Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cancer Cell Int ; 23(1): 279, 2023 Nov 18.
Article in English | MEDLINE | ID: mdl-37980483

ABSTRACT

BACKGROUND: Myeloid cells play an essential role in cancer metastasis. The phenotypic diversity of these cells during cancer development has attracted great interest; however, their functional heterogeneity and plasticity have limited their role as prognostic markers and therapeutic targets. METHODS: To identify markers associated with myeloid cells in metastatic tumours, we compared transcriptomic data from immune cells sorted from metastatic and non-metastatic mammary tumours grown in BALB/cJ mice. To assess the translational relevance of our in vivo findings, we assessed human breast cancer biopsies and evaluated the association between arginase 1 protein expression in breast cancer tissues with tumour characteristics and patient outcomes. RESULTS: Among the differentially expressed genes, arginase 1 (ARG1) showed a unique expression pattern in tumour-infiltrating myeloid cells that correlated with the metastatic capacity of the tumour. Even though ARG1-positive cells were found almost exclusively inside the metastatic tumour, ARG1 protein was also present in the plasma. In human breast cancer biopsies, the presence of ARG1-positive cells was strongly correlated with high-grade proliferating tumours, poor prognosis, and low survival. CONCLUSION: Our findings highlight the potential use of ARG1-positive myeloid cells as an independent prognostic marker to evaluate the risk of metastasis in breast cancer patients.

2.
Breast Cancer Res Treat ; 201(2): 339-350, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37450087

ABSTRACT

BACKGROUND AND AIMS: Amplification of S100A8 occurs in 10-30% of all breast cancers and has been linked to poorer prognosis. Similarly, the protein S100A8 is overexpressed in a roughly comparable proportion of breast cancers and is also found in infiltrating myeloid-lineage cells, again linked to poorer prognosis. We explore the relationship between these findings. METHODS: We examined S100A8 copy number (CN) alterations using fluorescence in situ hybridization in 475 primary breast cancers and 117 corresponding lymph nodes. In addition, we studied S100A8 protein expression using immunohistochemistry in 498 primary breast cancers from the same cohort. RESULTS: We found increased S100A8 CN (≥ 4) in tumor epithelial cells in 20% of the tumors, increased S100A8 protein expression in 15%, and ≥ 10 infiltrating S100A8 + polymorphonuclear cells in 19%. Both increased S100A8 CN and protein expression in cancer cells were associated with high Ki67 status, high mitotic count and high histopathological grade. We observed no association between increased S100A8 CN and S100A8 protein expression, and only a weak association (p = 0.09) between increased CN and number of infiltrating S100A8 + immune cells. Only S100A8 protein expression in cancer cells was associated with significantly worse prognosis. CONCLUSIONS: Amplification of S100A8 does not appear to be associated with S100A8 protein expression in breast cancer. S100A8 protein expression in tumor epithelial cells identifies a subgroup of predominantly non-luminal tumors with a high mean age at diagnosis and significantly worse prognosis. Finally, S100A8 alone is not a sufficient marker to identify infiltrating immune cells linked to worse prognosis.


Subject(s)
Breast Neoplasms , Female , Humans , Breast Neoplasms/pathology , Calgranulin A/genetics , Calgranulin A/metabolism , Cell Proliferation , Gene Dosage , In Situ Hybridization, Fluorescence , Prognosis
3.
Eur J Med Chem ; 255: 115344, 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37141705

ABSTRACT

The colony-stimulating factor 1 receptor (CSF1R) plays an important role in the regulation of many inflammatory processes, and overexpression of the kinase is implicated in several disease states. Identifying selective, small-molecule inhibitors of CSF1R may be a crucial step toward treating these disorders. Through modelling, synthesis, and a systematic structure-activity relationship study, we have identified a number of potent and highly selective purine-based inhibitors of CSF1R. The optimized 6,8-disubstituted antagonist, compound 9, has enzymatic IC50 of 0.2 nM, and displays a strong affinity toward the autoinhibited form of CSF1R, contrasting that of other previously reported inhibitors. As a result of its binding mode, the inhibitor shows excellent selectivity (Selectivity score: 0.06), evidenced by profiling towards a panel of 468 kinases. In cell-based assays, this inhibitor shows dose-dependent blockade of CSF1-mediated downstream signalling in murine bone marrow-derived macrophages (IC50 = 106 nM) as well as disruption of osteoclast differentiation at nanomolar levels. In vivo experiments, however, indicate that improve metabolic stability is needed in order to further progress this compound class.


Subject(s)
Macrophages , Osteoclasts , Animals , Mice , Receptor Protein-Tyrosine Kinases , Cell Differentiation , Purines/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor
4.
J Med Chem ; 66(10): 6959-6980, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37191268

ABSTRACT

Colony-stimulating factor-1 receptor (CSF1R) is a receptor tyrosine kinase that controls the differentiation and maintenance of most tissue-resident macrophages, and the inhibition of CSF1R has been suggested as a possible therapy for a range of human disorders. Herein, we present the synthesis, development, and structure-activity relationship of a series of highly selective pyrrolo[2,3-d]pyrimidines, showing subnanomolar enzymatic inhibition of this receptor and with excellent selectivity toward other kinases in the platelet-derived growth factor receptor (PDGFR) family. The crystal structure of the protein and 23 revealed that the binding conformation of the protein is DFG-out-like. The most promising compounds in this series were profiled for cellular potency and subjected to pharmacokinetic profiling and in vivo stability, indicating that this compound class could be relevant in a potential disease setting. Additionally, these compounds inhibited primarily the autoinhibited form of the receptor, contrasting the behavior of pexidartinib, which could explain the exquisite selectivity of these structures.


Subject(s)
Pyrimidines , Receptor Protein-Tyrosine Kinases , Humans , Structure-Activity Relationship , Pyrimidines/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry
5.
Cell Commun Signal ; 21(1): 50, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36882786

ABSTRACT

BACKGROUND: To our current understanding, solid tumors depend on suppressed local immune reactions, often elicited by the interaction between tumor cells and tumor microenvironment (TME) components. Despite an improved understanding of anti-cancer immune responses in the TME, it is still unclear how immuno-suppressive TME are formed and how some cancer cells survive and metastasize. METHODS: To identify the major adaptations that cancer cells undergo during tumor development and progression, we compared the transcriptome and proteome from metastatic 66cl4 and non-metastatic 67NR cell lines in culture versus their corresponding mouse mammary primary tumors. Using confocal microscopy, RT-qPCR, flow cytometry and western blotting, we studied the signaling pathway and the mechanisms involved. In addition, we used public gene expression data from human breast cancer biopsies to evaluate the correlation between gene expression and clinical outcomes in patients. RESULTS: We found that type I interferon (IFN-I) response was a key differentially regulated pathway between metastatic and non-metastatic cell lines and tumors. The IFN-I response was active in metastatic cancer cells in culture and markedly dampened when these cells formed primary tumors. Interestingly, the opposite was observed in non-metastatic cancer cells and tumors. Consistent with an active IFN-I response in culture, the metastatic cancer cells displayed elevated levels of cytosolic DNA from both mitochondria and ruptured micronuclei with concomitant activation of cGAS-STING signaling. Interestingly, decreased IFN-I-related gene expression in breast cancer biopsies correlated with an unfavourable prognosis in patients. CONCLUSION: Our findings show that IFN-I response is dampened in the tumors with the metastatic ability and lower IFN-I expression predicts poor prognosis in triple-negative and HER2 enriched breast cancer patients. This study highlights the possibility of reactivating the IFN-I response as a potential therapeutic strategy in breast cancer. Video Abstract.


Subject(s)
Breast Neoplasms , Interferon Type I , Humans , Animals , Mice , Female , Breast , Signal Transduction , Antibodies , Tumor Microenvironment
6.
Free Radic Biol Med ; 184: 170-184, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35381325

ABSTRACT

Many breast cancer patients are diagnosed with small, well-differentiated, hormone receptor-positive tumors. Risk of relapse is not easily identified in these patients, resulting in overtreatment. To identify metastasis-related gene expression patterns, we compared the transcriptomes of the non-metastatic 67NR and metastatic 66cl4 cell lines from the murine 4T1 mammary tumor model. The transcription factor nuclear factor, erythroid 2-like 2 (NRF2, encoded by NFE2L2) was constitutively activated in the metastatic cells and tumors, and correspondingly a subset of established NRF2-regulated genes was also upregulated. Depletion of NRF2 increased basal levels of reactive oxygen species (ROS) and severely reduced ability to form primary tumors and lung metastases. Consistently, a set of NRF2-controlled genes was elevated in breast cancer biopsies. Sixteen of these were combined into a gene expression signature that significantly improves the PAM50 ROR score, and is an independent, strong predictor of prognosis, even in hormone receptor-positive tumors.


Subject(s)
Breast Neoplasms , NF-E2-Related Factor 2 , Animals , Breast Neoplasms/pathology , Female , Humans , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplasm Recurrence, Local , Oxidative Stress , Reactive Oxygen Species/metabolism
7.
ACS Nano ; 14(7): 7832-7846, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32413260

ABSTRACT

Although the first nanomedicine was clinically approved more than two decades ago, nanoparticles' (NP) in vivo behavior is complex and the immune system's role in their application remains elusive. At present, only passive-targeting nanoformulations have been clinically approved, while more complicated active-targeting strategies typically fail to advance from the early clinical phase stage. This absence of clinical translation is, among others, due to the very limited understanding for in vivo targeting mechanisms. Dynamic in vivo phenomena such as NPs' real-time targeting kinetics and phagocytes' contribution to active NP targeting remain largely unexplored. To better understand in vivo targeting, monitoring NP accumulation and distribution at complementary levels of spatial and temporal resolution is imperative. Here, we integrate in vivo positron emission tomography/computed tomography imaging with intravital microscopy and flow cytometric analyses to study αvß3-integrin-targeted cyclic arginine-glycine-aspartate decorated liposomes and oil-in-water nanoemulsions in tumor mouse models. We observed that ligand-mediated accumulation in cancerous lesions is multifaceted and identified "NP hitchhiking" with phagocytes to contribute considerably to this intricate process. We anticipate that this understanding can facilitate rational improvement of nanomedicine applications and that immune cell-NP interactions can be harnessed to develop clinically viable nanomedicine-based immunotherapies.


Subject(s)
Nanoparticles , Neoplasms , Animals , Integrin alphaV , Integrin alphaVbeta3 , Lipids , Mice , Neoplasms/drug therapy , Phagocytes
8.
J Cachexia Sarcopenia Muscle ; 11(1): 195-207, 2020 02.
Article in English | MEDLINE | ID: mdl-31436048

ABSTRACT

BACKGROUND: The majority of patients with advanced cancer develop cachexia, a weight loss syndrome that severely reduces quality of life and limits survival. Our understanding of the underlying mechanisms that cause the condition is limited, and there are currently no treatment options that can completely reverse cachexia. Several tumour-derived factors and inflammatory mediators have been suggested to contribute to weight loss in cachectic patients. However, inconsistencies between studies are recurrent. Activin A and interleukin 6 (IL-6) are among the best studied factors that seem to be important, and several studies support their individual role in cachexia development. METHODS: We investigated the interplay between activin A and IL-6 in the cachexia-inducing TOV21G cell line, both in culture and in tumours in mice. We previously found that the human TOV21G cells secrete IL-6 that induces autophagy in reporter cells and cachexia in mice. Using this established cachexia cell model, we targeted autocrine activin A by genetic, chemical, and biological approaches. The secretion of IL-6 from the cancer cells was determined in both culture and tumour-bearing mice by a species-specific ELISA. Autophagy reporter cells were used to monitor the culture medium for autophagy-inducing activities, and muscle mass changes were evaluated in tumour-bearing mice. RESULTS: We show that activin A acts in an autocrine manner to promote the synthesis and secretion of IL-6 from cancer cells. By inhibiting activin A signalling, the production of IL-6 from the cancer cells is reduced by 40-50% (up to 42% reduction on protein level, P = 0.0048, and 48% reduction on mRNA level, P = 0.0308). Significantly reduced IL-6 secretion (P < 0.05) from the cancer cells is consistently observed when using biological, chemical, and genetic approaches to interfere with the autocrine activin A loop. Inhibiting activin signalling also reduces the ability of the cancer cells to accelerate autophagy in non-cancerous cells (up to 43% reduced autophagy flux, P = 0.0006). Coherent to the in vitro data, the use of an anti-activin receptor 2 antibody in cachectic tumour-bearing mice reduces serum levels of cancer cell-derived IL-6 by 62% (from 417 to 159 pg/mL, P = 0.03), and, importantly, it reverses cachexia and counteracts loss of all measured muscle groups (P < 0.0005). CONCLUSIONS: Our data support a functional link between activin A and IL-6 signalling pathways and indicate that interference with activin A-induced IL-6 secretion from the tumour has therapeutic potential for cancer-induced cachexia.


Subject(s)
Activins/metabolism , Autocrine Communication/physiology , Autophagy/genetics , Cachexia/genetics , Interleukin-6/metabolism , Ovarian Neoplasms/genetics , Animals , Disease Models, Animal , Female , Humans , Mice , Ovarian Neoplasms/pathology , Signal Transduction
9.
Cell Commun Signal ; 17(1): 140, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31694641

ABSTRACT

BACKGROUND: In breast cancer, activation of bone morphogenetic protein (BMP) signaling and elevated levels of BMP-antagonists have been linked to tumor progression and metastasis. However, the simultaneous upregulation of BMPs and their antagonist, and the fact that both promote tumor aggressiveness seems contradictory and is not fully understood. METHODS: We analyzed the transcriptomes of the metastatic 66cl4 and the non-metastatic 67NR cell lines of the 4T1 mouse mammary tumor model to search for factors that promote metastasis. CRISPR/Cas9 gene editing was used for mechanistic studies in the same cell lines. Furthermore, we analyzed gene expression patterns in human breast cancer biopsies obtained from public datasets to evaluate co-expression and possible relations to clinical outcome. RESULTS: We found that mRNA levels of the BMP-antagonist Grem1, encoding gremlin1, and the ligand Bmp4 were both significantly upregulated in cells and primary tumors of 66cl4 compared to 67NR. Depletion of gremlin1 in 66cl4 could impair metastasis to the lungs in this model. Furthermore, we found that expression of Grem1 correlated with upregulation of several stem cell markers in 66cl4 cells compared to 67NR cells. Both in the mouse model and in patients, expression of GREM1 associated with extracellular matrix organization, and formation, biosynthesis and modification of collagen. Importantly, high expression of GREM1 predicted poor prognosis in estrogen receptor negative breast cancer patients. Analyses of large patient cohorts revealed that amplification of genes encoding BMP-antagonists and elevation of the corresponding transcripts is evident in biopsies from more than half of the patients and much more frequent for the secreted BMP-antagonists than the intracellular inhibitors of SMAD signaling. CONCLUSION: In conclusion, our results show that GREM1 is associated with metastasis and predicts poor prognosis in ER-negative breast cancer patients. Gremlin1 could represent a novel target for therapy.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , RNA, Messenger/genetics , Receptors, Estrogen/metabolism , Survival Analysis , Trastuzumab , Zebrafish
10.
Prog Cardiovasc Dis ; 62(2): 193-202, 2019.
Article in English | MEDLINE | ID: mdl-30867130

ABSTRACT

The benefits of physical activity in cardiovascular diseases have long been appreciated. However, the molecular mechanisms that trigger and sustain the cardiac benefits of exercise are poorly understood, and it is anticipated that unveiling these mechanisms will identify novel therapeutic targets. In search of these mechanisms we took advantage of unbiased RNA-sequencing (RNA-seq) technology to discover cardiac gene targets whose expression is disrupted in heart failure (HF) and rescued by exercise in a rat model. Upon exhaustive validation in a separate rat cohort (qPCR) and human datasets, we shortlisted 16 targets for a cell-based screening, aiming to evaluate whether targeted disruption of these genes with silencing RNA would affect the abundance of a CVD biomarker (BNP, B-type natriuretic peptide) in human cardiomyocytes. Overall, these experiments showed that Proline Dehydrogenase (PRODH) expression is reduced in human failing hearts, rescued by exercise in a rat model of HF, and its targeted knockdown increases BNP expression in human cardiomyocytes. On the other hand, overexpression of PRODH increases the abundance of metabolism-related gene transcripts, and PRODH appears to be crucial to sustain normal mitochondrial function and maintenance of ATP levels in human cardiomyocytes in a hypoxic environment, as well as for redox homeostasis in both normoxic and hypoxic conditions. Altogether our findings show that PRODH is a novel molecular target of exercise in failing hearts and highlight its role in cardiomyocyte physiology, thereby proposing PRODH as a potential experimental target for gene therapy in HF.


Subject(s)
Exercise/physiology , Heart Failure , Proline Oxidase/metabolism , Animals , Biomarkers/metabolism , Disease Models, Animal , Gene Expression Profiling , Heart Failure/metabolism , Heart Failure/prevention & control , Humans , Mitochondria, Heart/metabolism , Rats , Signal Transduction
11.
J Proteome Res ; 18(3): 1237-1247, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30707844

ABSTRACT

Extracellular vesicles are emerging as biomarkers in breast cancer. Our recent report suggested that an intracellular granular staining pattern of the extracellular matrix protein nephronectin (NPNT) in breast tumor sections correlated with a poor prognosis. Furthermore, the results showed that NPNT is localized in extracellular vesicles derived from mouse breast cancer cells. In this study, we performed proteomic analysis that revealed that several proteins, including tumor-promoting molecules, are differentially expressed in the cargo of small extracellular vesicles (sEVs) derived from NPNT-expressing mouse breast cancer cells. We also identified three different forms of NPNT at 80, 60, and 20 kDa. We report that the native form of NPNT at 60 kDa becomes further glycosylated and is detected as the 80 kDa NPNT, which may be processed by matrix metalloproteinases to a shorter form of around 20 kDa, which has not previously been described. Although both 80 and 20 kDa NPNT are detected in sEVs derived from breast cancer cells, the 20 kDa form of NPNT is concentrated in sEVs. In summary, we show that a novel truncated form of NPNT is found in sEVs derived from breast cancer cells.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Extracellular Matrix Proteins/genetics , Proteomics , Animals , Breast Neoplasms/pathology , Disease Models, Animal , Extracellular Vesicles/genetics , Extracellular Vesicles/pathology , Female , Gene Expression Regulation, Neoplastic/genetics , Glycosylation , Humans , Mice , Protein Isoforms/genetics
12.
Redox Biol ; 20: 1-12, 2019 01.
Article in English | MEDLINE | ID: mdl-30253279

ABSTRACT

Age-related macular degeneration (AMD) is a multi-factorial disease that is the leading cause of irreversible and severe vision loss in the developed countries. It has been suggested that the pathogenesis of dry AMD involves impaired protein degradation in retinal pigment epithelial cells (RPE). RPE cells are constantly exposed to oxidative stress that may lead to the accumulation of damaged cellular proteins, DNA and lipids and evoke tissue deterioration during the aging process. The ubiquitin-proteasome pathway and the lysosomal/autophagosomal pathway are the two major proteolytic systems in eukaryotic cells. NRF-2 (nuclear factor-erythroid 2-related factor-2) and PGC-1α (peroxisome proliferator-activated receptor gamma coactivator-1 alpha) are master transcription factors in the regulation of cellular detoxification. We investigated the role of NRF-2 and PGC-1α in the regulation of RPE cell structure and function by using global double knockout (dKO) mice. The NRF-2/PGC-1α dKO mice exhibited significant age-dependent RPE degeneration, accumulation of the oxidative stress marker, 4-HNE (4-hydroxynonenal), the endoplasmic reticulum stress markers GRP78 (glucose-regulated protein 78) and ATF4 (activating transcription factor 4), and damaged mitochondria. Moreover, levels of protein ubiquitination and autophagy markers p62/SQSTM1 (sequestosome 1), Beclin-1 and LC3B (microtubule associated protein 1 light chain 3 beta) were significantly increased together with the Iba-1 (ionized calcium binding adaptor molecule 1) mononuclear phagocyte marker and an enlargement of RPE size. These histopathological changes of RPE were accompanied by photoreceptor dysmorphology and vision loss as revealed by electroretinography. Consequently, these novel findings suggest that the NRF-2/PGC-1α dKO mouse is a valuable model for investigating the role of proteasomal and autophagy clearance in the RPE and in the development of dry AMD.


Subject(s)
Genetic Predisposition to Disease , Macular Degeneration/genetics , Macular Degeneration/pathology , NF-E2-Related Factor 2/deficiency , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/deficiency , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Animals , Autophagy/genetics , Biomarkers , Disease Models, Animal , Electroretinography , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Genetic Association Studies , Immunohistochemistry , Lysosomes/metabolism , Lysosomes/ultrastructure , Macular Degeneration/diagnosis , Macular Degeneration/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/ultrastructure , Molecular Imaging , Mutation , Oxidative Stress/genetics , Phenotype , Photoreceptor Cells/metabolism , Protein Aggregation, Pathological , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/ultrastructure
13.
Autophagy ; 13(10): 1664-1678, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28820283

ABSTRACT

Inflammation is crucial in the defense against infections but must be tightly controlled to limit detrimental hyperactivation. Our diet influences inflammatory processes and omega-3 polyunsaturated fatty acids (n-3 PUFAs) have known anti-inflammatory effects. The balance of pro- and anti-inflammatory processes is coordinated by macrophages and macroautophagy/autophagy has recently emerged as a cellular process that dampens inflammation. Here we report that the n-3 PUFA docosahexaenoic acid (DHA) transiently induces cytosolic speckles of the autophagic receptor SQSTM1/p62 (sequestosome 1) (described as SQSTM1/p62-bodies) in macrophages. We suggest that the formation of SQSTM1/p62-bodies represents a fast mechanism of NFE2L2/Nrf2 (nuclear factor, erythroid 2 like 2) activation by recruitment of KEAP1 (kelch like ECH associated protein 1). Further, the autophagy receptor TAX1BP1 (Tax1 binding protein 1) and ubiquitin-editing enzyme TNFAIP3/A20 (TNF α induced protein 3) could be identified in DHA-induced SQSTM1/p62-bodies. Simultaneously, DHA strongly dampened the induction of pro-inflammatory genes including CXCL10 (C-X-C motif chemokine ligand 10) and we suggest that formation of SQSTM1/p62-bodies and activation of NFE2L2 leads to tolerance towards selective inflammatory stimuli. Finally, reduced CXCL10 levels were related to the improved clinical outcome in n-3 PUFA-supplemented heart-transplant patients and we propose CXCL10 as a robust marker for the clinical benefits mobilized by n-3 PUFA supplementation.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Inclusion Bodies/drug effects , Inflammation/prevention & control , Kelch-Like ECH-Associated Protein 1/metabolism , Macrophages/drug effects , NF-E2-Related Factor 2/metabolism , Sequestosome-1 Protein/metabolism , Animals , Autophagy/drug effects , Autophagy/physiology , Cells, Cultured , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Sequestosome-1 Protein/genetics , Transcriptional Activation/drug effects
14.
Sci Rep ; 7(1): 2046, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28515477

ABSTRACT

The majority of cancer patients with advanced disease experience weight loss, including loss of lean body mass. Severe weight loss is characteristic for cancer cachexia, a condition that significantly impairs functional status and survival. The underlying causes of cachexia are incompletely understood, and currently no therapeutic approach can completely reverse the condition. Autophagy coordinates lysosomal destruction of cytosolic constituents and is systemically induced by starvation. We hypothesized that starvation-mimicking signaling compounds secreted from tumor cells may cause a systemic acceleration of autophagy during cachexia. We found that IL-6 secreted by tumor cells accelerates autophagy in myotubes when complexed with soluble IL-6 receptor (trans-signaling). In lung cancer patients, were cachexia is prevalent, there was a significant correlation between elevated IL-6 expression in the tumor and poor prognosis of the patients. We found evidence for an autophagy-inducing bioactivity in serum from cancer patients and that this is clearly associated with weight loss. Importantly, the autophagy-inducing bioactivity was reduced by interference with IL-6 trans-signaling. Together, our findings suggest that IL-6 trans-signaling may be targeted in cancer cachexia.


Subject(s)
Autophagy , Cachexia/etiology , Cachexia/metabolism , Interleukin-6/metabolism , Neoplasms/complications , Neoplasms/metabolism , Signal Transduction , Animals , Biomarkers , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Interleukin-6/blood , Lung Neoplasms/complications , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Mice , Muscle, Skeletal/metabolism , Prognosis , Weight Loss
15.
Biosci Rep ; 37(2)2017 04 30.
Article in English | MEDLINE | ID: mdl-28356483

ABSTRACT

The organization of the Golgi apparatus (GA) is tightly regulated. Golgi stack scattering is observed in cellular processes such as apoptosis and mitosis, and has also been associated with disruption of cellular lipid metabolism and neurodegenerative diseases. Our studies show that depletion of the human N-α-acetyltransferase 30 (hNaa30) induces fragmentation of the Golgi stack in HeLa and CAL-62 cell lines. The GA associated GTPase ADP ribosylation factor related protein 1 (ARFRP1) was previously shown to require N-terminal acetylation for membrane association and based on its N-terminal sequence, it is likely to be a substrate of hNaa30. ARFRP1 is involved in endosome-to-trans-Golgi network (TGN) traffic. We observed that ARFRP1 shifted from a predominantly cis-Golgi and TGN localization to localizing both Golgi and non-Golgi vesicular structures in hNaa30-depleted cells. However, we did not observe loss of membrane association of ARFRP1. We conclude that hNaa30 depletion induces Golgi scattering and induces aberrant ARFRP1 Golgi localization.


Subject(s)
ADP-Ribosylation Factors/metabolism , Golgi Apparatus/metabolism , N-Terminal Acetyltransferase C/metabolism , ADP-Ribosylation Factors/analysis , Endosomes/metabolism , Gene Knockdown Techniques , Golgi Apparatus/ultrastructure , HeLa Cells , Humans , N-Terminal Acetyltransferase C/analysis , N-Terminal Acetyltransferase C/genetics , Protein Transport , trans-Golgi Network/metabolism , trans-Golgi Network/ultrastructure
16.
BMC Cancer ; 17(1): 68, 2017 01 21.
Article in English | MEDLINE | ID: mdl-28109268

ABSTRACT

BACKGROUND: The peptide hormone gastrin exerts a growth-promoting effect in both normal and malignant gastrointestinal tissue. Gastrin mediates its effect via the cholecystokinin 2 receptor (CCKBR/CCK2R). Although a substantial part of the gastric adenocarcinomas express gastrin and CCKBR, the role of gastrin in tumor development is not completely understood. Autophagy has been implicated in mechanisms governing cytoprotection, tumor growth, and contributes to chemoresistance. This study explores the role of autophagy in response to gastrin in gastric adenocarcinoma cell lines. METHODS: Immunoblotting, survival assays and the xCELLigence system were used to study gastrin induced autophagy. Chemical inhibitors of autophagy were utilized to assess the role of this process in the regulation of cellular responses induced by gastrin. Further, knockdown studies using siRNA and immunoblotting were performed to explore the signaling pathways that activate autophagy in response to gastrin treatment. RESULTS: We demonstrate that gastrin increases the expression of the autophagy markers MAP1LC3B-II and SQSTM1 in gastric adenocarcinoma cells. Gastrin induces autophagy via activation of the STK11-PRKAA2-ULK1 and that this signaling pathway is involved in increased migration and cell survival. Furthermore, gastrin mediated increase in survival of cells treated with cisplatin is partially dependent on induced autophagy. CONCLUSION: This study reveals a novel role of gastrin in the regulation of autophagy. It also opens up new avenues in the treatment of gastric cancer by targeting CCKBR mediated signaling and/or autophagy in combination with conventional cytostatic drugs.


Subject(s)
Adenocarcinoma/genetics , Gastrins/metabolism , Microtubule-Associated Proteins/genetics , Sequestosome-1 Protein/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/metabolism , Autophagy , Cell Line, Tumor , Cell Movement , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Receptor, Cholecystokinin B/metabolism , Signal Transduction , Stomach Neoplasms/metabolism
17.
J Steroid Biochem Mol Biol ; 165(Pt B): 228-235, 2017 01.
Article in English | MEDLINE | ID: mdl-27343990

ABSTRACT

While estrogens have been shown to modulate EGFR/HER-1 and HER-2/neu expression in experimental systems, the effects of estrogen deprivation on expression levels of the HER-receptors and the neuregulin (NRG)1 ligand in breast cancers remain unknown. Here, we measured EGFR/HER-1-4 and NRG1 mRNA in ER positive tumors from 85 postmenopausal breast cancer patients before and after two weeks (n=64) and three months (n=85) of primary treatment with an aromatase inhibitor (AI). In tumors lacking HER-2/neu amplification, quantitative real-time PCR analyses revealed EGFR/HER-1 and NRG1 to vary significantly between the three time points (before therapy, after 2 weeks and after 3 months on treatment; P≤0.001 for both). Pair-wise comparison revealed a significant increase in EGFR/HER-1 already during the first two weeks of treatment (P=0.049) with a further increase for both EGFR/HER-1 and NRG1 after 3 months on treatment (P≤0.001 and P=0.001 for both comparing values at 3 months to values at baseline and 2 weeks respectively). No difference between tumors responding versus non-responders was recorded. Further, no significant change in any parameter was observed among HER-2/neu amplified tumors. Analyzing components of the HER-2/neu PI3K/Akt downstream pathway, the PIK3CA H1047R mutation was associated with treatment response (P=0.035); however no association between either AKT phosphorylation status or PIK3CA gene mutations and EGFR/HER-1 or NRG1 expression levels were observed. Our results indicate primary AI treatment to modulate expression of HER-family members and the growth factor NRG1 in HER-2/neu non-amplified breast cancers in vivo. Potential implications to long term sensitivity warrants further investigations.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Neuregulin-1/metabolism , Anastrozole , Breast Neoplasms/drug therapy , DNA Mutational Analysis , Drug Administration Schedule , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Letrozole , Mutation , Nitriles/therapeutic use , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Treatment Outcome , Triazoles/therapeutic use
18.
Oncotarget ; 7(43): 70845-70856, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27683126

ABSTRACT

Cells degrade proteins either by proteasomes that clinically are targeted by for example bortezomib or carfilzomib, or by formation of autophagosomes and lysosomal degradation that can be inhibited by hydroxychloroquine (HCQ). Multiple myeloma is unique among cancers because proteasomal inhibition has good clinical effects. However, some multiple myeloma patients display intrinsic resistance to the treatment and most patients acquire resistance over time. We hypothesized that simultaneous targeting both arms of protein degradation could be a way to improve treatment of multiple myeloma. Here we tested the combined effects of the lysosomal inhibitor HCQ and clinically relevant proteasome inhibitors on myeloma cell lines and primary cells. Carfilzomib and bortezomib both induced immunoglobulin-containing aggregates in myeloma cells. HCQ significantly potentiated the effect of carfilzomib in both cell lines and in primary myeloma cells. In contrast, HCQ had little or no effects on the toxicity of bortezomib. Furthermore, cells adapted to tolerate high levels of carfilzomib could be re-sensitized to the drug by co-treatment with HCQ. Thus, we show that inhibition of lysosomal degradation can overcome carfilzomib resistance, suggesting that the role of autophagy in myeloma cells is dependent on type of proteasome inhibitor. In conclusion, attempts should be made to combine HCQ with carfilzomib in the treatment of multiple myeloma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Hydroxychloroquine/pharmacology , Multiple Myeloma/drug therapy , Oligopeptides/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy/drug effects , Bortezomib/pharmacology , Bortezomib/therapeutic use , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/therapeutic use , Humans , Hydroxychloroquine/therapeutic use , Lysosomes/drug effects , Macrolides/pharmacology , Microscopy, Confocal , Microscopy, Fluorescence , Multiple Myeloma/pathology , Oligopeptides/therapeutic use , Primary Cell Culture , Proteasome Endopeptidase Complex/drug effects , Proteolysis/drug effects , Sequestosome-1 Protein/metabolism
19.
Free Radic Biol Med ; 90: 158-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26585906

ABSTRACT

Polyunsaturated fatty acids (PUFAs) are important constituents of the diet and health benefits of omega-3/n-3 PUFAs, especially eicosapentaenoic acid (EPA, 20:5 n-3) and docosahexaenoic acid (DHA, 22:6 n-3) have been well documented in relation to several diseases. Increasing evidence suggests that n-3 PUFAs may have anticancer activity and improve the effect of conventional cancer therapy. The mechanisms behind these effects are still unclear and need to be elucidated. We have examined the DHA-induced stress response in two human colon cancer cell lines, SW620 and Caco-2. SW620 cells are growth-inhibited at early time points by DHA, while the growth of Caco-2 cells almost remains unaffected by the same treatment. Gene expression analysis of SW620 cells treated with DHA revealed changes at early time points; transcripts involved in oxidative stress and autophagy were among the first to be differentially expressed. We find that oxidative stress is induced in both cell lines, although at different time points and to different extent. DHA induced nuclear translocation of the oxidative stress sensor NFE2L2 in both cell lines, indicating an induction of an anti-oxidative response. However, vitamin E did not counteract ROS-production or the translocation of NFE2L2 to the nucleus. Neither vitamin E nor the antioxidants butylated hydoxyanisole (BHA) and butylated hydoxytoluene (BHT) did affect the growth inhibition in SW620 cells after DHA-treatment. Also, siRNA-mediated down-regulation of NFE2L2 did not sensitize SW620 and Caco-2 cells to DHA. These results indicate that oxidative stress response is not the cause of DHA-induced cytotoxicity in SW620 cells. Using biochemical and imaging based functional assays, we found a low basal level of autophagy and no increase in autophagic flux after adding DHA to the SW620 cells. However, Caco-2 cells displayed a higher level of autophagy, both in the absence and presence of DHA. Inhibition of autophagy by siRNA mediated knock down of ATG5 and ATG7 sensitized both SW620 and Caco-2 cells to DHA. Stimulation of autophagy by rapamycin in SW620 and Caco-2 cells resulted in decreased DHA-sensitivity and inhibition of autophagy in Caco-2 cells by chloroquine resulted in increased DHA-sensitivity. These results suggest that autophagy is important for the DHA sensitivity of colon cancer cells and imply possible therapeutic effects of this fatty acid against cancer cells with low autophagy.


Subject(s)
Autophagy/drug effects , Colonic Neoplasms/drug therapy , Docosahexaenoic Acids/pharmacology , Oxidative Stress/drug effects , Caco-2 Cells , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , NF-E2-Related Factor 2/physiology , Protein Folding
20.
Autophagy ; 11(9): 1636-51, 2015.
Article in English | MEDLINE | ID: mdl-26237736

ABSTRACT

Accumulation and aggregation of misfolded proteins is a hallmark of several diseases collectively known as proteinopathies. Autophagy has a cytoprotective role in diseases associated with protein aggregates. Age-related macular degeneration (AMD) is the most common neurodegenerative eye disease that evokes blindness in elderly. AMD is characterized by degeneration of retinal pigment epithelial (RPE) cells and leads to loss of photoreceptor cells and central vision. The initial phase associates with accumulation of intracellular lipofuscin and extracellular deposits called drusen. Epidemiological studies have suggested an inverse correlation between dietary intake of marine n-3 polyunsaturated fatty acids (PUFAs) and the risk of developing neurodegenerative diseases, including AMD. However, the disease-preventive mechanism(s) mobilized by n-3 PUFAs is not completely understood. In human retinal pigment epithelial cells we find that physiologically relevant doses of the n-3 PUFA docosahexaenoic acid (DHA) induce a transient increase in cellular reactive oxygen species (ROS) levels that activates the oxidative stress response regulator NFE2L2/NRF2 (nuclear factor, erythroid derived 2, like 2). Simultaneously, there is a transient increase in intracellular protein aggregates containing SQSTM1/p62 (sequestosome 1) and an increase in autophagy. Pretreatment with DHA rescues the cells from cell cycle arrest induced by misfolded proteins or oxidative stress. Cells with a downregulated oxidative stress response, or autophagy, respond with reduced cell growth and survival after DHA supplementation. These results suggest that DHA both induces endogenous antioxidants and mobilizes selective autophagy of misfolded proteins. Both mechanisms could be relevant to reduce the risk of developing aggregate-associate diseases such as AMD.


Subject(s)
Autophagy/drug effects , Cytoprotection/drug effects , Docosahexaenoic Acids/pharmacology , Epithelial Cells/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Protein Folding/drug effects , Retinal Pigment Epithelium/cytology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Aquatic Organisms/chemistry , Autophagy-Related Protein 5 , Cell Line , Cell Survival/drug effects , Cytoprotection/genetics , Epithelial Cells/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Protein Aggregates/drug effects , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...