Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Sci Signal ; 17(827): eade0580, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38470957

ABSTRACT

Intercellular communication between different cell types in solid tumors contributes to tumor growth and metastatic dissemination. The secretome of cancer-associated fibroblasts (CAFs) plays major roles in these processes. Using human mammary CAFs, we showed that CAFs with a myofibroblast phenotype released extracellular vesicles that transferred proteins to endothelial cells (ECs) that affected their interaction with immune cells. Mass spectrometry-based proteomics identified proteins transferred from CAFs to ECs, which included plasma membrane receptors. Using THY1 as an example of a transferred plasma membrane-bound protein, we showed that CAF-derived proteins increased the adhesion of a monocyte cell line to ECs. CAFs produced high amounts of matrix-bound EVs, which were the primary vehicles of protein transfer. Hence, our work paves the way for future studies that investigate how CAF-derived matrix-bound EVs influence tumor pathology by regulating the function of neighboring cancer, stromal, and immune cells.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Endothelial Cells , Neoplasms/metabolism , Cell Membrane , Cell Line , Fibroblasts/metabolism , Tumor Microenvironment , Cell Line, Tumor
3.
Nat Metab ; 5(12): 2148-2168, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38066114

ABSTRACT

Serine is a vital amino acid in tumorigenesis. While cells can perform de novo serine synthesis, most transformed cells rely on serine uptake to meet their increased biosynthetic requirements. Solute carriers (SLCs), a family of transmembrane nutrient transport proteins, are the gatekeepers of amino acid acquisition and exchange in mammalian cells and are emerging as anticancer therapeutic targets; however, the SLCs that mediate serine transport in cancer cells remain unknown. Here we perform an arrayed RNAi screen of SLC-encoding genes while monitoring amino acid consumption and cell proliferation in colorectal cancer cells using metabolomics and high-throughput imaging. We identify SLC6A14 and SLC25A15 as major cytoplasmic and mitochondrial serine transporters, respectively. We also observe that SLC12A4 facilitates serine uptake. Dual targeting of SLC6A14 and either SLC25A15 or SLC12A4 diminishes serine uptake and growth of colorectal cancer cells in vitro and in vivo, particularly in cells with compromised de novo serine biosynthesis. Our results provide insight into the mechanisms that contribute to serine uptake and intracellular handling.


Subject(s)
Colorectal Neoplasms , Membrane Transport Proteins , Animals , Membrane Transport Proteins/metabolism , Biological Transport , Amino Acids/metabolism , Serine/metabolism , Colorectal Neoplasms/genetics , Mammals/metabolism
4.
Nat Metab ; 5(8): 1303-1318, 2023 08.
Article in English | MEDLINE | ID: mdl-37580540

ABSTRACT

The genomic landscape of colorectal cancer (CRC) is shaped by inactivating mutations in tumour suppressors such as APC, and oncogenic mutations such as mutant KRAS. Here we used genetically engineered mouse models, and multimodal mass spectrometry-based metabolomics to study the impact of common genetic drivers of CRC on the metabolic landscape of the intestine. We show that untargeted metabolic profiling can be applied to stratify intestinal tissues according to underlying genetic alterations, and use mass spectrometry imaging to identify tumour, stromal and normal adjacent tissues. By identifying ions that drive variation between normal and transformed tissues, we found dysregulation of the methionine cycle to be a hallmark of APC-deficient CRC. Loss of Apc in the mouse intestine was found to be sufficient to drive expression of one of its enzymes, adenosylhomocysteinase (AHCY), which was also found to be transcriptionally upregulated in human CRC. Targeting of AHCY function impaired growth of APC-deficient organoids in vitro, and prevented the characteristic hyperproliferative/crypt progenitor phenotype driven by acute deletion of Apc in vivo, even in the context of mutant Kras. Finally, pharmacological inhibition of AHCY reduced intestinal tumour burden in ApcMin/+ mice indicating its potential as a metabolic drug target in CRC.


Subject(s)
Colorectal Neoplasms , Animals , Humans , Mice , Adenosylhomocysteinase/genetics , Adenosylhomocysteinase/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Metabolomics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
5.
Br J Cancer ; 127(10): 1773-1786, 2022 11.
Article in English | MEDLINE | ID: mdl-36115879

ABSTRACT

BACKGROUND: Cellular metabolism is an integral component of cellular adaptation to stress, playing a pivotal role in the resistance of cancer cells to various treatment modalities, including radiotherapy. In response to radiotherapy, cancer cells engage antioxidant and DNA repair mechanisms which mitigate and remove DNA damage, facilitating cancer cell survival. Given the reliance of these resistance mechanisms on amino acid metabolism, we hypothesised that controlling the exogenous availability of the non-essential amino acids serine and glycine would radiosensitise cancer cells. METHODS: We exposed colorectal, breast and pancreatic cancer cell lines/organoids to radiation in vitro and in vivo in the presence and absence of exogenous serine and glycine. We performed phenotypic assays for DNA damage, cell cycle, ROS levels and cell death, combined with a high-resolution untargeted LCMS metabolomics and RNA-Seq. RESULTS: Serine and glycine restriction sensitised a range of cancer cell lines, patient-derived organoids and syngeneic mouse tumour models to radiotherapy. Comprehensive metabolomic and transcriptomic analysis of central carbon metabolism revealed that amino acid restriction impacted not only antioxidant response and nucleotide synthesis but had a marked inhibitory effect on the TCA cycle. CONCLUSION: Dietary restriction of serine and glycine is a viable radio-sensitisation strategy in cancer.


Subject(s)
Pancreatic Neoplasms , Serine , Mice , Animals , Serine/metabolism , Glycine/pharmacology , Antioxidants/metabolism , Amino Acids
7.
Nat Metab ; 4(6): 693-710, 2022 06.
Article in English | MEDLINE | ID: mdl-35760868

ABSTRACT

Elevated production of collagen-rich extracellular matrix is a hallmark of cancer-associated fibroblasts (CAFs) and a central driver of cancer aggressiveness. Here we find that proline, a highly abundant amino acid in collagen proteins, is newly synthesized from glutamine in CAFs to make tumour collagen in breast cancer xenografts. PYCR1 is a key enzyme for proline synthesis and highly expressed in the stroma of breast cancer patients and in CAFs. Reducing PYCR1 levels in CAFs is sufficient to reduce tumour collagen production, tumour growth and metastatic spread in vivo and cancer cell proliferation in vitro. Both collagen and glutamine-derived proline synthesis in CAFs are epigenetically upregulated by increased pyruvate dehydrogenase-derived acetyl-CoA levels. PYCR1 is a cancer cell vulnerability and potential target for therapy; therefore, our work provides evidence that targeting PYCR1 may have the additional benefit of halting the production of a pro-tumorigenic extracellular matrix. Our work unveils new roles for CAF metabolism to support pro-tumorigenic collagen production.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Pyrroline Carboxylate Reductases/metabolism , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Glutamine/metabolism , Humans , Proline , delta-1-Pyrroline-5-Carboxylate Reductase
8.
Nat Commun ; 12(1): 6572, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34772930

ABSTRACT

Damaged or superfluous cells are typically eliminated by apoptosis. Although apoptosis is a cell-autonomous process, apoptotic cells communicate with their environment in different ways. Here we describe a mechanism whereby cells under apoptotic stress can promote survival of neighbouring cells. We find that upon apoptotic stress, cells release the growth factor FGF2, leading to MEK-ERK-dependent transcriptional upregulation of pro-survival BCL-2 proteins in a non-cell autonomous manner. This transient upregulation of pro-survival BCL-2 proteins protects neighbouring cells from apoptosis. Accordingly, we find in certain cancer types a correlation between FGF-signalling, BCL-2 expression and worse prognosis. In vivo, upregulation of MCL-1 occurs in an FGF-dependent manner during skin repair, which regulates healing dynamics. Importantly, either co-treatment with FGF-receptor inhibitors or removal of apoptotic stress restores apoptotic sensitivity to cytotoxic therapy and delays wound healing. These data reveal a pathway by which cells under apoptotic stress can increase resistance to cell death in surrounding cells. Beyond mediating cytotoxic drug resistance, this process also provides a potential link between tissue damage and repair.


Subject(s)
Apoptosis/drug effects , Cell Death/drug effects , Signal Transduction/drug effects , Animals , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation/drug effects , Wound Healing
9.
Mol Cell ; 81(11): 2290-2302.e7, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33831358

ABSTRACT

Cancer cells adapt their metabolism to support elevated energetic and anabolic demands of proliferation. Folate-dependent one-carbon metabolism is a critical metabolic process underpinning cellular proliferation supplying carbons for the synthesis of nucleotides incorporated into DNA and RNA. Recent research has focused on the nutrients that supply one-carbons to the folate cycle, particularly serine. Tryptophan is a theoretical source of one-carbon units through metabolism by IDO1, an enzyme intensively investigated in the context of tumor immune evasion. Using in vitro and in vivo pancreatic cancer models, we show that IDO1 expression is highly context dependent, influenced by attachment-independent growth and the canonical activator IFNγ. In IDO1-expressing cancer cells, tryptophan is a bona fide one-carbon donor for purine nucleotide synthesis in vitro and in vivo. Furthermore, we show that cancer cells release tryptophan-derived formate, which can be used by pancreatic stellate cells to support purine nucleotide synthesis.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/metabolism , Tumor Escape/drug effects , Allografts , Animals , Antineoplastic Agents/pharmacology , Carbon/immunology , Carbon/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Formates/immunology , Formates/metabolism , Gene Expression Regulation, Neoplastic , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Oximes/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Serine/immunology , Serine/metabolism , Serine/pharmacology , Signal Transduction , Sulfonamides/pharmacology , Tryptophan/immunology , Tryptophan/metabolism , Tryptophan/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
10.
Nat Commun ; 12(1): 366, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33446657

ABSTRACT

Many tumour cells show dependence on exogenous serine and dietary serine and glycine starvation can inhibit the growth of these cancers and extend survival in mice. However, numerous mechanisms promote resistance to this therapeutic approach, including enhanced expression of the de novo serine synthesis pathway (SSP) enzymes or activation of oncogenes that drive enhanced serine synthesis. Here we show that inhibition of PHGDH, the first step in the SSP, cooperates with serine and glycine depletion to inhibit one-carbon metabolism and cancer growth. In vitro, inhibition of PHGDH combined with serine starvation leads to a defect in global protein synthesis, which blocks the activation of an ATF-4 response and more broadly impacts the protective stress response to amino acid depletion. In vivo, the combination of diet and inhibitor shows therapeutic efficacy against tumours that are resistant to diet or drug alone, with evidence of reduced one-carbon availability. However, the defect in ATF4-response seen in vitro following complete depletion of available serine is not seen in mice, where dietary serine and glycine depletion and treatment with the PHGDH inhibitor lower but do not eliminate serine. Our results indicate that inhibition of PHGDH will augment the therapeutic efficacy of a serine depleted diet.


Subject(s)
Glycine/metabolism , Neoplasms/diet therapy , Serine/biosynthesis , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Glycine/analysis , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/physiopathology , Phosphoglycerate Dehydrogenase/metabolism , Serine/analysis
11.
Nat Metab ; 2(10): 1062-1076, 2020 10.
Article in English | MEDLINE | ID: mdl-32747794

ABSTRACT

Cancer cells have high demands for non-essential amino acids (NEAAs), which are precursors for anabolic and antioxidant pathways that support cell survival and proliferation. It is well-established that cancer cells consume the NEAA cysteine, and that cysteine deprivation can induce cell death; however, the specific factors governing acute sensitivity to cysteine starvation are poorly characterized. Here, we show that that neither expression of enzymes for cysteine synthesis nor availability of the primary precursor methionine correlated with acute sensitivity to cysteine starvation. We observed a strong correlation between efflux of the methionine-derived metabolite methylthioadenosine (MTA) and sensitivity to cysteine starvation. MTA efflux results from genetic deletion of methylthioadenosine phosphorylase (MTAP), which is frequently deleted in cancers. We show that MTAP loss upregulates polyamine metabolism which, concurrently with cysteine withdrawal, promotes elevated reactive oxygen species and prevents cell survival. Our results reveal an unexplored metabolic weakness at the intersection of polyamine and cysteine metabolism.


Subject(s)
Cysteine/metabolism , Metabolic Networks and Pathways , Neoplasms/metabolism , Polyamines/metabolism , Animals , Cell Line, Tumor , Cell Survival , Cysteine/deficiency , Female , Gene Knockout Techniques , Humans , Methionine/metabolism , Mice , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism , Reactive Oxygen Species , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Nutrients ; 12(8)2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32708052

ABSTRACT

Current nutritional recommendations are focused on energy, fat, carbohydrate, protein and vitamins. Less attention has been paid to the nutritional demand of one-carbon units for nucleotide and methionine synthesis. Here, we investigated the impact of sodium formate supplementation as a nutritional intervention to increase the dietary intake of one-carbon units. A cohort of six female and six male mice received 125 mM of sodium formate in the drinking water for three months. A control group of another six female and six male mice was also followed up for the same period of time. Tail vein blood samples were collected once a month and profiled with a haematology analyser. At the end of the study, blood and tissues were collected for metabolomics analysis and immune cell profiling. Formate supplementation had no significant physiological effect on male mice, except for a small decrease in body weight. Formate supplementation had no significant effect on the immune cell counts during the intervention or at the end of the study in either gender. In female mice, however, the body weight and spleen wet weight were significantly increased by formate supplementation, while the blood plasma levels of amino acids were decreased. Formate supplementation also increased the frequency of bifidobacteria, a probiotic bacterium, in the stools of female mice. We conclude that formate supplementation induces physiological changes in a gender-specific manner.


Subject(s)
Amino Acids/blood , Body Weight/drug effects , Dietary Supplements , Formates/pharmacology , Animals , Bifidobacterium/drug effects , Bifidobacterium/metabolism , Female , Formates/blood , Gastrointestinal Microbiome , Immune System/metabolism , Male , Mice , Phylogeny , Sample Size
13.
Cancer Res ; 80(11): 2325-2339, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32156779

ABSTRACT

The recurring association of specific genetic lesions with particular types of cancer is a fascinating and largely unexplained area of cancer biology. This is particularly true of clear cell renal cell carcinoma (ccRCC) where, although key mutations such as loss of VHL is an almost ubiquitous finding, there remains a conspicuous lack of targetable genetic drivers. In this study, we have identified a previously unknown protumorigenic role for the RUNX genes in this disease setting. Analysis of patient tumor biopsies together with loss-of-function studies in preclinical models established the importance of RUNX1 and RUNX2 in ccRCC. Patients with high RUNX1 (and RUNX2) expression exhibited significantly poorer clinical survival compared with patients with low expression. This was functionally relevant, as deletion of RUNX1 in ccRCC cell lines reduced tumor cell growth and viability in vitro and in vivo. Transcriptional profiling of RUNX1-CRISPR-deleted cells revealed a gene signature dominated by extracellular matrix remodeling, notably affecting STMN3, SERPINH1, and EPHRIN signaling. Finally, RUNX1 deletion in a genetic mouse model of kidney cancer improved overall survival and reduced tumor cell proliferation. In summary, these data attest to the validity of targeting a RUNX1-transcriptional program in ccRCC. SIGNIFICANCE: These data reveal a novel unexplored oncogenic role for RUNX genes in kidney cancer and indicate that targeting the effects of RUNX transcriptional activity could be relevant for clinical intervention in ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Core Binding Factor Alpha 2 Subunit/biosynthesis , Kidney Neoplasms/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Growth Processes , Cell Line, Tumor , Cell Movement/physiology , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Female , Gene Knockout Techniques , HEK293 Cells , Heterografts , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Mice , Mice, Nude , Prognosis , Transcriptome
14.
Haematologica ; 105(2): 375-386, 2020.
Article in English | MEDLINE | ID: mdl-31097632

ABSTRACT

RAS-signaling mutations induce the myelomonocytic differentiation and proliferation of hematopoietic stem and progenitor cells. Moreover, they are important players in the development of myeloid neoplasias. RAF kinase inhibitor protein (RKIP) is a negative regulator of RAS-signaling. As RKIP loss has recently been described in RAS-mutated myelomonocytic acute myeloid leukemia, we now aimed to analyze its role in myelomonocytic differentiation and RAS-driven leukemogenesis. Therefore, we initially analyzed RKIP expression during human and murine hematopoietic differentiation and observed that it is high in hematopoietic stem and progenitor cells and lymphoid cells but decreases in cells belonging to the myeloid lineage. By employing short hairpin RNA knockdown experiments in CD34+ umbilical cord blood cells and the undifferentiated acute myeloid leukemia cell line HL-60, we show that RKIP loss is indeed functionally involved in myelomonocytic lineage commitment and drives the myelomonocytic differentiation of hematopoietic stem and progenitor cells. These results could be confirmed in vivo, where Rkip deletion induced a myelomonocytic differentiation bias in mice by amplifying the effects of granulocyte macrophage-colony-stimulating factor. We further show that RKIP is of relevance for RAS-driven myelomonocytic leukemogenesis by demonstrating that Rkip deletion aggravates the development of a myeloproliferative disease in NrasG12D -mutated mice. Mechanistically, we demonstrate that RKIP loss increases the activity of the RAS-MAPK/ERK signaling module. Finally, we prove the clinical relevance of these findings by showing that RKIP loss is a frequent event in chronic myelomonocytic leukemia, and that it co-occurs with RAS-signaling mutations. Taken together, these data establish RKIP as novel player in RAS-driven myeloid leukemogenesis.


Subject(s)
Leukemia, Myeloid, Acute , Phosphatidylethanolamine Binding Protein , Animals , Cell Differentiation , Leukemia, Myeloid, Acute/genetics , Mice , Monocytes/metabolism , Phosphatidylethanolamine Binding Protein/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Signal Transduction
15.
Int J Mol Sci ; 20(14)2019 Jul 12.
Article in English | MEDLINE | ID: mdl-31336867

ABSTRACT

BACKGROUND: Cutaneous squamous cell carcinoma (cSCC) incidence continues to rise with increasing morbidity and mortality, with limited treatment options for advanced disease. Future improvements in targeted therapy will rely on advances in genomic/transcriptomic understanding and the use of model systems for basic research. We describe here the panel of 16 primary and metastatic cSCC cell lines developed and characterised over the past three decades in our laboratory in order to provide such a resource for future preclinical research and drug screening. METHODS: Primary keratinocytes were isolated from cSCC tumours and metastases, and cell lines were established. These were characterised using short tandem repeat (STR) profiling and genotyped by whole exome sequencing. Multiple in vitro assays were performed to document their morphology, growth characteristics, migration and invasion characteristics, and in vivo xenograft growth. RESULTS: STR profiles of the cSCC lines allow the confirmation of their unique identity. Phylogenetic trees derived from exome sequence analysis of the matched primary and metastatic lines provide insight into the genetic basis of disease progression. The results of in vivo and in vitro analyses allow researchers to select suitable cell lines for specific experimentation. CONCLUSIONS: There are few well-characterised cSCC lines available for widespread preclinical experimentation and drug screening. The described cSCC cell line panel provides a critical tool for in vitro and in vivo experimentation.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Skin Neoplasms/pathology , Animals , Biomarkers, Tumor , Biopsy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Gene Expression Profiling , Humans , Immunohistochemistry , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Mutation , Neoplasm Metastasis , Neoplasm Staging , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Xenograft Model Antitumor Assays
16.
Nat Commun ; 9(1): 1368, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29636461

ABSTRACT

Formate overflow coupled to mitochondrial oxidative metabolism\ has been observed in cancer cell lines, but whether that takes place in the tumor microenvironment is not known. Here we report the observation of serine catabolism to formate in normal murine tissues, with a relative rate correlating with serine levels and the tissue oxidative state. Yet, serine catabolism to formate is increased in the transformed tissue of in vivo models of intestinal adenomas and mammary carcinomas. The increased serine catabolism to formate is associated with increased serum formate levels. Finally, we show that inhibition of formate production by genetic interference reduces cancer cell invasion and this phenotype can be rescued by exogenous formate. We conclude that increased formate overflow is a hallmark of oxidative cancers and that high formate levels promote invasion via a yet unknown mechanism.


Subject(s)
Adenoma/metabolism , Formates/metabolism , Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Serine/metabolism , Adenoma/genetics , Adenoma/pathology , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Female , Formates/pharmacology , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Intestinal Mucosa/metabolism , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestines/pathology , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Animal/virology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/pathogenicity , Methotrexate/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Tumor Microenvironment/drug effects
17.
Cell Death Dis ; 8(12): 3206, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29238045

ABSTRACT

Oncolytic adenoviral mutants infect human malignant cells and replicate selectively within them. This induces direct cytotoxicity that can also trigger profound innate and adaptive immune responses. However, the mechanism by which adenoviruses produce cell death remains uncertain. We previously suggested that type 5 adenoviruses, including the E1A CR2 deletion mutant dl922-947, might induce a novel form of programmed death resembling necroptosis. Here we have investigated the roles of core necrosis proteins RIPK1, RIPK3 and MLKL in the cytotoxicity of dl922-947 and other adenovirus serotypes. By electron microscopy, we show that dl922-947 induces similar necrotic morphology as TSZ treatment (TNF-α, Smac mimetic, zVAD.fmk). However, dl922-947-mediated death is independent of TNF-α signalling, does not require RIPK1 and does not rely upon the presence of MLKL. However, inhibition of caspases, specifically caspase-8, induces necroptosis that is RIPK3 dependent and significantly enhances dl922-947 cytotoxicity. Moreover, using CRISPR/Cas9 gene editing, we demonstrate that the increase in cytotoxicity seen upon caspase inhibition is also MLKL dependent. Even in the absence of caspase inhibition, RIPK3 expression promotes dl922-947 and wild-type adenovirus type 5 efficacy both in vitro and in vivo. Together, these results suggest that adenovirus induces a form of programmed necrosis that differs from classical TSZ necroptosis.


Subject(s)
Adenoviruses, Human/genetics , DNA, Viral/genetics , Necrosis/genetics , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Adenoviruses, Human/metabolism , Adenoviruses, Human/pathogenicity , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Cell Line, Tumor , DNA, Viral/metabolism , Female , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Mice , Mice, Nude , Necrosis/etiology , Necrosis/metabolism , Necrosis/pathology , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Sequence Deletion , Signal Transduction , Thiazoles/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays
19.
EMBO J ; 36(16): 2373-2389, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28694244

ABSTRACT

Tumor progression alters the composition and physical properties of the extracellular matrix. Particularly, increased matrix stiffness has profound effects on tumor growth and metastasis. While endothelial cells are key players in cancer progression, the influence of tumor stiffness on the endothelium and the impact on metastasis is unknown. Through quantitative mass spectrometry, we find that the matricellular protein CCN1/CYR61 is highly regulated by stiffness in endothelial cells. We show that stiffness-induced CCN1 activates ß-catenin nuclear translocation and signaling and that this contributes to upregulate N-cadherin levels on the surface of the endothelium, in vitro This facilitates N-cadherin-dependent cancer cell-endothelium interaction. Using intravital imaging, we show that knockout of Ccn1 in endothelial cells inhibits melanoma cancer cell binding to the blood vessels, a critical step in cancer cell transit through the vasculature to metastasize. Targeting stiffness-induced changes in the vasculature, such as CCN1, is therefore a potential yet unappreciated mechanism to impair metastasis.


Subject(s)
Cell Communication , Endothelial Cells/physiology , Melanocytes/physiology , Cadherins/analysis , Cell Line , Cysteine-Rich Protein 61/analysis , Gene Expression Regulation , Humans , Mass Spectrometry , beta Catenin/analysis
20.
Nature ; 544(7650): 372-376, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28425994

ABSTRACT

The non-essential amino acids serine and glycine are used in multiple anabolic processes that support cancer cell growth and proliferation (reviewed in ref. 1). While some cancer cells upregulate de novo serine synthesis, many others rely on exogenous serine for optimal growth. Restriction of dietary serine and glycine can reduce tumour growth in xenograft and allograft models. Here we show that this observation translates into more clinically relevant autochthonous tumours in genetically engineered mouse models of intestinal cancer (driven by Apc inactivation) or lymphoma (driven by Myc activation). The increased survival following dietary restriction of serine and glycine in these models was further improved by antagonizing the anti-oxidant response. Disruption of mitochondrial oxidative phosphorylation (using biguanides) led to a complex response that could improve or impede the anti-tumour effect of serine and glycine starvation. Notably, Kras-driven mouse models of pancreatic and intestinal cancers were less responsive to depletion of serine and glycine, reflecting an ability of activated Kras to increase the expression of enzymes that are part of the serine synthesis pathway and thus promote de novo serine synthesis.


Subject(s)
Glycine/deficiency , Intestinal Neoplasms/diet therapy , Intestinal Neoplasms/metabolism , Lymphoma/diet therapy , Lymphoma/metabolism , Serine/deficiency , Animals , Antioxidants/metabolism , Biguanides/pharmacology , Cell Line, Tumor , Diet , Disease Models, Animal , Female , Food Deprivation , Glycine/metabolism , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Lymphoma/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Nutritional Status , Oxidative Phosphorylation/drug effects , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Serine/biosynthesis , Serine/metabolism , Serine/pharmacology , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...