Transcriptional Analysis Of a Tissue-Specific Factor VIII Knockout Model Demonstrates The Importance Of Endothelial Factor VIII Synthesis

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 27-27 ◽  
Author(s):  
Scot A. Fahs ◽  
Matthew T. Hille ◽  
Robert R. Montgomery

Abstract Definitively identifying the cells responsible for synthesis of coagulation factor VIII (F8) has proven to be a challenge. Transplantation studies demonstrate that as an organ liver is the major, but not exclusive source of plasma F8. Within the liver F8 expression has been variously attributed to hepatocytes, and/or liver sinusoidal endothelial cells, and/or Kupffer cells. Extrahepatic transcription of F8 mRNA appears to be nearly ubiquitous at a low level throughout the body. Previous studies have relied upon retrospective post-expression detection of F8 protein or mRNA using a variety of immunochemical, in situ, and cell isolation techniques, but continuing controversy speaks to the difficulties in localizing expression of a trace protein such as F8. We used a rather different, pre-emptive approach to address the question of F8 synthesis. We developed a conditional F8 knockout (KO) mouse model that allows inactivation of the F8 gene, thus preventing expression, in specific cell types. Exons 17/18 of the F8 gene were flanked by LoxP sites (floxed) resulting in their excision in cells expressing Cre recombinase. Tissue-specific Cre-expressing mouse strains were cross-bred with floxed (F8F) mice to generate tissue-specific F8-KO models. Embryonic Cre expression resulted in a new F8KOstrain displaying a severe hemophilia A phenotype. A hepatocyte-specific F8-KO has completely normal plasma F8 levels, while each of 3 endothelial cell (EC)-Cre models displays a reduced-F8 phenotype that correlates in severity with endothelial Cre efficiency. Presumably due to a shared hemangioblast progenitor, Cre is expressed with similar efficiency in both EC and hematopoietic cells in these models. Plasma F8 is undetectable in the most efficient EC-KO model. In contrast, a highly efficient hematopoietic F8-KO model presents with only modestly reduced F8 levels, likely due to off-target effects. RNA analysis revealed that the F8KO allele produces 2 alternatively spliced transcripts in roughly equivalent amounts. The 1st transcript represents the predicted exon 16/19 splicing event. In the 2nd transcript, 46bp at the 5’ end of intron 16 are retained due to the same cryptic splice site observed in the Kazazian exon 17-disrupted F8null model. Combined, the 2 F8KO allele transcripts are present at ∼1/8 to 1/5 of normal levels in the F8KO strain. No normal F8 transcripts are present. In the phenotypically normal hepatocyte-KO model ∼70% of total liver gDNA is converted to the F8KO allele, indicative of very efficient hepatocyte Cre activity, yet almost exclusively normal F8 mRNA is present, with only traces of F8KO message. This is consistent with endothelial synthesis as our further results indicate. For the 3 EC-KO models, plasma F8 levels were correlated with hepatic levels of normal F8 mRNA, and inversely correlated with F8KO transcripts. Excessive F8F to F8KOconversion in the hematopoietic-Cre model suggests variable loss of tissue-specificity. In the most efficient, functionally hemophilic EC-KO model, ∼20% of liver gDNA is converted to the F8KO allele, in good agreement with the expected number of hepatic EC, and F8KO mRNA is present at ∼10% of normal liver levels. With undetectable plasma F8, the continued production of normal F8 mRNA at a similar low level (∼10%) by the remaining 80% of Cre-negative, presumably non-endothelial hepatic cells, was unexpected. In addition to liver we found both normal and F8KO message only in kidney and perhaps brain. As expected, only F8KOmRNA was found in spleen and bone marrow, but the presence of exclusively normal mRNA in heart, intestine, testis, lung, and thymus, at relatively normal (low) levels, was surprising. The persistence of widespread transcriptional “expression” of F8, albeit in a functionally hemophilic mouse, is reminiscent of the near-ubiquitous presence of low level F8 transcription in normal mice. This low level transcription apparently does not support functional plasma F8 production, at least not in these EC-KO mice. In summary, our results support the hypothesis that synthesis of F8 is a function of endothelial cells, both in the liver and presumably elsewhere. Neither hepatocytes nor hematopoietic cells appear to contribute significantly to steady-state plasma F8 levels. Transcriptional analysis of normal and F8KO-specific transcripts provides further support for the localization of F8 expression to endothelial cells. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 26-26
Author(s):  
Lesley Everett ◽  
Audrey C.A. Cleuren ◽  
Rami Khoriaty ◽  
David Ginsburg

Abstract Combined deficiency of coagulation factors V and VIII (F5F8D) is an autosomal recessive bleeding disorder resulting from mutations in Lman1. This gene encodes a cargo receptor in the early secretory pathway that is responsible for the efficient secretion of factor V (FV) and factor VIII (FVIII) to the plasma. F5F8D is characterized by levels of both FV and FVIII reduced to ∼5-30% of normal. In contrast, Lman1 knockout mouse models of F5F8D exhibit FV and FVIII activities that are ∼50% of normal, relative to wildtype mice. Though FV and FVIII are synthesized at markedly different levels and potentially in different tissues, loss of the LMAN1 cargo receptor leads to parallel reductions in both FV and FVIII activity. FV is synthesized in hepatocytes (as well as megakaryocytes in the mouse). However, the primary cellular source of FVIII biosynthesis is controversial, with contradictory evidence supporting an endothelial or hepatocyte origin. We took advantage of the dependence of efficient FV and FVIII secretion on LMAN1 to examine the cellular source of each protein. FV and FVIII secretion profiles of conditional Lman1 knockout mice were characterized, relative to that of wildtype mice and ubiquitous Lman1 null mice (Lman1-/-). In order to generate mice with Lman1 expression specifically deleted in the endothelium or the hepatocytes, either a Tie2-Cre or Albumin-Cre transgene was crossed into Lman1 conditional mice (Lman1fl). FV and FVIII activity levels were measured by functional coagulation activity assays. Though Lman1fl/fl/Tie2-Cre+ mice (endothelial-specific knockout) exhibit normal plasma FV activity (99.9%) relative to wildtype mice (set to 100%), FVIII activity is reduced to 53.5% (p < 2.5 x 10-6). In contrast, Lman1fl/fl/Alb-Cre+ (hepatocyte-specific knockout) mice demonstrate normal FVIII activity (89.0%) and reduced FV activity (37.0%) (p < 1.4 x 10-10). To confirm endothelial cells as the biosynthetic source of FVIII, we took advantage of the previously reported RiboTag mouse (Sanz et al., 2009. PNAS 106(33):13939-44) to isolate endothelial cell RNA for qPCR analysis from various murine tissues. RiboTag mice carry a hemaglutinin-tagged ribosomal protein that can be used for cell-type specific immunoprecipitation of polyribosomes and subsequent RNA analysis when crossed with a Cre-recombinase expressing animal. qPCR analyses of endothelial cell RNA isolated from total liver lysates of five RiboTag/Tie2-Cre+ mice demonstrated 10-20 fold enrichment for gene transcripts that are known to be endothelial-specific, such as Cdhs (12.1 fold enrichment, p < 8.0 x 10-3), Vcam1 (13.4 fold enrichment, p <1.1 x 10-5), and Vwf (15.3 fold enrichment, p < 7.0 x 10-4), as well as for FVIII transcripts (11.4 fold enrichment, p < 4.0 x 10-5). In contrast, this analysis demonstrated a statistically significant depletion (5-10 fold) of transcripts from many known hepatocyte-specific genes, including multiple coagulation factor genes. Similar examination of kidney endothelial cell RNA also demonstrated enrichment for FVIII transcripts, thereby demonstrating that endothelial cells from multiple tissues and vascular beds contribute to the plasma FVIII pool in the mouse. These results explain the successful reversal of hemophilia A by both liver and kidney transplants. Taken together, these results definitively demonstrate that endothelial cells are the primary source of FVIII biosynthesis in the mouse, and that hepatocytes make no significant contribution to the plasma FVIII pool. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5015-5015
Author(s):  
Jaewoo Song ◽  
Juwon Kim ◽  
Jungwoo Han ◽  
Jin Seok Kim ◽  
Jihye Ha

Abstract Background: The needs for sensitive coagulation factor assays able to measure factor VIII (FVIII) and factor IX (FIX) in the range of 0.0 to 1.0 %, are continuously growing with diversification of hemophilia management. However, practical methods with sufficient analytical sensitivity available in clinical laboratory have not yet been introduced. We developed new coagulation factor assays applying various parameters derived from a turbidity based coagulometer and examined their ability to measure low-level FVIII and FIX and analytical resolution in that range. Method: We prepared 12 spiked samples with FVIII and FIX levels from 0.0 to 2.4 % and conducted conventional one-stage coagulation factor assays in repeat. We collected measured values of APTT, velocity and acceleration peaks of coagulation (peak 1 and peak 2) from each measurement. We also calculated values of peak 1 and peak 2 from the mathematical model of turbidity curves. From the measured values of these parameters we derived calibration formulae for coagulation factor assays, FVIIICT, FVIIIpeak1, FVIIIpeak2, FVIIIcalc1, FVIIIcalc2, FIXCT, FIXpeak1, FIXpeak2, FIXcalc1, and FIXcalc2. Results: The reliability interval (range of FVIII levels producing unequivocal results) of FVIIICT (the conventional FVIII assay) covered only 9 % of 0.0 to 1.0 % range. For new assays, the coverages were 54, 31, 55, and 65 % for FVIIIpeak1, FVIIIpeak2, FVIIIcalc1, and FVIIIcalc2 respectively. The resolution between immediate levels of spiked samples could be determined from modeled distributions or be checked simply by inspecting the actual assay result distributions. For FVIIIpeak1, 0.2 % and 0.6 % results stood apart from each other. For FVIIIcalc1 and FVIIIcalc2, 0.2, 0.4, and 0.6 % were distinguished from each other. When we measured recombinant human (rh) FVIII, the coverages were 7, 64, 52, 73, and 79 % for rhFVIIICT, rhFVIIIpeak1, rhFVIIIpeak2, rhFVIIIcalc1, and rhFVIIIcalc2 respectively. (rh)FVIIIpeak1, (rh)FVIIIcalc1, and (rh)FVIIIcalc2 particularly showed wide measurable ranges of guarantee. For FVIIIpeak1, 0.2 % and 0.6 % results stood apart from each other. For FVIIIcalc1 and FVIIIcalc2, 0.2, 0.4, and 0.6 % were distinguished from each other. rhFVIIIpeak1 and rhFVIIIcalc1 showed slightly better resolution than the former. rhFVIIIcalc2 was notable in that every 0.1, 0.2, 0.4, 0.6, 0.8 % result stood apart from each immediate level result. We could not determine certainty interval (the range of unequivocal values) of FIXCT and FIXpeak2 because the 0.0 % and 1.0 % ranges overlapped. Thus, the conventional FIX assay cannot measure between 0.0 and 1.0 %. FIXpeak1, FIXcalc1, and FIXcalc2 worked better and the certainty interval of unequivocal results could be determined between 0.0 and 1.0 %. The reliability interval was not available for any FIX assay. Results from rhFIX measurements were similar those of plasma FIX assays. Conclusion: We introduce new FVIII and FIX assays with superior analytical resolution in the range of 0.0 to 1.0 % in comparison to the conventional assays. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Morisada Hayakawa ◽  
Asuka Sakata ◽  
Hiroko Hayakawa ◽  
Hikari Matsumoto ◽  
Takafumi Hiramoto ◽  
...  

AbstractCoagulation factors are produced from hepatocytes, whereas production of coagulation factor VIII (FVIII) from primary tissues and cell species is still controversial. Here, we tried to characterize primary FVIII-producing organ and cell species using genetically engineered mice, in which enhanced green fluorescent protein (EGFP) was expressed instead of the F8 gene. EGFP-positive FVIII-producing cells existed only in thin sinusoidal layer of the liver and characterized as CD31high, CD146high, and lymphatic vascular endothelial hyaluronan receptor 1 (Lyve1)+. EGFP-positive cells can be clearly distinguished from lymphatic endothelial cells in the expression profile of the podoplanin− and C-type lectin-like receptor-2 (CLEC-2)+. In embryogenesis, EGFP-positive cells began to emerge at E14.5 and subsequently increased according to liver maturation. Furthermore, plasma FVIII could be abolished by crossing F8 conditional deficient mice with Lyve1-Cre mice. In conclusion, in mice, FVIII is only produced from endothelial cells exhibiting CD31high, CD146high, Lyve1+, CLEC-2+, and podoplanin− in liver sinusoidal endothelial cells.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3787-3787
Author(s):  
Pete Lollar ◽  
Ernest T. Parker ◽  
John F. Healey ◽  
Christopher B. Doering

Abstract Inhibitory polyclonal IgG antibodies (inhibitors) to factor VIII (fVIII) represent the most significant complication in patients with congenital hemophilia A. FVIII also is the most frequently targeted coagulation factor in autoimmunity. Antibodies recognizing epitopes in the fVIII A2 and C2 domains are present in most inhibitor patients. In the current study, we characterized the hydrodynamic properties of fVIII immune complexes formed by murine anti-human anti-A2 and anti-C2 fVIII monoclonal antibodies (MAbs) 4A4 and 3D12. 4A4 is representative of the most frequently identified group of anti-A2 MAbs identified in the murine hemophilia A immune response to human fVIII. 3D12 is a classical anti-C2 MAb that inhibits the binding of fVIII to von Willebrand factor (VWF) and phospholipid membranes. Velocity sedimentation of immune complexes formed by varying ratios of 4A4 and 3D12 with a high-expression fVIII construct designated ET3 was conducted at 55,000g and 20 °C by measuring protein absorbance at 280 nm in a Beckman XL-I analytical ultracentrifuge. Sedimentation coefficient (s20,w) distributions of fVIII, MAbs and immune complexes were determined using SEDFIT. The sedimentation coefficients of fVIII in the absence of MAbs and of the MAbs in the absence of fVIII were 7.7 S and 6.4 S, respectively. Under conditions of excess MAb (equimolar 4A4 and 3D12 each in five-fold molar excess over fVIII), a 10.3 S immune complex was observed, representing singly-ligated MAbs (Figure, red trace). Under conditions of excess fVIII (fVIII in four-fold molar excess over equimolar 4A4 and 3D12), 11.9 S doubly-ligated MAb complexes were observed (Figure, green trace). A mixture containing equimolar fVIII and 4A4/3D12 MAb binding sites produced a dominant 14.0 S species and a minor 18.8 S species, indicative of cross-linked 3D12-fVIII-4A4 immune complexes (Figure, blue trace). Indefinite association or immunoprecipitation was not observed. These results demonstrate that a biclonal, bivalent anti-fVIII antibody population can form higher-order immune complexes. These complexes may be a driving factor in the immune response to fVIII by promoting B cell activation and/or antigen presentation. Additionally, these results indicate that analytical ultracentrifugation is a useful tool to characterize fVIII immune complexes. Figure Figure. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 427-427
Author(s):  
Emily Bowers ◽  
Slaughter Anastasiya ◽  
Daniel Lucas-Alcaraz

Abstract In addition to eliminating host hematopoietic cells myeloablation also disrupts the blood vessels that sustain hematopoiesis. Regeneration of the bone marrow (BM) vasculature is necessary for hematopoietic recovery and survival after transplantation (Cell Stem Cell. 2009 Mar 6;4(3):263-74) but the mechanisms that drive vascular regeneration are not clear. We found that, fourteen days after lethal irradiation and transplantation, mice transplanted with 20x106 bone marrow nucleated cells (BMNC) had ~6-fold more CD45-Ter119-CD31+CD105+ endothelial cells (6.9x103 vs 0.96x103 EC/femur, p<0.001), 2-fold more blood vessels (195 vs 87 blood vessels/sternum, p<0.05) and ~2-fold less vascular leakage (4.8 vs 9.3 ng of Evans Blue/ml of BM extracellular fluid, p<0.001) than mice transplanted with 105 BMNC. Transplant experiments using GFP+donor BMNC revealed that all endothelial cells after transplantation were host derived. Because hematopoietic progenitors inhibit vascular regeneration via angiopoietin 1 (Elife2015 Mar 30;4:e05521) we hypothesized that mature hematopoietic cells mediated vascular recovery. To test this we adoptively transferred, B and T cells, monocytes and macrophages (MO), granulocytes and erythroid cells into lethally irradiated recipients previously transplanted with 105 donor BMNC. Only CD115-Gr1+ granulocytes promoted endothelial cell regeneration (2.5x103 for granulocyte treated mice vs 0.9x103 for PBS, 0.3x103 for B- and T-cell, 0.7 1x103 for MO and 0.3x103 EC/femur for erythroid cell-treated mice; p<0.01). Granulocyte transfer also promoted survival (granulocytes=100%, PBS=50% p<0.05), probably due to faster host platelets and red blood cells recovery (granulocytes= 4.5x107, PBS=2.1x107 platelets/ml of blood, p<0.001; granulocytes=4x109, PBS=6.2x109 RBC/ml of blood, p<0.01). Importantly, competitive BM transplants showed that granulocytes did not exhaust donor HSC. These demonstrate that granulocyte transfer is sufficient to promote survival and drive vascular and hematopoietic recovery after transplantation. We then generated Mrp8-cre:iDTR mice which allowed us to specifically ablate BM granulocytes via diphtheria toxin (DT) injection. We transplanted lethally irradiated WT recipients with 106 BMNC purified from C67BL/6 WT or Mrp8-cre:iDTR mice followed by DT treatment for 7 days. This led to granulocyte depletion (1.6x106 vs 0.4x106 p<0.001) and impaired endothelial cell recovery (5.7x103 vs 2.4.x103 p<0.05) in mice transplanted with Mrp8-cre:iDTR BMNC. These results demonstrate that donor granulocytes are necessary for vascular regeneration. We found that granulocytes produced high levels of the angiogenic cytokine TNFα. This cytokine signals via Tnfrsf1aand Tnfrsf1b. Tnfrsf1a was upregulated specifically in BM endothelial cells. After myeloablation with 5-fluorouracil Tnfa-/-mice have reduced survival (Tnfa-/-= 13% vs WT= 93%; p<0.001) and reduced endothelial cell numbers (WT=9x103, Tnfa-/-=4.1x103 EC/femur; p<0.05) indicating that TNFα is necessary for survival and vascular regeneration after myeloablation. To test whether granulocytes promoted vascular regeneration via TNFα we lethally irradiated and transplanted C57BL/6 recipients followed by treatment with PBS or adoptive transfer of 106 WT or Tnfa-/- granulocytes. Only WT granulocytes induced vascular recovery as demonstrated by quantification of endothelial cells (PBS=0.9 x103, WT granulocytes=5.24x103 and Tnfa-/- granulocytes=3.0x103 cells/femur, p<0.05) and blood vessel numbers (PBS=126, WT granulocytes=186 and Tnfa-/- granulocytes=84 vessels per sternum BM; p<0.05). Further, adoptive transfer of WT granulocytes promoted survival and vascular regeneration (WT+PBS=1.4x103 vs WT+granulocytes=2.6x103, p<0.05; Tnfrsf1a-/-:Tnfrsf1b-/- +PBS=0.8x103 vs Tnfrsf1a-/-:Tnfrsf1b-/-+granulocytes=0.7x103 EC/femur p=0.83) in WT but not Tnfrsf1a-/-:Tnfrsf1b-/-recipients after transplantation. These experiments demonstrate that granulocytes crosstalk directly with stromal cells (likely endothelial cells) via TNFα to drive vascular regeneration. We have identified a new type of cellular crosstalk in the microenvironment that drives regeneration. Our research also provides proof of principle for studies targeting BM granulocytes to enhance vascular recovery and survival after transplantation in patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2241-2241
Author(s):  
Eveline Bouwens ◽  
Maartje van den Biggelaar ◽  
Jan Voorberg ◽  
Koen Mertens

Abstract Abstract 2241 Recent studies have shown that factor VIII (FVIII) expressed in endothelial cells sorts with von Willebrand factor (VWF) to secretory Weibel-Palade bodies (WPBs). The sorting mechanism remains controversial although VWF is thought to be essential. However, mutations that lead to impaired FVIII-VWF complex assembly do not reduce the sorting efficiency of FVIII. As factor V (FV) and FVIII are highly homologous in structure, we addressed the possibility that FV sorts to WPBs as well. Our study was designed to identify domains in FVIII that are needed for sorting to WPBs by means of domain deletions and FVIII-FV domain exchange. As the C domains of FVIII contain membrane and VWF binding sites, we particularly focused on comparing the C domains of FVIII and FV. Blood outgrowth endothelial cells (BOECs) were transduced with lentiviral vectors encoding FV, FVIII deletion mutants, or FVIII-FV chimeras. We found by confocal microscopy and subcellular fractionations that FV displays a strong reduction in sorting efficiency (2% sorting efficiency) compared to FVIII (20% sorting efficiency). This indicates that sorting to WPBs is mediated by FVIII-specific structural elements. As the C domains of FVIII are implicated in membrane and VWF binding, these domains could drive sorting to WPBs. Therefore, we constructed FVIII variants lacking C domains to establish their role in WPB sorting. Quantitative determination of the sorting efficiencies demonstrates that the C1 domain is not of major importance for sorting to WPBs (10% sorting efficiency), whereas the C2 domain is (not detectable in WPB fractions). Moreover, exchanging the FVIII C domains for corresponding domains of FV also suggests that the C2 domain drives WPB sorting (3% sorting efficiency). This leads to the conclusion that FVIII sorting to WPBs is driven by FVIII-specific structural elements in both C domains, but in particular the C2 domain. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1223-1223
Author(s):  
Hiroshi Inaba ◽  
Keiko Shinozawa ◽  
Takeshi Hagiwara ◽  
Kagehiro Amano ◽  
Katsuyuki Fukutake

Abstract Abstract 1223 Introduction/Background: Hemophilia A is a congenital X-linked bleeding disorder caused by various mutations in the coagulation factor VIII gene (F8). However, recent studies have described that no genetic mutation could be found in the F8 of about 2% of hemophilia A patients, even after nucleotide sequencing including the entire coding region, exon/intron boundaries, and the 5'- and 3'-untranslated region (Vidal et al, 2001; Klopp et al, 2002). Factor VIII deficient mechanisms underlying this phenomenon remain unexplained. To further elucidate the mechanisms causing hemophilia A in these patients, we performed a detailed analysis of F8 mRNA. Materials and methods: F8 mRNA from a Japanese hemophilia A patient with undetectable mutations was analyzed. Total RNA was isolated from peripheral blood cells using a QIAamp® RNA Blood Mini Kit (Qiagen) or PAXgene® Blood RNA Kit (Qiagen). Both preparations were performed following the manufacturer's instructions. In order to analyze the F8 mRNA, we performed the cDNA-amplification in two rounds of PCR using the nested approach reported by El-Maarri et al (2005). The nucleotide sequences of primer used followed those of their report. OneStep RT-PCR Kit (Qiagen) and TaKaRa LA Taq ™ (TaKaRa) were used for first and second round PCR amplification, respectively. Ectopic F8 mRNA expression level was relatively quantified by a real-time PCR technique using 4 TaqMan gene expression assays (Hs00240767_m1 amplify exon 1–2 boundary, Hs01109548_m1 amplify exon 6–7 boundary, Hs01109541_m1 amplify exon 14–15 boundary, Hs01109543_m1 amplify exon 20–21 boundary; Applied Biosystems). Results: Because the size of the F8 mRNA is very large ∼9kb, the entire F8 cDNA was divided into four different regions: exons 1–8 (region A); exons 8–14 (region B); exons 14–22 (region C); and exons 19–26 (region D) and amplified in the first round. Then, each of four regions were further divided into two different regions (a total of 8 overlapping regions; region 1–8), and amplified in the second round. An abnormality was observed in the amplification. Although the PCR products of regions 1 and 2, (region A), were obtained, the products remaining in all later regions (regions 3–8) were not. A similar phenomenon was also confirmed in the semi-quantification of the mRNA. Though we were able to quantify the mRNA by using both exon 1–2 and 5–6 boundary amplifications, we were not able to quantify the mRNA using the 14–15 and 20–21 boundaries. These results suggested that the quantity of the mRNA decrease remarkably in the vicinity of exon 8 as a boundary. Further analysis of the mRNA showed that quantity of the mRNA is normal from exon 1 through 9. Nucleotide sequencing of intron 9 revealed a single nucleotide substitution, adenine to guanine transition, at 602bp downstream from the 3' end of exon 9. This transition has not been registered in any international database as a mutation or a polymorphism and was not found in the F8 from 124 Japanese. These results strongly suggest that the transition is very rare and may be involved in factor VIII deficiency in these patients. Analysis of the nucleotide sequence of the substitution by splicing site prediction software predicted the formation of a new acceptor splice site. This result suggested the existence of splice abnormality. However, further characterization is needed to elucidate the mechanism that causes the decrease in mRNA in the middle of the gene. Conclusion: The mechanism behind factor VIII deficiency in hemophilia A patients with undetectable mutations is very interesting and various possibilities are conceivable. This study provides the possibility that some causative genetic abnormality remains in a further unanalyzed F8 region, most likely deep inside the intron, of these patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 641-641
Author(s):  
Steffen Boettcher ◽  
Rahel Gerosa ◽  
Ramin Radpour ◽  
Markus G. Manz

Abstract Abstract 641 Severe systemic infections evoke a number of characteristic clinical signs such as fever, neutrophilia and the appearance of immature myeloid precursors in the circulation (left-shift). This reflects a well-regulated hematopoietic response program to enhance myeloid cell output during times of increased hematopoietic demand, a condition which is referred to as 'emergency myelopoiesis'. Important molecular components of the emergency myelopoiesis cascade, such as cytokines and transcription factors involved, have been elucidated. However, the initial steps of emergency myelopoiesis involving pathogen recognition and translation into accelerated bone marrow (BM) myelopoiesis have only been inferred from findings on Toll-like receptor (TLR) expression on immature hematopoietic stem and progenitor cells (HSPCs) as well as on mature hematopoietic cells (e.g. macrophages). Accordingly, it has been assumed that both immature as well as mature hematopoietic cells are involved in sensing infection and inducing emergency myelopoiesis directly and indirectly, respectively. Surprisingly, by generating reciprocal BM chimeric animals mice with TLR4−/− hematopoiesis on a wild-type (WT) nonhematopoietic background (TLR4−/−→WT mice) and WT hematopoiesis on a TLR4−/− nonhematopoietic background (WT→TLR4−/−mice), we demonstrated that LPS-Induced emergency myelopoiesis depends on TLR4-expressing nonhematopoietic cells (Boettcher et al., J Immunol. 2012 Jun 15;188(12):5824–8.). However, the precise identity and localization of the nonhematopoietic cell type crucial for sensing gramnegative infection-derived lipopolysaccharide (LPS) has remained elusive to date. We now have addressed this fundamental question using BM transplantation experiments and Cre-loxP recombination technology. BM chimeric mice with a myeloid differentiation primary response gene 88 (Myd88)-deficiency in the hematopoietic lineage (MYD88−/−→WT mice) showed a normal LPS response indistinguishable to control (WT→WT) mice, while knocked out Myd88 within the nonhematopoietic compartment (WT→MYD88−/− mice) led to a non-responsiveness towards LPS similar to controls (Myd88−/−→Myd88−/− mice). These results are in line with our earlier data, thus confirming the critical role of the TLR4/MYD88 pathway in nonhematopoietic cells for the induction of emergency myelopoiesis. In order to specifically delete TLR-MyYD88-downstream signaling in various nonhematopoietic cells including BM Nestin+ mesenchymal stem cells (MSCs) and their progeny, perivascular cells, endothelial cells, and hepatocytes, we generated Nes-Cre;Myd88fl/fl, Pdgfrb-Cre;Myd88fl/fl, Tek-Cre;Myd88fl/fl, and Alb-Cre;Myd88fl/fl mice, respectively. We observed a normal increase in the frequency of BM CD11b+Gr-1low immature myeloid precursors accompanied by a decrease of BM CD11b+Gr-1high mature myeloid cells upon LPS stimulation characteristic for efficient emergency myelopoiesis in Nes-Cre;Myd88fl/fl, Pdgfrb-Cre;Myd88fl/fl, and Alb-Cre;Myd88fl/fl mice as compared to control mice. Furthermore, we measured highly-elevated plasma G-CSF levels in these mouse strains upon LPS injection. Hence, intact TLR signaling in mesenchymal stromal cells incl. Nestin+ MSCs, perivascular cells as well as hepatocytes is dispensable for induction of emergency myelopoiesis. Strikingly, Tek-Cre;Myd88fl/fl mice were completely non-responsive towards LPS stimulation as assessed by the above-mentioned parameters. Our results thus demonstrate a fundamental and unanticipated role of the endothelium for sensing of systemically spread pathogens and subsequent stimulation of BM emergency myelopoiesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1474-1474 ◽  
Author(s):  
Michail Zaboikin ◽  
Tatiana Zaboikina ◽  
Carl E. Freter ◽  
Narasimhachar Srinivasakumar

Abstract Gene and cell therapy for hemophilia A requires the use of the appropriate target cell for genetic modification and, given the advances in genome editing, an approach that can be applied universally for the wide variety of genetic mutations in the coagulation factor VIII gene (F8) responsible for hemophilia A. Recent studies using two different conditional knockout mouse models showed that the principal, and possibly exclusive, source for FVIII in the circulation are endothelial cells (Everett LA et al. Blood 2014.123: 3697; Fahs SA et al. Blood 2014.123: 3706). Since endothelial cells are present in all the major organs previously thought to produce coagulation factor VIII (FVIII), these studies provide the basis for the earlier reports indicating different tissues (liver, lung, spleen, lymphatic tissue) as sources of FVIII. ECs conveniently express von Willebrand factor (vWF) that is essential for the stability of FVIII. The precursor of ECs, endothelial progenitor cells (EPCs), have been isolated from adult human peripheral blood and cord blood. EPCs can readily integrate into existing vascular system upon intravenous injection. EPCs are quite rare in peripheral blood (about 20 colony forming cells per 100 mL of blood). Moreover, EPCs derived from adult peripheral blood have lower proliferative potential than those obtained from cord blood (Ingram DA. Blood. 2004. 104: 2752). In contrast, induced pluripotent stem cells (iPSCs), that are more amenable for expansion, can be readily differentiated into EPCs. Studies have also shown that iPSC-derived EPCs when injected intrahepatically in mice integrated into liver sinusoids, resulted in therapeutic levels of FVIII production (Xu D et al. PNAS 2009. 106: 808). Here we describe an optimized in vitro differentiation protocol for derivation of EPCs from iPSCs. We have previously reported the generation and characterization of human iPSCs from lung fibroblasts (Srinivasakumar et al. PeerJ. 2013;1:e224). In this study we used human iPSCs generated from adult dermal fibroblasts using Yamanaka's non-integrating Epstein-Barr based episomal vectors. We used a step-wise differentiation protocol for obtaining EPCs that was a combination of a method for differentiation of iPSCs into hematopoietic progenitors (Fig A, Steps 1 & 2) to generate hemangioblasts (Niwa A et al. PLoS ONE 6(7): e22261), and a protocol for obtaining EPCs from peripheral blood (Step 3) (Mead LE et al. Current Protocols in Stem Cell Biol. 2C.1.1-2C.1.27). A sorting step after differentiation into hemangioblasts followed by a final round of sorting after step 3 yielded >90% pure population of EPC that exhibited the canonical cell surface markers: CD31 and CD34, and absence of CD45 (Fig B). The cells also took up fluorophore-conjugated acetylated LDL (acLDL-A488) that was inhibited with 50x excess of unlabeled acLDL (Fig C). Immunofluorescence staining for vWF revealed characteristic staining reminiscent of Weibel-Palade bodies in the cytoplasm (Fig D). The cells exhibited the typical tube formation ability in Matrigel (Fig E). Additional studies are needed to determine the proliferative potential of these cells and their ability to integrate into vasculature. To address the myriad mutations shown to be responsible for hemophilia A, we have designed high efficiency dimeric guide RNAs (as part of a separate study) (Zaboikin M et al. Manuscript in preparation) for use with the CRISPR/dCas9-Fok1 system (Tsai SQ et al. Nat Biotechnol. 2014. 32:569) for precise modification at the F8 locus downstream of the first coding exon. We also showed in that study the replacement of target sequence at the site with that of a donor template sequence with desired attributes. We hypothesize that using a donor template that encodes the F8 promoter driving a functional F8 cDNA for homology directed repair at the target double stranded break site will provide an universal solution for the large variety of mutations observed in hemophilia A. Results of genome editing of iPSCs using the above mentioned CRISPR/dCas9-Fok1 system (together with the donor template) followed by the differentiation of genetically modified iPSCs into EPCs will be presented. Figure Figure. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document