The overall goal of this program is to develop a strong experimental foundation for the correction of inherited disease of bone marrow-derived cells by genetic modification of hematopoietic stem cells. Current objectives are focused on the use of recombinant retroviral, lentiviral, and adeno-associated virus (AAV) vectors to achieve efficient transfer of functional genes into primitive hematopoietic stem cells while maintaining their maximal hematopoietic potential.
The specific aims are to 1) examine the relationship between cell cycle, fibronectin-mediated adhesion, and cytokine regulators on hematopoietic cell function and viral transduction; 2) determine whether recombinant viral vectors derived from AAV and lentivirus can be used for efficient and stable gene transfer in murine and human hematopoietic cells; 3) identify optimal strategies using viral-mediated gene transfer to correct the phenotype in two inherited blood disorders, X-linked chronic granulomatous disease (X-CGD) and Fanconi anemia group C (FanC); and 4) evaluate non- ablative conditioning regimens and selection of transduced cells using maker proteins or drug resistance genes. Experimental approaches include the use of in vitro culture systems, NOD/SCID mouse-human xenografts, and murine models of X-CGD and FanC previously generated by gene targeting approaches. The implementation of these aims will be shared among 4 projects and 3 core units. This proposal draws from a group of investigators with diverse but complementary experience in stem cell biology, retrovirus-, lentivirus-, and AAV-mediated gene transfer, molecular genetics, virology, bone marrow transplantation, and neonatology. Achievement of these goals will permit the translation of this basic work to the development of clinical protocols for effective viral-mediated gene transfer of genetic blood diseases. On a broader level, these studies should provide insight into the biologic behavior of hematopoietic stem cells and the ability to manipulate them ex vivo. GANT=P01HL31992-17 PROPOSED PROGRAM (Adapted from Applicant's Abstract) The long-term objectives of this Program Project application are to evaluate basic mechanisms and develop new treatments for acute lung injury. High concentrations of oxygen and septic lung injury are the primary models that will be evaluated. The proposed program consists of four projects and three core units. Project 1 will evaluate the efficacy of small molecular weight catalytic antioxidants in the treatment of both hyperoxic and LPS + sepsis-initiated lung injury. This project will also develop new antioxidant mimetics and explore their relationships with the antioxidant properties of heme oxygenase (HO). Project 2 will test the hypothesis that activation of extrinsic coagulation and disordered fibrin turnover are central elements in hyperoxic and septic lung injury. The efficacy of specific blockade of the initiating steps of extrinsic coagulation in reducing inflammation and acute lung injury will be tested using two new anticoagulant drugs that block tissue factor (TF) function and do not cause bleeding. Project 3 will evaluate the regulation and function of the extracellular superoxide dismutase (EC-SOD) in acute lung injury and determine the impact of cleavage of the C-terminal """"""""heparin binding"""""""" domain of this enzyme in determining its distribution and function. Project 4 will evaluate control of metabolic pathways and upregulation of lung cell glycolysis in modulating responses to acute injury. This project will test the hypothesis that adaptation to oxidant stress in the lung requires elevated expression of hexokinase (HK), a rate limiting step in glycolysis in the lung. The overall rationale for the Program Project is to use an interdisciplinary approach to define the cellular pathways and cellular adaptive responses involved in acute lung injury and to test new strategies for pharmacologic therapy that can be extended to the treatment of humans with ARDS and sepsis.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Program Projects (P01)
Project #
5P01HL053586-07
Application #
6363535
Study Section
Heart, Lung, and Blood Initial Review Group (HLBP)
Program Officer
Peterson, Charles M
Project Start
1994-12-01
Project End
2005-02-28
Budget Start
2001-04-06
Budget End
2002-02-28
Support Year
7
Fiscal Year
2001
Total Cost
$1,446,593
Indirect Cost
Name
Indiana University-Purdue University at Indianapolis
Department
Pediatrics
Type
Schools of Medicine
DUNS #
005436803
City
Indianapolis
State
IN
Country
United States
Zip Code
46202
Stein, Stefan; Scholz, Simone; Schwäble, Joachim et al. (2013) From bench to bedside: preclinical evaluation of a self-inactivating gammaretroviral vector for the gene therapy of X-linked chronic granulomatous disease. Hum Gene Ther Clin Dev 24:86-98
Song, Liujiang; Kauss, M Ariel; Kopin, Etana et al. (2013) Optimizing the transduction efficiency of capsid-modified AAV6 serotype vectors in primary human hematopoietic stem cells in vitro and in a xenograft mouse model in vivo. Cytotherapy 15:986-98
Liu, Ying; Ballman, Kimberly; Li, Deqiang et al. (2012) Impaired function of Fanconi anemia type C-deficient macrophages. J Leukoc Biol 91:333-40
Hawkins, Troy B; Dantzer, Jessica; Peters, Brandon et al. (2011) Identifying viral integration sites using SeqMap 2.0. Bioinformatics 27:720-2
Ou, Xuan; Chae, Hee-Don; Wang, Rui-Hong et al. (2011) SIRT1 deficiency compromises mouse embryonic stem cell hematopoietic differentiation, and embryonic and adult hematopoiesis in the mouse. Blood 117:440-50
Grez, Manuel; Reichenbach, Janine; Schwäble, Joachim et al. (2011) Gene therapy of chronic granulomatous disease: the engraftment dilemma. Mol Ther 19:28-35
Rohrabaugh, Sara L; Campbell, Timothy B; Hangoc, Giao et al. (2011) Ex vivo rapamycin treatment of human cord blood CD34+ cells enhances their engraftment of NSG mice. Blood Cells Mol Dis 46:318-20
Broxmeyer, Hal E; Lee, Man-Ryul; Hangoc, Giao et al. (2011) Hematopoietic stem/progenitor cells, generation of induced pluripotent stem cells, and isolation of endothelial progenitors from 21- to 23.5-year cryopreserved cord blood. Blood 117:4773-7
Rohrabaugh, Sara L; Hangoc, Giao; Kelley, Mark R et al. (2011) Mad2 haploinsufficiency protects hematopoietic progenitor cells subjected to cell-cycle stress in vivo and to inhibition of redox function of Ape1/Ref-1 in vitro. Exp Hematol 39:415-23
Liu, Ying; Timani, Khalid; Mantel, Charlie et al. (2011) TIP110/p110nrb/SART3/p110 regulation of hematopoiesis through CMYC. Blood 117:5643-51

Showing the most recent 10 out of 158 publications