Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 39(1): 64-73, 2021 01.
Article in English | MEDLINE | ID: mdl-32719479

ABSTRACT

Inducible expression of neoantigens in mice would enable the study of endogenous antigen-specific naïve T cell responses in disease and infection, but has been difficult to generate because leaky antigen expression in the thymus results in central T cell tolerance. Here we develop inversion-induced joined neoantigen (NINJA), using RNA splicing, DNA recombination and three levels of regulation to prevent leakiness and allow tight control over neoantigen expression. We apply NINJA to create tumor cell lines with inducible neoantigen expression, which could be used to study antitumor immunity. We also show that the genetic regulation in NINJA mice bypasses central and peripheral tolerance mechanisms and allows for robust endogenous CD8 and CD4 T cell responses on neoantigen induction in peripheral tissues. NINJA will enable studies of how T cells respond to defined neoantigens in the context of peripheral tolerance, transplantation, autoimmune diseases and cancer.


Subject(s)
Antigens, Neoplasm , Cell Engineering/methods , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/metabolism , Female , Humans , Mice , Organ Specificity/genetics , RNA Splicing/genetics , Tumor Cells, Cultured
2.
Elife ; 92020 07 10.
Article in English | MEDLINE | ID: mdl-32648540

ABSTRACT

Tumors are composed of many different cell types including cancer cells, fibroblasts, and immune cells. Dissecting functional metabolic differences between cell types within a mixed population can be challenging due to the rapid turnover of metabolites relative to the time needed to isolate cells. To overcome this challenge, we traced isotope-labeled nutrients into macromolecules that turn over more slowly than metabolites. This approach was used to assess differences between cancer cell and fibroblast metabolism in murine pancreatic cancer organoid-fibroblast co-cultures and tumors. Pancreatic cancer cells exhibited increased pyruvate carboxylation relative to fibroblasts, and this flux depended on both pyruvate carboxylase and malic enzyme 1 activity. Consequently, expression of both enzymes in cancer cells was necessary for organoid and tumor growth, demonstrating that dissecting the metabolism of specific cell populations within heterogeneous systems can identify dependencies that may not be evident from studying isolated cells in culture or bulk tissue.


Tumors contain a mixture of many different types of cells, including cancer cells and non-cancer cells. The interactions between these two groups of cells affect how the cancer cells use nutrients, which, in turn, affects how fast these cells grow and divide. Furthermore, different cell types may use nutrients in diverse ways to make other molecules ­ known as metabolites ­ that the cell needs to survive. Fibroblasts are a subset of non-cancer cells that are typically found in tumors and can help them form. Separating fibroblasts from cancer cells in a tumor takes a lot longer than the chemical reactions in each cell of the tumor that produce and use up nutrients, also known as the cell's metabolism. Therefore, measuring the levels of glucose (the sugar that is the main energy source for cells) and other metabolites in each tumor cell after separating them does not necessarily provide accurate information about the tumor cell's metabolism. This makes it difficult to study how cancer cells and fibroblasts use nutrients differently. Lau et al. have developed a strategy to study the metabolism of cancer cells and fibroblasts in tumors. Mice with tumors in their pancreas were provided glucose that had been labelled using biochemical techniques. As expected, when the cell processed the glucose, the label was transferred into metabolites that got used up very quickly. But the label also became incorporated into larger, more stable molecules, such as proteins. Unlike the small metabolites, these larger molecules do not change in the time it takes to separate the cancer cells from the fibroblasts. Lau et al. sorted cells from whole pancreatic tumors and analyzed large, stable molecules that can incorporate the label from glucose in cancer cells and fibroblasts. The experiments showed that, in cancer cells, these molecules were more likely to have labeling patterns that are characteristic of two specific enzymes called pyruvate carboxylase and malic enzyme 1. This suggests that these enzymes are more active in cancer cells. Lau et al. also found that pancreatic cancer cells needed these two enzymes to metabolize glucose and to grow into large tumors. Pancreatic cancer is one of the most lethal cancers and current therapies offer limited benefit to many patients. Therefore, it is important to develop new drugs to treat this disease. Understanding how cancer cells and non-cancer cells in pancreatic tumors use nutrients differently is important for developing drugs that only target cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Tumor Microenvironment/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL
3.
Cancer Metab ; 6: 6, 2018.
Article in English | MEDLINE | ID: mdl-29854399

ABSTRACT

BACKGROUND: Alternative splicing of the Pkm gene product generates the PKM1 and PKM2 isoforms of the glycolytic enzyme pyruvate kinase. PKM2 expression is associated with embryogenesis, tissue regeneration, and cancer. PKM2 is also the pyruvate kinase isoform expressed in most wild-type adult tissues, with PKM1 restricted primarily to skeletal muscle, heart, and brain. To interrogate the functional requirement for PKM2 during tumor initiation in an autochthonous mouse model for soft tissue sarcoma (STS), we used a conditional Pkm2 allele (Pkm2fl ) to abolish PKM2 expression. RESULTS: PKM2 deletion slowed tumor onset but did not abrogate eventual tumor outgrowth. PKM2-null sarcoma cells expressed PKM1 with tumors containing a high number of infiltrating PKM2 expressing stromal cells. End-stage PKM2-null tumors showed increased proliferation compared to tumors with a wild-type Pkm2 allele, and tumor metabolite analysis revealed metabolic changes associated with PKM2 loss. CONCLUSIONS: While PKM2 is not required for soft tissue sarcoma growth, PKM2 expression may facilitate initiation of this tumor type. Because these data differ from what has been observed in other cancer models where PKM2 has been deleted, they argue that the consequences of PKM2 loss during tumor initiation are dependent on the tumor type.

4.
Proc Natl Acad Sci U S A ; 114(28): E5625-E5634, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28652369

ABSTRACT

The extracellular microenvironment is an integral component of normal and diseased tissues that is poorly understood owing to its complexity. To investigate the contribution of the microenvironment to lung fibrosis and adenocarcinoma progression, two pathologies characterized by excessive stromal expansion, we used mouse models to characterize the extracellular matrix (ECM) composition of normal lung, fibrotic lung, lung tumors, and metastases. Using quantitative proteomics, we identified and assayed the abundance of 113 ECM proteins, which revealed robust ECM protein signatures unique to fibrosis, primary tumors, or metastases. These analyses indicated significantly increased abundance of several S100 proteins, including Fibronectin and Tenascin-C (Tnc), in primary lung tumors and associated lymph node metastases compared with normal tissue. We further showed that Tnc expression is repressed by the transcription factor Nkx2-1, a well-established suppressor of metastatic progression. We found that increasing the levels of Tnc, via CRISPR-mediated transcriptional activation of the endogenous gene, enhanced the metastatic dissemination of lung adenocarcinoma cells. Interrogation of human cancer gene expression data revealed that high TNC expression correlates with worse prognosis for lung adenocarcinoma, and that a three-gene expression signature comprising TNC, S100A10, and S100A11 is a robust predictor of patient survival independent of age, sex, smoking history, and mutational load. Our findings suggest that the poorly understood ECM composition of the fibrotic and tumor microenvironment is an underexplored source of diagnostic markers and potential therapeutic targets for cancer patients.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Proteomics/methods , Tenascin/physiology , Adenocarcinoma/metabolism , Animals , Annexin A2/metabolism , CRISPR-Cas Systems , Disease Progression , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Multivariate Analysis , Neoplasm Metastasis , Prognosis , S100 Proteins/metabolism , Thyroid Nuclear Factor 1/metabolism , Treatment Outcome , Tumor Microenvironment
5.
Nat Commun ; 7: 12685, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27585860

ABSTRACT

Although it has become increasingly clear that cancers display extensive cellular heterogeneity, the spatial growth dynamics of genetically distinct clones within developing solid tumours remain poorly understood. Here we leverage mosaic analysis with double markers (MADM) to trace subclonal populations retaining or lacking p53 within oncogenic Kras-initiated lung and pancreatic tumours. In both models, p53 constrains progression to advanced adenocarcinomas. Comparison of lineage-related p53 knockout and wild-type clones reveals a minor role of p53 in suppressing cell expansion in lung adenomas. In contrast, p53 loss promotes both the initiation and expansion of low-grade pancreatic intraepithelial neoplasia (PanINs), likely through differential expression of the p53 regulator p19ARF. Strikingly, lineage-related cells are often dispersed in lung adenomas and PanINs, contrasting with more contiguous growth of advanced subclones. Together, these results support cancer type-specific suppressive roles of p53 in early tumour progression and offer insights into clonal growth patterns during tumour development.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Lung Neoplasms/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Mice , Mice, Transgenic , Tumor Cells, Cultured
6.
Cell Rep ; 16(6): 1762-1773, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27477282

ABSTRACT

Deregulated cathepsin proteolysis occurs across numerous cancers, but in vivo substrates mediating tumorigenesis remain ill-defined. Applying 8-plex iTRAQ terminal amine isotopic labeling of substrates (TAILS), a systems-level N-terminome degradomics approach, we identified cathepsin B, H, L, S, and Z in vivo substrates and cleavage sites with the use of six different cathepsin knockout genotypes in the Rip1-Tag2 mouse model of pancreatic neuroendocrine tumorigenesis. Among 1,935 proteins and 1,114 N termini identified by TAILS, stable proteolytic products were identified in wild-type tumors compared with one or more different cathepsin knockouts (17%-44% of 139 cleavages). This suggests a lack of compensation at the substrate level by other cathepsins. The majority of neo-N termini (56%-83%) for all cathepsins was consistent with protein degradation. We validated substrates, including the glycolytic enzyme pyruvate kinase M2 associated with the Warburg effect, the ER chaperone GRP78, and the oncoprotein prothymosin-alpha. Thus, the identification of cathepsin substrates in tumorigenesis improves the understanding of cathepsin functions in normal physiology and cancer.


Subject(s)
Cathepsins/metabolism , Pancreatic Neoplasms/metabolism , Proteome/metabolism , Animals , Carcinogenesis/metabolism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Mice, Transgenic , Oncogene Proteins/metabolism , Protein Processing, Post-Translational , Proteomics/methods , Substrate Specificity/physiology
7.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G548-60, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27514475

ABSTRACT

Pancreatitis is an inflammatory disease of the pancreas characterized by dysregulated activity of digestive enzymes, necrosis, immune infiltration, and pain. Repeated incidence of pancreatitis is an important risk factor for pancreatic cancer. Legumain, a lysosomal cysteine protease, has been linked to inflammatory diseases such as atherosclerosis, stroke, and cancer. Until now, legumain activation has not been studied during pancreatitis. We used a fluorescently quenched activity-based probe to assess legumain activation during caerulein-induced pancreatitis in mice. We detected activated legumain by ex vivo imaging, confocal microscopy, and gel electrophoresis. Compared with healthy controls, legumain activity in the pancreas of caerulein-treated mice was increased in a time-dependent manner. Legumain was localized to CD68(+) macrophages and was not active in pancreatic acinar cells. Using a small-molecule inhibitor of legumain, we found that this protease is not essential for the initiation of pancreatitis. However, it may serve as a biomarker of disease, since patients with chronic pancreatitis show strongly increased legumain expression in macrophages. Moreover, the occurrence of legumain-expressing macrophages in regions of acinar-to-ductal metaplasia suggests that this protease may influence reprogramming events that lead to inflammation-induced pancreatic cancer.


Subject(s)
Cysteine Endopeptidases/metabolism , Macrophages/enzymology , Pancreatitis/enzymology , Animals , Ceruletide/toxicity , Cysteine Endopeptidases/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced
8.
Genes Dev ; 30(9): 1020-33, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27125672

ABSTRACT

Alternative splicing of the Pkm gene product generates the PKM1 and PKM2 isoforms of pyruvate kinase (PK), and PKM2 expression is closely linked to embryogenesis, tissue regeneration, and cancer. To interrogate the functional requirement for PKM2 during development and tissue homeostasis, we generated germline PKM2-null mice (Pkm2(-/-)). Unexpectedly, despite being the primary isoform expressed in most wild-type adult tissues, we found that Pkm2(-/-) mice are viable and fertile. Thus, PKM2 is not required for embryonic or postnatal development. Loss of PKM2 leads to compensatory expression of PKM1 in the tissues that normally express PKM2. Strikingly, PKM2 loss leads to spontaneous development of hepatocellular carcinoma (HCC) with high penetrance that is accompanied by progressive changes in systemic metabolism characterized by altered systemic glucose homeostasis, inflammation, and hepatic steatosis. Therefore, in addition to its role in cancer metabolism, PKM2 plays a role in controlling systemic metabolic homeostasis and inflammation, thereby preventing HCC by a non-cell-autonomous mechanism.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Energy Metabolism/genetics , Liver Neoplasms/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Animals , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/physiopathology , Cell Proliferation/genetics , Diet, High-Fat , Embryo, Mammalian , Embryonic Development/genetics , Female , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Growth and Development/genetics , Hepatocytes/cytology , Homeostasis/genetics , Liver Neoplasms/enzymology , Liver Neoplasms/physiopathology , Male , Mice , Protein Isoforms , Thyroid Hormone-Binding Proteins
9.
Genes Dev ; 30(2): 220-32, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26773004

ABSTRACT

Proteases are important for regulating multiple tumorigenic processes, including angiogenesis, tumor growth, and invasion. Elevated protease expression is associated with poor patient prognosis across numerous tumor types. Several multigene protease families have been implicated in cancer, including cysteine cathepsins. However, whether individual family members have unique roles or are functionally redundant remains poorly understood. Here we demonstrate stage-dependent effects of simultaneously deleting cathepsin B (CtsB) and CtsS in a murine pancreatic neuroendocrine tumor model. Early in tumorigenesis, the double knockout results in an additive reduction in angiogenic switching, whereas at late stages, several tumorigenic phenotypes are unexpectedly restored to wild-type levels. We identified CtsZ, which is predominantly supplied by tumor-associated macrophages, as the compensatory protease that regulates the acquired tumor-promoting functions of lesions deficient in both CtsB and CtsS. Thus, deletion of multiple cathepsins can lead to stage-dependent, compensatory mechanisms in the tumor microenvironment, which has potential implications for the clinical consideration of selective versus pan-family cathepsin inhibitors in cancer.


Subject(s)
Carcinoma, Neuroendocrine/enzymology , Cathepsins/genetics , Cathepsins/metabolism , Gene Deletion , Pancreatic Neoplasms/enzymology , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/physiopathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology
10.
Cancer Discov ; 6(2): 188-201, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26586766

ABSTRACT

UNLABELLED: The two unrelated miRNAs miR-143 and miR-145, coexpressed from the miR-143/145 cluster, have been proposed to act as tumor suppressors in human cancer, and therapeutic benefits of delivering miR-143 and miR-145 to tumors have been reported. In contrast, we found that tumor-specific deletion of miR-143/145 in an autochthonous mouse model of lung adenocarcinoma did not affect tumor development. This was consistent with the lack of endogenous miR-143/145 expression in normal and transformed lung epithelium. Surprisingly, miR-143/145 in the tumor microenvironment dramatically promoted tumor growth by stimulating the proliferation of endothelial cells. Loss of miR-143/145 in vivo led to derepression of the miR-145 target CAMK1D, an inhibitory kinase, which when overexpressed prevents mitotic entry of endothelial cells. As a consequence, tumors in miR-143/145-deficient animals exhibited diminished neoangiogenesis, increased apoptosis, and their expansion was limited by the tumor's ability to co-opt the alveolar vasculature. These findings demonstrate that stromal miR-143/145 promotes tumorigenesis and caution against the use of these miRNAs as agents in cancer therapeutics. SIGNIFICANCE: This study shows that miR-143/145 expressed from the tumor microenvironment stimulates neoangiogenesis and supports tumor expansion in the lung, demonstrating a surprising role for the putative tumor suppressor miRNA cluster in promoting tumorigenesis. We propose inhibition of miR-143/145 as a therapeutic avenue to modulate tumor neoangiogenesis.


Subject(s)
Lung Neoplasms/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Differentiation , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , MicroRNAs/metabolism , Neoplasms, Experimental , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Stromal Cells/cytology , Stromal Cells/metabolism , Tumor Microenvironment
11.
Genes Dev ; 29(17): 1850-62, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26341558

ABSTRACT

Despite the fact that the majority of lung cancer deaths are due to metastasis, the molecular mechanisms driving metastatic progression are poorly understood. Here, we present evidence that loss of Foxa2 and Cdx2 synergizes with loss of Nkx2-1 to fully activate the metastatic program. These three lineage-specific transcription factors are consistently down-regulated in metastatic cells compared with nonmetastatic cells. Knockdown of these three factors acts synergistically and is sufficient to promote the metastatic potential of nonmetastatic cells to that of naturally arising metastatic cells in vivo. Furthermore, silencing of these three transcription factors is sufficient to account for a significant fraction of the gene expression differences between the nonmetastatic and metastatic states in lung adenocarcinoma, including up-regulated expression of the invadopodia component Tks5long, the embryonal proto-oncogene Hmga2, and the epithelial-to-mesenchymal mediator Snail. Finally, analyses of tumors from a genetically engineered mouse model and patients show that low expression of Nkx2-1, Foxa2, and Cdx2 strongly correlates with more advanced tumors and worse survival. Our findings reveal that a large part of the complex transcriptional network in metastasis can be controlled by a small number of regulatory nodes that function redundantly, and loss of multiple nodes is required to fully activate the metastatic program.


Subject(s)
Adenocarcinoma/physiopathology , Hepatocyte Nuclear Factor 3-beta/metabolism , Homeodomain Proteins/metabolism , Lung Neoplasms/physiopathology , Neoplasm Metastasis/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma of Lung , Animals , Animals, Genetically Modified , CDX2 Transcription Factor , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Gene Silencing , Hepatocyte Nuclear Factor 3-beta/genetics , Homeodomain Proteins/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Mice , Mice, Nude , Nuclear Proteins/genetics , Proto-Oncogene Mas , Thyroid Nuclear Factor 1 , Transcription Factors/genetics
12.
Mol Cell Proteomics ; 14(8): 2213-28, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26081835

ABSTRACT

Extracellular cysteine cathepsins are known to drive cancer progression, but besides degradation of extracellular matrix proteins little is known about their physiological substrates and thus the molecular mechanisms they deploy. One of the major mechanisms used by other extracellular proteases to facilitate cancer progression is proteolytic release of the extracellular domains of transmembrane proteins or ectodomain shedding. Here we show using a mass spectrometry-based approach that cathepsins L and S act as sheddases and cleave extracellular domains of CAM adhesion proteins and transmembrane receptors from the surface of cancer cells. In cathepsin S-deficient mouse pancreatic cancers, processing of these cathepsin substrates is highly reduced, pointing to an essential role of cathepsins in extracellular shedding. In addition to influencing cell migration and invasion, shedding of surface proteins by extracellular cathepsins impacts intracellular signaling as demonstrated for regulation of Ras GTPase activity, thereby providing a putative mechanistic link between extracellular cathepsin activity and cancer progression. The MS data is available via ProteomeXchange with identifier PXD002192.


Subject(s)
Cathepsin B/metabolism , Cathepsin L/metabolism , Cathepsins/metabolism , Cell Membrane/metabolism , Neoplasms/metabolism , Proteomics/methods , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement , Humans , Macrophages/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Neoplasm Invasiveness , Protein Processing, Post-Translational , Substrate Specificity , ras Proteins/metabolism
13.
Genes Dev ; 28(19): 2134-50, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25274726

ABSTRACT

During the process of tumor progression, cancer cells can produce the requisite growth- and invasion-promoting factors and can also rely on noncancerous cells in the tumor microenvironment as an alternative, cell-extrinsic source. However, whether the cellular source influences the function of such tumor-promoting factors remains an open question. Here, we examined the roles of the cathepsin Z (CtsZ) protease, which is provided by both cancer cells and macrophages in pancreatic neuroendocrine tumors in humans and mice. We found that tumor proliferation was exclusively regulated by cancer cell-intrinsic functions of CtsZ, whereas tumor invasion required contributions from both macrophages and cancer cells. Interestingly, several of the tumor-promoting functions of CtsZ were not dependent on its described catalytic activity but instead were mediated via the Arg-Gly-Asp (RGD) motif in the enzyme prodomain, which regulated interactions with integrins and the extracellular matrix. Together, these results underscore the complexity of interactions within the tumor microenvironment and indicate that cellular source can indeed impact molecular function.


Subject(s)
Cathepsin Z/metabolism , Extracellular Matrix/metabolism , Macrophages/enzymology , Neoplasms/enzymology , Neoplasms/physiopathology , Animals , Cell Line, Tumor , Integrins/metabolism , Mice, Inbred C57BL , Neoplasm Invasiveness/physiopathology
14.
J Biol Chem ; 288(11): 7481-7491, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23297415

ABSTRACT

Immunologic adjuvants are critical components of vaccines, but it remains unclear how prototypical adjuvants enhance the adaptive immune response. Recent studies have shown that necrotic cells could trigger an immune response. Although most adjuvants have been shown to be cytotoxic, this activity has traditionally been considered a side effect. We set out to test the role of adjuvant-mediated cell death in immunity and found that alum, the most commonly used adjuvant worldwide, triggers a novel form of cell death in myeloid leukocytes characterized by cathepsin-dependent lysosome-disruption. We demonstrated that direct lysosome-permeabilization with a soluble peptide, Leu-Leu-OMe, mimics the alum-like form of necrotic cell death in terms of cathepsin dependence and cell-type specificity. Using a combination of a haploid genetic screen and cathepsin-deficient cells, we identified specific cathepsins that control lysosome-mediated necrosis. We identified cathepsin C as critical for Leu-Leu-OMe-induced cell death, whereas cathepsins B and S were required for alum-mediated necrosis. Consistent with a role of necrotic cell death in adjuvant effects, Leu-Leu-OMe replicated an alum-like immune response in vivo, characterized by dendritic cell activation, granulocyte recruitment, and production of Th2-associated antibodies. Strikingly, cathepsin C deficiency not only blocked Leu-Leu-OMe-mediated necrosis but also impaired Leu-Leu-OMe-enhanced immunity. Together our findings suggest that necrotic cell death is a powerful mediator of a Th2-associated immune response.


Subject(s)
Adjuvants, Immunologic/metabolism , Cathepsins/metabolism , Necrosis , Th2 Cells/cytology , Animals , Caspase 1/metabolism , Cathepsin C/pharmacology , Cell Death , Cell Line , Female , Granulocytes/cytology , Immune System , Immunity, Innate , Inflammation , Lysosomes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/chemistry , Signal Transduction , Spleen/cytology , Th2 Cells/metabolism
15.
Biol Chem ; 391(8): 937-45, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20731543

ABSTRACT

Proteases can regulate many aspects of tumor development as their actions, which include degradation of the extracellular matrix, proteolytic processing of chemokines and activation of other enzymes, influence several key tumorigenic processes. Members of one protease class, the cysteine cathepsins, have received increasing recognition for their involvement in cancer development, and numerous clinical studies have reported correlations between elevated cathepsin levels and malignant progression. This is also the case for cathepsin H, a member of the cysteine cathepsin family, and its utility as a prognostic marker has been analyzed extensively. However, there is limited information available on its specific functions in tumor development and progression. To gain further insight into the role of this protease in cancer, we crossed cathepsin H-deficient mice with the RIP1-Tag2 model of pancreatic islet carcinogenesis. Deletion of cathepsin H significantly impaired angiogenic switching of the pre-malignant hyperplastic islets and resulted in a reduction in the subsequent number of tumors that formed. Moreover, the tumor burden in cathepsin H null RT2 mice was significantly reduced, in association with defects in the blood vasculature and increased apoptosis. Thus, we demonstrate here for the first time important tumor-promoting roles for cathepsin H in vivo using a mouse model of human cancer.


Subject(s)
Cathepsin H/physiology , Insulinoma/metabolism , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/metabolism , Animals , Apoptosis , Cathepsin H/genetics , Disease Progression , GTPase-Activating Proteins/genetics , Gene Targeting , Immunohistochemistry , Insulinoma/blood supply , Insulinoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Tumor Burden
16.
Biochimie ; 92(11): 1618-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20447439

ABSTRACT

Proteolytic activity is required for several key processes in cancer development and progression, including tumor growth, invasion and metastasis. Accordingly, high levels of protease expression and activity have been found to correlate with malignant progression and poor patient prognosis in a wide variety of human cancers. Members of the papain family of cysteine cathepsins are among the protease classes that have been functionally implicated in cancer. Therefore, the discovery of effective cathepsin inhibitors has considerable potential for anti-cancer therapy. In this study we describe the identification of a novel, reversible cathepsin inhibitor, VBY-825, which has high potency against cathepsins B, L, S and V. VBY-825 was tested in a pre-clinical model of pancreatic islet cancer and found to significantly decrease tumor burden and tumor number. Thus, the identification of VBY-825 as a new and effective anti-tumor drug encourages the therapeutic application of cathepsin inhibitors in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cathepsins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Hydrocarbons, Fluorinated/pharmacology , Pancreatic Neoplasms/drug therapy , Protease Inhibitors/pharmacology , Sulfones/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Biological Availability , Cell Proliferation/drug effects , Cyclopropanes , Disease Models, Animal , Drug Design , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Mice , Neoplasm Invasiveness , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacokinetics , Tumor Burden/drug effects
17.
Genes Dev ; 24(3): 241-55, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20080943

ABSTRACT

Innate immune cells can constitute a substantial proportion of the cells within the tumor microenvironment and have been associated with tumor malignancy in patients and animal models of cancer; however, the mechanisms by which they modulate cancer progression are incompletely understood. Here, we show that high levels of cathepsin protease activity are induced in the majority of macrophages in the microenvironment of pancreatic islet cancers, mammary tumors, and lung metastases during malignant progression. We further show that tumor-associated macrophage (TAM)-supplied cathepsins B and S are critical for promoting pancreatic tumor growth, angiogenesis, and invasion in vivo, and markedly enhance the invasiveness of cancer cells in culture. Finally, we demonstrate that interleukin-4 (IL-4) is responsible for inducing cathepsin activity in macrophages in vitro and in vivo. Together, these data establish IL-4 as an important regulator, and cathepsin proteases as critical mediators, of the cancer-promoting functions of TAMs.


Subject(s)
Cathepsins/metabolism , Interleukin-4/metabolism , Macrophages/enzymology , Neoplasm Invasiveness , Neoplasms/enzymology , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Female , Lung Neoplasms/pathology , Macrophages/metabolism , Mammary Neoplasms, Experimental , Mice , Mice, Transgenic , Neoplasms/pathology , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/pathology
18.
Cell Cycle ; 8(17): 2837-43, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19667758

ABSTRACT

Several studies have recently reported that the cyclin dependent kinase (cdk) 6 oncogene plays a role in differentiation of a variety of cell types. This novel function expands the previously understood function of cdk6 as a regulator of G(1) phase of the cell cycle. The proposed mechanisms of these functions both require nuclear localization. That is, cdk6 phosphorylation of the retinoblastoma protein (pRb) to regulate cell cycle, and the recently proposed transcriptional regulation to block differentiation, are both nuclear functions that predict nuclear localization of the kinase. This report provides a thorough analysis of cdk6 localization and compares the localization of a commonly used mutant cdk6 to the corrected wildtype sequence as recorded in GenBank. The widely shared mutant of cdk6 contains a tyrosine residue at amino acid 224 (instead of an aspartic acid) introducing a potential phosphorylation site to the cdk6 sequence. Results indicate a majority of cdk6 is localized to the cytoplasm with concentrations of cdk6 in the edges of the cytoplasm and in the cytoplasmic extensions of cells. The results of this study may help to better understand the emerging roles of cdk6 in cell cycle control, differentiation and cancer.


Subject(s)
Cyclin-Dependent Kinase 6/analysis , Animals , Astrocytes/cytology , Base Sequence , Cell Differentiation , Cell Line , Cyclin-Dependent Kinase 6/metabolism , G1 Phase , Mice , Mutagenesis, Site-Directed , NIH 3T3 Cells , Phosphorylation , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Transfection
19.
Cancer Res ; 69(10): 4537-44, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19435903

ABSTRACT

Tumors initiate angiogenesis primarily by secreting vascular endothelial growth factor (VEGF-A(164)). The first new vessels to form are greatly enlarged, pericyte-poor sinusoids, called mother vessels (MV), that originate from preexisting venules. We postulated that the venular enlargement necessary to form MV would require a selective degradation of their basement membranes, rigid structures that resist vascular expansion. To identify the specific proteases responsible for MV formation, we induced angiogenesis in mouse tissues with an adenoviral vector expressing VEGF-A(164) (Ad-VEGF-A(164)) or with VEGF-A-secreting TA3/St mammary tumors. We found that MV formation resulted from greatly increased activity of cathepsins (B>S>L) in venules transitioning into MV, as well as from a reciprocal decrease in the expression of several cysteine protease inhibitors (CPI), stefin A and cystatins B and C, by these same venules. Using a fluorescence probe that selectively binds cellular sites of cathepsin protease activity in vivo, we showed that increased cathepsin activity was localized exclusively to perivenular cells, not to venule endothelial cells. CPI strikingly inhibited angiogenesis in the Matrigel assay, and Ad-VEGF-A(164)-induced angiogenesis was reduced by approximately 50% in cathepsin B-null mice. Thus, VEGF-A, whether expressed by interstitial cells infected with an adenoviral vector or by tumor cells, upsets the normal cathepsin-CPI balance in nearby venules, leading to degradation of their basement membranes, an important first step in angiogenesis.


Subject(s)
Cathepsins/genetics , Cysteine Proteinase Inhibitors/pharmacology , Neoplasms/blood supply , Neovascularization, Pathologic/physiopathology , Vascular Endothelial Growth Factor A/pharmacology , Venules/physiology , Animals , Cathepsins/antagonists & inhibitors , Cystatin A/genetics , Cystatin A/pharmacology , Cystatin B/deficiency , Cystatin B/pharmacology , Mice , Mice, Knockout , Mice, Nude , Microcirculation/drug effects , Microcirculation/physiology , Polymerase Chain Reaction , Venules/drug effects
20.
Chem Biol ; 14(5): 499-511, 2007 May.
Article in English | MEDLINE | ID: mdl-17524981

ABSTRACT

The papain-family cathepsins are cysteine proteases that are emerging as promising therapeutic targets for a number of human disease conditions ranging from osteoporosis to cancer. Relatively few selective inhibitors for this family exist, and the in vivo selectivity of most existing compounds is unclear. We present here the synthesis of focused libraries of epoxysuccinyl-based inhibitors and their screening in crude tissue extracts. We identified a number of potent inhibitors that display selectivity for endogenous cathepsin targets both in vitro and in vivo. Importantly, the selectivity patterns observed in crude extracts were generally retained in vivo, as assessed by active-site labeling of tissues from treated animals. Overall, this study identifies several important compound classes and highlights the use of activity-based probes to assess pharmacodynamic properties of small-molecule inhibitors in vivo.


Subject(s)
Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacology , Epoxy Compounds/chemical synthesis , Epoxy Compounds/pharmacology , Papain/antagonists & inhibitors , Animals , Chromatography, High Pressure Liquid , Cysteine Proteinase Inhibitors/pharmacokinetics , Drug Design , Drug Evaluation, Preclinical , Epoxy Compounds/pharmacokinetics , Indicators and Reagents , Injections, Intraperitoneal , Intubation, Gastrointestinal , Male , Mass Spectrometry , Mice , Peptide Library , Proteomics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...