resulted, as expected, not only in enhanced hepatocyte apoptosis in response to Fas signals, but also in decreased levels of IL-6, Stat3, and Hamp. In contrast, overexpression of FLIPL protected hepatocytes against agonistic anti-Fas antibody-mediated apoptosis and increased the levels of IL-6 and Stat3, thereby maintaining the expression of Hamp in an NF-κB-dependent fashion. In vivo overexpression of FLIPL in the liver via hydrodynamic transfection, similarly, interfered with Fas-initiated apoptosis and prevented down-regulation of IL-6, Stat3, and Hamp. These data indicate that Fas-dependent signals alter the regulation of iron homeostasis and suggest that signals initiated by Fas may contribute to peritransplantation iron accumulation.


Subject(s)
Hepcidins/metabolism , Iron Overload/etiology , T-Lymphocytes/transplantation , fas Receptor/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/antagonists & inhibitors , CASP8 and FADD-Like Apoptosis Regulating Protein/biosynthesis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Fas Ligand Protein/metabolism , Female , Hepatocytes/immunology , Hepatocytes/metabolism , Hepcidins/biosynthesis , Hepcidins/genetics , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , Iron Overload/genetics , Iron Overload/metabolism , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transfection , Transplantation, Homologous , Treatment Outcome , fas Receptor/immunology
13.
Am J Physiol Gastrointest Liver Physiol ; 305(1): G25-34, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23639813

ABSTRACT

A disintegrin and metalloproteinase 17 (ADAM17), or tumor necrosis factor (TNF)-α-converting enzyme, is a key metalloproteinase and physiological convertase for a number of putative targets that play critical roles in cytokine and growth factor signaling. These interdependent pathways are essential components of the signaling network that links liver function with the compensatory growth that occurs during liver regeneration following 2/3 partial hepatectomy (PH) or chemically induced hepatotoxicity. Despite identification of many soluble factors needed for efficient liver regeneration, very little is known about how such ligands are regulated in the liver. To directly study the role of ADAM17 in the liver, we employed two cell-specific ADAM17 knockout (KO) mouse models. Using lipopolysaccharide (LPS) as a robust stimulus for TNF release, we found attenuated levels of circulating TNF in myeloid-specific ADAM17 KO mice (ADAM17 m-KO) and, unexpectedly, in mice with hepatocyte-specific ADAM17 deletion (ADAM17 h-KO), indicating that ADAM17 expression in both cell types plays a role in TNF shedding. After 2/3 PH, induction of TNF, TNFR1, and amphiregulin (AR) was significantly attenuated in ADAM17 h-KO mice, implicating ADAM17 as the primary sheddase for these factors in the liver. Surprisingly, the extent and timing of hepatocyte proliferation were not affected after PH or carbon tetrachloride injection in ADAM17 h-KO or ADAM17 m-KO mice. We conclude that ADAM17 regulates TNF, TNFR1, and AR in the liver, and its expression in both hepatocytes and myeloid cells is important for TNF regulation after LPS injury or 2/3 PH, but is not required for liver regeneration.


Subject(s)
ADAM Proteins/metabolism , Inflammation/metabolism , Liver Regeneration/physiology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Amphiregulin , Animals , EGF Family of Proteins , Gene Expression Regulation/physiology , Glycoproteins/genetics , Glycoproteins/metabolism , Hepatectomy , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/genetics
14.
Biochem Biophys Res Commun ; 437(1): 146-50, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23810393

ABSTRACT

Tuberin is a negative regulator of mTOR pathway. To investigate the function of tuberin during liver regeneration, we performed 70% hepatectomy on wild-type and Tsc2+/- mice. We found the tuberin phosphorylation correlated with mTOR activation during early liver regeneration in wild-type mice. However, liver regeneration in the Tsc2+/- mice was not enhanced. Instead, the Tsc2+/- livers failed to accumulate lipid bodies, and this was accompanied by increased mortality. These findings suggest that tuberin plays a critical role in liver energy balance by regulating hepatocellular lipid accumulation during early liver regeneration. These effects may influence the role of mTORC1 on cell growth and proliferation.


Subject(s)
Heterozygote , Lipid Metabolism , Liver Regeneration , Liver/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Body Weight , Enzyme Activation , Hepatectomy , Liver/pathology , Liver/surgery , Male , Mice , Mice, Inbred C57BL , Organ Size , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Time Factors , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency
15.
Nucleic Acids Res ; 39(11): e73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21459851

ABSTRACT

Herein, a detailed protocol for a random mutation capture (RMC) assay to measure nuclear point mutation frequency in mouse tissue is described. This protocol is a simplified version of the original method developed for human tissue that is easier to perform, yet retains a high sensitivity of detection. In contrast to assays relying on phenotypic selection of reporter genes in transgenic mice, the RMC assay allows direct detection of mutations in endogenous genes in any mouse strain. Measuring mutation frequency within an intron of a transcribed gene, we show this assay to be highly reproducible. We analyzed mutation frequencies from the liver tissue of animals with a mutation within the intrinsic exonuclease domains of the two major DNA polymerases, δ and ε. These mice exhibited significantly higher mutation frequencies than did wild-type animals. A comparison with a previous analysis of these genotypes in Big Blue mice revealed the RMC assay to be more sensitive than the Big Blue assay for this application. As RMC does not require analysis of a particular gene, simultaneous analysis of mutation frequency at multiple genetic loci is feasible. This assay provides a versatile alternative to transgenic mouse models for the study of mutagenesis in vivo.


Subject(s)
DNA Mutational Analysis , Point Mutation , Animals , DNA Polymerase II/genetics , DNA Polymerase III/genetics , Genome , Mice , Mice, Inbred C57BL , Mutagenesis , Polymerase Chain Reaction , Reproducibility of Results
16.
Front Immunol ; 14: 1170462, 2023.
Article in English | MEDLINE | ID: mdl-37207206

ABSTRACT

MHC class I "single-chain trimer" molecules, coupling MHC heavy chain, ß2-microglobulin, and a specific peptide into a single polypeptide chain, are widely used in research. To more fully understand caveats associated with this design that may affect its use for basic and translational studies, we evaluated a set of engineered single-chain trimers with combinations of stabilizing mutations across eight different classical and non-classical human class I alleles with 44 different peptides, including a novel human/murine chimeric design. While, overall, single-chain trimers accurately recapitulate native molecules, care was needed in selecting designs for studying peptides longer or shorter than 9-mers, as single-chain trimer design could affect peptide conformation. In the process, we observed that predictions of peptide binding were often discordant with experiment and that yields and stabilities varied widely with construct design. We also developed novel reagents to improve the crystallizability of these proteins and confirmed novel modes of peptide presentation.


Subject(s)
Histocompatibility Antigens Class I , Peptides , Humans , Mice , Animals , Histocompatibility Antigens Class I/genetics , Peptides/metabolism , Epitopes/chemistry
17.
J Cell Physiol ; 227(4): 1319-25, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21604268

ABSTRACT

Sorafenib increases survival rate of patients with advanced hepatocellular carcinoma (HCC). The mechanism underlying this effect is not completely understood. In this work we have analyzed the effects of sorafenib on autocrine proliferation and survival of different human HCC cell lines. Our results indicate that sorafenib in vitro counteracts autocrine growth of different tumor cells (Hep3B, HepG2, PLC-PRF-5, SK-Hep1). Arrest in S/G2/M cell cycle phases were observed coincident with cyclin D1 down-regulation. However, sorafenib's main anti-tumor activity seems to occur through cell death induction which correlated with caspase activation, increase in the percentage of hypodiploid cells, activation of BAX and BAK and cytochrome c release from mitochondria to cytosol. In addition, we observed a rise in mRNA and protein levels of the pro-apoptotic "BH3-domain only" PUMA and BIM, as well as decreased protein levels of the anti-apoptotic MCL1 and survivin. PUMA targeting knock-down, by using specific siRNAs, inhibited sorafenib-induced apoptotic features. Moreover, we obtained evidence suggesting that sorafenib also sensitizes HCC cells to the apoptotic activity of transforming growth factor-ß (TGF-ß) through the intrinsic pathway and to tumor necrosis factor-α (TNF) through the extrinsic pathway. Interestingly, sensitization to sorafenib-induced apoptosis is characteristic of liver tumor cells, since untransformed hepatocytes did not respond to sorafenib inducing apoptosis, either alone or in combination with TGF-ß or TNF. Indeed, sorafenib effectiveness in delaying HCC late progression might be partly related to a selectively sensitization of HCC cells to apoptosis by disrupting autocrine signals that protect them from adverse conditions and pro-apoptotic physiological cytokines.


Subject(s)
Benzenesulfonates/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Pyridines/pharmacology , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Autocrine Communication/drug effects , Autocrine Communication/physiology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/physiopathology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Knockdown Techniques , Humans , Liver Neoplasms/pathology , Liver Neoplasms/physiopathology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Sorafenib , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
18.
Hepatology ; 54(2): 597-608, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21574169

ABSTRACT

UNLABELLED: Partial hepatectomy (PH) consistently results in an early increase of circulating interleukin-6 (IL-6), which is thought to play a major role in liver regeneration. Activation of this cytokine after PH requires the adaptor protein, MyD88, but the specific MyD88-related receptors involved remain unidentified. It is also unknown whether the magnitude of IL-6 elevation determines the extent of subsequent hepatocyte proliferation. Here, we uncovered artifacts in the assessment of circulating IL-6 levels when using cardiac puncture in mice after PH. By using retro-orbital bleed sampling, we show that the circulating levels of IL-6 after PH were not directly correlated with the extent of hepatocyte DNA synthesis in individual mice. The IL-6 increase after PH was attenuated in all lipopolysaccharide-hyporesponsive mouse strains studied (e.g., C3H/HeJ, Tlr4 null, Cd14 null, Tlr2,4,9 null, and Tlr2,4-Caspase1 null) and was severely abrogated in Myd88 null mice. Despite attenuated IL-6 levels, Tlr4 null mice showed normal signaling downstream of IL-6 and normal hepatocyte proliferation. In contrast, Myd88 null mice showed severe impairments in signal transducer and activator of transcription 3 phosphorylation and Socs3 induction, but had enhanced and prolonged extracellular signal-related kinase 1 and 2 phosphorylation in the first 6 hours after PH. Unexpectedly, these changes were associated with accelerated initiation of hepatocyte proliferation, as assessed by hepatocyte bromodeoxyuridine incorporation, phospho-histone H3 immunostaining, and cyclin E and A protein expression. CONCLUSION: TLR-4 signaling contributes to IL-6 activation after PH, but the Tlr4-independent component appears sufficient for ensuring intact signaling downstream of IL-6. The lack of correlation between IL-6 levels and hepatocyte proliferation after PH, and the accelerated start of hepatocyte proliferation in Myd88 null mice despite abrogated cytokine activation, may highlight relevant antiproliferative effects of IL-6 signaling, possibly via Socs3, in the regulation of liver regeneration.


Subject(s)
Interleukin-6/physiology , Liver Regeneration/physiology , Myeloid Differentiation Factor 88/physiology , Toll-Like Receptor 4/physiology , Animals , Hepatectomy , Mice
19.
Am J Public Health ; 102(12): 2220-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23078487

ABSTRACT

A diverse panel convened in June 2011 to explore a dilemma in human research: some traits may make individuals or communities particularly vulnerable to a variety of harms in research; however, well-intended efforts to protect these vulnerable individuals and communities from harm may actually generate a series of new harms. We have presented a consensus statement forged by the panel through discussion during a 2-day meeting and the article-writing process. We have identified practical problems that sometimes arise in connection with providing additional safeguards for groups labeled as vulnerable and offered recommendations on how we might better balance concerns for protection with concerns for justice and participant autonomy.


Subject(s)
Human Experimentation/standards , Vulnerable Populations , Government Regulation , Human Experimentation/ethics , Human Experimentation/legislation & jurisprudence , Humans , Informed Consent , Risk Assessment , United States
20.
Cancer J ; 28(5): 407-415, 2022.
Article in English | MEDLINE | ID: mdl-36165730

ABSTRACT

ABSTRACT: Despite the availability of prophylactic human papillomavirus (HPV) vaccines, there is a growing incidence of HPV-associated head and neck squamous cell carcinomas (HPV-HNSCC) worldwide. The viral etiology of HPV-HNSCC provides an opportunity to develop personalized immune-based therapies, which target the unique viral- or tumor-specific proteins. Novel HPV-targeted immunotherapeutic approaches in clinical development are reviewed. Early results from these trials highlight new opportunities and potential challenges ahead. Immunotherapies for HPV-associated HNSCCs will require a tailored combinatorial approach based on preexisting mechanisms of host immune resistance. As the field continues to identify the relevant HPV types 16 and 18 immunogenic epitopes that are presented by diverse HLA class I alleles, improved HPV-targeted biologics and clinical monitoring tools can be developed and applied to a broader cancer patient population.


Subject(s)
Alphapapillomavirus , Biological Products , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Vaccines , Epitopes , Humans , Oropharyngeal Neoplasms/etiology , Oropharyngeal Neoplasms/therapy , Papillomaviridae , Papillomavirus Infections/complications , Papillomavirus Infections/epidemiology , Papillomavirus Infections/therapy , Squamous Cell Carcinoma of Head and Neck
SELECTION OF CITATIONS
SEARCH DETAIL