Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell ; 176(1-2): 98-112.e14, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30633912

ABSTRACT

The ability of circulating tumor cells (CTCs) to form clusters has been linked to increased metastatic potential. Yet biological features and vulnerabilities of CTC clusters remain largely unknown. Here, we profile the DNA methylation landscape of single CTCs and CTC clusters from breast cancer patients and mouse models on a genome-wide scale. We find that binding sites for stemness- and proliferation-associated transcription factors are specifically hypomethylated in CTC clusters, including binding sites for OCT4, NANOG, SOX2, and SIN3A, paralleling embryonic stem cell biology. Among 2,486 FDA-approved compounds, we identify Na+/K+ ATPase inhibitors that enable the dissociation of CTC clusters into single cells, leading to DNA methylation remodeling at critical sites and metastasis suppression. Thus, our results link CTC clustering to specific changes in DNA methylation that promote stemness and metastasis and point to cluster-targeting compounds to suppress the spread of cancer.


Subject(s)
Breast Neoplasms/genetics , Neoplasm Metastasis/genetics , Neoplastic Cells, Circulating/pathology , Animals , Breast Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , DNA Methylation/physiology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred NOD , Nanog Homeobox Protein/metabolism , Neoplasm Metastasis/physiopathology , Neoplastic Cells, Circulating/metabolism , Octamer Transcription Factor-3/metabolism , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
2.
Nature ; 607(7917): 156-162, 2022 07.
Article in English | MEDLINE | ID: mdl-35732738

ABSTRACT

The metastatic spread of cancer is achieved by the haematogenous dissemination of circulating tumour cells (CTCs). Generally, however, the temporal dynamics that dictate the generation of metastasis-competent CTCs are largely uncharacterized, and it is often assumed that CTCs are constantly shed from growing tumours or are shed as a consequence of mechanical insults1. Here we observe a striking and unexpected pattern of CTC generation dynamics in both patients with breast cancer and mouse models, highlighting that most spontaneous CTC intravasation events occur during sleep. Further, we demonstrate that rest-phase CTCs are highly prone to metastasize, whereas CTCs generated during the active phase are devoid of metastatic ability. Mechanistically, single-cell RNA sequencing analysis of CTCs reveals a marked upregulation of mitotic genes exclusively during the rest phase in both patients and mouse models, enabling metastasis proficiency. Systemically, we find that key circadian rhythm hormones such as melatonin, testosterone and glucocorticoids dictate CTC generation dynamics, and as a consequence, that insulin directly promotes tumour cell proliferation in vivo, yet in a time-dependent manner. Thus, the spontaneous generation of CTCs with a high proclivity to metastasize does not occur continuously, but it is concentrated within the rest phase of the affected individual, providing a new rationale for time-controlled interrogation and treatment of metastasis-prone cancers.


Subject(s)
Breast Neoplasms , Neoplasm Metastasis , Sleep , Animals , Breast Neoplasms/pathology , Cell Count , Cell Proliferation , Disease Models, Animal , Female , Glucocorticoids , Humans , Insulin , Melatonin , Mice , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , RNA-Seq , Single-Cell Analysis , Testosterone , Time Factors
3.
Nature ; 573(7774): 439-444, 2019 09.
Article in English | MEDLINE | ID: mdl-31485072

ABSTRACT

Metastasis is the major driver of death in patients with cancer. Invasion of surrounding tissues and metastasis have been proposed to initiate following loss of the intercellular adhesion protein, E-cadherin1,2, on the basis of inverse correlations between in vitro migration and E-cadherin levels3. However, this hypothesis is inconsistent with the observation that most breast cancers are invasive ductal carcinomas and express E-cadherin in primary tumours and metastases4. To resolve this discrepancy, we tested the genetic requirement for E-cadherin in metastasis using mouse and human models of both luminal and basal invasive ductal carcinomas. Here we show that E-cadherin promotes metastasis in diverse models of invasive ductal carcinomas. While loss of E-cadherin increased invasion, it also reduced cancer cell proliferation and survival, circulating tumour cell number, seeding of cancer cells in distant organs and metastasis outgrowth. Transcriptionally, loss of E-cadherin was associated with upregulation of genes involved in transforming growth factor-ß (TGFß), reactive oxygen species and apoptosis signalling pathways. At the cellular level, disseminating E-cadherin-negative cells exhibited nuclear enrichment of SMAD2/3, oxidative stress and increased apoptosis. Colony formation of E-cadherin-negative cells was rescued by inhibition of TGFß-receptor signalling, reactive oxygen accumulation or apoptosis. Our results reveal that E-cadherin acts as a survival factor in invasive ductal carcinomas during the detachment, systemic dissemination and seeding phases of metastasis by limiting reactive oxygen-mediated apoptosis. Identifying molecular strategies to inhibit E-cadherin-mediated survival in metastatic breast cancer cells may have potential as a therapeutic approach for breast cancer.


Subject(s)
Antigens, CD , Breast Neoplasms/pathology , Cadherins , Carcinoma, Ductal, Breast/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Animals , Antigens, CD/metabolism , Breast Neoplasms/metabolism , Cadherins/metabolism , Carcinoma, Ductal, Breast/metabolism , Female , Humans , Mice , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism
4.
Nature ; 566(7745): 553-557, 2019 02.
Article in English | MEDLINE | ID: mdl-30728496

ABSTRACT

A better understanding of the features that define the interaction between cancer cells and immune cells is important for the development of new cancer therapies1. However, focus is often given to interactions that occur within the primary tumour and its microenvironment, whereas the role of immune cells during cancer dissemination in patients remains largely uncharacterized2,3. Circulating tumour cells (CTCs) are precursors of metastasis in several types of cancer4-6, and are occasionally found within the bloodstream in association with non-malignant cells such as white blood cells (WBCs)7,8. The identity and function of these CTC-associated WBCs, as well as the molecular features that define the interaction between WBCs and CTCs, are unknown. Here we isolate and characterize individual CTC-associated WBCs, as well as corresponding cancer cells within each CTC-WBC cluster, from patients with breast cancer and from mouse models. We use single-cell RNA sequencing to show that in the majority of these cases, CTCs were associated with neutrophils. When comparing the transcriptome profiles of CTCs associated with neutrophils against those of CTCs alone, we detect a number of differentially expressed genes that outline cell cycle progression, leading to more efficient metastasis formation. Further, we identify cell-cell junction and cytokine-receptor pairs that define CTC-neutrophil clusters, representing key vulnerabilities of the metastatic process. Thus, the association between neutrophils and CTCs drives cell cycle progression within the bloodstream and expands the metastatic potential of CTCs, providing a rationale for targeting this interaction in treatment of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cell Cycle , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Neutrophils/pathology , Animals , Breast Neoplasms/therapy , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Exons/genetics , Female , Gene Expression Profiling , Humans , Intercellular Junctions , Mice , Mutation/genetics , Neoplasm Metastasis/genetics , Neoplastic Cells, Circulating/metabolism , Neutrophils/metabolism , Sequence Analysis, RNA , Exome Sequencing
5.
Br J Cancer ; 125(1): 23-27, 2021 07.
Article in English | MEDLINE | ID: mdl-33762721

ABSTRACT

Circulating tumour cell (CTC) clusters have been proposed to be major players in the metastatic spread of breast cancer, particularly during advanced disease stages. Yet, it is unclear whether or not they manifest in early breast cancer, as their occurrence in patients with metastasis-free primary disease has not been thoroughly evaluated. In this study, exploiting nanostructured titanium oxide-coated slides for shear-free CTC identification, we detect clustered CTCs in the curative setting of multiple patients with early breast cancer prior to surgical treatment, highlighting their presence already at early disease stages. These results spotlight an important aspect of metastasis biology and the possibility to intervene with anti-cluster therapeutics already during the early manifestation of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Titanium/chemistry , Breast Neoplasms/surgery , Case-Control Studies , Cell Line, Tumor , Female , Humans , Nanostructures , Neoplasm Metastasis , Neoplasm Staging
6.
Breast Cancer Res ; 20(1): 141, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30458879

ABSTRACT

BACKGROUND: The presence of circulating tumor cells (CTCs) in patients with breast cancer correlates to a bad prognosis. Yet, CTCs are detectable in only a minority of patients with progressive breast cancer, and factors that influence the abundance of CTCs remain elusive. METHODS: We conducted CTC isolation and enumeration in a selected group of 73 consecutive patients characterized by progressive invasive breast cancer, high tumor load and treatment discontinuation at the time of CTC isolation. CTCs were quantified with the Parsortix microfluidic device. Clinicopathological variables, blood counts at the time of CTC isolation and detailed treatment history prior to blood sampling were evaluated for each patient. RESULTS: Among 73 patients, we detected at least one CTC per 7.5 ml of blood in 34 (46%). Of these, 22 (65%) had single CTCs only, whereas 12 (35%) featured both single CTCs and CTC clusters. Treatment with the monoclonal antibody denosumab correlated with the absence of CTCs, both when considering all patients and when considering only those with bone metastasis. We also found that low red blood cell count was associated with the presence of CTCs, whereas high CA 15-3 tumor marker, high mean corpuscular volume, high white blood cell count and high mean platelet volume associated specifically with CTC clusters. CONCLUSIONS: In addition to blood count correlatives to single and clustered CTCs, we found that denosumab treatment associates with most patients lacking CTCs from their peripheral circulation. Prospective studies will be needed to validate the involvement of denosumab in the prevention of CTC generation.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Denosumab/pharmacology , Erythrocytes , Neoplastic Cells, Circulating/drug effects , Aged , Antineoplastic Agents/therapeutic use , Breast/pathology , Breast Neoplasms/blood , Breast Neoplasms/pathology , Cell Count/methods , Denosumab/therapeutic use , Disease Progression , Female , Humans , Microfluidic Analytical Techniques/methods , Middle Aged , Neoplasm Invasiveness/pathology , Prognosis , Retrospective Studies
7.
Br J Cancer ; 119(4): 487-491, 2018 08.
Article in English | MEDLINE | ID: mdl-30065256

ABSTRACT

Human glioblastoma (GBM) is a highly aggressive, invasive and hypervascularised malignant brain cancer. Individual circulating tumour cells (CTCs) are sporadically found in GBM patients, yet it is unclear whether multicellular CTC clusters are generated in this disease and whether they can bypass the physical hurdle of the blood-brain barrier.  Here, we assessed CTC presence and composition at multiple time points in 13 patients with progressing GBM during an open-label phase 1/2a study with the microtubule inhibitor BAL101553. We observe CTC clusters ranging from 2 to 23 cells and present at multiple sampling time points in a GBM patient with pleomorphism and extensive necrosis, throughout disease progression. Exome sequencing of GBM CTC clusters highlights variants in 58 cancer-associated genes including ATM, PMS2, POLE, APC, XPO1, TFRC, JAK2, ERBB4 and ALK. Together, our findings represent the first evidence of the presence of CTC clusters in GBM.


Subject(s)
Benzimidazoles/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplastic Cells, Circulating/pathology , Oxadiazoles/administration & dosage , Animals , Benzimidazoles/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Count , Cluster Analysis , Disease Progression , Female , Gene Regulatory Networks/drug effects , Genetic Variation , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice , Mutation , Neoplastic Cells, Circulating/chemistry , Neoplastic Cells, Circulating/drug effects , Oxadiazoles/pharmacology , Exome Sequencing , Xenograft Model Antitumor Assays
8.
Lab Invest ; 96(9): 972-80, 2016 09.
Article in English | MEDLINE | ID: mdl-27428080

ABSTRACT

Notch signaling pathways have recently been implicated in the pathogenesis of metabolic diseases. However, the role of hepatic Notch signaling in glucose and lipid metabolism remains unclear and needs further investigation as it might be a candidate therapeutic target in metabolic diseases such as nonalcoholic steatohepatitis (NASH) and nonalcoholic fatty liver disease (NAFLD). We used hepatocyte-specific Notch1 knockout (KO) mice and liver biopsies from NASH and NAFLD patients to analyze the role of Notch1 in glucose and lipid metabolism. Hepatocyte-specific Notch1 KO mice were fed with a high fat diet (HFD) or a regular diet (RD). We assessed the metabolic phenotype, glucose and insulin tolerance tests, and liver histology. Hepatic mRNA expression was profiled by Affymetrix Mouse Gene arrays and validated by quantitative reverse transcription PCR (qPCR). Akt phosphorylation was visualized by immunoblotting. Gene expression was analyzed in liver biopsies from NASH, NAFLD, and control patients by qPCR. We found that Notch1 KO mice had elevated fasting glucose. Gene expression analysis showed an upregulation of glucose-6-phosphatase, involved in the final step of gluconeogenesis and glucose release from glycogenolysis, and perilipin-5, a regulator of hepatic lipid accumulation. When fed with an HFD KO mice developed overt diabetes and hepatic steatosis. Akt was highly phosphorylated in KO animals and the Foxo1 target gene expression was altered. Accordingly, a reduction in Notch1 and increase in glucose-6-phosphatase and perilipin-5 expression was observed in liver biopsies from NAFLD/NASH compared with controls. Notch1 is a regulator of hepatic glucose and lipid homeostasis. Hepatic impairment of Notch1 expression may be involved in the pathogenesis of human NAFLD/NASH.


Subject(s)
Diabetes Mellitus/genetics , Fatty Liver/genetics , Genetic Predisposition to Disease/genetics , Glucose-6-Phosphatase/genetics , Perilipin-1/genetics , Receptor, Notch1/genetics , Animals , Diabetes Mellitus/etiology , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Gene Expression Profiling/methods , Hepatocytes/metabolism , Humans , Immunoblotting , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/deficiency , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
9.
Cancer Res ; 82(4): 681-694, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34916221

ABSTRACT

Blood-borne metastasis of breast cancer involves a series of tightly regulated sequential steps, including the growth of a primary tumor lesion, intravasation of circulating tumor cells (CTC), and adaptation in various distant metastatic sites. The genes orchestrating each of these steps are poorly understood in physiologically relevant contexts, owing to the rarity of experimental models that faithfully recapitulate the biology, growth kinetics, and tropism of human breast cancer. Here, we conducted an in vivo loss-of-function CRISPR screen in newly derived CTC xenografts, unique in their ability to spontaneously mirror the human disease, and identified specific genetic dependencies for each step of the metastatic process. Validation experiments revealed sensitivities to inhibitors that are already available, such as PLK1 inhibitors, to prevent CTC intravasation. Together, these findings present a new tool to reclassify driver genes involved in the spread of human cancer, providing insights into the biology of metastasis and paving the way to test targeted treatment approaches. SIGNIFICANCE: A loss-of-function CRISPR screen in human CTC-derived xenografts identifies genes critical for individual steps of the metastatic cascade, suggesting novel drivers and treatment opportunities for metastatic breast cancers.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Neoplastic Cells, Circulating/metabolism , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Metastasis , Neoplastic Cells, Circulating/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , RNA-Seq/methods , Survival Analysis , Xenograft Model Antitumor Assays/methods , Polo-Like Kinase 1
10.
Microsyst Nanoeng ; 8: 130, 2022.
Article in English | MEDLINE | ID: mdl-36561926

ABSTRACT

Cancer patients with advanced disease are characterized by intrinsic challenges in predicting drug response patterns, often leading to ineffective treatment. Current clinical practice for treatment decision-making is commonly based on primary or secondary tumour biopsies, yet when disease progression accelerates, tissue biopsies are not performed on a regular basis. It is in this context that liquid biopsies may offer a unique window to uncover key vulnerabilities, providing valuable information about previously underappreciated treatment opportunities. Here, we present MyCTC chip, a novel microfluidic device enabling the isolation, culture and drug susceptibility testing of cancer cells derived from liquid biopsies. Cancer cell capture is achieved through a label-free, antigen-agnostic enrichment method, and it is followed by cultivation in dedicated conditions, allowing on-chip expansion of captured cells. Upon growth, cancer cells are then transferred to drug screen chambers located within the same device, where multiple compounds can be tested simultaneously. We demonstrate MyCTC chip performance by means of spike-in experiments with patient-derived breast circulating tumour cells, enabling >95% capture rates, as well as prospective processing of blood from breast cancer patients and ascites fluid from patients with ovarian, tubal and endometrial cancer, where sensitivity to specific chemotherapeutic agents was identified. Together, we provide evidence that MyCTC chip may be used to identify personalized drug response patterns in patients with advanced metastatic disease and with limited treatment opportunities.

11.
Cancers (Basel) ; 13(19)2021 Oct 02.
Article in English | MEDLINE | ID: mdl-34638450

ABSTRACT

Circulating tumor cells (CTCs) are promising diagnostic and prognostic tools for clinical use. In several cancers, including colorectal and breast, the CTC load has been associated with a therapeutic response as well as progression-free and overall survival. However, counting and isolating CTCs remains sub-optimal because they are currently largely identified by epithelial markers such as EpCAM. New, complementary CTC surface markers are therefore urgently needed. We previously demonstrated that a splice variant of CD44, CD44 variable alternative exon 6 (CD44v6), is highly and specifically expressed by CTC cell lines derived from blood samples in colorectal cancer (CRC) patients. Two different approaches-immune detection coupled with magnetic beads and fluorescence-activated cell sorting-were optimized to purify CTCs from patient blood samples based on high expressions of CD44v6. We revealed the potential of the CD44v6 as a complementary marker to EpCAM to detect and purify CTCs in colorectal cancer blood samples. Furthermore, this marker is not restricted to colorectal cancer since CD44v6 is also expressed on CTCs from breast cancer patients. Overall, these results strongly suggest that CD44v6 could be useful to enumerate and purify CTCs from cancers of different origins, paving the way to more efficacious combined markers that encompass CTC heterogeneity.

12.
Sci Rep ; 10(1): 10181, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32576883

ABSTRACT

Circulating tumor cells (CTCs) are derivatives of solid cancerous lesions that detach from the tumor mass and enter the blood circulation. CTCs are considered to be the precursors of metastasis in several cancer types. They are present in the blood of cancer patients as single cells or clusters, with the latter being associated with a higher metastatic potential. Methods to eliminate CTCs from the bloodstream are currently lacking. Here, we took advantage of the lower shear stress-resistance of cancer cells compared to blood cells, and developed a device that can eliminate cancer cells without blood damage. The device consists of an axial pump and a coupled rotating throttle, controllable to prevent local blood flow impairment, yet maintaining a constant shear performance. When processing cancer cells through our device, we observe cancer cell-cluster disruption and viability reduction of single cancer cells, without noticeable effects on human blood cells. When injecting cancer cell-containing samples into tumor-free recipient mice, processed samples fail to generate metastasis. Together, our data show that a selective disruption of cancer cells is possible while preserving blood cells, paving the way towards the development of novel, implantable tools for CTC disruption and metastasis prevention.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Animals , Cell Count/methods , Cell Line, Tumor , Female , Humans , Mice , Neoplasm Metastasis/pathology
13.
Nat Commun ; 6: 7305, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26041499

ABSTRACT

Brain regions, such as the cortex and retina, are composed of layers of uniform thickness. The molecular mechanism that controls this uniformity is not well understood. Here we show that during mouse postnatal development the timed expression of Rncr4, a retina-specific long noncoding RNA, regulates the similarly timed processing of pri-miR-183/96/182, which is repressed at an earlier developmental stage by RNA helicase Ddx3x. Shifting the timing of mature miR-183/96/182 accumulation or interfering with Ddx3x expression leads to the disorganization of retinal architecture, with the photoreceptor layer being most affected. We identify Crb1, a component of the adhesion belt between glial and photoreceptor cells, as a link between Rncr4-regulated miRNA metabolism and uniform retina layering. Our results suggest that the precise timing of glia-neuron interaction controlled by noncoding RNAs and Ddx3x is important for the even distribution of cells across layers.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/genetics , Neuroglia/metabolism , Neurons/metabolism , RNA Helicases/metabolism , RNA, Long Noncoding/genetics , Retina/growth & development , Animals , Blotting, Northern , Blotting, Western , DEAD-box RNA Helicases , Gene Regulatory Networks , HEK293 Cells , Humans , Immunohistochemistry , Mice , MicroRNAs/metabolism , Microscopy, Confocal , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , RNA, Long Noncoding/metabolism , Real-Time Polymerase Chain Reaction , Retina/metabolism
14.
J Clin Invest ; 124(4): 1568-81, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569457

ABSTRACT

The use of pegylated interferon-α (pegIFN-α) has replaced unmodified recombinant IFN-α for the treatment of chronic viral hepatitis. While the superior antiviral efficacy of pegIFN-α is generally attributed to improved pharmacokinetic properties, the pharmacodynamic effects of pegIFN-α in the liver have not been studied. Here, we analyzed pegIFN-α-induced signaling and gene regulation in paired liver biopsies obtained prior to treatment and during the first week following pegIFN-α injection in 18 patients with chronic hepatitis C. Despite sustained high concentrations of pegIFN-α in serum, the Jak/STAT pathway was activated in hepatocytes only on the first day after pegIFN-α administration. Evaluation of liver biopsies revealed that pegIFN-α induces hundreds of genes that can be classified into four clusters based on different temporal expression profiles. In all clusters, gene transcription was mainly driven by IFN-stimulated gene factor 3 (ISGF3). Compared with conventional IFN-α therapy, pegIFN-α induced a broader spectrum of gene expression, including many genes involved in cellular immunity. IFN-induced secondary transcription factors did not result in additional waves of gene expression. Our data indicate that the superior antiviral efficacy of pegIFN-α is not the result of prolonged Jak/STAT pathway activation in hepatocytes, but rather is due to induction of additional genes that are involved in cellular immune responses.


Subject(s)
Interferon-alpha/pharmacology , Janus Kinases/metabolism , Liver/drug effects , Liver/metabolism , Polyethylene Glycols/pharmacology , STAT Transcription Factors/metabolism , Adult , Aged , Antiviral Agents/pharmacology , Endopeptidases/genetics , Endopeptidases/metabolism , Female , Gene Expression/drug effects , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/metabolism , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Interferon alpha-2 , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Janus Kinases/genetics , Kinetics , Liver/immunology , Male , Middle Aged , Recombinant Proteins/pharmacology , STAT Transcription Factors/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitin Thiolesterase
15.
J Exp Med ; 211(5): 857-68, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24752298

ABSTRACT

The molecular mechanisms that link IFN-λ3 genotypes to differential induction of interferon (IFN)-stimulated genes (ISGs) in the liver of patients with chronic hepatitis C (CHC) are not known. We measured the expression of IFN-λ and of the specific IFN-λ receptor chain (IFN-λR1) in 122 liver biopsies of patients with CHC and 53 control samples. The IFN-λ3 genotype was not associated with differential expression of IFN-λ, but rather IFN-λR1. In a series of 30 primary human hepatocyte (PHH) samples, IFN-λR1 expression was low but could be induced with IFN-α. IFN-α-induced IFN-λR1 expression was significantly stronger in PHHs carrying the minor IFN-λ3 allele. The analysis of liver biopsies of patients with CHC revealed a strong association of high IFN-λR1 expression with elevated ISG expression, with IFN-λ3 minor alleles, and with nonresponse to pegylated IFN-α and ribavirin. The findings provide a missing link between the IFN-λ3 genotype and the associated phenotype of treatment nonresponse.


Subject(s)
Hepatitis C, Chronic/metabolism , Interferons/metabolism , Liver/metabolism , Receptors, Interferon/metabolism , Biopsy , Blotting, Western , Case-Control Studies , DNA Primers/genetics , Fluorescent Antibody Technique , Genotype , Humans , In Situ Hybridization, Fluorescence , Interferon-alpha/therapeutic use , Microscopy, Confocal , Polymorphism, Single Nucleotide/genetics , Real-Time Polymerase Chain Reaction , Switzerland
16.
Nat Commun ; 5: 5699, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25534433

ABSTRACT

Hepatitis C virus (HCV) infections are the major cause of chronic liver disease, cirrhosis and hepatocellular carcinoma worldwide. Both spontaneous and treatment-induced clearance of HCV depend on genetic variation within the interferon-lambda locus, but until now no clear causal relationship has been established. Here we demonstrate that an amino-acid substitution in the IFNλ4 protein changing a proline at position 70 to a serine (P70S) substantially alters its antiviral activity. Patients harbouring the impaired IFNλ4-S70 variant display lower interferon-stimulated gene (ISG) expression levels, better treatment response rates and better spontaneous clearance rates, compared with patients coding for the fully active IFNλ4-P70 variant. Altogether, these data provide evidence supporting a role for the active IFNλ4 protein as the driver of high hepatic ISG expression as well as the cause of poor HCV clearance.


Subject(s)
Hepacivirus/physiology , Hepatitis C/immunology , Interleukins/immunology , Acetyltransferases/genetics , Acetyltransferases/immunology , Cell Line , Cytokines/genetics , Cytokines/immunology , Hepacivirus/genetics , Hepatitis C/genetics , Humans , Interleukins/genetics , Membrane Proteins/genetics , Membrane Proteins/immunology , Oxidoreductases Acting on CH-CH Group Donors , Proteins/genetics , Proteins/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Ubiquitins/genetics , Ubiquitins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL