Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
STAR Protoc ; 5(1): 102754, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38096060

ABSTRACT

Characterization of isolated extracellular vesicles and particles (EVPs) is crucial for determining functions and biomarker potential. Here, we present a protocol to analyze size, number, morphology, and EVP protein cargo and to validate EVP proteins in both humans and mice. We describe steps for nanoparticle tracking analysis, transmission electron microscopy, single-EVP immunodetection, EVP proteomic mass spectrometry and bioinformatic analysis, and EVP protein validation by ExoELISA and western blot analysis. This allows for EVP cross-validation across different platforms. For complete details on the use and execution of this protocol, please refer to Hoshino et al.1.


Subject(s)
Extracellular Vesicles , Proteomics , Humans , Animals , Mice , Blotting, Western , Computational Biology , Mass Spectrometry
2.
Nature ; 618(7964): 374-382, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225988

ABSTRACT

Cancer alters the function of multiple organs beyond those targeted by metastasis1,2. Here we show that inflammation, fatty liver and dysregulated metabolism are hallmarks of systemically affected livers in mouse models and in patients with extrahepatic metastasis. We identified tumour-derived extracellular vesicles and particles (EVPs) as crucial mediators of cancer-induced hepatic reprogramming, which could be reversed by reducing tumour EVP secretion via depletion of Rab27a. All EVP subpopulations, exosomes and principally exomeres, could dysregulate hepatic function. The fatty acid cargo of tumour EVPs-particularly palmitic acid-induced secretion of tumour necrosis factor (TNF) by Kupffer cells, generating a pro-inflammatory microenvironment, suppressing fatty acid metabolism and oxidative phosphorylation, and promoting fatty liver formation. Notably, Kupffer cell ablation or TNF blockade markedly decreased tumour-induced fatty liver generation. Tumour implantation or pre-treatment with tumour EVPs diminished cytochrome P450 gene expression and attenuated drug metabolism in a TNF-dependent manner. We also observed fatty liver and decreased cytochrome P450 expression at diagnosis in tumour-free livers of patients with pancreatic cancer who later developed extrahepatic metastasis, highlighting the clinical relevance of our findings. Notably, tumour EVP education enhanced side effects of chemotherapy, including bone marrow suppression and cardiotoxicity, suggesting that metabolic reprogramming of the liver by tumour-derived EVPs may limit chemotherapy tolerance in patients with cancer. Our results reveal how tumour-derived EVPs dysregulate hepatic function and their targetable potential, alongside TNF inhibition, for preventing fatty liver formation and enhancing the efficacy of chemotherapy.


Subject(s)
Extracellular Vesicles , Fatty Acids , Fatty Liver , Liver , Pancreatic Neoplasms , Animals , Mice , Cytochrome P-450 Enzyme System/genetics , Extracellular Vesicles/metabolism , Fatty Acids/metabolism , Fatty Liver/drug therapy , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Liver/metabolism , Liver/pathology , Liver/physiopathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Liver Neoplasms/secondary , Humans , Inflammation/metabolism , Palmitic Acid/metabolism , Kupffer Cells , Oxidative Phosphorylation , rab27 GTP-Binding Proteins/deficiency
3.
Methods Mol Biol ; 2628: 291-300, 2023.
Article in English | MEDLINE | ID: mdl-36781793

ABSTRACT

Plasma extracellular vesicles and particles (EVPs) are enriched in biomolecules that reflect individuals' physiological and pathological states. Several studies have demonstrated the potential of human plasma EVPs as a novel liquid biopsy. Here we describe a protocol for human plasma EVPs isolation and proteomic characterization. We isolated human plasma EVPs by the classical ultracentrifugation method and performed mass spectrometry-based proteomic profiling. Using this protocol, researchers can reveal the plasma EVPs proteome and explore the clinical application of plasma EVPs proteins for developing disease biomarkers.


Subject(s)
Extracellular Vesicles , Proteomics , Humans , Proteomics/methods , Mass Spectrometry , Ultracentrifugation , Blood Proteins/metabolism , Extracellular Vesicles/metabolism , Proteome/metabolism
4.
Nat Commun ; 13(1): 6513, 2022 10 31.
Article in English | MEDLINE | ID: mdl-36316305

ABSTRACT

Tumors initiate by mutations in cancer cells, and progress through interactions of the cancer cells with non-malignant cells of the tumor microenvironment. Major players in the tumor microenvironment are cancer-associated fibroblasts (CAFs), which support tumor malignancy, and comprise up to 90% of the tumor mass in pancreatic cancer. CAFs are transcriptionally rewired by cancer cells. Whether this rewiring is differentially affected by different mutations in cancer cells is largely unknown. Here we address this question by dissecting the stromal landscape of BRCA-mutated and BRCA Wild-type pancreatic ductal adenocarcinoma. We comprehensively analyze pancreatic cancer samples from 42 patients, revealing different CAF subtype compositions in germline BRCA-mutated vs. BRCA Wild-type tumors. In particular, we detect an increase in a subset of immune-regulatory clusterin-positive CAFs in BRCA-mutated tumors. Using cancer organoids and mouse models we show that this process is mediated through activation of heat-shock factor 1, the transcriptional regulator of clusterin. Our findings unravel a dimension of stromal heterogeneity influenced by germline mutations in cancer cells, with direct implications for clinical research.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Clusterin , Heat Shock Transcription Factors , Pancreatic Neoplasms , Animals , Mice , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/pathology , Clusterin/genetics , Clusterin/metabolism , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment/genetics , Humans , Pancreatic Neoplasms
5.
Cancers (Basel) ; 14(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35740692

ABSTRACT

Cancer-related mortality is primarily a consequence of metastatic dissemination and associated complications. Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies and tends to metastasize early, especially in the liver. Emerging evidence suggests that organs that develop metastases exhibit microscopic changes that favor metastatic growth, collectively known as "pre-metastatic niches". By definition, a pre-metastatic niche is chronologically established before overt metastatic outgrowth, and its generation involves the release of tumor-derived secreted factors that modulate cells intrinsic to the recipient organ, as well as recruitment of additional cells from tertiary sites, such as bone marrow-all orchestrated by the primary tumor. The pre-metastatic niche is characterized by tumor-promoting inflammation with tumor-supportive and immune-suppressive features, remodeling of the extracellular matrix, angiogenic modulation and metabolic alterations that support growth of disseminated tumor cells. In this paper, we review the current state of knowledge of the hepatic pre-metastatic niche in PDAC and attempt to create a framework to guide future diagnostic and therapeutic studies.

6.
STAR Protoc ; 2(1): 100225, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33786456

ABSTRACT

We developed a modified protocol, based on differential ultracentrifugation (dUC), to isolate extracellular vesicles and particles (specifically exomeres) (EVPs) from various human and murine sources, including cell lines, surgically resected tumors and adjacent tissues, and bodily fluids, such as blood, lymphatic fluid, and bile. The diversity of these samples requires robust and highly reproducible protocols and refined isolation technology, such as asymmetric-flow field-flow fractionation (AF4). Our isolation protocol allows for preparation of EVPs for various downstream applications, including proteomic profiling. For complete details on the use and execution of this protocol, please refer to Hoshino et al. (2020).


Subject(s)
Body Fluids/chemistry , Centrifugation, Density Gradient , Extracellular Vesicles/chemistry , Fractionation, Field Flow , Proteomics , Animals , Cell Line , Humans , Mice
7.
Cell ; 182(4): 1044-1061.e18, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32795414

ABSTRACT

There is an unmet clinical need for improved tissue and liquid biopsy tools for cancer detection. We investigated the proteomic profile of extracellular vesicles and particles (EVPs) in 426 human samples from tissue explants (TEs), plasma, and other bodily fluids. Among traditional exosome markers, CD9, HSPA8, ALIX, and HSP90AB1 represent pan-EVP markers, while ACTB, MSN, and RAP1B are novel pan-EVP markers. To confirm that EVPs are ideal diagnostic tools, we analyzed proteomes of TE- (n = 151) and plasma-derived (n = 120) EVPs. Comparison of TE EVPs identified proteins (e.g., VCAN, TNC, and THBS2) that distinguish tumors from normal tissues with 90% sensitivity/94% specificity. Machine-learning classification of plasma-derived EVP cargo, including immunoglobulins, revealed 95% sensitivity/90% specificity in detecting cancer. Finally, we defined a panel of tumor-type-specific EVP proteins in TEs and plasma, which can classify tumors of unknown primary origin. Thus, EVP proteins can serve as reliable biomarkers for cancer detection and determining cancer type.


Subject(s)
Biomarkers, Tumor/metabolism , Extracellular Vesicles/metabolism , Neoplasms/diagnosis , Animals , Biomarkers, Tumor/blood , Cell Line , HSC70 Heat-Shock Proteins/metabolism , Humans , Machine Learning , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Neoplasms/metabolism , Proteome/analysis , Proteome/metabolism , Proteomics/methods , Sensitivity and Specificity , Tetraspanin 29/metabolism , rap GTP-Binding Proteins/metabolism
8.
Nat Cell Biol ; 21(11): 1403-1412, 2019 11.
Article in English | MEDLINE | ID: mdl-31685984

ABSTRACT

The development of effective therapies against brain metastasis is currently hindered by limitations in our understanding of the molecular mechanisms driving it. Here we define the contributions of tumour-secreted exosomes to brain metastatic colonization and demonstrate that pre-conditioning the brain microenvironment with exosomes from brain metastatic cells enhances cancer cell outgrowth. Proteomic analysis identified cell migration-inducing and hyaluronan-binding protein (CEMIP) as elevated in exosomes from brain metastatic but not lung or bone metastatic cells. CEMIP depletion in tumour cells impaired brain metastasis, disrupting invasion and tumour cell association with the brain vasculature, phenotypes rescued by pre-conditioning the brain microenvironment with CEMIP+ exosomes. Moreover, uptake of CEMIP+ exosomes by brain endothelial and microglial cells induced endothelial cell branching and inflammation in the perivascular niche by upregulating the pro-inflammatory cytokines encoded by Ptgs2, Tnf and Ccl/Cxcl, known to promote brain vascular remodelling and metastasis. CEMIP was elevated in tumour tissues and exosomes from patients with brain metastasis and predicted brain metastasis progression and patient survival. Collectively, our findings suggest that targeting exosomal CEMIP could constitute a future avenue for the prevention and treatment of brain metastasis.


Subject(s)
Brain Neoplasms/genetics , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Hyaluronoglucosaminidase/genetics , Neovascularization, Pathologic/genetics , Tumor Microenvironment/genetics , Animals , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemokine CCL1/genetics , Chemokine CCL1/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Exosomes/pathology , Humans , Hyaluronoglucosaminidase/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/mortality , Neovascularization, Pathologic/pathology , Signal Transduction , Survival Analysis , Tumor Burden , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
9.
Sci Adv ; 4(2): eaao0665, 2018 02.
Article in English | MEDLINE | ID: mdl-29503865

ABSTRACT

The ribosome, the integration point for protein synthesis in the cell, is conventionally considered a homogeneous molecular assembly that only passively contributes to gene expression. Yet, epigenetic features of the ribosomal DNA (rDNA) operon and changes in the ribosome's molecular composition have been associated with disease phenotypes, suggesting that the ribosome itself may possess inherent regulatory capacity. Analyzing whole-genome sequencing data from the 1000 Genomes Project and the Mouse Genomes Project, we find that rDNA copy number varies widely across individuals, and we identify pervasive intra- and interindividual nucleotide variation in the 5S, 5.8S, 18S, and 28S ribosomal RNA (rRNA) genes of both human and mouse. Conserved rRNA sequence heterogeneities map to functional centers of the assembled ribosome, variant rRNA alleles exhibit tissue-specific expression, and ribosomes bearing variant rRNA alleles are present in the actively translating ribosome pool. These findings provide a critical framework for exploring the possibility that the expression of genomically encoded variant rRNA alleles gives rise to physically and functionally heterogeneous ribosomes that contribute to mammalian physiology and human disease.


Subject(s)
Alleles , Gene Expression Regulation , Mutation/genetics , Organ Specificity/genetics , RNA, Ribosomal/genetics , Animals , Base Sequence , Chromosomes, Human/genetics , Conserved Sequence/genetics , DNA, Ribosomal/genetics , Evolution, Molecular , Gene Dosage , Gene Expression Profiling , Genome, Human , HEK293 Cells , Humans , Mice , Operon/genetics , Protein Biosynthesis , Protein Subunits/genetics , RNA Processing, Post-Transcriptional/genetics , Ribosomes/metabolism
10.
Nat Cell Biol ; 20(3): 332-343, 2018 03.
Article in English | MEDLINE | ID: mdl-29459780

ABSTRACT

The heterogeneity of exosomal populations has hindered our understanding of their biogenesis, molecular composition, biodistribution and functions. By employing asymmetric flow field-flow fractionation (AF4), we identified two exosome subpopulations (large exosome vesicles, Exo-L, 90-120 nm; small exosome vesicles, Exo-S, 60-80 nm) and discovered an abundant population of non-membranous nanoparticles termed 'exomeres' (~35 nm). Exomere proteomic profiling revealed an enrichment in metabolic enzymes and hypoxia, microtubule and coagulation proteins as well as specific pathways, such as glycolysis and mTOR signalling. Exo-S and Exo-L contained proteins involved in endosomal function and secretion pathways, and mitotic spindle and IL-2/STAT5 signalling pathways, respectively. Exo-S, Exo-L and exomeres each had unique N-glycosylation, protein, lipid, DNA and RNA profiles and biophysical properties. These three nanoparticle subsets demonstrated diverse organ biodistribution patterns, suggesting distinct biological functions. This study demonstrates that AF4 can serve as an improved analytical tool for isolating extracellular vesicles and addressing the complexities of heterogeneous nanoparticle subpopulations.


Subject(s)
Cell Fractionation/methods , Exosomes/metabolism , Nanoparticles , Neoplasms/metabolism , Proteins/metabolism , Animals , Biomarkers/metabolism , DNA/genetics , DNA/metabolism , Energy Metabolism , Exosomes/classification , Exosomes/genetics , Exosomes/pathology , Female , Glycomics , Glycosylation , HCT116 Cells , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Metabolomics , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/pathology , PC-3 Cells , Phenotype , Proteomics , RNA/genetics , RNA/metabolism , Signal Transduction , Tissue Distribution
11.
HPB (Oxford) ; 20(7): 597-604, 2018 07.
Article in English | MEDLINE | ID: mdl-29339034

ABSTRACT

BACKGROUND: Exosomes are nanovesicles that have been shown to mediate carcinogenesis in pancreatic ductal adenocarcinoma (PDAC). Given the direct communication of pancreatic duct fluid with the tumor and its relative accessibility, we aimed to determine the feasibility of isolating and characterizing exosomes from pancreatic duct fluid. METHODS: Pancreatic duct fluid was collected from 26 patients with PDAC (n = 13), intraductal papillary mucinous neoplasm (IPMN) (n = 8) and other benign pancreatic diseases (n = 5) at resection. Exosomes were isolated by serial ultracentrifugation, proteins were identified by mass spectrometry, and their expression was evaluated by immunohistochemistry. RESULTS: Exosomes were isolated from all specimens with a mean concentration of 5.9 ± 1 × 108 particles/mL and most frequent size of 138 ± 9 nm. Among the top 35 proteins that were significantly associated with PDAC, multiple carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) and extracellular matrix (ECM) proteins were identified. Interestingly, CEACAM 1/5 expression by immunohistochemistry was seen only on tumor epithelia whereas tenascin C positivity was restricted to stroma, suggesting that both tumor and stromal cells contributed to exosomes. CONCLUSION: This is the first study showing that exosome isolation is feasible from pancreatic duct fluid, and that exosomal proteins may be utilized to diagnose patients with PDAC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/chemistry , Cell Adhesion Molecules/analysis , Exosomes/chemistry , Extracellular Matrix Proteins/analysis , Pancreatic Ducts/chemistry , Pancreatic Intraductal Neoplasms/chemistry , Pancreatic Juice/chemistry , Pancreatic Neoplasms/chemistry , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/pathology , Feasibility Studies , Female , Humans , Immunohistochemistry , Male , Mass Spectrometry , Middle Aged , Pancreatic Ducts/pathology , Pancreatic Intraductal Neoplasms/pathology , Pancreatic Neoplasms/pathology , Pilot Projects , Predictive Value of Tests , Ultracentrifugation
12.
Oncotarget ; 7(13): 16396-408, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26918939

ABSTRACT

Pancreatic stellate cells (PSCs) are the key precursor cells for cancer-associated fibroblasts (CAFs) in pancreatic tumor stroma. In this study, we explored miRNA as therapeutic targets in tumor stroma and found miR-199a-3p and miR-214-3p induced in patient-derived pancreatic CAFs and TGF-ß-activated human PSCs (hPSCs). Inhibition of miR-199a/-214 using hairpin inhibitors significantly inhibited TGFß-induced differentiation markers (e.g. α-SMA, collagen, PDGFßR), migration and proliferation. Furthermore, heterospheroids of Panc-1 and hPSCs attained smaller size with hPSCs transfected with anti-miR-199a/-214 compared to control anti-miR. The conditioned medium obtained from TGFß-activated hPSCs induced tumor cell growth and endothelial cell tube formation. Interestingly, these inductions were abrogated in hPSCs transfected with anti-miR-199a or miR-214. Moreover, IPA analyses revealed signaling pathways related to miR-199a (TP53, mTOR, Smad1) and miR-214 (PTEN, Bax, ING4). Taken together, this study reveals miR-199a-3p and miR-214-3p as major regulators of PSC activation and PSC-induced pro-tumoral effects, representing them as key therapeutic targets in pancreatic cancer.


Subject(s)
MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/metabolism , Antagomirs/genetics , Apoptosis/genetics , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Situ Hybridization , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transforming Growth Factor beta1/pharmacology
13.
Nature ; 527(7578): 329-35, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26524530

ABSTRACT

Ever since Stephen Paget's 1889 hypothesis, metastatic organotropism has remained one of cancer's greatest mysteries. Here we demonstrate that exosomes from mouse and human lung-, liver- and brain-tropic tumour cells fuse preferentially with resident cells at their predicted destination, namely lung fibroblasts and epithelial cells, liver Kupffer cells and brain endothelial cells. We show that tumour-derived exosomes uptaken by organ-specific cells prepare the pre-metastatic niche. Treatment with exosomes from lung-tropic models redirected the metastasis of bone-tropic tumour cells. Exosome proteomics revealed distinct integrin expression patterns, in which the exosomal integrins α6ß4 and α6ß1 were associated with lung metastasis, while exosomal integrin αvß5 was linked to liver metastasis. Targeting the integrins α6ß4 and αvß5 decreased exosome uptake, as well as lung and liver metastasis, respectively. We demonstrate that exosome integrin uptake by resident cells activates Src phosphorylation and pro-inflammatory S100 gene expression. Finally, our clinical data indicate that exosomal integrins could be used to predict organ-specific metastasis.


Subject(s)
Brain/metabolism , Exosomes/metabolism , Integrins/metabolism , Liver/metabolism , Lung/metabolism , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Tropism , Animals , Biomarkers/metabolism , Brain/cytology , Cell Line, Tumor , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genes, src , Humans , Integrin alpha6beta1/metabolism , Integrin alpha6beta4/antagonists & inhibitors , Integrin alpha6beta4/metabolism , Integrin beta Chains/metabolism , Integrin beta4/metabolism , Integrins/antagonists & inhibitors , Kupffer Cells/cytology , Kupffer Cells/metabolism , Liver/cytology , Lung/cytology , Mice , Mice, Inbred C57BL , Organ Specificity , Phosphorylation , Receptors, Vitronectin/antagonists & inhibitors , Receptors, Vitronectin/metabolism , S100 Proteins/genetics
14.
World J Gastroenterol ; 21(6): 1775-83, 2015 Feb 14.
Article in English | MEDLINE | ID: mdl-25684942

ABSTRACT

AIM: To investigate whether nitrite administered prior to ischemia/reperfusion (I/R) reduces liver injury. METHODS: Thirty-six male Sprague-Dawley rats were randomized to 3 groups, including sham operated (n = 8), 45-min segmental ischemia of the left liver lobe (IR, n = 14) and ischemia/reperfusion (I/R) preceded by the administration of 480 nmol of nitrite (n = 14). Serum transaminases were measured after 4 h of reperfusion. Liver microdialysate (MD) was sampled in 30-min intervals and analyzed for glucose, lactate, pyruvate and glycerol as well as the total nitrite and nitrate (NOx). The NOx was measured in serum. RESULTS: Aspartate aminotransferase (AST) at the end of reperfusion was higher in the IR group than in the nitrite group (40 ± 6.8 µkat/L vs 22 ± 2.6 µkat/L, P = 0.022). Similarly, alanine aminotransferase (ALT) was also higher in the I/R group than in the nitrite group (34 ± 6 µkat vs 14 ± 1.5 µkat, P = 0.0045). The NOx in MD was significantly higher in the nitrite group than in the I/R group (10.1 ± 2.9 µmol/L vs 3.2 ± 0.9 µmol/L, P = 0.031) after the administration of nitrite. During ischemia, the levels decreased in both groups and then increased again during reperfusion. At the end of reperfusion, there was a tendency towards a higher NOx in the I/R group than in the nitrite group (11.6 ± 0.7 µmol/L vs 9.2 ± 1.1 µmol/L, P = 0.067). Lactate in MD was significantly higher in the IR group than in the nitrite group (3.37 ± 0.18 mmol/L vs 2.8 ± 0.12 mmol/L, P = 0.01) during ischemia and the first 30 min of reperfusion. During the same period, glycerol was also higher in the IRI group than in the nitrite group (464 ± 38 µmol/L vs 367 ± 31 µmol/L, P = 0.049). With respect to histology, there were more signs of tissue damage in the I/R group than in the nitrite group, and 29% of the animals in the I/R group exhibited necrosis compared with none in the nitrite group. Inducible nitric oxide synthase transcription increased between early ischemia (t = 15) and the end of reperfusion in both groups. CONCLUSION: Nitrite administered before liver ischemia in the rat liver reduces anaerobic metabolism and cell necrosis, which could be important in the clinical setting.


Subject(s)
Liver/blood supply , Liver/drug effects , Reperfusion Injury/prevention & control , Sodium Nitrite/administration & dosage , Animals , Biomarkers/blood , Cytoprotection , Disease Models, Animal , Energy Metabolism/drug effects , Injections, Intravenous , Liver/metabolism , Liver/pathology , Liver/surgery , Male , Necrosis , Rats, Sprague-Dawley , Reperfusion Injury/blood , Reperfusion Injury/pathology , Time Factors
15.
World J Gastroenterol ; 20(28): 9506-12, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25071345

ABSTRACT

AIM: To study the effects of preconditioning on inducible nitric oxide synthase (iNOS) and interleukin 1 (IL-1) receptor transcription in rat liver ischemia/reperfusion injury (IRI). METHODS: Seventy-two male rats were randomized into 3 groups: the one-hour segmental ischemia (IRI, n = 24) group, the ischemic preconditioning (IPC, n = 24) group or the remote ischemic preconditioning (R-IPC, n = 24) group. The IPC and R-IPC were performed as 10 min of ischemia and 10 min of reperfusion. The iNOS and the IL-1 receptor mRNA in the liver tissue was analyzed with real time PCR. The total Nitrite and Nitrate (NOx) in continuously sampled microdialysate (MD) from the liver was analyzed. In addition, the NOx levels in the serum were analyzed. RESULTS: After 4 h of reperfusion, the iNOS mRNA was significantly higher in the R-IPC (ΔCt: 3.44 ± 0.57) group than in the IPC (ΔCt: 5.86 ± 0.82) group (P = 0.025). The IL-1 receptor transcription activity was reduced in the IPC group (ΔCt: 1.88 ± 0.53 to 4.81 ± 0.21), but not in the R-IPC group, during reperfusion (P = 0.027). In the MD, a significant drop in the NOx levels was noted in the R-IPC group (12.3 ± 2.2 to 4.7 ± 1.2 µmol/L) at the end of ischemia compared with the levels in early ischemia (P = 0.008). A similar trend was observed in the IPC group (11.8 ± 2.1 to 6.4 ± 1.5 µmol/L), although this difference was not statistically significant. The levels of NOx rose quickly during reperfusion in both groups. CONCLUSION: IPC, but not R-IPC, reduces iNOS and IL-1 receptor transcription during early reperfusion, indicating a lower inflammatory reaction. NOx is consumed in the ischemic liver lobe.


Subject(s)
Hindlimb/blood supply , Ischemic Preconditioning/methods , Liver/blood supply , Liver/enzymology , Nitric Oxide Synthase Type II/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control , Transcription, Genetic , Animals , Disease Models, Animal , Down-Regulation , Inflammation Mediators/metabolism , Liver/pathology , Male , Microdialysis , Nitrates/blood , Nitric Oxide Synthase Type II/genetics , Nitrites/blood , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Time Factors
16.
PLoS One ; 8(12): e84815, 2013.
Article in English | MEDLINE | ID: mdl-24376848

ABSTRACT

The role of the epithelial-mesenchymal transition (EMT) in cancer has been studied extensively in vitro, but involvement of the EMT in tumorigenesis in vivo is largely unknown. We investigated the potential of microRNAs as clinical markers and analyzed participation of the EMT-associated microRNA-200-ZEB-E-cadherin pathway in cancer progression. Expression of the microRNA-200 family was quantified by real-time RT-PCR analysis of fresh-frozen and microdissected formalin-fixed paraffin-embedded primary colorectal tumors, normal colon mucosa, and matched liver metastases. MicroRNA expression was validated by in situ hybridization and after in vitro culture of the malignant cells. To assess EMT as a predictive marker, factors considered relevant in colorectal cancer were investigated in 98 primary breast tumors from a treatment-randomized study. Associations between the studied EMT-markers were found in primary breast tumors and in colorectal liver metastases. MicroRNA-200 expression in epithelial cells was lower in malignant mucosa than in normal mucosa, and was also decreased in metastatic compared to non-metastatic colorectal cancer. Low microRNA-200 expression in colorectal liver metastases was associated with bad prognosis. In breast cancer, low levels of microRNA-200 were related to reduced survival and high expression of microRNA-200 was predictive of benefit from radiotheraphy. MicroRNA-200 was associated with ER positive status, and inversely correlated to HER2 and overactivation of the PI3K/AKT pathway, that was associated with high ZEB1 mRNA expression. Our findings suggest that the stability of microRNAs makes them suitable as clinical markers and that the EMT-related microRNA-200-ZEB-E-cadherin signaling pathway is connected to established clinical characteristics and can give useful prognostic and treatment-predictive information in progressive breast and colorectal cancers.


Subject(s)
Breast Neoplasms/physiopathology , Colorectal Neoplasms/physiopathology , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Signal Transduction/physiology , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cadherins/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Laser Capture Microdissection , Liver Neoplasms/secondary , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1
17.
PLoS One ; 8(8): e70874, 2013.
Article in English | MEDLINE | ID: mdl-23951028

ABSTRACT

The interplay between the tumor cells and the surrounding stroma creates inflammation, which promotes tumor growth and spread. The inflammation is a hallmark for pancreatic adenocarcinoma (PDAC) and is to high extent driven by IL-1α. IL-1α is expressed and secreted by the tumor cells and exerting its effect on the stroma, i.e. cancer associated fibroblasts (CAF), which in turn produce massive amount of inflammatory and immune regulatory factors. IL-1 induces activation of transcription factors such as nuclear factor-κß (NF-κß), but also activator protein 1 (AP-1) via the small G-protein Ras. Dysregulation of Ras pathways are common in cancer as this oncogene is the most frequently mutated in many cancers. In contrast, the signaling events leading up to the expression of IL-1α by tumor cells are not well elucidated. Our aim was to examine the signaling cascade involved in the induction of IL-1α expression in PDAC. We found p38MAPK, activated by the K-Ras signaling pathway, to be involved in the expression of IL-1α by PDAC as blocking this pathway decreased both the gene and protein expression of IL-1α. Blockage of the P38MAPK signaling in PDAC also dampened the ability of the tumor cell to induce inflammation in CAFs. In addition, the IL-1α autocrine signaling regulated the migratory capacity of PDAC cells. Taken together, the blockage of signaling pathways leading to IL-1α expression and/or neutralization of IL-1α in the PDAC microenvironment should be taken into consideration as possible treatment or complement to existing treatment of this cancer.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Movement/genetics , Interleukin-1alpha/genetics , MAP Kinase Signaling System , Cell Communication/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Cancer Res ; 73(4): 1287-97, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23243022

ABSTRACT

Platelet-derived growth factor (PDGF) receptor signaling is a major functional determinant of cancer-associated fibroblasts (CAF). Elevated expression of PDGF receptors on stromal CAFs is associated with metastasis and poor prognosis, but mechanism(s) that underlie these connections are not understood. Here, we report the identification of the secreted glycoprotein stanniocalcin-1 (STC1) as a mediator of metastasis by PDGF receptor function in the setting of colorectal cancer. PDGF-stimulated fibroblasts increased migration and invasion of cocultured colorectal cancer cells in an STC1-dependent manner. Analyses of human colorectal cancers revealed significant associations between stromal PDGF receptor and STC1 expression. In an orthotopic mouse model of colorectal cancer, tumors formed in the presence of STC1-deficient fibroblasts displayed reduced intravasation of tumor cells along with fewer and smaller distant metastases formed. Our results reveal a mechanistic basis for understanding the contribution of PDGF-activated CAFs to cancer metastasis.


Subject(s)
Colorectal Neoplasms/metabolism , Fibroblasts/metabolism , Glycoproteins/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cells, Cultured , Coculture Techniques , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Fibroblasts/drug effects , Gene Expression Profiling , Glycoproteins/genetics , HCT116 Cells , HT29 Cells , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oligonucleotide Array Sequence Analysis , Platelet-Derived Growth Factor/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
19.
Scand J Gastroenterol ; 47(2): 225-36, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22242616

ABSTRACT

OBJECTIVE: N-acetylcysteine (NAC) is an antioxidative molecule known to protect liver tissue from oxygen radical species generated during ischemia and reperfusion (IR). Nutritional and toxicology studies have shown that NAC also improves glucose metabolism and glycogen stores. We hypothesized that NAC improves glycogenesis and that impaired glycogenesis is a key element in IR injury. MATERIAL AND METHODS: In an experimental model, 80 min of segmental liver ischemia was induced in 16 pigs and the reperfusion was followed for 360 min. Eight animals received NAC 150 mg/kg as a bolus injection followed by an infusion of NAC 50 mg/kg/h intravenously. RESULTS: AST and leukocyte density were lower in the NAC-treated animals, unrelated to the glutathione levels or apoptosis. Glycogen stores returned to a higher degree in the NAC-treated animals and microdialysis revealed lower levels of lactate during the reperfusion phase. Nitrite/Nitrate levels in the NAC group were lower in both serum and microdialysates, indicating that NAC scavenges radical nitrosative species. CONCLUSIONS: NAC treatment improves glycogenesis after liver IR injury and reduces the level of intraparenchymal lactate during reperfusion, possibly due to the scavenging of radical nitrosative species.


Subject(s)
Acetylcysteine/pharmacology , Free Radical Scavengers/pharmacology , Glycogen/biosynthesis , Liver/metabolism , Reperfusion Injury/metabolism , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Apoptosis , Aspartate Aminotransferases/blood , Glutathione/metabolism , Lactic Acid/metabolism , Leukocyte Count , Liver/physiopathology , Male , Microdialysis , Neutrophils , Nitrates/metabolism , Nitrites/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Swine
20.
J Surg Res ; 176(1): 55-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21962739

ABSTRACT

BACKGROUND: Ischemic preconditioning (IPC) of the liver decreases liver injury secondary to ischemia and reperfusion. An attractive alternative to IPC is remote ischemic preconditioning (R-IPC), but these two methods have not previously been compared. MATERIAL AND METHODS: Eighty-seven rats were randomized into four groups: sham operated (n = 15), 1 h segmental ischemia (IRI, n = 24), preceded by IPC (n = 24), or R-IPC (n = 24) (to the left hindleg). IPC and R-IPC were performed with 10 min ischemia and 10 min of reperfusion. Analyses of liver microdialysate (MD), serum transaminase levels, and liver histology were made. RESULTS: Rats treated with IPC and R-IPC had significantly lower AST, 71.5 (19.6) IU/L respective 96.6 (12.4) at 4 h reperfusion than those subjected to IRI alone, 155 (20.9), P = 0.0004 and P = 0.04 respectively. IPC also had lower ALT levels, 41.6 (11.3) IU/L than had IRI 107.4 (15.5), P = 0.003. The MD glycerol was significantly higher during ischemia in the R-IPC [759 (84) µM] and the IRI [732 (67)] groups than in the IPC 514 (70) group, P = 0.022 and P = 0.046 respectively. The MD glucose after ischemia was lower in the IPC group 7.1 (1.2) than in the IRI group 12.7 (1.6), P = 0.005. Preconditioning to the liver caused an direct increase in lactate, glucose and glycerol in the ischemic segment compared with the control segment an effect not seen in the R-IPC and IRI groups. CONCLUSIONS: IPC affects glucose metabolism in the rat liver, observed with MD. IPC reduces liver cell injury during ischemic and reperfusion in rats. R-IPC performed over the same length of time as IPC does not have the same effect as the latter on ALT levels and MD glycerol; this may suggest that R-IPC does not offer the same protection as IPC in this setting of rat liver IRI.


Subject(s)
Ischemic Preconditioning/methods , Liver/blood supply , Liver/metabolism , Microdialysis/methods , Reperfusion Injury/prevention & control , Animals , Blood Glucose/metabolism , Glycerol/metabolism , Lactates/metabolism , Liver/pathology , Male , Models, Animal , Pyruvates/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Transaminases/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...