Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Respir Res ; 24(1): 254, 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37880678

ABSTRACT

BACKGROUND: Fibroblast activation protein-α (FAPα) is a marker of activated fibroblasts that can be selectively targeted by an inhibitor (FAPI) and visualised by PET/CT imaging. We evaluated whether the measurement of FAPα in bronchoalveolar lavage fluids (BALF) and the uptake of FAPI by PET/CT could be used as biomarkers of fibrogenesis. METHODS: The dynamics of lung uptake of 18F-labeled FAPI ([18F]FAPI-74) was assessed in the bleomycin mouse model at various time points and using different concentrations of bleomycin by PET/CT. FAPα was measured in BALFs from these bleomycin-treated and control mice. FAPα levels were also assessed in BALFs from controls and patients with idiopathic pulmonary fibrosis (IPF). RESULTS: Bleomycin-treated mice presented a significantly higher uptake of [18F]FAPI-74 during lung fibrinogenesis (days 10 and 16 after instillation) compared to control mice. No significant difference was observed at initial inflammatory phase (3 days) and when fibrosis was already established (28 days). [18F]FAPI-74 tracer was unable to show a dose-response to bleomycin treatment. On the other hand, BALF FAPα levels were steeply higher in bleomycin-treated mice at day 10 and a significant dose-response effect was observed. Moreover, FAPα levels were strongly correlated with lung fibrosis as measured by the modified Aschroft histological analysis, hydroxyproline and the percentage of weight loss. Importantly, higher levels of FAPα were observed in IPF patients where the disease was progressing as compared to stable patients and controls. Moreover, patients with FAPα BALF levels higher than 192.5 pg/mL presented a higher risk of progression, transplantation or death compared to patients with lower levels. CONCLUSIONS: Our preclinical data highlight a specific increase of [18F]FAPI-74 lung uptake during the fibrotic phase of the bleomycin murine model. The measurement of FAPα in BALF appears to be a promising marker of the fibrotic activity in preclinical models of lung fibrosis and in IPF patients. Further studies are required to confirm the role of FAPα in BALF as biomarker of IPF activity and assess the relationship between FAPα levels in BALF and [18F]FAPI-74 uptake on PET/CT in patients with fibrotic lung disease.


Subject(s)
Idiopathic Pulmonary Fibrosis , Positron Emission Tomography Computed Tomography , Humans , Mice , Animals , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/diagnostic imaging , Idiopathic Pulmonary Fibrosis/drug therapy , Fibrosis , Bronchoalveolar Lavage Fluid , Bleomycin/adverse effects
2.
Mol Cancer Ther ; 22(11): 1343-1351, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37578807

ABSTRACT

Adjusting the molecular size, the valency and the pharmacokinetics of drug conjugates are as many leverages to improve their therapeutic window, notably by affecting tumor penetration, renal clearance, and short systemic exposure. In that regard, small tumor-targeting ligands are gaining attention. In this study, we demonstrate the benefits of the small Nanofitin alternative scaffolds (7 kDa) as selective tumor-targeting modules for the generation of drug conjugates, focusing on Nanofitins B10 and D8 directed against the EGFR. Owing to their small size and monovalent format, the two Nanofitins displayed a fast and deep tumor penetration in EGFR-positive A431 xenografts in BALB/c nude mice after intravenous administration, yielding to a targeting of respectively 67.9% ± 14.1 and 98.9% ± 0.7 of the tumor cells as demonstrated by IHC. Conjugation with the monomethyl auristatin E toxin provided homogeneous Nanofitin-drug conjugates, with an overall yield of ≥97%, for in vivo assessment in a curative xenograft model using bioluminescent, EGFR-positive, A431 cells in BALB/c nude mice. Internalization was found critical for efficient release of the toxin. Hence, the intravenous administration of the D8-based construct showed significant antitumor effect in vivo as determined by monitoring tumor volumes and bioluminescence levels over 2 months.


Subject(s)
ErbB Receptors , Neoplasms , Humans , Animals , Mice , Heterografts , Mice, Nude , Neoplasms/drug therapy , Cell Line, Tumor , Xenograft Model Antitumor Assays
3.
Nat Commun ; 13(1): 7057, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36396935

ABSTRACT

The thyroid gland captures iodide in order to synthesize hormones that act on almost all tissues and are essential for normal growth and metabolism. Low plasma levels of thyroid hormones lead to hypothyroidism, which is one of the most common disorder in humans and is not always satisfactorily treated by lifelong hormone replacement. Therefore, in addition to the lack of in vitro tractable models to study human thyroid development, differentiation and maturation, functional human thyroid organoids could pave the way to explore new therapeutic approaches. Here we report the generation of transplantable thyroid organoids derived from human embryonic stem cells capable of restoring plasma thyroid hormone in athyreotic mice as a proof of concept for future therapeutic development.


Subject(s)
Hypothyroidism , Organoids , Humans , Animals , Mice , Hypothyroidism/therapy , Embryonic Stem Cells , Thyroid Hormones
4.
Cell Metab ; 34(1): 125-139.e8, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986331

ABSTRACT

Concerted alteration of immune and metabolic homeostasis underlies several inflammation-related pathologies, ranging from metabolic syndrome to infectious diseases. Here, we explored the coordination of nucleic acid-dependent inflammatory responses and metabolic homeostasis. We reveal that the STING (stimulator of interferon genes) protein regulates metabolic homeostasis through inhibition of the fatty acid desaturase 2 (FADS2) rate-limiting enzyme in polyunsaturated fatty acid (PUFA) desaturation. STING ablation and agonist-mediated degradation increased FADS2-associated desaturase activity and led to accumulation of PUFA derivatives that drive thermogenesis. STING agonists directly activated FADS2-dependent desaturation, promoting metabolic alterations. PUFAs in turn inhibited STING, thereby regulating antiviral responses and contributing to resolving STING-associated inflammation. Thus, we have unveiled a negative regulatory feedback loop between STING and FADS2 that fine-tunes inflammatory responses. Our results highlight the role of metabolic alterations in human pathologies associated with aberrant STING activation and STING-targeting therapies.


Subject(s)
Fatty Acid Desaturases , Metabolic Syndrome , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Fatty Acids, Unsaturated/metabolism , Humans , Inflammation , Lipid Metabolism
5.
Mol Imaging ; 2021: 6677847, 2021.
Article in English | MEDLINE | ID: mdl-33746630

ABSTRACT

Molecular imaging holds great promise in the noninvasive monitoring of several diseases with nanoparticles (NPs) being considered an efficient imaging tool for cancer, central nervous system, and heart- or bone-related diseases and for disorders of the mononuclear phagocytic system (MPS). In the present study, we used an iron-based nanoformulation, already established as an MRI/SPECT probe, as well as to load different biomolecules, to investigate its potential for nuclear planar and tomographic imaging of several target tissues following its distribution via different administration routes. Iron-doped hydroxyapatite NPs (FeHA) were radiolabeled with the single photon γ-emitting imaging agent [99mTc]TcMDP. Administration of the radioactive NPs was performed via the following four delivery methods: (1) standard intravenous (iv) tail vein, (2) iv retro-orbital injection, (3) intratracheal (it) instillation, and (4) intrarectal installation (pr). Real-time, live, fast dynamic screening studies were performed on a dedicated bench top, mouse-sized, planar SPECT system from t = 0 to 1 hour postinjection (p.i.), and consequently, tomographic SPECT/CT imaging was performed, for up to 24 hours p.i. The administration routes that have been studied provide a wide range of possible target tissues, for various diseases. Studies can be optimized following this workflow, as it is possible to quickly assess more parameters in a small number of animals (injection route, dosage, and fasting conditions). Thus, such an imaging protocol combines the strengths of both dynamic planar and tomographic imaging, and by using iron-based NPs of high biocompatibility along with the appropriate administration route, a potential diagnostic or therapeutic effect could be attained.


Subject(s)
Nanoparticles , Animals , Magnetic Iron Oxide Nanoparticles , Mice , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Workflow
6.
Nucl Med Biol ; 93: 74-80, 2021 02.
Article in English | MEDLINE | ID: mdl-33422771

ABSTRACT

INTRODUCTION: Cabozantinib is a tyrosine kinase inhibitor (TKI) approved for the treatment of medullary thyroid cancer, renal cell carcinoma and hepatocellular carcinoma, and is currently in clinical trials for the treatment of prostate cancer and others. It exerts its therapeutic effect mainly through inhibition of the tyrosine kinases MET (hepatocyte growth factor receptor) and VEGFR2 (vascular endothelial growth factor receptor 2), in addition to several other kinases involved in cancer. PET imaging with TKIs such as [18F]cabozantinib could potentially aid in cancer diagnosis and guide treatment. This study aims to evaluate the utility of [18F]cabozantinib as a PET imaging probe in PC3 tumor xenografted mice. METHODS: [18F]cabozantinib was evaluated in non-tumor and tumor bearing (PC3 xenografted) male mice by ex vivo biodistribution studies and in vivo µPET imaging. Pretreatment studies were performed in the tumor bearing mice with the MET inhibitor PF04217903. Mouse plasma was analyzed with HPLC to quantify radiometabolites. To further evaluate the binding specificity of [18F]cabozantinib, in vitro autoradiography studies on heart and PC3 tumor sections were performed in the presence of authentic cabozantinib or specific MET and VEGFR2 inhibitors. RESULTS: Tissue distribution studies in non-tumor bearing mice revealed slow blood clearance, absence of brain uptake and a high myocardial uptake. In the tumor bearing mice, tumor uptake was low (0.58 ± 0.20% ID/g at 30 min post tracer injection), which was confirmed by µPET imaging. No differences in tissue distribution and kinetics were observed in both biodistributions and µPET studies after pretreatment with the MET inhibitor PF04217903. At 30 min post tracer injection, 60 ± 3% of the recovered radioactivity in plasma in non-tumor bearing mice was present as intact tracer. [18F]cabozantinib binding in vitro to heart and tumor tissues was partly blocked in the presence of selective MET and VEGFR2 inhibitors (up to 40% block). The fraction of non-specific binding was relatively high for both tissues (66% for heart and 39% for tumor). CONCLUSION: [18F]cabozantinib exhibits non-favorable properties as a PET imaging probe, demonstrated by slow excretion kinetics along with low tumor uptake and high non-specific binding in tumor and heart tissue. The results reflect cabozantinibs multi-kinase activity, making PET imaging of tumor specific kinase expression with [18F]cabozantinib challenging.


Subject(s)
Anilides , Fluorine Radioisotopes , Positron-Emission Tomography/methods , Pyridines , Anilides/pharmacokinetics , Animals , Disease Models, Animal , Male , Mice , Pyridines/pharmacokinetics , Tissue Distribution
7.
Theranostics ; 11(4): 1626-1640, 2021.
Article in English | MEDLINE | ID: mdl-33408771

ABSTRACT

Colorectal cancer (CRC) cells are traditionally considered unresponsive to TGFß due to mutations in the receptors and/or downstream signaling molecules. TGFß influences CRC cells only indirectly via stromal cells, such as cancer-associated fibroblasts. However, CRC cell ability to directly respond to TGFß currently remains unexplored. This represents a missed opportunity for diagnostic and therapeutic interventions. Methods: We examined whether cancer cells from primary CRC and liver metastases respond to TGFß by inducing TGFß-induced protein ig-h3 (TGFBI) expression, and the contribution of canonical and non-canonical TGFß signaling pathways to this effect. We then investigated in vitro and in vivo TGFBI impact on metastasis formation and angiogenesis. Using patient serum samples and an orthotopic mouse model of CRC liver metastases we assessed the diagnostic/tumor targeting value of novel antibodies against TGFBI. Results: Metastatic CRC cells, such as circulating tumor cells, directly respond to TGFß. These cells were characterized by the absence of TGFß receptor mutations and the frequent presence of p53 mutations. The pro-tumorigenic program orchestrated by TGFß in CRC cells was mediated through TGFBI, the expression of which was positively regulated by non-canonical TGFß signaling cascades. TGFBI inhibition was sufficient to significantly reduce liver metastasis formation in vivo. Moreover, TGFBI pro-tumorigenic function was linked to its ability to stimulate angiogenesis. TGFBI levels were higher in serum samples from untreated patients with CRC than in patients who were receiving chemotherapy. A radiolabeled anti-TGFBI antibody selectively targeted metastatic lesions in vivo, underscoring its diagnostic and therapeutic potential. Conclusions: TGFß signaling in CRC cells directly contributes to their metastatic potential and stromal cell-independence. Proteins downstream of activated TGFß, such as TGFBI, represent novel diagnostic and therapeutic targets for more specific anti-metastatic therapies.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/blood supply , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Extracellular Matrix Proteins/genetics , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice , Neovascularization, Pathologic/metabolism , Prognosis , Signal Transduction , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Eur J Nucl Med Mol Imaging ; 48(6): 1773-1784, 2021 06.
Article in English | MEDLINE | ID: mdl-33398412

ABSTRACT

PURPOSE: This preclinical study aims to evaluate the extent to which a change in prostate-specific membrane antigen (PSMA) expression of castration-resistant prostate cancer (CRPC) following standard treatment is reflected in [18F]JK-PSMA-7 PET/CT. METHODS: Castrated mice supplemented with testosterone implant were xenografted with human LNCaP CRPC. After appropriate tumour growth, androgen deprivation therapy (ADT) was carried out by the removal of the implant followed by a single injection of docetaxel (400 µg/20-g mouse) 2 weeks later. [18F]JK-PSMA-7 PET/CT were performed before ADT, then before and at days 12, 26, 47 and 69 after docetaxel administration. The [18F]JK-PSMA-7 PET data were compared to corresponding unspecific metabolic [18F]FDG PET/CT and ex vivo quantification of PSMA expression estimated by flow cytometry on repeated tumour biopsies. RESULTS: ADT alone had no early effect on LNCaP tumours that pursued their progression. Until day 12 post-docetaxel, the [18F]JK-PSMA7 uptake was significantly higher than that of [18F]FDG, indicating the persistence of PSMA expression at those time points. From day 26 onwards when the tumours were rapidly expanding, both [18F]JK-PSMA7 and [18F]FDG uptake continuously decreased although the decrease in [18F]JK-PSMA uptake was markedly faster. The fraction of PSMA-positive cells in tumour biopsies decreased similarly over time to reach a non-specific level after the same time period. CONCLUSION: Applying PSMA-based imaging for therapy monitoring in patients with CRPC should be considered with caution since a reduction in [18F]JK-PSMA-7 PET uptake after successive ADT and chemotherapy may be related to downregulation of PSMA expression in dedifferentiated and rapidly proliferating tumour cells.


Subject(s)
Prostatic Neoplasms , Androgen Antagonists , Animals , Fluorodeoxyglucose F18 , Heterografts , Humans , Male , Mice , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/drug therapy
9.
Respir Res ; 20(1): 10, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30646908

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is characterized by a progressive and irreversible respiratory failure. Non-invasive markers of disease activity are essential for prognosis and evaluation of early response to anti-fibrotic treatments. OBJECTIVES: The aims of this study were to determine whether fluorodeoxyglucose ([18F]-FDG) lung uptake is reduced after initiation of pirfenidone or nintedanib and to assess its possible use as a prognostic factor. METHODS: [18F]-FDG PET/CT was performed in IPF patients and in a murine model of pulmonary fibrosis. PET/CTs were performed at day 8 and day 15 post-instillation of bleomycin in pirfenidone- or vehicule-treated mice. In IPF patients, PET-CT was performed before and 3 months after the initiation of pirfenidone or nintedanib. RESULTS: In bleomycin-treated mice, pirfenidone significantly reduced the [18F]-FDG uptake compared to vehicule-treated mice at day 15 (p < 0.001), whereas no difference was observed at day 8 after bleomycin administration. In IPF patients, [18F]-FDG lung uptake before and after 3 months of treatment by nintedanib (n = 11) or pirfenidone (n = 14) showed no significant difference regardless the antifibrotic treatment. Moreover, no difference was noticed between patients with progressive or non-progressive disease at one year of follow up. CONCLUSIONS: Pirfenidone significantly reduces the lung [18F]-FDG uptake during the fibrotic phase in a mouse model of IPF. However, these preclinical data were not confirmed in IPF patients 3 months after the initiation of antifibrotic therapy. [18F]-FDG seems therefore not useful in clinical practice to assess the early response of IPF patients to nintedanib or pirfenidone.


Subject(s)
Fluorodeoxyglucose F18 , Idiopathic Pulmonary Fibrosis/diagnostic imaging , Idiopathic Pulmonary Fibrosis/drug therapy , Indoles/therapeutic use , Positron Emission Tomography Computed Tomography/methods , Pyridones/therapeutic use , Aged , Animals , Antineoplastic Agents/therapeutic use , Female , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prospective Studies
10.
Oncogene ; 37(9): 1237-1250, 2018 03.
Article in English | MEDLINE | ID: mdl-29242606

ABSTRACT

Cancer research is increasingly dependent of patient-derived xenograft model (PDX). However, a major point of concern regarding the PDX model remains the replacement of the human stroma with murine counterpart. In the present work we aimed at clarifying the significance of the human-to-murine stromal replacement for the fidelity of colorectal cancer (CRC) and liver metastasis (CRC-LM) PDX model. We have conducted a comparative metabolic analysis between 6 patient tumors and corresponding PDX across 4 generations. Metabolic signatures of cancer cells and stroma were measured separately by MALDI-imaging, while metabolite changes in entire tumors were quantified using mass spectrometry approach. Measurement of glucose metabolism was also conducted in vivo using [18F]-fluorodeoxyglucose (FDG) and positron emission tomography (PET). In CRC/CRC-LM PDX model, human stroma was entirely replaced at the second generation. Despite this change, MALDI-imaging demonstrated that the metabolic profiles of both stromal and cancer cells remained stable for at least four generations in comparison to the original patient material. On the tumor level, profiles of 86 water-soluble metabolites as well as 93 lipid mediators underlined the functional stability of the PDX model. In vivo PET measurement of glucose uptake (reflecting tumor glucose metabolism) supported the ex vivo observations. Our data show for the first time that CRC/CRC-LM PDX model maintains the functional stability at the metabolic level despite the early replacement of the human stroma by murine cells. The findings demonstrate that human cancer cells actively educate murine stromal cells during PDX development to adopt the human-like phenotype.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Colorectal Neoplasms/metabolism , Disease Models, Animal , Glucose/metabolism , Liver Neoplasms/metabolism , Metabolome , Stromal Cells/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Cohort Studies , Colorectal Neoplasms/pathology , Female , Humans , Liver Neoplasms/secondary , Male , Mice , Mice, Inbred NOD , Mice, SCID , Phenotype , Stromal Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Oncotarget ; 8(34): 56185-56198, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28915583

ABSTRACT

Trastuzumab remains an important drug in the management of human epidermal growth factor receptor 2 (HER2) overexpressing breast cancer (BC). Several studies reported resistance mechanisms to trastuzumab, including impaired HER2-accessibility caused by mucin 4 (MUC4). Previously, we demonstrated an increase of Zirconium-89-radiolabeled-trastuzumab (89Zr-Trastuzumab) accumulation when MUC4-overexpressing BC-cells were challenged with the mucolytic drug N-Acetylcysteine (NAC). Hereby, using the same approach we investigated whether tumor exposure to NAC would also enhance trastuzumab-efficacy. Dual SKBr3 (HER2+/MUC4-, sensitive to trastuzumab) and JIMT1 (HER2+/MUC4+, resistant to trastuzumab) HER2-BC-bearing-xenografts were treated with trastuzumab and NAC. Treatment was monitored by molecular imaging evaluating HER2-accessibility/activity (89Zr-Trastuzumab HER2-immunoPET) and glucose metabolism (18F-FDG-PET/CT), as well as tumor volume and the expression of key proteins. In the MUC4-positive JIMT1-tumors, the NAC-trastuzumab combination resulted in improved tumor-growth control compared to trastuzumab alone; with smaller tumor volume/weight, lower 18F-FDG uptake, lower %Ki67 and pAkt-expression. NAC reduced MUC4-expression, but did not affect HER2-expression or the trastuzumab-sensitivity of the MUC4-negative SKBr3-tumors. These findings suggest that improving HER2-accessibility by reducing MUC4-masking with the mucolytic drug NAC, results in a higher anti-tumor effect of trastuzumab. This provides a rationale for the potential benefit of this approach to possibly treat a subset of HER2-positive BC overexpressing MUC4.

12.
Bioconjug Chem ; 28(9): 2361-2371, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28825794

ABSTRACT

Epidermal growth-factor receptor (EGFR) is involved in cell growth and proliferation and is over-expressed in malignant tissues. Although anti-EGFR-based immunotherapy became a standard of care for patients with EGFR-positive tumors, this strategy of addressing cancer tumors by targeting EGFR with monoclonal antibodies is less-developed for patient diagnostic and monitoring. Indeed, antibodies exhibit a slow blood clearance, which is detrimental for positron emission tomography (PET) imaging. New molecular probes are proposed to overcome such limitations for patient monitoring, making use of low-molecular-weight protein scaffolds as alternatives to antibodies, such as Nanofitins with better pharmacokinetic profiles. Anti-EGFR Nanofitin B10 was reformatted by genetic engineering to exhibit a unique cysteine moiety at its C-terminus, which allows the development of a fast and site-specific radiolabeling procedure with 18F-4-fluorobenzamido-N-ethylamino-maleimide (18F-FBEM). The in vivo tumor targeting and imaging profile of the anti-EGFR Cys-B10 Nanofitin was investigated in a double-tumor xenograft model by static small-animal PET at 2 h after tail-vein injection of the radiolabeled Nanofitin 18F-FBEM-Cys-B10. The image showed that the EGFR-positive tumor (A431) is clearly delineated in comparison to the EGFR-negative tumor (H520) with a significant tumor-to-background contrast. 18F-FBEM-Cys-B10 demonstrated a significantly higher retention in A431 tumors than in H520 tumors at 2.5 h post-injection with a A431-to-H520 uptake ratio of 2.53 ± 0.18 and a tumor-to-blood ratio of 4.55 ± 0.63. This study provides the first report of Nanofitin scaffold used as a targeted PET radiotracer for in vivo imaging of EGFR-positive tumor, with the anti-EGFR B10 Nanofitin used as proof-of-concept. The fast generation of specific Nanofitins via a fully in vitro selection process, together with the excellent imaging features of the Nanofitin scaffold, could facilitate the development of valuable PET-based companion diagnostics.


Subject(s)
Antibodies, Monoclonal/chemistry , Cysteine/chemistry , ErbB Receptors/analysis , Maleimides/chemistry , Neoplasms/diagnosis , Positron-Emission Tomography/methods , Animals , Antibodies, Monoclonal/pharmacokinetics , Cell Line, Tumor , Cysteine/pharmacokinetics , Female , Humans , Maleimides/pharmacokinetics , Mice, Inbred BALB C , Mice, Nude
13.
JCI Insight ; 2(11)2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28570274

ABSTRACT

Tristetraprolin (TTP, encoded by the Zfp36 gene) regulates the mRNA stability of several important cytokines. Due to the critical role of this RNA-binding protein in the control of inflammation, TTP deficiency leads to the spontaneous development of a complex inflammatory syndrome. So far, this phenotype has been largely attributed to dysregulated production of TNF and IL­23 by myeloid cells, such as macrophages or DCs. Here, we generated mice with conditional deletion of TTP in keratinocytes (Zfp36fl/flK14-Cre mice, referred to herein as Zfp36ΔEP mice). Unlike DC-restricted (CD11c-Cre) or myeloid cell-restricted (LysM-Cre) TTP ablation, these mice developed exacerbated inflammation in the imiquimod-induced psoriasis model. Furthermore, Zfp36ΔEP mice progressively developed a spontaneous pathology with systemic inflammation, psoriatic-like skin lesions, and dactylitis. Finally, we provide evidence that keratinocyte-derived TNF production drives these different pathological features. In summary, these findings expand current views on the initiation of psoriasis and related arthritis by revealing the keratinocyte-intrinsic role of TTP.

14.
Oncotarget ; 7(37): 59519-59534, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27517747

ABSTRACT

Small nucleolar RNAs (snoRNAs) are emerging as a novel class of proto-oncogenes and tumor suppressors; their involvement in tumorigenesis remains unclear. The box C/D snoRNAs U3 and U8 are upregulated in breast cancers. Here we characterize the function of human U3 and U8 in ribosome biogenesis, nucleolar structure, and tumorigenesis. We show in breast (MCF-7) and lung (H1944) cancer cells that U3 and U8 are required for pre-rRNA processing reactions leading, respectively, to synthesis of the small and large ribosomal subunits. U3 or U8 depletion triggers a remarkably potent p53-dependent anti-tumor stress response involving the ribosomal proteins uL5 (RPL11) and uL18 (RPL5). Interestingly, the nucleolar structure is more sensitive to perturbations in lung cancer than in breast cancer cells. We reveal in a mouse xenograft model that the tumorigenic potential of cancer cells is reduced in the case of U3 suppression and totally abolished upon U8 depletion. Tumors derived from U3-knockdown cells displayed markedly lower metabolic volume and activity than tumors derived from aggressive control cancer cells. Unexpectedly, metabolic tracer uptake by U3-suppressed tumors appeared more heterogeneous, indicating distinctive tumor growth properties that may reflect non-conventional regulatory functions of U3 (or fragments derived from it) in mRNA metabolism.


Subject(s)
Breast Neoplasms/genetics , Conserved Sequence/genetics , Lung Neoplasms/genetics , RNA Precursors/genetics , RNA, Ribosomal/genetics , RNA, Small Nucleolar/genetics , Animals , Carcinogenesis , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , RNA Processing, Post-Transcriptional , RNA, Small Interfering/genetics , Ribosomal Proteins/metabolism , Xenograft Model Antitumor Assays
15.
J Nucl Med ; 56(1): 127-32, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25537989

ABSTRACT

UNLABELLED: Idiopathic pulmonary fibrosis is characterized by a progressive and irreversible respiratory failure. Validated noninvasive methods able to assess disease activity are essential for prognostic purposes as well as for the evaluation of emerging antifibrotic treatments. METHODS: C57BL/6 mice were used in a murine model of pulmonary fibrosis induced by an intratracheal instillation of bleomycin (control mice were instilled with a saline solution). At different times after instillation, PET/CT with (18)F-FDG- or (18)F-4-fluorobenzamido-N-ethylamino-maleimide ((18)F-FBEM)-labeled leukocytes was performed to assess metabolic activity and leukocyte recruitment, respectively. RESULTS: In bleomycin-treated mice, a higher metabolic activity was measured on (18)F-FDG PET/CT scans from day 7 to day 24 after instillation, with a peak of activity measured at day 14. Of note, lung mean standardized uptake values correlated with bleomycin doses, histologic score of fibrosis, lung hydroxyproline content, and weight loss. Moreover, during the inflammatory phase of the model (day 7), but not the fibrotic phase (day 23), bleomycin-treated mice presented with an enhanced leukocyte recruitment as assessed by (18)F-FBEM-labeled leukocyte PET/CT. Autoradiographic analysis of lung sections and CD45 immunostaining confirm the higher and early recruitment of leukocytes in bleomycin-treated mice, compared with control mice. CONCLUSION: (18)F-FDG- and (18)F-FBEM-labeled leukocyte PET/CT enable monitoring of metabolic activity and leukocyte recruitment in a mouse model of pulmonary fibrosis. Implications for preclinical evaluation of antifibrotic therapy are expected.


Subject(s)
Fluorodeoxyglucose F18 , Leukocytes/immunology , Leukocytes/metabolism , Maleimides , Positron-Emission Tomography , Pulmonary Fibrosis/metabolism , Tomography, X-Ray Computed , Animals , Biological Transport/drug effects , Bleomycin/adverse effects , Body Weight/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fibrosis , Fluorodeoxyglucose F18/metabolism , Leukocytes/diagnostic imaging , Lung/drug effects , Lung/immunology , Maleimides/metabolism , Mice , Pulmonary Fibrosis/diagnostic imaging , Pulmonary Fibrosis/pathology , Staining and Labeling
16.
Contrast Media Mol Imaging ; 8(5): 409-16, 2013.
Article in English | MEDLINE | ID: mdl-23740811

ABSTRACT

We used [(18)F]-4-fluorobenzamido-N-ethylamino-maleimide ([(18)F]-FBEM) to radiolabel cells ex vivo for in vivo positron emission tomography (PET) in order to assess cell trafficking in mice. In contrast to commonly used imaging agents, [(18)F]-FBEM forms a covalent bond with thiol groups present on the cells surface. The stability of the probe in aqueous medium was tested at different pH values and cross-experiment showed that thiol-labeling efficiency was retained (at least) up to pH 9. The labeling procedure did not affect significantly the cell viability. To illustrate the procedure, PET images of living mice injected intravenously with labeled T lymphocytes were obtained. They showed the expected cell homing in the spleen that was absent in mice injected with free label.


Subject(s)
Cell Tracking , Maleimides , Sulfhydryl Compounds/isolation & purification , T-Lymphocytes/ultrastructure , Animals , Cell Line, Tumor , Fluorine Radioisotopes , Maleimides/administration & dosage , Maleimides/chemistry , Mice , Mice, Nude , Positron-Emission Tomography , Radioisotopes , Staining and Labeling , Sulfhydryl Compounds/chemistry , Surface Properties , T-Lymphocytes/chemistry , Tissue Distribution
17.
Blood ; 109(6): 2630-3, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17105817

ABSTRACT

Mdm2 and Mdm4 are critical negative regulators of the p53 tumor suppressor. Mdm4-null mutants are severely anemic and exhibit impaired proliferation of the fetal liver erythroid lineage cells. This phenotype may indicate a cell-intrinsic function of Mdm4 in erythropoiesis. In contrast, red blood cell count was nearly normal in mice engineered to express low levels of Mdm2, suggesting that Mdm2 might be dispensable for red cell production. Here, we further explore the tissue-specific functions of Mdm2 and Mdm4 in the erythroid lineage by intercrossing conditional Mdm4 and Mdm2 alleles to an erythroid-specific Cre (Er-GFP-Cre) knock-in allele. Our data show that Mdm2 is required for rescuing erythroid progenitors from p53-mediated apoptosis during primitive erythropoiesis. In contrast, Mdm4 is only required for the high erythropoietic rate during embryonic definitive erythropoiesis. Thus, in this particular cellular context, Mdm4 only contributes to p53 regulation at a specific phase of the differentiation program.


Subject(s)
Erythrocytes/cytology , Erythrocytes/metabolism , Erythropoiesis , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Aging , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Ubiquitin-Protein Ligases/genetics
18.
Proc Natl Acad Sci U S A ; 103(9): 3232-7, 2006 Feb 28.
Article in English | MEDLINE | ID: mdl-16492744

ABSTRACT

The Mdm2 and Mdm4 oncoproteins are key negative regulators of the p53 tumor suppressor. However, their physiological contributions to the regulation of p53 stability and activity remain highly controversial. Here, we combined a p53 knock-in allele, in which p53 is silenced by a transcriptional stop element flanked by loxP sites, with the mdm2- and mdm4-null alleles. This approach allows Cre-mediated conditional p53 expression in tissues in vivo and cells in vitro lacking Mdm2, Mdm4, or both. Using this strategy, we show that Mdm2 and Mdm4 are essential in a nonredundant manner for preventing p53 activity in the same cell type, irrespective of the proliferation/differentiation status of the cells. Although Mdm2 prevents accumulation of the p53 protein, Mdm4 contributes to the overall inhibition of p53 activity independent of Mdm2. We propose a model in which Mdm2 is critical for the regulation of p53 levels and Mdm4 is critical for the fine-tuning of p53 transcriptional activity, both proteins acting synergistically to keep p53 in check.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Proliferation , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/deficiency , Proto-Oncogene Proteins c-mdm2/genetics , Stem Cells/cytology , Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
19.
Cell Cycle ; 4(12): 1703-5, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16258284

ABSTRACT

The p53 tumor suppressor functions as a sequence-specific DNA-binding transcription factor that promotes antiproliferative responses, including cell cycle checkpoints, cellular senescence and apoptosis. The precise nature of the p53 transcriptional programs and the complex mechanisms that govern whether or not a cell dies in response to p53 activation remain elusive. We have recently reported the identification of a new direct p53 target, Ptprv, encoding a transmembrane tyrosine phosphatase. Ptprv expression is dramatically and preferentially increased in cells undergoing p53-dependent cell cycle exit, but not in cells undergoing p53-mediated apoptosis. Importantly, while p53-induced apoptosis is intact in mice lacking Ptprv, Ptprv-null cells are defective in G1 checkpoint control. In addition, we report herein that Ptprv is induced at high cell density and mediates contact inhibition of cell growth. Together, the data suggest that Ptprv is a potent inhibitor of cell proliferation and a critical mediator of p53-induced cell cycle exit.


Subject(s)
Cell Cycle , Protein Tyrosine Phosphatases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Fibroblasts/cytology , Mice , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3
20.
EMBO J ; 24(17): 3093-103, 2005 Sep 07.
Article in English | MEDLINE | ID: mdl-16107883

ABSTRACT

In response to DNA damage, p53 activates a G1 cell cycle checkpoint, in part through induction of the cyclin-dependent kinase inhibitor p21(Waf1/Cip1). Here we report the identification of a new direct p53 target, Ptprv (or ESP), encoding a transmembrane tyrosine phosphatase. Ptprv transcription is dramatically and preferentially increased in cultured cells undergoing p53-dependent cell cycle arrest, but not in cells undergoing p53-mediated apoptosis. This observation was further confirmed in vivo using a Ptprv null-reporter mouse line. A p53-responsive element is present in the Ptprv promoter and p53 is recruited to this site in vivo. Importantly, while p53-dependent apoptosis is intact in mice lacking Ptprv, Ptprv-null fibroblasts and epithelial cells of the small intestine are defective in G1 checkpoint control. Thus, Ptprv is a new direct p53 target and a key mediator of p53-induced cell cycle arrest. Finally, Ptprv loss enhances the formation of epidermal papillomas after exposure to chemical carcinogens, suggesting that Ptprv acts to suppress tumor formation in vivo.


Subject(s)
G1 Phase/physiology , Papilloma/metabolism , Protein Tyrosine Phosphatases/metabolism , Skin Neoplasms/metabolism , Tumor Suppressor Protein p53/physiology , 9,10-Dimethyl-1,2-benzanthracene , Amino Acid Sequence , Animals , Apoptosis , Carcinogens , Cells, Cultured , DNA Damage , Embryo, Mammalian/metabolism , Epithelial Cells/metabolism , Fibroblasts/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Papilloma/chemically induced , Promoter Regions, Genetic , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3 , Skin Neoplasms/chemically induced , Transcription, Genetic , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...