270 results on '"P. Alvin"'
Search Results
2. Methylation Biology of a Blood-Based MDS Risk Stratification Test
- Author
-
Shi, Alvin, Ali, Abdullah Mahmood, Cann, Gordon, Shaknovich, Rita, Venn, Oliver, Raza, Azra, and Liu, Qinwen
- Abstract
Background:Myelodysplastic syndromes (MDS) are a heterogeneous group of myeloid malignancies associated with a myriad of deleterious outcomes and a 5-year relative survival of 37%. MDS risk stratification is key for optimal treatment decisions. We previously demonstrated that methylation-based markers may help stratify MDS risk and achieved comparable performance relative to IPSS-R (the Revised International Prognostic Scoring System) by assessing the risk prediction ability of a classifier trained with features from either Bone Marrow (BM) whole-genome bisulfite sequencing (WGBS) or serum targeted methylation (TM) sequencing. Here, we elucidate the biological underpinnings of the methylation-based classifier and explore the survival stratification performance of our previously reported classifier.
- Published
- 2023
- Full Text
- View/download PDF
3. Serpin targets in hemostasis/kinin formation
- Author
-
Schmaier, Alvin H.
- Published
- 2019
- Full Text
- View/download PDF
4. Serpin targets in hemostasis/kinin formation
- Author
-
Schmaier, Alvin H.
- Published
- 2019
- Full Text
- View/download PDF
5. SRSF2P95Hand FLT3ITDSynergistically Drives Leukemic Transformation in Zebrafish
- Author
-
Tu, Lingge, Chun-fung, SIN, Chen, Yanan, He, Fangfang, Wang, Dandan, Chan, Hoi Yi, Tsui, Sze Pui, Ma, Alvin Chun Hang, Sun, Xuan, and Leung, Anskar
- Published
- 2022
- Full Text
- View/download PDF
6. Pathogenetic Role of RNA Splicing and NRASMutation Combination in Chronic Myelomonocytic Leukemia
- Author
-
Chen, Yanan, Chun-Fung, Sin, Tu, Lingge, Chan, Hoi Yi, He, Fangfang, Wang, Dandan, Tsui, Sze Pui, Ma, Alvin Chun Hang, Sun, Xuan, and Leung, Anskar
- Published
- 2022
- Full Text
- View/download PDF
7. Pathogenetic Role of RNA Splicing and NRAS Mutation Combination in Chronic Myelomonocytic Leukemia
- Author
-
Chen, Yanan, Chun-Fung, Sin, Tu, Lingge, Chan, Hoi Yi, He, Fangfang, Wang, Dandan, Tsui, Sze Pui, Ma, Alvin Chun Hang, Sun, Xuan, and Leung, Anskar
- Published
- 2022
- Full Text
- View/download PDF
8. SRSF2P95H and FLT3ITD Synergistically Drives Leukemic Transformation in Zebrafish
- Author
-
Tu, Lingge, Chun-fung, SIN, Chen, Yanan, He, Fangfang, Wang, Dandan, Chan, Hoi Yi, Tsui, Sze Pui, Ma, Alvin Chun Hang, Sun, Xuan, and Leung, Anskar
- Published
- 2022
- Full Text
- View/download PDF
9. Functions of idh1 and its mutation in the regulation of developmental hematopoiesis in zebrafish
- Author
-
Shi, Xiangguo, He, Bai-Liang, Ma, Alvin C. H., Guo, Yuhan, Chi, Yali, Man, Cheuk Him, Zhang, Wenqing, Zhang, Yiyue, Wen, Zilong, Cheng, Tao, and Leung, Anskar Y. H.
- Abstract
Isocitrate dehydrogenase 1 mutation (IDH1-R132H) was recently identified in acute myeloid leukemia with normal cytogenetics. The mutant enzyme is thought to convert α-ketoglutarate to the pathogenic 2-hydroxyglutarate (2-HG) that affects DNA methylation via inhibition of ten-eleven translocation 2. However, the role of wild-type IDH1 in normal hematopoiesis and its relevance to acute myeloid leukemia is unknown. Here we showed that zebrafish idh1 (zidh1) knockdown by morpholino and targeted mutagenesis by transcription activator–like effector nuclease might induce blockade in myeloid differentiation, as evident by an increase in pu.1 and decrease in mpo, l-plastin, and mpeg1 expression, and significantly reduce definitive hematopoiesis. Morpholino knockdown of zidh2 also induced a blockade in myeloid differentiation but definitive hematopoiesis was not affected. The hematopoietic phenotype of zidh1 knockdown was not rescuable by zidh2 messenger RNA, suggesting nonredundant functions. Overexpression of human IDH1-R132H or its zebrafish ortholog resulted in 2-HG elevation and expansion of myelopoiesis in zebrafish embryos. A human IDH1-R132H–specific inhibitor (AGI-5198) significantly ameliorated both hematopoietic and 2-HG responses in human but not zebrafish IDH1 mutant expression. The results provided important insights to the role of zidh1 in myelopoiesis and definitive hematopoiesis and of IDH1-R132H in leukemogenesis.
- Published
- 2015
- Full Text
- View/download PDF
10. Functions of idh1 and its mutation in the regulation of developmental hematopoiesis in zebrafish
- Author
-
Shi, Xiangguo, He, Bai-Liang, Ma, Alvin C.H., Guo, Yuhan, Chi, Yali, Man, Cheuk Him, Zhang, Wenqing, Zhang, Yiyue, Wen, Zilong, Cheng, Tao, and Leung, Anskar Y.H.
- Abstract
Isocitrate dehydrogenase 1 mutation (IDH1-R132H) was recently identified in acute myeloid leukemia with normal cytogenetics. The mutant enzyme is thought to convert α-ketoglutarate to the pathogenic 2-hydroxyglutarate (2-HG) that affects DNA methylation via inhibition of ten-eleven translocation 2. However, the role of wild-type IDH1 in normal hematopoiesis and its relevance to acute myeloid leukemia is unknown. Here we showed that zebrafish idh1(zidh1) knockdown by morpholino and targeted mutagenesis by transcription activator–like effector nuclease might induce blockade in myeloid differentiation, as evident by an increase in pu.1and decrease in mpo, l-plastin, and mpeg1expression, and significantly reduce definitive hematopoiesis. Morpholino knockdown of zidh2also induced a blockade in myeloid differentiation but definitive hematopoiesis was not affected. The hematopoietic phenotype of zidh1knockdown was not rescuable by zidh2messenger RNA, suggesting nonredundant functions. Overexpression of human IDH1-R132H or its zebrafish ortholog resulted in 2-HG elevation and expansion of myelopoiesis in zebrafish embryos. A human IDH1-R132H–specific inhibitor (AGI-5198) significantly ameliorated both hematopoietic and 2-HG responses in human but not zebrafish IDH1 mutant expression. The results provided important insights to the role of zidh1 in myelopoiesis and definitive hematopoiesis and of IDH1-R132H in leukemogenesis.
- Published
- 2015
- Full Text
- View/download PDF
11. Reduced thrombosis in Klkb1−/− mice is mediated by increased Mas receptor, prostacyclin, Sirt1, and KLF4 and decreased tissue factor
- Author
-
Stavrou, Evi X., Fang, Chao, Merkulova, Alona, Alhalabi, Omar, Grobe, Nadja, Antoniak, Silvio, Mackman, Nigel, and Schmaier, Alvin H.
- Abstract
The precise mechanism for reduced thrombosis in prekallikrein null mice (Klkb1−/−) is unknown. Klkb1−/− mice have delayed carotid artery occlusion times on the rose bengal and ferric chloride thrombosis models. Klkb1−/− plasmas have long-activated partial thromboplastin times and defective contact activation–induced thrombin generation that partially corrects upon prolonged incubation. However, in contact activation–induced pulmonary thromboembolism by collagen/epinephrine or long-chain polyphosphate, Klkb1−/− mice, unlike F12−/− mice, do not have survival advantage. Klkb1−/− mice have reduced plasma BK levels and renal B2R mRNA. They also have increased expression of the renal receptor Mas and plasma prostacyclin. Increased prostacyclin is associated with elevated aortic vasculoprotective transcription factors Sirt1 and KLF4. Treatment of Klkb1−/− mice with the Mas antagonist A-779, COX-2 inhibitor nimesulide, or Sirt1 inhibitor splitomicin lowers plasma prostacyclin and normalizes arterial thrombosis times. Treatment of normal mice with the Mas agonist AVE0991 reduces thrombosis. Klkb1−/− mice have reduced aortic tissue factor (TF) mRNA, antigen, and activity. In sum, Klkb1−/− mice have a novel mechanism for thrombosis protection in addition to reduced contact activation. This pathway arises when bradykinin delivery to vasculature is compromised and mediated by increased receptor Mas, prostacyclin, Sirt1, and KLF4, leading to reduced vascular TF.
- Published
- 2015
- Full Text
- View/download PDF
12. Reduced thrombosis in Klkb1−/−mice is mediated by increased Mas receptor, prostacyclin, Sirt1, and KLF4 and decreased tissue factor
- Author
-
Stavrou, Evi X., Fang, Chao, Merkulova, Alona, Alhalabi, Omar, Grobe, Nadja, Antoniak, Silvio, Mackman, Nigel, and Schmaier, Alvin H.
- Abstract
The precise mechanism for reduced thrombosis in prekallikrein null mice (Klkb1−/−) is unknown. Klkb1−/−mice have delayed carotid artery occlusion times on the rose bengal and ferric chloride thrombosis models. Klkb1−/−plasmas have long-activated partial thromboplastin times and defective contact activation–induced thrombin generation that partially corrects upon prolonged incubation. However, in contact activation–induced pulmonary thromboembolism by collagen/epinephrine or long-chain polyphosphate, Klkb1−/−mice, unlike F12−/−mice, do not have survival advantage. Klkb1−/−mice have reduced plasma BK levels and renal B2R mRNA. They also have increased expression of the renal receptor Mas and plasma prostacyclin. Increased prostacyclin is associated with elevated aortic vasculoprotective transcription factors Sirt1 and KLF4. Treatment of Klkb1−/−mice with the Mas antagonist A-779, COX-2 inhibitor nimesulide, or Sirt1 inhibitor splitomicin lowers plasma prostacyclin and normalizes arterial thrombosis times. Treatment of normal mice with the Mas agonist AVE0991 reduces thrombosis. Klkb1−/−mice have reduced aortic tissue factor (TF) mRNA, antigen, and activity. In sum, Klkb1−/−mice have a novel mechanism for thrombosis protection in addition to reduced contact activation. This pathway arises when bradykinin delivery to vasculature is compromised and mediated by increased receptor Mas, prostacyclin, Sirt1, and KLF4, leading to reduced vascular TF.
- Published
- 2015
- Full Text
- View/download PDF
13. The thromboprotective effect of bortezomib is dependent on the transcription factor Kruppel-like factor 2 (KLF2)
- Author
-
Nayak, Lalitha, Shi, Hong, Atkins, G. Brandon, Lin, Zhiyong, Schmaier, Alvin H., and Jain, Mukesh K.
- Abstract
Multiple myeloma confers a high risk for vascular thrombosis, a risk that is increased by treatment with immunomodulatory agents. Strikingly, inclusion of the proteasome inhibitor bortezomib reduces thrombotic risk, yet the molecular basis for this observation remains unknown. Here, we show that bortezomib prolongs thrombosis times in the carotid artery photochemical injury assay in normal mice. Cell-based studies show that bortezomib increases expression of the transcription factor Kruppel-like factor 2 (KLF2) in multiple cell types. Global postnatal overexpression of KLF2 (GL-K2-TG) increased time to thrombosis, and global postnatal deletion of KLF2 (GL-K2-KO) conferred an antiparallel effect. Finally, studies in GL-K2-KO mice showed that the thromboprotective effect of bortezomib is KLF2 dependent. These findings identify a transcriptional basis for the antithrombotic effects of bortezomib.
- Published
- 2014
- Full Text
- View/download PDF
14. The thromboprotective effect of bortezomib is dependent on the transcription factor Kruppel-like factor 2 (KLF2)
- Author
-
Nayak, Lalitha, Shi, Hong, Atkins, G. Brandon, Lin, Zhiyong, Schmaier, Alvin H., and Jain, Mukesh K.
- Abstract
Multiple myeloma confers a high risk for vascular thrombosis, a risk that is increased by treatment with immunomodulatory agents. Strikingly, inclusion of the proteasome inhibitor bortezomib reduces thrombotic risk, yet the molecular basis for this observation remains unknown. Here, we show that bortezomib prolongs thrombosis times in the carotid artery photochemical injury assay in normal mice. Cell-based studies show that bortezomib increases expression of the transcription factor Kruppel-like factor 2 (KLF2) in multiple cell types. Global postnatal overexpression of KLF2(GL-K2-TG) increased time to thrombosis, and global postnatal deletion of KLF2(GL-K2-KO) conferred an antiparallel effect. Finally, studies in GL-K2-KO mice showed that the thromboprotective effect of bortezomib is KLF2dependent. These findings identify a transcriptional basis for the antithrombotic effects of bortezomib.
- Published
- 2014
- Full Text
- View/download PDF
15. Functions of flt3 in zebrafish hematopoiesis and its relevance to human acute myeloid leukemia
- Author
-
He, Bai-Liang, Shi, Xiangguo, Man, Cheuk Him, Ma, Alvin C. H., Ekker, Stephen C., Chow, Howard C. H., So, Chi Wai Eric, Choi, William W. L., Zhang, Wenqing, Zhang, Yiyue, and Leung, Anskar Y. H.
- Abstract
FMS-like tyrosine kinase 3 (FLT3) is expressed in human hematopoietic stem and progenitor cells (HSPCs) but its role during embryogenesis is unclear. In acute myeloid leukemia (AML), internal tandem duplication (ITD) of FLT3 at the juxtamembrane (JMD) and tyrosine kinase (TKD) domains (FLT3-ITD+) occurs in 30% of patients and is associated with inferior clinical prognosis. TKD mutations (FLT3-TKD+) occur in 5% of cases. We made use of zebrafish to examine the role of flt3 in developmental hematopoiesis and model human FLT3-ITD+ and FLT3-TKD+ AML. Zebrafish flt3 JMD and TKD were remarkably similar to their mammalian orthologs. Morpholino knockdown significantly reduced the expression of l-plastin (pan-leukocyte), csf1r, and mpeg1 (macrophage) as well as that of c-myb (definitive HSPCs), lck, and rag1 (T-lymphocyte). Expressing human FLT3-ITD in zebrafish embryos resulted in expansion and clustering of myeloid cells (pu.1+, mpo+, and cebpα+) which were ameliorated by AC220 and associated with stat5, erk1/2, and akt phosphorylation. Human FLT3-TKD (D835Y) induced significant, albeit modest, myeloid expansion resistant to AC220. This study provides novel insight into the role of flt3 during hematopoiesis and establishes a zebrafish model of FLT3-ITD+ and FLT3-TKD+ AML that may facilitate high-throughput screening of novel and personalized agents.
- Published
- 2014
- Full Text
- View/download PDF
16. Functions of flt3 in zebrafish hematopoiesis and its relevance to human acute myeloid leukemia
- Author
-
He, Bai-Liang, Shi, Xiangguo, Man, Cheuk Him, Ma, Alvin C.H., Ekker, Stephen C., Chow, Howard C.H., So, Chi Wai Eric, Choi, William W.L., Zhang, Wenqing, Zhang, Yiyue, and Leung, Anskar Y.H.
- Abstract
FMS-like tyrosine kinase 3 (FLT3) is expressed in human hematopoietic stem and progenitor cells (HSPCs) but its role during embryogenesis is unclear. In acute myeloid leukemia (AML), internal tandem duplication (ITD) of FLT3 at the juxtamembrane (JMD) and tyrosine kinase (TKD) domains (FLT3-ITD+) occurs in 30% of patients and is associated with inferior clinical prognosis. TKD mutations (FLT3-TKD+) occur in 5% of cases. We made use of zebrafish to examine the role of flt3 in developmental hematopoiesis and model human FLT3-ITD+and FLT3-TKD+AML. Zebrafish flt3 JMD and TKD were remarkably similar to their mammalian orthologs. Morpholino knockdown significantly reduced the expression of l-plastin(pan-leukocyte), csf1r, and mpeg1(macrophage) as well as that of c-myb(definitive HSPCs), lck, and rag1(T-lymphocyte). Expressing human FLT3-ITD in zebrafish embryos resulted in expansion and clustering of myeloid cells (pu.1+, mpo+, and cebpα+) which were ameliorated by AC220 and associated with stat5, erk1/2, and akt phosphorylation. Human FLT3-TKD (D835Y) induced significant, albeit modest, myeloid expansion resistant to AC220. This study provides novel insight into the role of flt3 during hematopoiesis and establishes a zebrafish model of FLT3-ITD+and FLT3-TKD+AML that may facilitate high-throughput screening of novel and personalized agents.
- Published
- 2014
- Full Text
- View/download PDF
17. Molecular mechanisms underlying synergistic adhesion of sickle red blood cells by hypoxia and low nitric oxide bioavailability
- Author
-
Gutsaeva, Diana R., Montero-Huerta, Pedro, Parkerson, James B., Yerigenahally, Shobha D., Ikuta, Tohru, and Head, C. Alvin
- Abstract
The molecular mechanisms by which nitric oxide (NO) bioavailability modulates the clinical expression of sickle cell disease (SCD) remain elusive. We investigated the effect of hypoxia and NO bioavailability on sickle red blood cell (sRBC) adhesion using mice deficient for endothelial NO synthase (eNOS) because their NO metabolite levels are similar to those of SCD mice but without hypoxemia. Whereas sRBC adhesion to endothelial cells in eNOS-deficient mice was synergistically upregulated at the onset of hypoxia, leukocyte adhesion was unaffected. Restoring NO metabolite levels to physiological levels markedly reduced sRBC adhesion to levels seen under normoxia. These results indicate that sRBC adherence to endothelial cells increases in response to hypoxia prior to leukocyte adherence, and that low NO bioavailability synergistically upregulates sRBC adhesion under hypoxia. Although multiple adhesion molecules mediate sRBC adhesion, we found a central role for P-selectin in sRBC adhesion. Hypoxia and low NO bioavailability upregulated P-selectin expression in endothelial cells in an additive manner through p38 kinase pathways. These results demonstrate novel cellular and signaling mechanisms that regulate sRBC adhesion under hypoxia and low NO bioavailability. Importantly, these findings point us toward new molecular targets to inhibit cell adhesion in SCD.
- Published
- 2014
- Full Text
- View/download PDF
18. Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A
- Author
-
Mahlangu, Johnny, Powell, Jerry S., Ragni, Margaret V., Chowdary, Pratima, Josephson, Neil C., Pabinger, Ingrid, Hanabusa, Hideji, Gupta, Naresh, Kulkarni, Roshni, Fogarty, Patrick, Perry, David, Shapiro, Amy, Pasi, K. John, Apte, Shashikant, Nestorov, Ivan, Jiang, Haiyan, Li, Shuanglian, Neelakantan, Srividya, Cristiano, Lynda M., Goyal, Jaya, Sommer, Jurg M., Dumont, Jennifer A., Dodd, Nigel, Nugent, Karen, Vigliani, Gloria, Luk, Alvin, Brennan, Aoife, and Pierce, Glenn F.
- Abstract
This phase 3 pivotal study evaluated the safety, efficacy, and pharmacokinetics of a recombinant FVIII Fc fusion protein (rFVIIIFc) for prophylaxis, treatment of acute bleeding, and perioperative hemostatic control in 165 previously treated males aged ≥12 years with severe hemophilia A. The study had 3 treatment arms: arm 1, individualized prophylaxis (25-65 IU/kg every 3-5 days, n = 118); arm 2, weekly prophylaxis (65 IU/kg, n = 24); and arm 3, episodic treatment (10-50 IU/kg, n = 23). A subgroup compared recombinant FVIII (rFVIII) and rFVIIIFc pharmacokinetics. End points included annualized bleeding rate (ABR), inhibitor development, and adverse events. The terminal half-life of rFVIIIFc (19.0 hours) was extended 1.5-fold vs rFVIII (12.4 hours; P < .001). Median ABRs observed in arms 1, 2, and 3 were 1.6, 3.6, and 33.6, respectively. In arm 1, the median weekly dose was 77.9 IU/kg; approximately 30% of subjects achieved a 5-day dosing interval (last 3 months on study). Across arms, 87.3% of bleeding episodes resolved with 1 injection. Adverse events were consistent with those expected in this population; no subjects developed inhibitors. rFVIIIFc was well-tolerated, had a prolonged half-life compared with rFVIII, and resulted in low ABRs when dosed prophylactically 1 to 2 times per week. This trial was registered at www.clinicaltrials.gov as #NCT01181128.
- Published
- 2014
- Full Text
- View/download PDF
19. Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A
- Author
-
Mahlangu, Johnny, Powell, Jerry S., Ragni, Margaret V., Chowdary, Pratima, Josephson, Neil C., Pabinger, Ingrid, Hanabusa, Hideji, Gupta, Naresh, Kulkarni, Roshni, Fogarty, Patrick, Perry, David, Shapiro, Amy, Pasi, K. John, Apte, Shashikant, Nestorov, Ivan, Jiang, Haiyan, Li, Shuanglian, Neelakantan, Srividya, Cristiano, Lynda M., Goyal, Jaya, Sommer, Jurg M., Dumont, Jennifer A., Dodd, Nigel, Nugent, Karen, Vigliani, Gloria, Luk, Alvin, Brennan, Aoife, and Pierce, Glenn F.
- Abstract
This phase 3 pivotal study evaluated the safety, efficacy, and pharmacokinetics of a recombinant FVIII Fc fusion protein (rFVIIIFc) for prophylaxis, treatment of acute bleeding, and perioperative hemostatic control in 165 previously treated males aged ≥12 years with severe hemophilia A. The study had 3 treatment arms: arm 1, individualized prophylaxis (25-65 IU/kg every 3-5 days, n = 118); arm 2, weekly prophylaxis (65 IU/kg, n = 24); and arm 3, episodic treatment (10-50 IU/kg, n = 23). A subgroup compared recombinant FVIII (rFVIII) and rFVIIIFc pharmacokinetics. End points included annualized bleeding rate (ABR), inhibitor development, and adverse events. The terminal half-life of rFVIIIFc (19.0 hours) was extended 1.5-fold vs rFVIII (12.4 hours; P< .001). Median ABRs observed in arms 1, 2, and 3 were 1.6, 3.6, and 33.6, respectively. In arm 1, the median weekly dose was 77.9 IU/kg; approximately 30% of subjects achieved a 5-day dosing interval (last 3 months on study). Across arms, 87.3% of bleeding episodes resolved with 1 injection. Adverse events were consistent with those expected in this population; no subjects developed inhibitors. rFVIIIFc was well-tolerated, had a prolonged half-life compared with rFVIII, and resulted in low ABRs when dosed prophylactically 1 to 2 times per week. This trial was registered at www.clinicaltrials.govas #NCT01181128.
- Published
- 2014
- Full Text
- View/download PDF
20. Prolylcarboxypeptidase promotes angiogenesis and vascular repair
- Author
-
Adams, Gregory N., Stavrou, Evi X., Fang, Chao, Merkulova, Alona, Alaiti, M. Amer, Nakajima, Kohsuke, Morooka, Toshifumi, Merkulov, Sergei, LaRusch, Gretchen A., Simon, Daniel I, Jain, Mukesh K., and Schmaier, Alvin H.
- Abstract
Prolylcarboxypeptidase (PRCP) is associated with leanness, hypertension, and thrombosis. PRCP-depleted mice have injured vessels with reduced Kruppel-like factor (KLF)2, KLF4, endothelial nitric oxide synthase (eNOS), and thrombomodulin. Does PRCP influence vessel growth, angiogenesis, and injury repair? PRCP depletion reduced endothelial cell growth, whereas transfection of hPRCP cDNA enhanced cell proliferation. Transfection of hPRCP cDNA, or an active site mutant (hPRCPmut) rescued reduced cell growth after PRCP siRNA knockdown. PRCP-depleted cells migrated less on scratch assay and murine PRCPgt/gt aortic segments had reduced sprouting. Matrigel plugs in PRCPgt/gt mice had reduced hemoglobin content and angiogenic capillaries by platelet endothelial cell adhesion molecule (PECAM) and NG2 immunohistochemistry. Skin wounds on PRCPgt/gt mice had delayed closure and reepithelialization with reduced PECAM staining, but increased macrophage infiltration. After limb ischemia, PRCPgt/gt mice also had reduced reperfusion of the femoral artery and angiogenesis. On femoral artery wire injury, PRCPgt/gt mice had increased neointimal formation, CD45 staining, and Ki-67 expression. Alternatively, combined PRCPgt/gt and MRP-14−/− mice were protected from wire injury with less neointimal thickening, leukocyte infiltration, and cellular proliferation. PRCP regulates cell growth, angiogenesis, and the response to vascular injury. Combined with its known roles in blood pressure and thrombosis control, PRCP is positioned as a key regulator of vascular homeostasis.
- Published
- 2013
- Full Text
- View/download PDF
21. Prolylcarboxypeptidase promotes angiogenesis and vascular repair
- Author
-
Adams, Gregory N., Stavrou, Evi X., Fang, Chao, Merkulova, Alona, Alaiti, M. Amer, Nakajima, Kohsuke, Morooka, Toshifumi, Merkulov, Sergei, LaRusch, Gretchen A., Simon, Daniel I, Jain, Mukesh K., and Schmaier, Alvin H.
- Abstract
Prolylcarboxypeptidase (PRCP) is associated with leanness, hypertension, and thrombosis. PRCP-depleted mice have injured vessels with reduced Kruppel-like factor (KLF)2, KLF4, endothelial nitric oxide synthase (eNOS), and thrombomodulin. Does PRCP influence vessel growth, angiogenesis, and injury repair? PRCP depletion reduced endothelial cell growth, whereas transfection of hPRCPcDNA enhanced cell proliferation. Transfection of hPRCPcDNA, or an active site mutant (hPRCPmut) rescued reduced cell growth after PRCP siRNA knockdown. PRCP-depleted cells migrated less on scratch assay and murine PRCPgt/gtaortic segments had reduced sprouting. Matrigel plugs in PRCPgt/gtmice had reduced hemoglobin content and angiogenic capillaries by platelet endothelial cell adhesion molecule (PECAM) and NG2 immunohistochemistry. Skin wounds on PRCPgt/gtmice had delayed closure and reepithelialization with reduced PECAM staining, but increased macrophage infiltration. After limb ischemia, PRCPgt/gtmice also had reduced reperfusion of the femoral artery and angiogenesis. On femoral artery wire injury, PRCPgt/gtmice had increased neointimal formation, CD45 staining, and Ki-67 expression. Alternatively, combined PRCPgt/gtand MRP-14−/−mice were protected from wire injury with less neointimal thickening, leukocyte infiltration, and cellular proliferation. PRCP regulates cell growth, angiogenesis, and the response to vascular injury. Combined with its known roles in blood pressure and thrombosis control, PRCP is positioned as a key regulator of vascular homeostasis.
- Published
- 2013
- Full Text
- View/download PDF
22. Angiotensin 1-7 and Mas decrease thrombosis in Bdkrb2−/− mice by increasing NO and prostacyclin to reduce platelet spreading and glycoprotein VI activation
- Author
-
Fang, Chao, Stavrou, Evi, Schmaier, Alec A., Grobe, Nadja, Morris, Mariana, Chen, Andrew, Nieman, Marvin T., Adams, Gregory N., LaRusch, Gretchen, Zhou, Yihua, Bilodeau, Matthew L., Mahdi, Fakhri, Warnock, Mark, and Schmaier, Alvin H.
- Abstract
Bradykinin B2 receptor–deleted mice (Bdkrb2−/−) have delayed carotid artery thrombosis times and prolonged tail bleeding time resulting from elevated angiotensin II (AngII) and angiotensin receptor 2 (AT2R) producing increased plasma nitric oxide (NO) and prostacyclin. Bdkrb2−/− also have elevated plasma angiotensin-(1-7) and messenger RNA and protein for its receptor Mas. Blockade of Mas with its antagonist A-779 in Bdkrb2−/− shortens thrombosis times (58 ± 4 minutes to 38 ± 4 minutes) and bleeding times (170 ± 13 seconds to 88 ± 8 seconds) and lowers plasma nitrate (22 ± 4 μM to 15 ± 5 μM), and 6-keto-PGF1α (259 ± 103 pg/mL to 132 ± 58 pg/mL). Bdkrb2−/− platelets express increased NO, guanosine 3′,5′-cyclic monophosphate, and cyclic adenosine monophosphate with reduced spreading on collagen, collagen peptide GFOGER, or fibrinogen. In vivo A-779 or combined L-NAME and nimesulide treatment corrects it. Bdkrb2−/− platelets have reduced collagen-related peptide–induced integrin α2bβ3 activation and P-selectin expression that are partially corrected by in vivo A-779, nimesulide, or L-NAME. Bone marrow transplantations show that the platelet phenotype and thrombosis time depends on the host rather than donor bone marrow progenitors. Transplantation of wild-type bone marrow into Bdkrb2−/− hosts produces platelets with a spreading defect and delayed thrombosis times. In Bdkrb2−/−, combined AT2R and Mas overexpression produce elevated plasma prostacyclin and NO leading to acquired platelet function defects and thrombosis delay.
- Published
- 2013
- Full Text
- View/download PDF
23. Angiotensin 1-7 and Mas decrease thrombosis in Bdkrb2−/−mice by increasing NO and prostacyclin to reduce platelet spreading and glycoprotein VI activation
- Author
-
Fang, Chao, Stavrou, Evi, Schmaier, Alec A., Grobe, Nadja, Morris, Mariana, Chen, Andrew, Nieman, Marvin T., Adams, Gregory N., LaRusch, Gretchen, Zhou, Yihua, Bilodeau, Matthew L., Mahdi, Fakhri, Warnock, Mark, and Schmaier, Alvin H.
- Abstract
Bradykinin B2 receptor–deleted mice (Bdkrb2−/−) have delayed carotid artery thrombosis times and prolonged tail bleeding time resulting from elevated angiotensin II (AngII) and angiotensin receptor 2 (AT2R) producing increased plasma nitric oxide (NO) and prostacyclin. Bdkrb2−/−also have elevated plasma angiotensin-(1-7) and messenger RNA and protein for its receptor Mas. Blockade of Mas with its antagonist A-779 in Bdkrb2−/−shortens thrombosis times (58 ± 4 minutes to 38 ± 4 minutes) and bleeding times (170 ± 13 seconds to 88 ± 8 seconds) and lowers plasma nitrate (22 ± 4 μM to 15 ± 5 μM), and 6-keto-PGF1α(259 ± 103 pg/mL to 132 ± 58 pg/mL). Bdkrb2−/−platelets express increased NO, guanosine 3′,5′-cyclic monophosphate, and cyclic adenosine monophosphate with reduced spreading on collagen, collagen peptide GFOGER, or fibrinogen. In vivo A-779 or combined L-NAME and nimesulide treatment corrects it. Bdkrb2−/−platelets have reduced collagen-related peptide–induced integrin α2bβ3activation and P-selectin expression that are partially corrected by in vivo A-779, nimesulide, or L-NAME. Bone marrow transplantations show that the platelet phenotype and thrombosis time depends on the host rather than donor bone marrow progenitors. Transplantation of wild-type bone marrow into Bdkrb2−/−hosts produces platelets with a spreading defect and delayed thrombosis times. In Bdkrb2−/−, combined AT2R and Mas overexpression produce elevated plasma prostacyclin and NO leading to acquired platelet function defects and thrombosis delay.
- Published
- 2013
- Full Text
- View/download PDF
24. VEGF-A recruits a proangiogenic MMP-9–delivering neutrophil subset that induces angiogenesis in transplanted hypoxic tissue
- Author
-
Christoffersson, Gustaf, Vågesjö, Evelina, Vandooren, Jennifer, Lidén, Majken, Massena, Sara, Reinert, Rachel B., Brissova, Marcela, Powers, Alvin C., Opdenakker, Ghislain, and Phillipson, Mia
- Abstract
Recruitment and retention of leukocytes at a site of blood vessel growth are crucial for proper angiogenesis and subsequent tissue perfusion. Although critical for many aspects of regenerative medicine, the mechanisms of leukocyte recruitment to and actions at sites of angiogenesis are not fully understood. In this study, we investigated the signals attracting leukocytes to avascular transplanted pancreatic islets and leukocyte actions at the engraftment site. Expression of the angiogenic stimulus VEGF-A by mouse pancreatic islets was elevated shortly after syngeneic transplantation to muscle. High levels of leukocytes, predominantly CD11b+/Gr-1+/CXCR4hi neutrophils, were observed at the site of engraftment, whereas VEGF-A–deficient islets recruited only half of the amount of leukocytes when transplanted. Acute VEGF-A exposure of muscle increased leukocyte extravasation but not the levels of SDF-1α. VEGF-A–recruited neutrophils expressed 10 times higher amounts of MMP-9 than neutrophils recruited to an inflammatory stimulus. Revascularization of islets transplanted to MMP-9–deficient mice was impaired because blood vessels initially failed to penetrate grafts, and after 2 weeks vascularity was still disturbed. This study demonstrates that VEGF-A recruits a proangiogenic circulating subset of CD11b+/Gr-1+ neutrophils that are CXCR4hi and deliver large amounts of the effector protein MMP-9, required for islet revascularization and functional integration after transplantation.
- Published
- 2012
- Full Text
- View/download PDF
25. VEGF-A recruits a proangiogenic MMP-9–delivering neutrophil subset that induces angiogenesis in transplanted hypoxic tissue
- Author
-
Christoffersson, Gustaf, Vågesjö, Evelina, Vandooren, Jennifer, Lidén, Majken, Massena, Sara, Reinert, Rachel B., Brissova, Marcela, Powers, Alvin C., Opdenakker, Ghislain, and Phillipson, Mia
- Abstract
Recruitment and retention of leukocytes at a site of blood vessel growth are crucial for proper angiogenesis and subsequent tissue perfusion. Although critical for many aspects of regenerative medicine, the mechanisms of leukocyte recruitment to and actions at sites of angiogenesis are not fully understood. In this study, we investigated the signals attracting leukocytes to avascular transplanted pancreatic islets and leukocyte actions at the engraftment site. Expression of the angiogenic stimulus VEGF-A by mouse pancreatic islets was elevated shortly after syngeneic transplantation to muscle. High levels of leukocytes, predominantly CD11b+/Gr-1+/CXCR4hineutrophils, were observed at the site of engraftment, whereas VEGF-A–deficient islets recruited only half of the amount of leukocytes when transplanted. Acute VEGF-A exposure of muscle increased leukocyte extravasation but not the levels of SDF-1α. VEGF-A–recruited neutrophils expressed 10 times higher amounts of MMP-9 than neutrophils recruited to an inflammatory stimulus. Revascularization of islets transplanted to MMP-9–deficient mice was impaired because blood vessels initially failed to penetrate grafts, and after 2 weeks vascularity was still disturbed. This study demonstrates that VEGF-A recruits a proangiogenic circulating subset of CD11b+/Gr-1+neutrophils that are CXCR4hiand deliver large amounts of the effector protein MMP-9, required for islet revascularization and functional integration after transplantation.
- Published
- 2012
- Full Text
- View/download PDF
26. In vivo roles of factor XII
- Author
-
Renné, Thomas, Schmaier, Alvin H., Nickel, Katrin F., Blombäck, Margareta, and Maas, Coen
- Abstract
Coagulation factor XII (FXII, Hageman factor, EC = 3.4.21.38) is the zymogen of the serine protease, factor XIIa (FXIIa). FXII is converted to FXIIa through autoactivation induced by “contact” to charged surfaces. FXIIa is of crucial importance for fibrin formation in vitro, but deficiency in the protease is not associated with excessive bleeding. For decades, FXII was considered to have no function for coagulation in vivo. Our laboratory developed the first murine knockout model of FXII. Consistent with their human counterparts, FXII−/− mice have a normal hemostatic capacity. However, thrombus formation in FXII−/− mice is largely defective, and the animals are protected from experimental cerebral ischemia and pulmonary embolism. This murine model has created new interest in FXII because it raises the possibility for safe anticoagulation, which targets thrombosis without influence on hemostasis. We recently have identified platelet polyphosphate (an inorganic polymer) and mast cell heparin as in vivo FXII activators with implications on the initiation of thrombosis and edema during hypersensitivity reactions. Independent of its protease activity, FXII exerts mitogenic activity with implications for angiogenesis. The goal of this review is to summarize the in vivo functions of FXII, with special focus to its functions in thrombosis and vascular biology.
- Published
- 2012
- Full Text
- View/download PDF
27. In vivo roles of factor XII
- Author
-
Renné, Thomas, Schmaier, Alvin H., Nickel, Katrin F., Blombäck, Margareta, and Maas, Coen
- Abstract
Coagulation factor XII (FXII, Hageman factor, EC = 3.4.21.38) is the zymogen of the serine protease, factor XIIa (FXIIa). FXII is converted to FXIIa through autoactivation induced by “contact” to charged surfaces. FXIIa is of crucial importance for fibrin formation in vitro, but deficiency in the protease is not associated with excessive bleeding. For decades, FXII was considered to have no function for coagulation in vivo. Our laboratory developed the first murine knockout model of FXII. Consistent with their human counterparts, FXII−/−mice have a normal hemostatic capacity. However, thrombus formation in FXII−/−mice is largely defective, and the animals are protected from experimental cerebral ischemia and pulmonary embolism. This murine model has created new interest in FXII because it raises the possibility for safe anticoagulation, which targets thrombosis without influence on hemostasis. We recently have identified platelet polyphosphate (an inorganic polymer) and mast cell heparin as in vivo FXII activators with implications on the initiation of thrombosis and edema during hypersensitivity reactions. Independent of its protease activity, FXII exerts mitogenic activity with implications for angiogenesis. The goal of this review is to summarize the in vivo functions of FXII, with special focus to its functions in thrombosis and vascular biology.
- Published
- 2012
- Full Text
- View/download PDF
28. A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma
- Author
-
Siegel, David S., Martin, Thomas, Wang, Michael, Vij, Ravi, Jakubowiak, Andrzej J., Lonial, Sagar, Trudel, Suzanne, Kukreti, Vishal, Bahlis, Nizar, Alsina, Melissa, Chanan-Khan, Asher, Buadi, Francis, Reu, Frederic J., Somlo, George, Zonder, Jeffrey, Song, Kevin, Stewart, A. Keith, Stadtmauer, Edward, Kunkel, Lori, Wear, Sandra, Wong, Alvin F., Orlowski, Robert Z., and Jagannath, Sundar
- Abstract
Carfilzomib is a next-generation, selective proteasome inhibitor being evaluated for the treatment of relapsed and refractory multiple myeloma. In this open-label, single-arm phase 2 study (PX-171-003-A1), patients received single-agent carfilzomib 20 mg/m2 intravenously twice weekly for 3 of 4 weeks in cycle 1, then 27 mg/m2 for ≤ 12 cycles. The primary endpoint was overall response rate (≥ partial response). Secondary endpoints included clinical benefit response rate (≥ minimal response), duration of response, progression-free survival, overall survival, and safety. A total of 266 patients were evaluable for safety, 257 for efficacy; 95% were refractory to their last therapy; 80% were refractory or intolerant to both bortezomib and lenalidomide. Patients had median of 5 prior lines of therapy, including bortezomib, lenalidomide, and thalidomide. Overall response rate was 23.7% with median duration of response of 7.8 months. Median overall survival was 15.6 months. Adverse events (AEs) were manageable without cumulative toxicities. Common AEs were fatigue (49%), anemia (46%), nausea (45%), and thrombocytopenia (39%). Thirty-three patients (12.4%) experienced peripheral neuropathy, primarily grades 1 or 2. Thirty-three patients (12.4%) withdrew because of an AE. Durable responses and an acceptable tolerability profile in this heavily pretreated population demonstrate the potential of carfilzomib to offer meaningful clinical benefit. This trial was registered at www.clinicaltrials.gov as #NCT00511238.
- Published
- 2012
- Full Text
- View/download PDF
29. A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma
- Author
-
Siegel, David S., Martin, Thomas, Wang, Michael, Vij, Ravi, Jakubowiak, Andrzej J., Lonial, Sagar, Trudel, Suzanne, Kukreti, Vishal, Bahlis, Nizar, Alsina, Melissa, Chanan-Khan, Asher, Buadi, Francis, Reu, Frederic J., Somlo, George, Zonder, Jeffrey, Song, Kevin, Stewart, A. Keith, Stadtmauer, Edward, Kunkel, Lori, Wear, Sandra, Wong, Alvin F., Orlowski, Robert Z., and Jagannath, Sundar
- Abstract
Carfilzomib is a next-generation, selective proteasome inhibitor being evaluated for the treatment of relapsed and refractory multiple myeloma. In this open-label, single-arm phase 2 study (PX-171-003-A1), patients received single-agent carfilzomib 20 mg/m2intravenously twice weekly for 3 of 4 weeks in cycle 1, then 27 mg/m2for ≤ 12 cycles. The primary endpoint was overall response rate (≥ partial response). Secondary endpoints included clinical benefit response rate (≥ minimal response), duration of response, progression-free survival, overall survival, and safety. A total of 266 patients were evaluable for safety, 257 for efficacy; 95% were refractory to their last therapy; 80% were refractory or intolerant to both bortezomib and lenalidomide. Patients had median of 5 prior lines of therapy, including bortezomib, lenalidomide, and thalidomide. Overall response rate was 23.7% with median duration of response of 7.8 months. Median overall survival was 15.6 months. Adverse events (AEs) were manageable without cumulative toxicities. Common AEs were fatigue (49%), anemia (46%), nausea (45%), and thrombocytopenia (39%). Thirty-three patients (12.4%) experienced peripheral neuropathy, primarily grades 1 or 2. Thirty-three patients (12.4%) withdrew because of an AE. Durable responses and an acceptable tolerability profile in this heavily pretreated population demonstrate the potential of carfilzomib to offer meaningful clinical benefit. This trial was registered at www.clinicaltrials.govas #NCT00511238.
- Published
- 2012
- Full Text
- View/download PDF
30. An open-label, single-arm, phase 2 (PX-171-004) study of single-agent carfilzomib in bortezomib-naive patients with relapsed and/or refractory multiple myeloma
- Author
-
Vij, Ravi, Wang, Michael, Kaufman, Jonathan L., Lonial, Sagar, Jakubowiak, Andrzej J., Stewart, A. Keith, Kukreti, Vishal, Jagannath, Sundar, McDonagh, Kevin T., Alsina, Melissa, Bahlis, Nizar J., Reu, Frederic J., Gabrail, Nashat Y., Belch, Andrew, Matous, Jeffrey V., Lee, Peter, Rosen, Peter, Sebag, Michael, Vesole, David H., Kunkel, Lori A., Wear, Sandra M., Wong, Alvin F., Orlowski, Robert Z., and Siegel, David S.
- Abstract
Carfilzomib is a selective proteasome inhibitor that binds irreversibly to its target. In phase 1 studies, carfilzomib elicited promising responses and an acceptable toxicity profile in patients with relapsed and/or refractory multiple myeloma (R/R MM). In the present phase 2, multicenter, open-label study, 129 bortezomib-naive patients with R/R MM (median of 2 prior therapies) were separated into Cohort 1, scheduled to receive intravenous carfilzomib 20 mg/m2for all treatment cycles, and Cohort 2, scheduled to receive 20 mg/m2for cycle 1 and then 27 mg/m2for all subsequent cycles. The primary end point was an overall response rate (≥ partial response) of 42.4% in Cohort 1 and 52.2% in Cohort 2. The clinical benefit response (overall response rate + minimal response) was 59.3% and 64.2% in Cohorts 1 and 2, respectively. Median duration of response was 13.1 months and not reached, and median time to progression was 8.3 months and not reached, respectively. The most common treatment-emergent adverse events were fatigue (62.0%) and nausea (48.8%). Single-agent carfilzomib elicited a low incidence of peripheral neuropathy—17.1% overall (1 grade 3; no grade 4)—in these pretreated bortezomib-naive patients. The results of the present study support the use of carfilzomib in R/R MM patients. This trial is registered at www.clinicaltrials.govas NCT00530816.
- Published
- 2012
- Full Text
- View/download PDF
31. An open-label, single-arm, phase 2 (PX-171-004) study of single-agent carfilzomib in bortezomib-naive patients with relapsed and/or refractory multiple myeloma
- Author
-
Vij, Ravi, Wang, Michael, Kaufman, Jonathan L., Lonial, Sagar, Jakubowiak, Andrzej J., Stewart, A. Keith, Kukreti, Vishal, Jagannath, Sundar, McDonagh, Kevin T., Alsina, Melissa, Bahlis, Nizar J., Reu, Frederic J., Gabrail, Nashat Y., Belch, Andrew, Matous, Jeffrey V., Lee, Peter, Rosen, Peter, Sebag, Michael, Vesole, David H., Kunkel, Lori A., Wear, Sandra M., Wong, Alvin F., Orlowski, Robert Z., and Siegel, David S.
- Abstract
Carfilzomib is a selective proteasome inhibitor that binds irreversibly to its target. In phase 1 studies, carfilzomib elicited promising responses and an acceptable toxicity profile in patients with relapsed and/or refractory multiple myeloma (R/R MM). In the present phase 2, multicenter, open-label study, 129 bortezomib-naive patients with R/R MM (median of 2 prior therapies) were separated into Cohort 1, scheduled to receive intravenous carfilzomib 20 mg/m2 for all treatment cycles, and Cohort 2, scheduled to receive 20 mg/m2 for cycle 1 and then 27 mg/m2 for all subsequent cycles. The primary end point was an overall response rate (≥ partial response) of 42.4% in Cohort 1 and 52.2% in Cohort 2. The clinical benefit response (overall response rate + minimal response) was 59.3% and 64.2% in Cohorts 1 and 2, respectively. Median duration of response was 13.1 months and not reached, and median time to progression was 8.3 months and not reached, respectively. The most common treatment-emergent adverse events were fatigue (62.0%) and nausea (48.8%). Single-agent carfilzomib elicited a low incidence of peripheral neuropathy—17.1% overall (1 grade 3; no grade 4)—in these pretreated bortezomib-naive patients. The results of the present study support the use of carfilzomib in R/R MM patients. This trial is registered at www.clinicaltrials.gov as NCT00530816.
- Published
- 2012
- Full Text
- View/download PDF
32. Sorafenib treatment of FLT3-ITD+acute myeloid leukemia: favorable initial outcome and mechanisms of subsequent nonresponsiveness associated with the emergence of a D835 mutation
- Author
-
Man, Cheuk Him, Fung, Tsz Kan, Ho, Christa, Han, Heron H.C., Chow, Howard C.H., Ma, Alvin C.H., Choi, William W.L., Lok, Si, Cheung, Alice M.S., Eaves, Connie, Kwong, Yok Lam, and Leung, Anskar Y.H.
- Abstract
Internal tandem duplication (ITD) of the fms-related tyrosine kinase-3 (FLT3) gene occurs in 30% of acute myeloid leukemias (AMLs) and confers a poor prognosis. Thirteen relapsed or chemo-refractory FLT3-ITD+AML patients were treated with sorafenib (200-400 mg twice daily). Twelve patients showed clearance or near clearance of bone marrow myeloblasts after 27 (range 21-84) days with evidence of differentiation of leukemia cells. The sorafenib response was lost in most patients after 72 (range 54-287) days but the FLT3 and downstream effectors remained suppressed. Gene expression profiling showed that leukemia cells that have become sorafenib resistant expressed several genes including ALDH1A1, JAK3, and MMP15, whose functions were unknown in AML. Nonobese diabetic/severe combined immunodeficiency mice transplanted with leukemia cells from patients before and during sorafenib resistance recapitulated the clinical results. Both ITD and tyrosine kinase domain mutations at D835 were identified in leukemia initiating cells (LICs) from samples before sorafenib treatment. LICs bearing the D835 mutant have expanded during sorafenib treatment and dominated during the subsequent clinical resistance. These results suggest that sorafenib have selected more aggressive sorafenib-resistant subclones carrying both FLT3-ITD and D835 mutations, and might provide important leads to further improvement of treatment outcome with FLT3 inhibitors.
- Published
- 2012
- Full Text
- View/download PDF
33. Sorafenib treatment of FLT3-ITD+ acute myeloid leukemia: favorable initial outcome and mechanisms of subsequent nonresponsiveness associated with the emergence of a D835 mutation
- Author
-
Man, Cheuk Him, Fung, Tsz Kan, Ho, Christa, Han, Heron H. C., Chow, Howard C. H., Ma, Alvin C. H., Choi, William W. L., Lok, Si, Cheung, Alice M. S., Eaves, Connie, Kwong, Yok Lam, and Leung, Anskar Y. H.
- Abstract
Internal tandem duplication (ITD) of the fms-related tyrosine kinase-3 (FLT3) gene occurs in 30% of acute myeloid leukemias (AMLs) and confers a poor prognosis. Thirteen relapsed or chemo-refractory FLT3-ITD+ AML patients were treated with sorafenib (200-400 mg twice daily). Twelve patients showed clearance or near clearance of bone marrow myeloblasts after 27 (range 21-84) days with evidence of differentiation of leukemia cells. The sorafenib response was lost in most patients after 72 (range 54-287) days but the FLT3 and downstream effectors remained suppressed. Gene expression profiling showed that leukemia cells that have become sorafenib resistant expressed several genes including ALDH1A1, JAK3, and MMP15, whose functions were unknown in AML. Nonobese diabetic/severe combined immunodeficiency mice transplanted with leukemia cells from patients before and during sorafenib resistance recapitulated the clinical results. Both ITD and tyrosine kinase domain mutations at D835 were identified in leukemia initiating cells (LICs) from samples before sorafenib treatment. LICs bearing the D835 mutant have expanded during sorafenib treatment and dominated during the subsequent clinical resistance. These results suggest that sorafenib have selected more aggressive sorafenib-resistant subclones carrying both FLT3-ITD and D835 mutations, and might provide important leads to further improvement of treatment outcome with FLT3 inhibitors.
- Published
- 2012
- Full Text
- View/download PDF
34. Safety and prolonged activity of recombinant factor VIII Fc fusion protein in hemophilia A patients
- Author
-
Powell, Jerry S., Josephson, Neil C., Quon, Doris, Ragni, Margaret V., Cheng, Gregory, Li, Ella, Jiang, Haiyan, Li, Lian, Dumont, Jennifer A., Goyal, Jaya, Zhang, Xin, Sommer, Jurg, McCue, Justin, Barbetti, Margaret, Luk, Alvin, and Pierce, Glenn F.
- Abstract
Current factor VIII (FVIII) products display a half-life (t1/2) of ∼ 8-12 hours, requiring frequent intravenous injections for prophylaxis and treatment of patients with hemophilia A. rFVIIIFc is a recombinant fusion protein composed of a single molecule of FVIII covalently linked to the Fc domain of human IgG1 to extend circulating rFVIII t1/2. This first-in-human study in previously treated subjects with severe hemophilia A investigated safety and pharmacokinetics of rFVIIIFc. Sixteen subjects received a single dose of rFVIII at 25 or 65 IU/kg followed by an equal dose of rFVIIIFc. Most adverse events were unrelated to study drug. None of the study subjects developed anti-rFVIIIFc antibodies or inhibitors. Across dose levels, compared with rFVIII, rFVIIIFc showed 1.54- to 1.70-fold longer elimination t1/2, 1.49- to 1.56-fold lower clearance, and 1.48- to 1.56-fold higher total systemic exposure. rFVIII and rFVIIIFc had comparable dose-dependent peak plasma concentrations and recoveries. Time to 1% FVIII activity above baseline was ∼ 1.53- to 1.68-fold longer than rFVIII across dose levels. Each subject showed prolonged exposure to rFVIIIFc relative to rFVIII. Thus, rFVIIIFc may offer a viable therapeutic approach to achieve prolonged hemostatic protection and less frequent dosing in patients with hemophilia A. This trial was registered at www.clinicaltrials.gov as NCT01027377.
- Published
- 2012
- Full Text
- View/download PDF
35. Safety and prolonged activity of recombinant factor VIII Fc fusion protein in hemophilia A patients
- Author
-
Powell, Jerry S., Josephson, Neil C., Quon, Doris, Ragni, Margaret V., Cheng, Gregory, Li, Ella, Jiang, Haiyan, Li, Lian, Dumont, Jennifer A., Goyal, Jaya, Zhang, Xin, Sommer, Jurg, McCue, Justin, Barbetti, Margaret, Luk, Alvin, and Pierce, Glenn F.
- Abstract
Current factor VIII (FVIII) products display a half-life (t1/2) of ∼ 8-12 hours, requiring frequent intravenous injections for prophylaxis and treatment of patients with hemophilia A. rFVIIIFc is a recombinant fusion protein composed of a single molecule of FVIII covalently linked to the Fc domain of human IgG1to extend circulating rFVIII t1/2. This first-in-human study in previously treated subjects with severe hemophilia A investigated safety and pharmacokinetics of rFVIIIFc. Sixteen subjects received a single dose of rFVIII at 25 or 65 IU/kg followed by an equal dose of rFVIIIFc. Most adverse events were unrelated to study drug. None of the study subjects developed anti-rFVIIIFc antibodies or inhibitors. Across dose levels, compared with rFVIII, rFVIIIFc showed 1.54- to 1.70-fold longer elimination t1/2, 1.49- to 1.56-fold lower clearance, and 1.48- to 1.56-fold higher total systemic exposure. rFVIII and rFVIIIFc had comparable dose-dependent peak plasma concentrations and recoveries. Time to 1% FVIII activity above baseline was ∼ 1.53- to 1.68-fold longer than rFVIII across dose levels. Each subject showed prolonged exposure to rFVIIIFc relative to rFVIII. Thus, rFVIIIFc may offer a viable therapeutic approach to achieve prolonged hemostatic protection and less frequent dosing in patients with hemophilia A. This trial was registered at www.clinicaltrials.govas NCT01027377.
- Published
- 2012
- Full Text
- View/download PDF
36. Recombinant factor IX-Fc fusion protein (rFIXFc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia B patients
- Author
-
Shapiro, Amy D., Ragni, Margaret V., Valentino, Leonard A., Key, Nigel S., Josephson, Neil C., Powell, Jerry S., Cheng, Gregory, Thompson, Arthur R., Goyal, Jaya, Tubridy, Karen L., Peters, Robert T., Dumont, Jennifer A., Euwart, Donald, Li, Lian, Hallén, Bengt, Gozzi, Peter, Bitonti, Alan J., Jiang, Haiyan, Luk, Alvin, and Pierce, Glenn F.
- Abstract
Current factor IX (FIX) products display a half-life (t1/2) of ∼ 18 hours, requiring frequent intravenous infusions for prophylaxis and treatment in patients with hemophilia B. This open-label, dose-escalation trial in previously treated adult subjects with hemophilia B examined the safety and pharmacokinetics of rFIXFc. rFIXFc is a recombinant fusion protein composed of FIX and the Fc domain of human IgG1, to extend circulating time. Fourteen subjects received a single dose of rFIXFc; 1 subject each received 1, 5, 12.5, or 25 IU/kg, and 5 subjects each received 50 or 100 IU/kg. rFIXFc was well tolerated, and most adverse events were mild or moderate in intensity. No inhibitors were detected in any subject. Dose-proportional increases in rFIXFc activity and Ag exposure were observed. With baseline subtraction, mean activity terminal t1/2 and mean residence time for rFIXFc were 56.7 and 71.8 hours, respectively. This is ∼ 3-fold longer than that reported for current rFIX products. The incremental recovery of rFIXFc was 0.93 IU/dL per IU/kg, similar to plasma-derived FIX. These results show that rFIXFc may offer a viable therapeutic approach to achieve prolonged hemostatic protection and less frequent dosing in patients with hemophilia B. The trial was registered at www.clinicaltrials.gov as NCT00716716.
- Published
- 2012
- Full Text
- View/download PDF
37. Recombinant factor IX-Fc fusion protein (rFIXFc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia B patients
- Author
-
Shapiro, Amy D., Ragni, Margaret V., Valentino, Leonard A., Key, Nigel S., Josephson, Neil C., Powell, Jerry S., Cheng, Gregory, Thompson, Arthur R., Goyal, Jaya, Tubridy, Karen L., Peters, Robert T., Dumont, Jennifer A., Euwart, Donald, Li, Lian, Hallén, Bengt, Gozzi, Peter, Bitonti, Alan J., Jiang, Haiyan, Luk, Alvin, and Pierce, Glenn F.
- Abstract
Current factor IX (FIX) products display a half-life (t1/2) of ∼ 18 hours, requiring frequent intravenous infusions for prophylaxis and treatment in patients with hemophilia B. This open-label, dose-escalation trial in previously treated adult subjects with hemophilia B examined the safety and pharmacokinetics of rFIXFc. rFIXFc is a recombinant fusion protein composed of FIX and the Fc domain of human IgG1, to extend circulating time. Fourteen subjects received a single dose of rFIXFc; 1 subject each received 1, 5, 12.5, or 25 IU/kg, and 5 subjects each received 50 or 100 IU/kg. rFIXFc was well tolerated, and most adverse events were mild or moderate in intensity. No inhibitors were detected in any subject. Dose-proportional increases in rFIXFc activity and Ag exposure were observed. With baseline subtraction, mean activity terminal t1/2and mean residence time for rFIXFc were 56.7 and 71.8 hours, respectively. This is ∼ 3-fold longer than that reported for current rFIX products. The incremental recovery of rFIXFc was 0.93 IU/dL per IU/kg, similar to plasma-derived FIX. These results show that rFIXFc may offer a viable therapeutic approach to achieve prolonged hemostatic protection and less frequent dosing in patients with hemophilia B. The trial was registered at www.clinicaltrials.govas NCT00716716.
- Published
- 2012
- Full Text
- View/download PDF
38. Bone microstructural changes revealed by high-resolution peripheral quantitative computed tomography imaging and elevated DKK1 and MIP-1α levels in patients with MGUS
- Author
-
Ng, Alvin C., Khosla, Sundeep, Charatcharoenwitthaya, Natthinee, Kumar, Shaji K., Achenbach, Sara J., Holets, Margaret F., McCready, Louise K., Melton, L. Joseph, Kyle, Robert A., Rajkumar, S. Vincent, and Drake, Matthew T.
- Abstract
Recent population-based studies demonstrate an increased fracture risk with monoclonal gammopathy of undetermined significance (MGUS). The etiology of this increased risk remains unclear, however, because areal bone mineral density (aBMD) measurements by dual-energy x-ray absorptiometry cannot assess bone microstructural properties critical to determining bone quality and strength. To better define the skeletal effects of MGUS, we performed aBMD and high-resolution peripheral quantitative computed tomography volumetric bone mineral density (vBMD) measurements in 50 MGUS patients (20 females, 30 males; mean ± SEM age, 70.5 ± 1.4 years) and 100 matched control subjects. Relative to controls, MGUS patients had decreased aBMD at the femoral neck (P= .05) and total femur (P< .05) but no differences at other sites. In contrast, high-resolution peripheral quantitative computed tomography showed markedly diminished cortical thickness (P< .05) and increased endocortical area (P< .01). Average vBMD (P< .01), cortical vBMD (P< .001), and trabecular thickness (P< .01) were all significantly decreased in MGUS patients, suggestive of impaired bone formation. Serum levels of the Wnt pathway inhibitor Dickkopf-related protein 1 (P< .001) and osteoclast-activating factor MIP-1α (P< .05) also were significantly elevated in MGUS patients. Our data provide the first evidence of altered bone microstructure in MGUS and suggest that cytokines elevated in osteolytic myeloma also may be associated with bone loss in MGUS.
- Published
- 2011
- Full Text
- View/download PDF
39. Bone microstructural changes revealed by high-resolution peripheral quantitative computed tomography imaging and elevated DKK1 and MIP-1α levels in patients with MGUS
- Author
-
Ng, Alvin C., Khosla, Sundeep, Charatcharoenwitthaya, Natthinee, Kumar, Shaji K., Achenbach, Sara J., Holets, Margaret F., McCready, Louise K., Melton, L. Joseph, Kyle, Robert A., Rajkumar, S. Vincent, and Drake, Matthew T.
- Abstract
Recent population-based studies demonstrate an increased fracture risk with monoclonal gammopathy of undetermined significance (MGUS). The etiology of this increased risk remains unclear, however, because areal bone mineral density (aBMD) measurements by dual-energy x-ray absorptiometry cannot assess bone microstructural properties critical to determining bone quality and strength. To better define the skeletal effects of MGUS, we performed aBMD and high-resolution peripheral quantitative computed tomography volumetric bone mineral density (vBMD) measurements in 50 MGUS patients (20 females, 30 males; mean ± SEM age, 70.5 ± 1.4 years) and 100 matched control subjects. Relative to controls, MGUS patients had decreased aBMD at the femoral neck (P = .05) and total femur (P < .05) but no differences at other sites. In contrast, high-resolution peripheral quantitative computed tomography showed markedly diminished cortical thickness (P < .05) and increased endocortical area (P < .01). Average vBMD (P < .01), cortical vBMD (P < .001), and trabecular thickness (P < .01) were all significantly decreased in MGUS patients, suggestive of impaired bone formation. Serum levels of the Wnt pathway inhibitor Dickkopf-related protein 1 (P < .001) and osteoclast-activating factor MIP-1α (P < .05) also were significantly elevated in MGUS patients. Our data provide the first evidence of altered bone microstructure in MGUS and suggest that cytokines elevated in osteolytic myeloma also may be associated with bone loss in MGUS.
- Published
- 2011
- Full Text
- View/download PDF
40. Methionine aminopeptidase 2 is required for HSC initiation and proliferation
- Author
-
Ma, Alvin C. H., Fung, Tsz K., Lin, Rachel H. C., Chung, Martin I. S., Yang, Dan, Ekker, Stephen C., and Leung, Anskar Y. H.
- Abstract
In a chemical screening, we tested the antiangiogenic effects of fumagillin derivatives and identified fumagillin as an inhibitor of definitive hematopoiesis in zebrafish embryos. Fumagillin is known to target methionine aminopeptidase II (MetAP2), an enzyme whose function in hematopoiesis is unknown. We investigated the role of MetAP2 in hematopoiesis by using zebrafish embryo and human umbilical cord blood models. Zebrafish metap2 was expressed ubiquitously during early embryogenesis and later in the somitic region, the caudal hematopoietic tissue, and pronephric duct. metap2 was inhibited by morpholino and fumagillin treatment, resulting in increased mpo expression at 18 hours postfertilization and reduced c-myb expression along the ventral wall of dorsal aorta at 36 hours postfertilization. It also disrupted intersegmental vessels in Tg(fli1:gfp) embryos without affecting development of major axial vasculatures. Inhibition of MetAP2 in CB CD34+ cells by fumagillin had no effect on overall clonogenic activity but significantly reduced their engraftment into immunodeficient nonobese diabetes/severe combined immunodeficiency mice. metap2 knock-down in zebrafish and inhibition by fumagillin in zebrafish and human CB CD34+ cells inhibited Calmodulin Kinase II activity and induced ERK phosphorylation. This study demonstrated a hithertoundescribed role of MetAP2 in definitive hematopoiesis and a possible link to noncanonical Wnt and ERK signaling.
- Published
- 2011
- Full Text
- View/download PDF
41. Methionine aminopeptidase 2 is required for HSC initiation and proliferation
- Author
-
Ma, Alvin C.H., Fung, Tsz K., Lin, Rachel H.C., Chung, Martin I.S., Yang, Dan, Ekker, Stephen C., and Leung, Anskar Y.H.
- Abstract
In a chemical screening, we tested the antiangiogenic effects of fumagillin derivatives and identified fumagillin as an inhibitor of definitive hematopoiesis in zebrafish embryos. Fumagillin is known to target methionine aminopeptidase II (MetAP2), an enzyme whose function in hematopoiesis is unknown. We investigated the role of MetAP2 in hematopoiesis by using zebrafish embryo and human umbilical cord blood models. Zebrafish metap2was expressed ubiquitously during early embryogenesis and later in the somitic region, the caudal hematopoietic tissue, and pronephric duct. metap2 was inhibited by morpholino and fumagillin treatment, resulting in increased mpoexpression at 18 hours postfertilization and reduced c-mybexpression along the ventral wall of dorsal aorta at 36 hours postfertilization. It also disrupted intersegmental vessels in Tg(fli1:gfp) embryos without affecting development of major axial vasculatures. Inhibition of MetAP2 in CB CD34+cells by fumagillin had no effect on overall clonogenic activity but significantly reduced their engraftment into immunodeficient nonobese diabetes/severe combined immunodeficiency mice. metap2knock-down in zebrafish and inhibition by fumagillin in zebrafish and human CB CD34+cells inhibited Calmodulin Kinase II activity and induced ERK phosphorylation. This study demonstrated a hithertoundescribed role of MetAP2 in definitive hematopoiesis and a possible link to noncanonical Wnt and ERK signaling.
- Published
- 2011
- Full Text
- View/download PDF
42. Long-term restoration of the human T-cell compartment after thymectomy during infancy: a role for thymic regeneration?
- Author
-
van Gent, Rogier, Schadenberg, Alvin W.L., Otto, Sigrid A., Nievelstein, Rutger A.J., Sieswerda, Gertjan T., Haas, Felix, Miedema, Frank, Tesselaar, Kiki, Jansen, Nicolaas J.G., and Borghans, José A.M.
- Abstract
Thymectomy during early childhood is generally thought to have serious consequences for the establishment of the T-cell compartment. In the present study, we investigated the composition of the T-cell pool in the first 3 decades after thymectomy during infancy due to cardiac surgery. In the first 5 years after thymectomy, naive and total CD4+and CD8+T-cell numbers in the blood and T-cell receptor excision circle (TREC) levels in CD4+T cells were significantly lower than in healthy age-matched controls. In the first years after thymectomy, plasma IL-7 levels were significantly elevated and peripheral T-cell proliferation levels were increased by ∼ 2-fold. From 5 years after thymectomy onward, naive CD4+and CD8+T-cell counts and TRECs were within the normal range. Because TREC levels are expected to decline continuously in the absence of thymic output, we investigated whether normalization of the naive T-cell pool could be due to regeneration of thymic tissue. In the majority of individuals who had been thymectomized during infancy, thymic tissue could indeed be identified on magnetic resonance imaging scans. Whereas thymectomy has severe effects on the establishment of the naive T-cell compartment during early childhood, our data suggest that functional regrowth of thymic tissue can limit its effects in subsequent years.
- Published
- 2011
- Full Text
- View/download PDF
43. Long-term restoration of the human T-cell compartment after thymectomy during infancy: a role for thymic regeneration?
- Author
-
van Gent, Rogier, Schadenberg, Alvin W. L., Otto, Sigrid A., Nievelstein, Rutger A. J., Sieswerda, Gertjan T., Haas, Felix, Miedema, Frank, Tesselaar, Kiki, Jansen, Nicolaas J. G., and Borghans, José A. M.
- Abstract
Thymectomy during early childhood is generally thought to have serious consequences for the establishment of the T-cell compartment. In the present study, we investigated the composition of the T-cell pool in the first 3 decades after thymectomy during infancy due to cardiac surgery. In the first 5 years after thymectomy, naive and total CD4+ and CD8+ T-cell numbers in the blood and T-cell receptor excision circle (TREC) levels in CD4+ T cells were significantly lower than in healthy age-matched controls. In the first years after thymectomy, plasma IL-7 levels were significantly elevated and peripheral T-cell proliferation levels were increased by ∼ 2-fold. From 5 years after thymectomy onward, naive CD4+ and CD8+ T-cell counts and TRECs were within the normal range. Because TREC levels are expected to decline continuously in the absence of thymic output, we investigated whether normalization of the naive T-cell pool could be due to regeneration of thymic tissue. In the majority of individuals who had been thymectomized during infancy, thymic tissue could indeed be identified on magnetic resonance imaging scans. Whereas thymectomy has severe effects on the establishment of the naive T-cell compartment during early childhood, our data suggest that functional regrowth of thymic tissue can limit its effects in subsequent years.
- Published
- 2011
- Full Text
- View/download PDF
44. Murine prolylcarboxypeptidase depletion induces vascular dysfunction with hypertension and faster arterial thrombosis
- Author
-
Adams, Gregory N., LaRusch, Gretchen A., Stavrou, Evi, Zhou, Yihua, Nieman, Marvin T., Jacobs, Gretta H., Cui, Yingjie, Lu, Yuan, Jain, Mukesh K., Mahdi, Fakhri, Shariat-Madar, Zia, Okada, Yoshio, D'Alecy, Louis G., and Schmaier, Alvin H.
- Abstract
Prolylcarboxypeptidase (PRCP) activates prekallikrein to plasma kallikrein, leading to bradykinin liberation, and degrades angiotensin II. We now identify PRCP as a regulator of blood vessel homeostasis. β-Galactosidase staining in PRCPgt/gtmice reveals expression in kidney and vasculature. Invasive telemetric monitorings show that PRCPgt/gtmice have significantly elevated blood pressure. PRCPgt/gtmice demonstrate shorter carotid artery occlusion times in 2 models, and their plasmas have increased thrombin generation times. Pharmacologic inhibition of PRCP with Z-Pro-Prolinal or plasma kallikrein with soybean trypsin inhibitor, Pro-Phe-Arg-chloromethylketone or PKSI 527 also shortens carotid artery occlusion times. Aortic and renal tissues have uncoupled eNOS and increased reactive oxygen species (ROS) in PRCPgt/gtmice as detected by dihydroethidium or Amplex Red fluorescence or lucigenin luminescence. The importance of ROS is evidenced by the fact that treatment of PRCPgt/gtmice with antioxidants (mitoTEMPO, apocynin, Tempol) abrogates the hypertensive, prothrombotic phenotype. Mechanistically, our studies reveal that PRCPgt/gtaortas express reduced levels of Kruppel-like factors 2 and 4, thrombomodulin, and eNOS mRNA, suggesting endothelial cell dysfunction. Further, PRCP siRNA treatment of endothelial cells shows increased ROS and uncoupled eNOS and decreased protein C activation because of thrombomodulin inactivation. Collectively, our studies identify PRCP as a novel regulator of vascular ROS and homeostasis.
- Published
- 2011
- Full Text
- View/download PDF
45. Murine prolylcarboxypeptidase depletion induces vascular dysfunction with hypertension and faster arterial thrombosis
- Author
-
Adams, Gregory N., LaRusch, Gretchen A., Stavrou, Evi, Zhou, Yihua, Nieman, Marvin T., Jacobs, Gretta H., Cui, Yingjie, Lu, Yuan, Jain, Mukesh K., Mahdi, Fakhri, Shariat-Madar, Zia, Okada, Yoshio, D'Alecy, Louis G., and Schmaier, Alvin H.
- Abstract
Prolylcarboxypeptidase (PRCP) activates prekallikrein to plasma kallikrein, leading to bradykinin liberation, and degrades angiotensin II. We now identify PRCP as a regulator of blood vessel homeostasis. β-Galactosidase staining in PRCPgt/gt mice reveals expression in kidney and vasculature. Invasive telemetric monitorings show that PRCPgt/gt mice have significantly elevated blood pressure. PRCPgt/gt mice demonstrate shorter carotid artery occlusion times in 2 models, and their plasmas have increased thrombin generation times. Pharmacologic inhibition of PRCP with Z-Pro-Prolinal or plasma kallikrein with soybean trypsin inhibitor, Pro-Phe-Arg-chloromethylketone or PKSI 527 also shortens carotid artery occlusion times. Aortic and renal tissues have uncoupled eNOS and increased reactive oxygen species (ROS) in PRCPgt/gt mice as detected by dihydroethidium or Amplex Red fluorescence or lucigenin luminescence. The importance of ROS is evidenced by the fact that treatment of PRCPgt/gt mice with antioxidants (mitoTEMPO, apocynin, Tempol) abrogates the hypertensive, prothrombotic phenotype. Mechanistically, our studies reveal that PRCPgt/gt aortas express reduced levels of Kruppel-like factors 2 and 4, thrombomodulin, and eNOS mRNA, suggesting endothelial cell dysfunction. Further, PRCP siRNA treatment of endothelial cells shows increased ROS and uncoupled eNOS and decreased protein C activation because of thrombomodulin inactivation. Collectively, our studies identify PRCP as a novel regulator of vascular ROS and homeostasis.
- Published
- 2011
- Full Text
- View/download PDF
46. Inhibition of cell adhesion by anti–P-selectin aptamer: a new potential therapeutic agent for sickle cell disease
- Author
-
Gutsaeva, Diana R., Parkerson, James B., Yerigenahally, Shobha D., Kurz, Jeffrey C., Schaub, Robert G., Ikuta, Tohru, and Head, C. Alvin
- Abstract
Adhesive interactions between circulating sickle red blood cells (RBCs), leukocytes, and endothelial cells are major pathophysiologic events in sickle cell disease (SCD). To develop new therapeutics that efficiently inhibit adhesive interactions, we generated an anti–P-selectin aptamer and examined its effects on cell adhesion using knockout-transgenic SCD model mice. Aptamers, single-stranded oligonucleotides that bind molecular targets with high affinity and specificity, are emerging as new therapeutics for cardiovascular and hematologic disorders. In vitro studies found that the anti–P-selectin aptamer exhibits high specificity to mouse P-selectin but not other selectins. SCD mice were injected with the anti–P-selectin aptamer, and cell adhesion was observed under hypoxia. The anti–P-selectin aptamer inhibited the adhesion of sickle RBCs and leukocytes to endothelial cells by 90% and 80%, respectively. The anti–P-selectin aptamer also increased microvascular flow velocities and reduced the leukocyte rolling flux. SCD mice treated with the anti–P-selectin aptamer demonstrated a reduced mortality rate associated with the experimental procedures compared with control mice. These results demonstrate that anti–P-selectin aptamer efficiently inhibits the adhesion of both sickle RBCs and leukocytes to endothelial cells in SCD model mice, suggesting a critical role for P-selectin in cell adhesion. Anti–P-selectin aptamer may be useful as a novel therapeutic agent for SCD.
- Published
- 2011
- Full Text
- View/download PDF
47. Inhibition of cell adhesion by anti–P-selectin aptamer: a new potential therapeutic agent for sickle cell disease
- Author
-
Gutsaeva, Diana R., Parkerson, James B., Yerigenahally, Shobha D., Kurz, Jeffrey C., Schaub, Robert G., Ikuta, Tohru, and Head, C. Alvin
- Abstract
Adhesive interactions between circulating sickle red blood cells (RBCs), leukocytes, and endothelial cells are major pathophysiologic events in sickle cell disease (SCD). To develop new therapeutics that efficiently inhibit adhesive interactions, we generated an anti–P-selectin aptamer and examined its effects on cell adhesion using knockout-transgenic SCD model mice. Aptamers, single-stranded oligonucleotides that bind molecular targets with high affinity and specificity, are emerging as new therapeutics for cardiovascular and hematologic disorders. In vitro studies found that the anti–P-selectin aptamer exhibits high specificity to mouse P-selectin but not other selectins. SCD mice were injected with the anti–P-selectin aptamer, and cell adhesion was observed under hypoxia. The anti–P-selectin aptamer inhibited the adhesion of sickle RBCs and leukocytes to endothelial cells by 90% and 80%, respectively. The anti–P-selectin aptamer also increased microvascular flow velocities and reduced the leukocyte rolling flux. SCD mice treated with the anti–P-selectin aptamer demonstrated a reduced mortality rate associated with the experimental procedures compared with control mice. These results demonstrate that anti–P-selectin aptamer efficiently inhibits the adhesion of both sickle RBCs and leukocytes to endothelial cells in SCD model mice, suggesting a critical role for P-selectin in cell adhesion. Anti–P-selectin aptamer may be useful as a novel therapeutic agent for SCD.
- Published
- 2011
- Full Text
- View/download PDF
48. Factor XII stimulates ERK1/2 and Akt through uPAR, integrins, and the EGFR to initiate angiogenesis
- Author
-
LaRusch, Gretchen A., Mahdi, Fakhri, Shariat-Madar, Zia, Adams, Gregory, Sitrin, Robert G., Zhang, Wan Ming, McCrae, Keith R., and Schmaier, Alvin H.
- Abstract
Factor XII (FXII) and high molecular weight kininogen (HK) mutually block each other's binding to the urokinase plasminogen activator receptor (uPAR). We investigated if FXII stimulates cells by interacting with uPAR. FXII (3-62nM) with 0.05mM Zn2+ induces extracellular signal-related kinase 1/2 (ERK1/2; mitogen-activated protein kinase 44 [MAPK44] andMAPK42) and Akt (Ser473) phosphorylation in endothelial cells. FXII-induced phosphorylation of ERK1/2 or Akt is a zymogen activity, not an enzymatic event. ERK1/2 or Akt phosphorylation is blocked upstream by PD98059 or Wortmannin or LY294002, respectively. An uPAR signaling region for FXII is on domain 2 adjacent to uPAR's integrin binding site. Cleaved HK or peptides from HK's domain 5 blocks FXII-induced ERK1/2 and Akt phosphorylation. A β1 integrin peptide that binds uPAR, antibody 6S6 to β1 integrin, or the epidermal growth factor receptor (EGFR) inhibitor AG1478 blocks FXII-induced phosphorylation of ERK1/2 and Akt. FXII induces endothelial cell proliferation and 5-bromo-2′deoxy-uridine incorporation. FXII stimulates aortic sprouting in normal but not uPAR-deficient mouse aorta. FXII produces angiogenesis in matrigel plugs in normal but not uPAR-deficient mice. FXII knockout mice have reduced constitutive and wound-induced blood vessel number. In sum, FXII initiates signaling mediated by uPAR, β1 integrin, and the EGFR to induce human umbilical vein endothelial cell proliferation, growth, and angiogenesis.
- Published
- 2010
- Full Text
- View/download PDF
49. Factor XII stimulates ERK1/2 and Akt through uPAR, integrins, and the EGFR to initiate angiogenesis
- Author
-
LaRusch, Gretchen A., Mahdi, Fakhri, Shariat-Madar, Zia, Adams, Gregory, Sitrin, Robert G., Zhang, Wan Ming, McCrae, Keith R., and Schmaier, Alvin H.
- Abstract
Factor XII (FXII) and high molecular weight kininogen (HK) mutually block each other's binding to the urokinase plasminogen activator receptor (uPAR). We investigated if FXII stimulates cells by interacting with uPAR. FXII (3-62nM) with 0.05mM Zn2+induces extracellular signal-related kinase 1/2 (ERK1/2; mitogen-activated protein kinase 44 [MAPK44] andMAPK42) and Akt (Ser473) phosphorylation in endothelial cells. FXII-induced phosphorylation of ERK1/2 or Akt is a zymogen activity, not an enzymatic event. ERK1/2 or Akt phosphorylation is blocked upstream by PD98059 or Wortmannin or LY294002, respectively. An uPAR signaling region for FXII is on domain 2 adjacent to uPAR's integrin binding site. Cleaved HK or peptides from HK's domain 5 blocks FXII-induced ERK1/2 and Akt phosphorylation. A β1integrin peptide that binds uPAR, antibody 6S6 to β1integrin, or the epidermal growth factor receptor (EGFR) inhibitor AG1478 blocks FXII-induced phosphorylation of ERK1/2 and Akt. FXII induces endothelial cell proliferation and 5-bromo-2′deoxy-uridine incorporation. FXII stimulates aortic sprouting in normal but not uPAR-deficient mouse aorta. FXII produces angiogenesis in matrigel plugs in normal but not uPAR-deficient mice. FXII knockout mice have reduced constitutive and wound-induced blood vessel number. In sum, FXII initiates signaling mediated by uPAR, β1integrin, and the EGFR to induce human umbilical vein endothelial cell proliferation, growth, and angiogenesis.
- Published
- 2010
- Full Text
- View/download PDF
50. Deletion of murine kininogen gene 1 (mKng1) causes loss of plasma kininogen and delays thrombosis
- Author
-
Merkulov, Sergei, Zhang, Wan-Ming, Komar, Anton A., Schmaier, Alvin H., Barnes, Ellen, Zhou, Yihua, Lu, Xincheng, Iwaki, Takayuki, Castellino, Francis J., Luo, Guangbin, and McCrae, Keith R.
- Abstract
High-molecular-weight kininogen (HK) plays an important role in the assembly of the plasma kallikrein-kinin system. While the human genome contains a single copy of the kininogen gene, 3 copies exist in the rat (1 encoding K-kininogen and 2 encoding T-kininogen). Here, we confirm that the mouse genome contains 2 homologous kininogen genes, mKng1 and mKng2, and demonstrate that these genes are expressed in a tissue-specific manner. To determine the roles of these genes in murine development and physiology, we disrupted mKng1, which is expressed primarily in the liver. mKng1−/− mice were viable, but lacked plasma HK and low-molecular-weight kininogen (LK), as well as ΔmHK-D5, a novel kininogen isoform that lacks kininogen domain 5. Moreover, despite normal tail vein bleeding times, mKng1−/− mice displayed a significantly prolonged time to carotid artery occlusion following Rose Bengal administration and laser-induced arterial injury. These results suggest that a single gene, mKng1, is responsible for production of plasma kininogen, and that plasma HK contributes to induced arterial thrombosis in mice.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.