Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Thromb Haemost ; 20(3): 648-660, 2022 03.
Article in English | MEDLINE | ID: mdl-34847278

ABSTRACT

BACKGROUND: Trousseau sign was the first demonstration of a close relationship between cancer and thrombosis. Currently, venous thromboembolism (VTE) is five to six times more likely to occur in cancer patients, whereas there is a greater risk of cancer diagnoses following thromboses. In considering novel players, factor VIII (FVIII), an essential coagulation cofactor with emerging extracoagulative functions, has been identified as an independent VTE risk factor in cancer; however, the basis of this increase is unknown. OBJECTIVE: To investigate the possible direct expression and secretion of FVIII by cancer cells. METHODS: Bladder cancer, with a high VTE risk, and normal bladder tissue and epithelium, were used to investigate FVIII. Factor VIII protein and secretion were examined in bladder cancer cell lines. Expanding to other cancers, the Cancer Cell line Encyclopedia database was used to analyze FVIII, tissue factor, FV, FVII, FIX, FX, and von Willebrand factor (VWF) mRNA in 811 cell lines subdivided according to origin. Factor VIII protein synthesis, secretion, and bioactivity were investigated in a profile of cancer cell lines of differing origins. RESULTS AND CONCLUSIONS: Although expressed in the normal bladder epithelium, FVIII mRNA and protein were higher in matched bladder neoplasms, with synthesis and secretion of bioactive FVIII evident in bladder cancer cells. This can be extended to other cancer cell lines, with a pattern reflecting the tumor origin, and that is independent of VWF and other relevant players in the coagulation cascade. Here, evidence is provided of a possible independent role for FVIII in cancer-related pathophysiology.


Subject(s)
Factor VIII/metabolism , Hemostatics , Neoplasms , Blood Coagulation , Factor VIII/genetics , Humans , von Willebrand Factor/metabolism
2.
J Thromb Haemost ; 18(5): 1050-1064, 2020 05.
Article in English | MEDLINE | ID: mdl-32078252

ABSTRACT

BACKGROUND: We have identified a synonymous F8 variation in a severe hemophilia A (HA) patient who developed inhibitors following factor VIII (FVIII) prophylaxis. The unreported c.6273 G > A variant targets the consensus splicing site of exon 21. OBJECTIVES: To determine the impact of c.6273 G > A nucleotide substitution on F8 splicing and its translated protein. METHODS: Patient peripheral blood mononuclear cells were isolated and differentiated into monocyte-derived macrophages (MDMs). FVIII distribution in cell compartments was evaluated by immunofluorescence. The splicing of mutated exon 21 was assessed by exon trapping. Identified FVIII splicing variants were generated by site-directed mutagenesis, inserted into a lentiviral vector (LV) to transduce Chinese hamster ovary (CHO) cells, and inject into B6/129 HA-mice. FVIII activity was assessed by activated partial thromboplastin time, whereas anti-FVIII antibodies and FVIII antigen, by ELISA. RESULTS: HA-MDMs demonstrated a predominant retention of FVIII around the endoplasmic reticulum. Exon trapping revealed the production of two isoforms: one retaining part of intron 21 and the other skipping exon 21. These variants, predicted to truncate FVIII in the C1 domain, were detected in the patient. CHO cells transduced with the two FVIII transcripts confirmed protein retention and absence of the C2 domain. HA mice injected with LV carrying FVIII mutants, partially recovered FVIII activity without the appearance of anti-FVIII antibodies. CONCLUSIONS: Herein, we demonstrate the aberrant impact of a FVIII synonymous mutation on its transcription, activity, and pathological outcomes. Our data underline the importance of increasing the knowledge regarding the functional consequences of F8 mutations and their link to inhibitor development and an effective replacement therapy.


Subject(s)
Hemophilia A , Animals , CHO Cells , Cricetinae , Cricetulus , Factor VIII/genetics , Factor VIII/metabolism , Hemophilia A/genetics , Humans , Leukocytes, Mononuclear/metabolism , Mice , RNA Splicing
3.
Blood Adv ; 3(5): 825-838, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862611

ABSTRACT

Here we describe a successful gene therapy approach for hemophilia A (HA), using the natural F8 promoter (pF8) to direct gene replacement to factor VIII (FVIII)-secreting cells. The promoter sequence and the regulatory elements involved in the modulation of F8 expression are still poorly characterized and biased by the historical assumption that FVIII expression is mainly in hepatocytes. Bioinformatic analyses have highlighted an underestimated complexity in gene expression at this locus, suggesting an activation of pF8 in more cell types than those previously expected. C57Bl/6 mice injected with a lentiviral vector expressing green fluorescent protein (GFP) under the pF8 (lentiviral vector [LV].pF8.GFP) confirm the predominant GFP expression in liver sinusoidal endothelial cells, with a few positive cells detectable also in hematopoietic organs. Therapeutic gene delivery (LV.pF8.FVIII) in hemophilic C57/Bl6 and 129-Bl6 mice successfully corrected the bleeding phenotype, rescuing up to 25% FVIII activity, using a codon-optimized FVIII, with sustained activity for the duration of the experiment (1 year) without inhibitor formation. Of note, LV.pF8.FVIII delivery in FVIII-immunized HA mice resulted in the complete reversion of the inhibitor titer with the recovery of therapeutic FVIII activity. Depletion of regulatory T cells (Tregs) in LV-treated mice allowed the formation of anti-FVIII antibodies, indicating a role for Tregs in immune tolerance induction. The significant blood loss reduction observed in all LV.pF8.FVIII-treated mice 1 year after injection confirmed the achievement of a long-term phenotypic correction. Altogether, our results highlight the potency of pF8-driven transgene expression to correct the bleeding phenotype in HA, as well as potentially in other diseases in which an endothelial-specific expression is required.


Subject(s)
Factor VIII/administration & dosage , Genetic Therapy/methods , Hemophilia A/therapy , Animals , Disease Models, Animal , Factor VIII/genetics , Factor VIII/therapeutic use , Green Fluorescent Proteins , Immune Tolerance , Lentivirus , Mice , Promoter Regions, Genetic , T-Lymphocytes, Regulatory/immunology
4.
Am J Pathol ; 186(3): 539-51, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26773351

ABSTRACT

Kupffer cells (KC) play major roles in immunity and tissue injury or repair. Because recapitulation of KC biology and function within liver will allow superior insights into their functional repertoire, we studied the efficacy of the cell transplantation approach for this purpose. Mouse KC were isolated from donor livers, characterized, and transplanted into syngeneic recipients. To promote cell engraftment through impairments in native KC, recipients were preconditioned with gadolinium chloride. The targeting, fate, and functionality of transplanted cells were evaluated. The findings indicated that transplanted KC engrafted and survived in recipient livers throughout the study period of 3 months. Transplanted KC expressed macrophage functions, including phagocytosis and cytokine expression, with or without genetic modifications using lentiviral vectors. This permitted studies of whether transplanted KC could affect outcomes in the context of acetaminophen hepatotoxicity or hepatic ischemia-reperfusion injury. Transplanted KC exerted beneficial effects in these injury settings. The benefits resulted from cytoprotective factors including vascular endothelial growth factor. In conclusion, transplanted adult KC were successfully targeted and engrafted in the liver with retention of innate immune and tissue repair functions over the long term. This will provide excellent opportunities to address critical aspects in the biogenesis, fate, and function of KC within their native liver microenvironment and to develop the cell and gene therapy potential of KC transplantation.


Subject(s)
Kupffer Cells/physiology , Kupffer Cells/transplantation , Macrophages/physiology , Reperfusion Injury/therapy , Acetaminophen/adverse effects , Animals , Chemical and Drug Induced Liver Injury/pathology , Cytokines/metabolism , Disease Models, Animal , Gadolinium , Genetic Therapy , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/physiology , Phagocytosis , Reperfusion Injury/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Haematologica ; 100(7): 881-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25911555

ABSTRACT

A large fraction of factor VIII in blood originates from liver sinusoidal endothelial cells although extrahepatic sources also contribute to plasma factor VIII levels. Identification of cell-types other than endothelial cells with the capacity to synthesize and release factor VIII will be helpful for therapeutic approaches in hemophilia A. Recent cell therapy and bone marrow transplantation studies indicated that Küpffer cells, monocytes and mesenchymal stromal cells could synthesize factor VIII in sufficient amount to ameliorate the bleeding phenotype in hemophilic mice. To further establish the role of blood cells in expressing factor VIII, we studied various types of mouse and human hematopoietic cells. We identified factor VIII in cells isolated from peripheral and cord blood, as well as bone marrow. Co-staining for cell type-specific markers verified that factor VIII was expressed in monocytes, macrophages and megakaryocytes. We additionally verified that factor VIII was expressed in liver sinusoidal endothelial cells and endothelial cells elsewhere, e.g., in the spleen, lungs and kidneys. Factor VIII was well expressed in sinusoidal endothelial cells and Küpffer cells isolated from human liver, whereas by comparison isolated human hepatocytes expressed factor VIII at very low levels. After transplantation of CD34(+) human cord blood cells into NOD/SCIDγNull-hemophilia A mice, fluorescence activated cell sorting of peripheral blood showed >40% donor cells engrafted in the majority of mice. In these animals, plasma factor VIII activity 12 weeks after cell transplantation was up to 5% and nine of 12 mice survived after a tail clip-assay. In conclusion, hematopoietic cells, in addition to endothelial cells, express and secrete factor VIII: this information should offer further opportunities for understanding mechanisms of factor VIII synthesis and replenishment.


Subject(s)
Cord Blood Stem Cell Transplantation , Endothelial Cells/metabolism , Factor VIII/biosynthesis , Hemophilia A/therapy , Hemorrhage/prevention & control , Kupffer Cells/metabolism , Animals , Blood Coagulation/genetics , Disease Models, Animal , Endothelial Cells/pathology , Factor VIII/genetics , Factor VIII/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Gene Expression Regulation , Graft Survival , Hemophilia A/blood , Hemophilia A/genetics , Hemophilia A/pathology , Hemorrhage/blood , Hemorrhage/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Kupffer Cells/pathology , Liver/metabolism , Liver/pathology , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes/metabolism , Monocytes/pathology , Phenotype , Transplantation, Heterologous
6.
Nat Commun ; 3: 755, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22453828

ABSTRACT

Endosomal functions are contingent on the integrity of the organelle-limiting membrane, whose disruption induces inflammation and cell death. Here we show that phagocytosis of ultrahigh molecular weight polyethylene particles induces damage to the endosomal-limiting membrane and results in the leakage of cathepsins into the cytosol and NLRP3-inflammasome activation. Annexin A2 recruitment to damaged organelles is shown by two-dimensional DIGE protein profiling, endosomal fractionation, confocal analysis of endogenous and annexin A2-GFP transfected cells, and immunogold labelling. Binding experiments, using fluorescent liposomes, confirms annexin A2 recruitment to endosomes containing phagocytosed polyethylene particles. Finally, an increase in cytosolic cathepsins, NLRP3-inflammasome activation, and IL-1 production is seen in dendritic cells from annexin A2-null mice, following exposure to polyethylene particles. Together, the results indicate a functional role of annexin A2 binding to endosomal membranes following organelle destabilization.


Subject(s)
Annexin A2/metabolism , Carrier Proteins/metabolism , Cathepsins/metabolism , Intracellular Membranes/ultrastructure , Phagocytosis , Animals , Annexin A2/genetics , Carrier Proteins/biosynthesis , Dendritic Cells/metabolism , Endosomes/metabolism , Green Fluorescent Proteins/genetics , Humans , Inflammasomes/metabolism , Interleukin-1/biosynthesis , Intracellular Membranes/metabolism , Joint Prosthesis , Liposomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microspheres , NLR Family, Pyrin Domain-Containing 3 Protein , Polyethylenes
SELECTION OF CITATIONS
SEARCH DETAIL
...