79 results on '"Imren S"'
Search Results
2. P1482: PATIENTS WITH SEVERE SICKLE CELL DISEASE ON STANDARD OF CARE TREATMENT ARE VERY UNLIKELY TO BECOME VOC FREE FOR ONE YEAR: A COHORT STUDY OF MEDICAID ENROLLEES
- Author
-
Mahesri, M., primary, Lee, S. B., additional, Levin, R., additional, Imren, S., additional, Zhang, L., additional, Beukelman, T., additional, Titievsky, L., additional, and Desai, R. J., additional
- Published
- 2022
- Full Text
- View/download PDF
3. CO197 Vaso-Occlusion Definitions Can Impact Efficacy Assessments in Clinical Studies of Sickle Cell Disease
- Author
-
Frangoul, H., Imren, S., Xuan, F., Li, N., Rubin, JL., Hobbs, B., and Locatelli, F.
- Published
- 2024
- Full Text
- View/download PDF
4. CO118 Sickle Cell Disease Outcomes in Patients Following Allogeneic Hematopoietic Stem Cell Transplantation: A Systematic Literature Review
- Author
-
Nair, R., Li, N., Imren, S., Kohli, P., Dejoie-Brewer, M.., Lach, K., Zhu, L., and Dudzic, M.
- Published
- 2024
- Full Text
- View/download PDF
5. CO78 Transfusion Outcomes Following Allogeneic Hematopoietic Stem Cell Transplantation in Patients With Transfusion-Dependent Beta-Thalassemia: A Systematic Literature Review
- Author
-
Nair, R., Li, N., Imren, S., Kohli, P., Dejoie-Brewer, M.., Lach, K., Zhu, L., and Dudzic, M.
- Published
- 2024
- Full Text
- View/download PDF
6. Meis1 disrupts the genomic imprint of Dlk1 in a NUP98-HOXD13 leukemia model
- Author
-
Argiropoulos, B, Palmqvist, L, Imren, S, Miller, M, Rouhi, A, Mager, D L, and Humphries, R K
- Published
- 2010
- Full Text
- View/download PDF
7. Ex vivo expansion of normal and chronic myeloid leukemic stem cells without functional alteration using a NUP98HOXA10homeodomain fusion gene
- Author
-
Sloma, I, Imren, S, Beer, P A, Zhao, Y, Lecault, V, Leung, D, Raghuram, K, Brimacombe, C, Lambie, K, Piret, J, Hansen, C, Humphries, R K, and Eaves, C J
- Published
- 2013
- Full Text
- View/download PDF
8. TAMAM (Turkish American Marmara Multichannel) Project in Marmara Sea
- Author
-
OKAY, SEDA, ÇİFCİ, GÜNAY, Barın, B, DEMİRBAĞ, MUSTAFA EMİN, Imren, S, KURT, HÜLYA, Shillington, D, Seeber, L, Steckler, M, Sorlien, C, GÜRÇAY, SAVAŞ, DONDURUR, DERMAN, ATGIN, ORHAN, and ÖZEL, ÖZKAN
- Published
- 2019
9. The ʼhematopoietic PBX1 interacting proteinʼ (HPIP), a novel member of the homeobox regulatory network, amplifies early human hematopoietic progenitor cells: 523
- Author
-
Buske, C., Feuring-Buske, M., Abramovich, C., Imren, S., Hiddemann, W., and Humphries, R. K.
- Published
- 2002
10. PROGRESS TOWARDS GENE THERAPY OF SICKLE CELL DISEASE
- Author
-
Humphries, R K, Imren, S, Kalberer, C P, Faulkes, S, Eaves, C J, Pawliuk, R, and Leboulch, P.
- Published
- 2000
11. The validity and the reliability of Turkish version of children's sleep habits questionnaire [Çocuk uyku alişkanliklari anketinin türkçe geçerlili?i ve güvenilirli?i]
- Author
-
Fiş N.P., Arman A., Ay P., Topuzo?lu A., Güler A.S., Gökçe Imren S., Ersu R., Berkem M., and Fiş, N.P., Marmara Üniv., Tip Fak., Çocuk Psikiyatrisi ABD, Turkey -- Arman, A., Marmara Üniv., Tip Fak., Çocuk Psikiyatrisi ABD, Turkey -- Ay, P., Halk Sa?li?i ABD, Turkey -- Topuzo?lu, A., Halk Sa?li?i ABD, Turkey -- Güler, A.S., Marmara Üniv., Tip Fak., Çocuk Psikiyatrisi ABD, Turkey, Cumhuriyet Üniversitesi, Tip Fakültesi, Çocuk Psikiyatrisi ABD, Sivas, Turkey -- Gökçe Imren, S., Marmara Üniv., Tip Fak., Çocuk Psikiyatrisi ABD, Turkey, Cumhuriyet Üniversitesi, Tip Fakültesi, Çocuk Psikiyatrisi ABD, Sivas, Turkey -- Ersu, R., Pediatrik Gö?üs Hastaliklari BD, Istanbul, Turkey -- Berkem, M., Marmara Üniv., Tip Fak., Çocuk Psikiyatrisi ABD, Turkey
- Subjects
Questionnaire ,Sleep problems ,Child ,Reliability ,Validity - Abstract
Objective: The objective of this study was to examine the validity and the reliability of Turkish version of Children's Sleep Habits Questionnaire (CSHQ)-Abbreviated Form. Methods: The sample consisted of 1749 1st, 2nd, 3rd, and 4th grade elementary schoolchildren. The parents were asked to complete the CSHQ, sociodemographic form, and a question list assessing the behavioral and emotional problems of children. Internal consistency, test-retest reliability, and construct validity of the CSHQ were investigated. Results: Cronbach's alpha coefficient was determined as 0.78. The test retest correlation coefficient was 0.81 (p0.05), yet the scores tended to increase with decreasing socio-economic status (p, Fiş, N. P.; Marmara Üniv., Hastanesi Çocuk Psikiyatrisi ABD, Tophanelio?lu Cad. No. 13-15 Altunizade, 34660 Üsküdar-Istanbul, Turkey; email: nepfis@yahoo.com
- Published
- 2010
12. Ex vivo expansion of normal and chronic myeloid leukemic stem cells without functional alteration using a NUP98HOXA10homeodomain fusion gene
- Author
-
Sloma, I, primary, Imren, S, additional, Beer, P A, additional, Zhao, Y, additional, Lecault, V, additional, Leung, D, additional, Raghuram, K, additional, Brimacombe, C, additional, Lambie, K, additional, Piret, J, additional, Hansen, C, additional, Humphries, R K, additional, and Eaves, C J, additional
- Published
- 2012
- Full Text
- View/download PDF
13. Does methylphenidate improve social impairment in ADHD?
- Author
-
Gokce Imren, S., primary, Gumustas, F., additional, Guler, A.S., additional, and Yazgan, Y., additional
- Published
- 2012
- Full Text
- View/download PDF
14. Ex vivo expansion of hematopoietic stem cells for use in nonmyeloablative transplantation
- Author
-
Bakovic, S., primary, Ohta, H., additional, Imren, S., additional, Cavilla, B., additional, Eaves, C.J., additional, and Humphries, R.K., additional
- Published
- 2008
- Full Text
- View/download PDF
15. Improved purification of hematopoietic stem cells based on their elevated aldehyde dehydrogenase activity
- Author
-
Christ, O., primary, Lucke, K., additional, Imren, S., additional, Leung, K., additional, Hamilton, M., additional, Eaves, A., additional, Smith, C., additional, and Eaves, C., additional
- Published
- 2007
- Full Text
- View/download PDF
16. Preselection of retrovirally transduced bone marrow avoids subsequent stem cell gene silencing and age-dependent extinction of expression of human beta -globin in engrafted mice
- Author
-
Kalberer, C. P., primary, Pawliuk, R., additional, Imren, S., additional, Bachelot, T., additional, Takekoshi, K. J., additional, Fabry, M., additional, Eaves, C. J., additional, London, I. M., additional, Humphries, R. K., additional, and Leboulch, P., additional
- Published
- 2000
- Full Text
- View/download PDF
17. Protease inhibitors: Role and potential therapeutic use in human cancer
- Author
-
DeClerck, Y.A., primary and Imren, S., additional
- Published
- 1994
- Full Text
- View/download PDF
18. How do healthcare professionals interview patients to assess suicide risk?
- Author
-
Rose McCabe, Imren Sterno, Stefan Priebe, Rebecca Barnes, and Richard Byng
- Subjects
Suicide ,Risk ,Communication ,Assessment ,Conversation analysis ,Mixed methods ,Psychiatry ,RC435-571 - Abstract
Abstract Background There is little evidence on how professionals communicate to assess suicide risk. This study analysed how professionals interview patients about suicidal ideation in clinical practice. Methods Three hundred nineteen video-recorded outpatient visits in U.K. secondary mental health care were screened. 83 exchanges about suicidal ideation were identified in 77 visits. A convenience sample of 6 cases in 46 primary care visits was also analysed. Depressive symptoms were assessed. Questions and responses were qualitatively analysed using conversation analysis. χ 2 tested whether questions were influenced by severity of depression or influenced patients’ responses. Results A gateway closed question was always asked inviting a yes/no response. 75% of questions were negatively phrased, communicating an expectation of no suicidal ideation, e.g., “No thoughts of harming yourself?”. 25% were positively phrased, communicating an expectation of suicidal ideation, e.g., “Do you feel life is not worth living?”. Comparing these two question types, patients were significantly more likely to say they were not suicidal when the question was negatively phrased but were not more likely to say they were suicidal when positively phrased (χ 2 = 7.2, df = 1, p = 0.016). 25% patients responded with a narrative rather than a yes/no, conveying ambivalence. Here, psychiatrists tended to pursue a yes/no response. When the patient responded no to the gateway question, the psychiatrist moved on to the next topic. A similar pattern was identified in primary care. Conclusions Psychiatrists tend to ask patients to confirm they are not suicidal using negative questions. Negatively phrased questions bias patients’ responses towards reporting no suicidal ideation.
- Published
- 2017
- Full Text
- View/download PDF
19. Functional cloning and characterization of a novel nonhomeodomain protein that inhibits the binding of PBX1-HOX complexes to DNA.
- Author
-
Abramovich, C, Shen, W F, Pineault, N, Imren, S, Montpetit, B, Largman, C, and Humphries, R K
- Abstract
PBX1 is a homeodomain protein that functions in complexes with other homeodomain-containing proteins to regulate gene expression during developmental and/or differentiation processes. A yeast two-hybrid screen of a fetal liver-hematopoietic cDNA library using PBX1a as bait led to the discovery of a novel non-homeodomain-containing protein that interacts with PBX1 as well as PBX2 and PBX3. RNA analysis revealed it to be expressed in CD34(+) hematopoietic cell populations enriched in primitive progenitors, as is PBX1; search of the expressed sequence tag data base indicated that it is also expressed in other early embryonic as well as adult tissues. The full-length cDNA encodes a 731-amino acid protein that has no significant homology to known proteins. This protein that we have termed hematopoietic PBX-interacting protein (HPIP) is mainly localized in the cytosol and in small amounts in the nucleus. The region of PBX that interacts with HPIP includes both the homeodomain and immediate N-terminal flanking sequences. Strikingly, electrophoretic mobility shift assays revealed that HPIP inhibits the ability of PBX-HOX heterodimers to bind to target sequences. Moreover, HPIP strongly inhibits the transactivation activity of E2A-PBX. Together these findings suggest that HPIP is a new regulator of PBX function.
- Published
- 2000
- Full Text
- View/download PDF
20. Overexpression of tissue inhibitor of metalloproteinases-2 retroviral-mediated gene transfer in vivo inhibits tumor growth and invasion
- Author
-
Imren S, Donald Kohn, Shimada H, Blavier L, and Ya, Declerck
- Subjects
Tissue Inhibitor of Metalloproteinase-2 ,DNA, Complementary ,Gene Transfer Techniques ,Gene Expression ,Mice, Nude ,Proteins ,3T3 Cells ,Neoplasms, Experimental ,Transfection ,Rats ,Mice ,Retroviridae ,Transduction, Genetic ,Protein Biosynthesis ,Animals ,Humans ,Female ,Neoplasm Invasiveness ,Cell Division ,Neoplasm Transplantation - Abstract
We have demonstrated previously that overexpression of tissue inhibitor of metalloproteinases-2 (TIMP-2), an inhibitor of matrix-degrading metalloproteinases, not only inhibits the invasive and metastatic behavior of tumor cells but also significantly decreases tumor growth in vivo (Y. A. DeClerck et at, Cancer Res., 52: 701-708, 1992). This latter effect was found to be dependent on the ability of TIMP-2 to prevent the degradation of the collagen matrix (A. M. Montgomery et al., Cancer Res., 54: 5467-5473, 1994). In this report, we have overexpressed TIMP-2 in tumor tissue by retroviral-mediated gene transfer into tumor cells by co-injecting s.c. in nude mice tumorigenic c-Ha-ras-transfected rat embryo fibroblasts with irradiated packaging cells producing high titer retroviral vectors containing the human TIMP-2 cDNA. The growth rate of tumors derived from cells co-injected with the TIMP-2 vector producer cells was significantly slower than the growth rate of tumors derived from cells co-injected with packaging cells producing a retrovirus containing the Escherichia coli beta-galactosidase gene. The transduction efficiency was estimated at 13%, and the production of a functional human TIMP-2 in tumor cells transduced with the TIMP-2-containing vector was documented. Furthermore, histological analysis of tumors derived from tumor cells co-injected with the TIMP-2 vector producer cells revealed the presence of a thick connective tissue capsule and a lack of local invasion. The data indicate that retroviral-mediated transduction of TIMP-2 cDNA into a limited population of tumor cells in vivo is sufficient to increase the accumulation of connective tissue proteins in tumor tissue, to inhibit growth, and to prevent local invasion.
21. GPR56 identifies primary human acute myeloid leukemia cells with high repopulating potential in vivo
- Author
-
Pabst C, Bergeron A, Vp, Lavallée, Yeh J, Gendron P, Gl, Norddahl, Krosl J, Boivin I, Deneault E, Simard J, Imren S, Geneviève Boucher, Eppert K, Herold T, Sk, Bohlander, Humphries K, Lemieux S, Hébert J, Sauvageau G, and Barabé F
22. Overexpression of tissue inhibitor of metalloproteinases-2 by retroviral-mediated gene transfer in vivo inhibits tumor growth and invasion
- Author
-
Imren, S., Kohn, D.B., Shimada, H., Blavier, L., and Declerck, Y.A.
- Subjects
Gene therapy -- Research -- Physiological aspects ,Metalloenzymes -- Physiological aspects -- Research ,Health ,Physiological aspects ,Research - Abstract
According to the authors' abstract of an article published in Proceedings of the National Academy of Sciences of the United States of America, 'Particle-mediated (gene gun) in vivo delivery of [...]
- Published
- 1996
23. Infrequent Resolution of Vaso-Occlusive Crises in Routine Clinical Care Among Patients Mimicking the Exa-Cel Trial Population: A Cohort Study of Medicaid Enrollees.
- Author
-
Mahesri M, Lee SB, Levin R, Imren S, Zhang L, Beukelman T, Titievsky L, and Desai RJ
- Subjects
- Humans, Male, Female, United States, Adult, Adolescent, Young Adult, Child, Cohort Studies, Medicaid, Anemia, Sickle Cell therapy
- Abstract
The CRISPR-based gene editing therapy exagamglogene autotemcel (exa-cel) recently received FDA approval for patients with severe sickle cell disease (SCD). The approval was based on a phase III trial (CLIMB SCD 121), which showed 97% efficacy of this treatment in eliminating vaso occlusive crises (VOCs) for 12 consecutive months. To help contextualize results from this trial, we aimed to investigate the proportion of patients with severe SCD who remain VOC-free for a 1-year period in routine clinical care. Using Medicaid claims data (2000-2018), we identified a cohort of patients, 12-35 years old with severe SCD, defined by ≥ 2 VOCs per year for 2 consecutive years, who met other exa-cel trial inclusion criteria to mimic a trial-like population. A VOC was identified using ICD diagnosis codes during hospitalization and ER visits. The primary outcome was the proportion of patients with no VOCs during a 1-year follow-up. A total of 7,425 patients with severe SCD [mean (SD) age: 20.5 (6.0) years, 54.6% females, 84% African Americans], had a mean of 5.2 VOCs, 5.1 ER visits and 3.5 hospitalizations per year during the baseline period. The proportion of patients with no VOCs during the 1-year follow-up was 7.7% (95% confidence interval: 7.1%-8.3%). In conclusion, less than one in 12 patients with severe SCD achieved VOC-free status within 1 year in routine clinical care. These findings suggest that the high efficacy observed for exa-cel in the trial, if replicated in routine clinical care, could translate into a significant public health impact., (© 2024 The Author(s). Clinical Pharmacology & Therapeutics © 2024 American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2024
- Full Text
- View/download PDF
24. CBFA2T3-GLIS2 model of pediatric acute megakaryoblastic leukemia identifies FOLR1 as a CAR T cell target.
- Author
-
Le Q, Hadland B, Smith JL, Leonti A, Huang BJ, Ries R, Hylkema TA, Castro S, Tang TT, McKay CN, Perkins L, Pardo L, Sarthy J, Beckman AK, Williams R, Idemmili R, Furlan S, Ishida T, Call L, Srivastava S, Loeb AM, Milano F, Imren S, Morris SM, Pakiam F, Olson JM, Loken MR, Brodersen L, Riddell SR, Tarlock K, Bernstein ID, Loeb KR, and Meshinchi S
- Subjects
- Humans, Mice, Animals, Child, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen genetics, Immunotherapy, Adoptive, Female, Male, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion immunology, Oncogene Proteins, Fusion metabolism, Leukemia, Megakaryoblastic, Acute immunology, Leukemia, Megakaryoblastic, Acute genetics, Leukemia, Megakaryoblastic, Acute pathology
- Published
- 2024
- Full Text
- View/download PDF
25. Exagamglogene Autotemcel for Severe Sickle Cell Disease.
- Author
-
Frangoul H, Locatelli F, Sharma A, Bhatia M, Mapara M, Molinari L, Wall D, Liem RI, Telfer P, Shah AJ, Cavazzana M, Corbacioglu S, Rondelli D, Meisel R, Dedeken L, Lobitz S, de Montalembert M, Steinberg MH, Walters MC, Eckrich MJ, Imren S, Bower L, Simard C, Zhou W, Xuan F, Morrow PK, Hobbs WE, and Grupp SA
- Subjects
- Adolescent, Adult, Child, Female, Humans, Male, Young Adult, Antigens, CD34, Busulfan therapeutic use, CRISPR-Cas Systems, Gene Editing, Hematopoietic Stem Cells, Repressor Proteins, Transplantation Conditioning, Cell- and Tissue-Based Therapy methods, Myeloablative Agonists therapeutic use, Europe, North America, Anemia, Sickle Cell complications, Anemia, Sickle Cell genetics, Anemia, Sickle Cell therapy, Fetal Hemoglobin biosynthesis, Fetal Hemoglobin genetics, Hematopoietic Stem Cell Transplantation
- Abstract
Background: Exagamglogene autotemcel (exa-cel) is a nonviral cell therapy designed to reactivate fetal hemoglobin synthesis by means of ex vivo clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing of autologous CD34+ hematopoietic stem and progenitor cells (HSPCs) at the erythroid-specific enhancer region of BCL11A ., Methods: We conducted a phase 3, single-group, open-label study of exa-cel in patients 12 to 35 years of age with sickle cell disease who had had at least two severe vaso-occlusive crises in each of the 2 years before screening. CD34+ HSPCs were edited with the use of CRISPR-Cas9. Before the exa-cel infusion, patients underwent myeloablative conditioning with pharmacokinetically dose-adjusted busulfan. The primary end point was freedom from severe vaso-occlusive crises for at least 12 consecutive months. A key secondary end point was freedom from inpatient hospitalization for severe vaso-occlusive crises for at least 12 consecutive months. The safety of exa-cel was also assessed., Results: A total of 44 patients received exa-cel, and the median follow-up was 19.3 months (range, 0.8 to 48.1). Neutrophils and platelets engrafted in each patient. Of the 30 patients who had sufficient follow-up to be evaluated, 29 (97%; 95% confidence interval [CI], 83 to 100) were free from vaso-occlusive crises for at least 12 consecutive months, and all 30 (100%; 95% CI, 88 to 100) were free from hospitalizations for vaso-occlusive crises for at least 12 consecutive months (P<0.001 for both comparisons against the null hypothesis of a 50% response). The safety profile of exa-cel was generally consistent with that of myeloablative busulfan conditioning and autologous HSPC transplantation. No cancers occurred., Conclusions: Treatment with exa-cel eliminated vaso-occlusive crises in 97% of patients with sickle cell disease for a period of 12 months or more. (CLIMB SCD-121; ClinicalTrials.gov number, NCT03745287.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
26. CBFA2T3-GLIS2 model of pediatric acute megakaryoblastic leukemia identifies FOLR1 as a CAR T cell target.
- Author
-
Le Q, Hadland B, Smith JL, Leonti A, Huang BJ, Ries R, Hylkema TA, Castro S, Tang TT, McKay CN, Perkins L, Pardo L, Sarthy J, Beckman AK, Williams R, Idemmili R, Furlan S, Ishida T, Call L, Srivastava S, Loeb AM, Milano F, Imren S, Morris SM, Pakiam F, Olson JM, Loken MR, Brodersen L, Riddell SR, Tarlock K, Bernstein ID, Loeb KR, and Meshinchi S
- Subjects
- Animals, Child, Child, Preschool, Humans, Infant, Disease Models, Animal, T-Lymphocytes, Transcriptome, Xenograft Model Antitumor Assays, Folate Receptor 1 genetics, Folate Receptor 1 metabolism, Immunotherapy, Adoptive, Leukemia, Megakaryoblastic, Acute genetics, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism
- Abstract
The CBFA2T3-GLIS2 (C/G) fusion is a product of a cryptic translocation primarily seen in infants and early childhood and is associated with dismal outcome. Here, we demonstrate that the expression of the C/G oncogenic fusion protein promotes the transformation of human cord blood hematopoietic stem and progenitor cells (CB HSPCs) in an endothelial cell coculture system that recapitulates the transcriptome, morphology, and immunophenotype of C/G acute myeloid leukemia (AML) and induces highly aggressive leukemia in xenograft models. Interrogating the transcriptome of C/G-CB cells and primary C/G AML identified a library of C/G-fusion-specific genes that are potential targets for therapy. We developed chimeric antigen receptor (CAR) T cells directed against one of the targets, folate receptor α (FOLR1), and demonstrated their preclinical efficacy against C/G AML using in vitro and xenograft models. FOLR1 is also expressed in renal and pulmonary epithelium, raising concerns for toxicity that must be addressed for the clinical application of this therapy. Our findings underscore the role of the endothelial niche in promoting leukemic transformation of C/G-transduced CB HSPCs. Furthermore, this work has broad implications for studies of leukemogenesis applicable to a variety of oncogenic fusion-driven pediatric leukemias, providing a robust and tractable model system to characterize the molecular mechanisms of leukemogenesis and identify biomarkers for disease diagnosis and targets for therapy.
- Published
- 2022
- Full Text
- View/download PDF
27. Correction: Effective drug treatment identified by in vivo screening in a transplantable patient-derived xenograft model of chronic myelomonocytic leukemia.
- Author
-
Kloos A, Mintzas K, Winckler L, Gabdoulline R, Alwie Y, Jyotsana N, Kattre N, Schottmann R, Scherr M, Gupta C, Adams FF, Schwarzer A, Heckl D, Schambach A, Imren S, Humphries RK, Ganser A, Thol F, and Heuser M
- Published
- 2021
- Full Text
- View/download PDF
28. Effective drug treatment identified by in vivo screening in a transplantable patient-derived xenograft model of chronic myelomonocytic leukemia.
- Author
-
Kloos A, Mintzas K, Winckler L, Gabdoulline R, Alwie Y, Jyotsana N, Kattre N, Schottmann R, Scherr M, Gupta C, Adams FF, Schwarzer A, Heckl D, Schambach A, Imren S, Humphries RK, Ganser A, Thol F, and Heuser M
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Azacitidine pharmacology, Clonal Evolution, Disease Models, Animal, Drug Synergism, Female, GTP Phosphohydrolases genetics, Humans, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic mortality, Leukemia, Myelomonocytic, Chronic pathology, Membrane Proteins genetics, Mice, Mutation, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Pyridones pharmacology, Pyridones therapeutic use, Pyrimidinones pharmacology, Pyrimidinones therapeutic use, RNA, Small Interfering genetics, Receptor, Notch1 genetics, Antineoplastic Agents pharmacology, Drug Evaluation, Preclinical methods, Drug Evaluation, Preclinical standards, Leukemia, Myelomonocytic, Chronic drug therapy, Xenograft Model Antitumor Assays methods
- Abstract
To establish novel and effective treatment combinations for chronic myelomonocytic leukemia (CMML) preclinically, we hypothesized that supplementation of CMML cells with the human oncogene Meningioma 1 (MN1) promotes expansion and serial transplantability in mice, while maintaining the functional dependencies of these cells on their original genetic profile. Using lentiviral expression of MN1 for oncogenic supplementation and transplanting transduced primary mononuclear CMML cells into immunocompromised mice, we established three serially transplantable CMML-PDX models with disease-related gene mutations that recapitulate the disease in vivo. Ectopic MN1 expression was confirmed to enhance the proliferation of CMML cells, which otherwise did not engraft upon secondary transplantation. Furthermore, MN1-supplemented CMML cells were serially transplantable into recipient mice up to 5 generations. This robust engraftment enabled an in vivo RNA interference screening targeting CMML-related mutated genes including NRAS, confirming that their functional relevance is preserved in the presence of MN1. The novel combination treatment with azacitidine and the MEK-inhibitor trametinib additively inhibited ERK-phosphorylation and thus depleted the signal from mutated NRAS. The combination treatment significantly prolonged survival of CMML mice compared to single-agent treatment. Thus, we identified the combination of azacitidine and trametinib as an effective treatment in NRAS-mutated CMML and propose its clinical development.
- Published
- 2020
- Full Text
- View/download PDF
29. Comprehensive Transcriptome Profiling of Cryptic CBFA2T3-GLIS2 Fusion-Positive AML Defines Novel Therapeutic Options: A COG and TARGET Pediatric AML Study.
- Author
-
Smith JL, Ries RE, Hylkema T, Alonzo TA, Gerbing RB, Santaguida MT, Eidenschink Brodersen L, Pardo L, Cummings CL, Loeb KR, Le Q, Imren S, Leonti AR, Gamis AS, Aplenc R, Kolb EA, Farrar JE, Triche TJ Jr, Nguyen C, Meerzaman D, Loken MR, Oehler VG, Bolouri H, and Meshinchi S
- Subjects
- Adult, CD56 Antigen genetics, Child, Preschool, Female, Follow-Up Studies, Humans, Infant, Infant, Newborn, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Prognosis, RNA, Messenger, Receptors, GABA-A genetics, Young Adult, Biomarkers, Tumor genetics, Gene Expression Profiling, Leukemia, Myeloid, Acute genetics, MicroRNAs genetics, Mutation, Oncogene Proteins, Fusion genetics
- Abstract
Purpose: A cryptic inv(16)(p13.3q24.3) encoding the CBFA2T3-GLIS2 fusion is associated with poor outcome in infants with acute megakaryocytic leukemia. We aimed to broaden our understanding of the pathogenesis of this fusion through transcriptome profiling., Experimental Design: Available RNA from children and young adults with de novo acute myeloid leukemia (AML; N = 1,049) underwent transcriptome sequencing (mRNA and miRNA). Transcriptome profiles for those with the CBFA2T3-GLIS2 fusion ( N = 24) and without ( N = 1,025) were contrasted to define fusion-specific miRNAs, genes, and pathways. Clinical annotations defined distinct fusion-associated disease characteristics and outcomes., Results: The CBFA2T3-GLIS2 fusion was restricted to infants <3 years old ( P < 0.001), and the presence of this fusion was highly associated with adverse outcome ( P < 0.001) across all morphologic classifications. Further, there was a striking paucity of recurrent cooperating mutations, and transduction of cord blood stem cells with this fusion was sufficient for malignant transformation. CBFA2T3-GLIS2 positive cases displayed marked upregulation of genes with cell membrane/extracellular matrix localization potential, including NCAM1 and GABRE . Additionally, miRNA profiling revealed significant overexpression of mature miR-224 and miR-452 , which are intronic miRNAs transcribed from the GABRE locus. Gene-set enrichment identified dysregulated Hippo, TGFβ, and hedgehog signaling, as well as NCAM1 (CD56) interaction pathways. Therapeutic targeting of fusion-positive leukemic cells with CD56-directed antibody-drug conjugate caused significant cytotoxicity in leukemic blasts., Conclusions: The CBFA2T3-GLIS2 fusion defines a highly refractory entity limited to infants that appears to be sufficient for malignant transformation. Transcriptome profiling elucidated several highly targetable genes and pathways, including the identification of CD56, providing a highly plausible target for therapeutic intervention., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
30. Expansion of primitive human hematopoietic stem cells by culture in a zwitterionic hydrogel.
- Author
-
Bai T, Li J, Sinclair A, Imren S, Merriam F, Sun F, O'Kelly MB, Nourigat C, Jain P, Delrow JJ, Basom RS, Hung HC, Zhang P, Li B, Heimfeld S, Jiang S, and Delaney C
- Subjects
- Animals, Antigens, CD34 metabolism, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, Cell Culture Techniques, Cell Proliferation drug effects, Coculture Techniques, Fetal Blood cytology, Fetal Blood metabolism, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells metabolism, Humans, Mice, Reactive Oxygen Species metabolism, Cell Differentiation drug effects, Cell- and Tissue-Based Therapy, Hematopoietic Stem Cells cytology, Hydrogels pharmacology
- Abstract
The ability to expand hematopoietic stem and progenitor cells (HSPCs) ex vivo is critical to fully realize the potential of HSPC-based therapies. In particular, the application of clinically effective therapies, such as cord blood transplantation, has been impeded because of limited HSPC availability. Here, using 3D culture of human HSPCs in a degradable zwitterionic hydrogel, we achieved substantial expansion of phenotypically primitive CD34
+ cord blood and bone-marrow-derived HSPCs. This culture system led to a 73-fold increase in long-term hematopoietic stem cell (LT-HSC) frequency, as demonstrated by limiting dilution assays, and the expanded HSPCs were capable of hematopoietic reconstitution for at least 24 weeks in immunocompromised mice. Both the zwitterionic characteristics of the hydrogel and the 3D format were important for HSPC self-renewal. Mechanistically, the impact of 3D zwitterionic hydrogel culture on mitigating HSPC differentiation and promoting self-renewal might result from an inhibition of excessive reactive oxygen species (ROS) production via suppression of O2 -related metabolism. HSPC expansion using zwitterionic hydrogels has the potential to facilitate the clinical application of hematopoietic-stem-cell therapies.- Published
- 2019
- Full Text
- View/download PDF
31. Hepatic leukemia factor is a novel leukemic stem cell regulator in DNMT3A, NPM1, and FLT3-ITD triple-mutated AML.
- Author
-
Garg S, Reyes-Palomares A, He L, Bergeron A, Lavallée VP, Lemieux S, Gendron P, Rohde C, Xia J, Jagdhane P, Müller-Tidow C, Lipka DB, Imren S, Humphries RK, Waskow C, Vick B, Jeremias I, Richard-Carpentier G, Hébert J, Sauvageau G, Zaugg JB, Barabé F, and Pabst C
- Subjects
- Animals, Biomarkers, Cell Cycle genetics, Cell Line, Tumor, Computational Biology methods, DNA Methyltransferase 3A, Disease Models, Animal, Gene Duplication, Gene Expression Profiling, Humans, Immunophenotyping, Mice, Transgenic, Mutation, Nucleophosmin, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Tandem Repeat Sequences, Transcription Initiation Site, Transcriptome, Basic-Leucine Zipper Transcription Factors metabolism, DNA (Cytosine-5-)-Methyltransferases genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Neoplastic Stem Cells metabolism, Nuclear Proteins genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
FLT3, DNMT3A , and NPM1 are the most frequently mutated genes in cytogenetically normal acute myeloid leukemia (AML), but little is known about how these mutations synergize upon cooccurrence. Here we show that triple-mutated AML is characterized by high leukemia stem cell (LSC) frequency, an aberrant leukemia-specific GPR56
high CD34low immunophenotype, and synergistic upregulation of Hepatic Leukemia Factor ( HLF ). Cell sorting based on the LSC marker GPR56 allowed isolation of triple-mutated from DNMT3A/NPM1 double-mutated subclones. Moreover, in DNMT3A R882-mutated patients, CpG hypomethylation at the HLF transcription start site correlated with high HLF mRNA expression, which was itself associated with poor survival. Loss of HLF via CRISPR/Cas9 significantly reduced the CD34+ GPR56+ LSC compartment of primary human triple-mutated AML cells in serial xenotransplantation assays. HLF knockout cells were more actively cycling when freshly harvested from mice, but rapidly exhausted when reintroduced in culture. RNA sequencing of primary human triple-mutated AML cells after shRNA-mediated HLF knockdown revealed the NOTCH target Hairy and Enhancer of Split 1 ( HES1 ) and the cyclin-dependent kinase inhibitor CDKN1C/p57 as novel targets of HLF, potentially mediating these effects. Overall, our data establish HLF as a novel LSC regulator in this genetically defined high-risk AML subgroup., (© 2019 by The American Society of Hematology.)- Published
- 2019
- Full Text
- View/download PDF
32. UM171 Enhances Lentiviral Gene Transfer and Recovery of Primitive Human Hematopoietic Cells.
- Author
-
Ngom M, Imren S, Maetzig T, Adair JE, Knapp DJHF, Chagraoui J, Fares I, Bordeleau ME, Sauvageau G, Leboulch P, Eaves C, and Humphries RK
- Abstract
Enhanced gene transfer efficiencies and higher yields of transplantable transduced human hematopoietic stem cells are continuing goals for improving clinical protocols that use stemcell-based gene therapies. Here, we examined the effect of the HSC agonist UM171 on these endpoints in both in vitro and in vivo systems. Using a 22-hr transduction protocol, we found that UM171 significantly enhances both the lentivirus-mediated transduction and yield of CD34
+ and CD34+ CD45RA- hematopoietic cells from human cord blood to give a 6-fold overall higher recovery of transduced hematopoietic stem cells, including cells with long-term lympho-myeloid repopulating activity in immunodeficient mice. The ability of UM171 to enhance gene transfer to primitive cord blood hematopoietic cells extended to multiple lentiviral pseudotypes, gamma retroviruses, and non-integrating lentiviruses and to adult bone marrow cells. UM171, thus, provides an interesting reagent for improving the ex vivo production of gene-modified cells and for reducing requirements of virus for a broad range of applications.- Published
- 2018
- Full Text
- View/download PDF
33. A Lentiviral Fluorescent Genetic Barcoding System for Flow Cytometry-Based Multiplex Tracking.
- Author
-
Maetzig T, Ruschmann J, Lai CK, Ngom M, Imren S, Rosten P, Norddahl GL, von Krosigk N, Sanchez Milde L, May C, Selich A, Rothe M, Dhillon I, Schambach A, and Humphries RK
- Subjects
- Cell Differentiation, Codon, Flow Cytometry, Gene Order, Gene Transfer Techniques, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Homeodomain Proteins genetics, Humans, Luminescent Proteins metabolism, MicroRNAs genetics, Reproducibility of Results, Transduction, Genetic, Cell Tracking methods, Gene Expression, Genes, Reporter, Genetic Vectors genetics, Lentivirus genetics, Luminescent Proteins genetics
- Abstract
Retroviral integration site analysis and barcoding have been instrumental for multiplex clonal fate mapping, although their use imposes an inherent delay between sample acquisition and data analysis. Monitoring of multiple cell populations in real time would be advantageous, but multiplex assays compatible with flow cytometric tracking of competitive growth behavior are currently limited. We here describe the development and initial validation of three generations of lentiviral fluorescent genetic barcoding (FGB) systems that allow the creation of 26, 14, or 6 unique labels. Color-coded populations could be tracked in multiplex in vitro assays for up to 28 days by flow cytometry using all three vector systems. Those involving lower levels of multiplexing eased color-code generation and the reliability of vector expression and enabled functional in vitro and in vivo studies. In proof-of-principle experiments, FGB vectors facilitated in vitro multiplex screening of microRNA (miRNA)-induced growth advantages, as well as the in vivo recovery of color-coded progeny of murine and human hematopoietic stem cells. This novel series of FGB vectors provides new tools for assessing comparative growth properties in in vitro and in vivo multiplexing experiments, while simultaneously allowing for a reduction in sample numbers by up to 26-fold., (Copyright © 2017 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
34. Real Time Immunophenotyping of Leukocyte Subsets Early after Double Cord Blood Transplantation Predicts Graft Function.
- Author
-
Li J, Nicoud I, Blake J, Oliver D, Cox E, Heimfeld S, Milano F, Imren S, and Delaney C
- Subjects
- Adolescent, Adult, Child, Cord Blood Stem Cell Transplantation standards, Delayed Graft Function, Female, Flow Cytometry, Humans, Kinetics, Leukocytes immunology, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear immunology, Male, Middle Aged, Prognosis, Prospective Studies, Young Adult, Cord Blood Stem Cell Transplantation methods, Graft Survival, Immunophenotyping, Leukocytes cytology
- Abstract
Cord blood transplantation (CBT) recipients are at increased risk for delayed engraftment and primary graft failure, complications that are often indistinguishable early post-transplantation. Current assays fail to accurately identify recipients with slow hematopoietic recovery and distinguish them from those with pending graft failure. To address this, we prospectively examined the kinetics of immune cell subset recovery in the peripheral blood of 39 patients on days +7 and +14 after double-unit CBT (dCBT) by multiparametric flow cytometry analysis, which we term real-time immunophenotyping (RTIP). RTIP analysis at day +14 revealed distinctive patterns of reconstitution and, importantly, identified patients with slow hematopoietic recovery who went on to engraft. Strikingly, higher absolute numbers of circulating monocytes and natural killer cells at day +14 were predictive of engraftment, but only the absolute number of circulating monocytes was significantly correlated with time to engraftment. This is the first evidence that RTIP on patient peripheral blood mononuclear cells early after dCBT is technically feasible and can be used as a "signature" for predicting the kinetics of hematopoietic recovery. Furthermore, RTIP is a time- and cost-efficient methodology that has the potential to become a clinically feasible diagnostic tool to guide therapeutic interventions in high-risk patients; therefore, its utility should be evaluated in a large cohort of patients., (Copyright © 2017 The American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
35. Notch-Expanded Murine Hematopoietic Stem and Progenitor Cells Mitigate Death from Lethal Radiation and Convey Immune Tolerance in Mismatched Recipients.
- Author
-
Milano F, Merriam F, Nicoud I, Li J, Gooley TA, Heimfeld S, Imren S, and Delaney C
- Subjects
- Acute Radiation Syndrome immunology, Acute Radiation Syndrome metabolism, Animals, Cells, Cultured, Disease Models, Animal, Female, Hematopoietic Stem Cells immunology, Hematopoietic Stem Cells metabolism, Intracellular Signaling Peptides and Proteins genetics, Male, Membrane Proteins genetics, Mice, Inbred BALB C, Mice, Inbred C3H, Recovery of Function, Signal Transduction, Time Factors, Acute Radiation Syndrome therapy, Cell Proliferation, Graft Survival, Hematopoiesis, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells radiation effects, Histocompatibility, Histocompatibility Antigens immunology, Intracellular Signaling Peptides and Proteins metabolism, Membrane Proteins metabolism, Skin Transplantation, Transplantation Tolerance
- Abstract
The hematopoietic syndrome of acute radiation syndrome (h-ARS) is characterized by severe bone marrow aplasia, resulting in a significant risk for bleeding, infections, and death. To date, clinical management of h-ARS is limited to supportive care dictated by the level of radiation exposure, with a high incidence of mortality in those exposed to high radiation doses. The ideal therapeutic agent would be an immediately available, easily distributable single-agent therapy capable of rapid in vivo hematopoietic reconstitution until recovery of autologous hematopoiesis occurs. Using a murine model of h-ARS, we herein demonstrate that infusion of ex vivo expanded murine hematopoietic stem and progenitor cells (HSPCs) into major histocompatibility complex mismatched recipient mice exposed to a lethal dose of ionizing radiation (IR) led to rapid myeloid recovery and improved survival. Survival benefit was significant in a dose-dependent manner even when infusion of the expanded cell therapy was delayed 3 days after lethal IR exposure. Most surviving mice (80%) demonstrated long-term in vivo persistence of donor T cells at low levels, and none had evidence of graft versus host disease. Furthermore, survival of donor-derived skin grafts was significantly prolonged in recipients rescued from h-ARS by infusion of the mismatched expanded cell product. These findings provide evidence that ex vivo expanded mismatched HSPCs can provide rapid, high-level hematopoietic reconstitution, mitigate IR-induced mortality, and convey donor-specific immune tolerance in a murine h-ARS model. Stem Cells Translational Medicine 2017;6:566-575., (© 2016 The Authors Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.)
- Published
- 2017
- Full Text
- View/download PDF
36. UV-inactivated HSV-1 potently activates NK cell killing of leukemic cells.
- Author
-
Samudio I, Rezvani K, Shaim H, Hofs E, Ngom M, Bu L, Liu G, Lee JT, Imren S, Lam V, Poon GF, Ghaedi M, Takei F, Humphries K, Jia W, and Krystal G
- Subjects
- Cell Degranulation immunology, Cell Movement immunology, Female, Humans, Interleukin-15 immunology, Interleukin-2 immunology, Jurkat Cells, Male, NF-kappa B immunology, Protein Kinase C immunology, Signal Transduction immunology, Toll-Like Receptor 2 immunology, Herpesvirus 1, Human immunology, Immunity, Cellular, Killer Cells, Natural immunology, Leukemia immunology, Ultraviolet Rays, Virus Inactivation radiation effects
- Abstract
Herein we demonstrate that oncolytic herpes simplex virus-1 (HSV-1) potently activates human peripheral blood mononuclear cells (PBMCs) to lyse leukemic cell lines and primary acute myeloid leukemia samples, but not healthy allogeneic lymphocytes. Intriguingly, we found that UV light-inactivated HSV-1 (UV-HSV-1) is equally effective in promoting PBMC cytolysis of leukemic cells and is 1000- to 10 000-fold more potent at stimulating innate antileukemic responses than UV-inactivated cytomegalovirus, vesicular stomatitis virus, reovirus, or adenovirus. Mechanistically, UV-HSV-1 stimulates PBMC cytolysis of leukemic cells, partly via Toll-like receptor-2/protein kinase C/nuclear factor-κB signaling, and potently stimulates expression of CD69, degranulation, migration, and cytokine production in natural killer (NK) cells, suggesting that surface components of UV-HSV-1 directly activate NK cells. Importantly, UV-HSV-1 synergizes with interleukin-15 (IL-15) and IL-2 in inducing activation and cytolytic activity of NK cells. Additionally, UV-HSV-1 stimulates glycolysis and fatty acid oxidation-dependent oxygen consumption in NK cells, but only glycolysis is required for their enhanced antileukemic activity. Last, we demonstrate that T cell-depleted human PBMCs exposed to UV-HSV-1 provide a survival benefit in a murine xenograft model of human acute myeloid leukemia (AML). Taken together, our results support the preclinical development of UV-HSV-1 as an adjuvant, alone or in combination with IL-15, for allogeneic donor mononuclear cell infusions to treat AML., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
37. GPR56 identifies primary human acute myeloid leukemia cells with high repopulating potential in vivo.
- Author
-
Pabst C, Bergeron A, Lavallée VP, Yeh J, Gendron P, Norddahl GL, Krosl J, Boivin I, Deneault E, Simard J, Imren S, Boucher G, Eppert K, Herold T, Bohlander SK, Humphries K, Lemieux S, Hébert J, Sauvageau G, and Barabé F
- Subjects
- Adult, Animals, Cell Separation, Cells, Cultured, HEK293 Cells, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute mortality, Mice, Mice, Inbred NOD, Mice, Transgenic, Neoplastic Stem Cells physiology, Receptors, G-Protein-Coupled physiology, Survival Analysis, Biomarkers, Tumor metabolism, Cell Proliferation, Leukemia, Myeloid, Acute pathology, Neoplastic Stem Cells pathology, Receptors, G-Protein-Coupled metabolism
- Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous hematologic malignancy, which is initiated and driven by a rare fraction of leukemia stem cells (LSCs). Despite the difficulties of identifying a common LSC phenotype, there is increasing evidence that high expression of stem cell gene signatures is associated with poor clinical outcome. Identification of functionally distinct subpopulations in this disease is therefore crucial to dissecting the molecular machinery underlying LSC self-renewal. Here, we combined next-generation sequencing technology with in vivo assessment of LSC frequencies and identified the adhesion G protein-coupled receptor 56 (GPR56) as a novel and stable marker for human LSCs for the majority of AML samples. High GPR56 expression was significantly associated with high-risk genetic subgroups and poor outcome. Analysis of GPR56 in combination with CD34 expression revealed engraftment potential of GPR56(+)cells in both the CD34(-)and CD34(+)fractions, thus defining a novel LSC compartment independent of the CD34(+)CD38(-)LSC phenotype., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
38. Modeling de novo leukemogenesis from human cord blood with MN1 and NUP98HOXD13.
- Author
-
Imren S, Heuser M, Gasparetto M, Beer PA, Norddahl GL, Xiang P, Chen L, Berg T, Rhyasen GW, Rosten P, Park G, Moon Y, Weng AP, Eaves CJ, and Humphries RK
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Mice, Inbred NOD, Mice, SCID, Mice, Transgenic, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Oncogene Proteins, Fusion genetics, Trans-Activators, Tumor Suppressor Proteins genetics, Cell Transformation, Neoplastic metabolism, Fetal Blood metabolism, Leukemia, Myeloid, Acute metabolism, Neoplasms, Experimental metabolism, Oncogene Proteins, Fusion metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Leukemic transformation of human cells is a complex process. Here we show that forced expression of MN1 in primitive human cord blood cells maintained on stromal cells in vitro induces a transient, but not serially transplantable, myeloproliferation in engrafted mice. However, cotransduction of an activated HOX gene (NUP98HOXD13) with MN1 induces a serially transplantable acute myeloid leukemia (AML). Further characterization of the leukemic cells generated from the dually transduced cells showed the activation of stem cell gene expression signatures also found in primary human AML. These findings show a new forward genetic model of human leukemogenesis and further highlight the relevance of homeobox transcription factors in the transformation process., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
39. Cord blood expansion. Pyrimidoindole derivatives are agonists of human hematopoietic stem cell self-renewal.
- Author
-
Fares I, Chagraoui J, Gareau Y, Gingras S, Ruel R, Mayotte N, Csaszar E, Knapp DJ, Miller P, Ngom M, Imren S, Roy DC, Watts KL, Kiem HP, Herrington R, Iscove NN, Humphries RK, Eaves CJ, Cohen S, Marinier A, Zandstra PW, and Sauvageau G
- Subjects
- Animals, Cell Culture Techniques, Fetal Blood cytology, Fetal Blood physiology, Genetic Therapy methods, Hematopoiesis physiology, Hematopoietic Stem Cell Transplantation methods, Hematopoietic Stem Cells physiology, Humans, Immunocompromised Host, Indoles chemistry, Mice, Pyrimidines chemistry, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Fetal Blood drug effects, Hematopoiesis drug effects, Hematopoietic Stem Cells drug effects, Indoles pharmacology, Pyrimidines pharmacology, Receptors, Aryl Hydrocarbon antagonists & inhibitors, Regeneration drug effects
- Abstract
The small number of hematopoietic stem and progenitor cells in cord blood units limits their widespread use in human transplant protocols. We identified a family of chemically related small molecules that stimulates the expansion ex vivo of human cord blood cells capable of reconstituting human hematopoiesis for at least 6 months in immunocompromised mice. The potent activity of these newly identified compounds, UM171 being the prototype, is independent of suppression of the aryl hydrocarbon receptor, which targets cells with more-limited regenerative potential. The properties of UM171 make it a potential candidate for hematopoietic stem cell transplantation and gene therapy., (Copyright © 2014, American Association for the Advancement of Science.)
- Published
- 2014
- Full Text
- View/download PDF
40. Varying levels of aldehyde dehydrogenase activity in adult murine marrow hematopoietic stem cells are associated with engraftment and cell cycle status.
- Author
-
Gasparetto M, Sekulovic S, Zakaryan A, Imren S, Kent DG, Humphries RK, Vasiliou V, and Smith C
- Subjects
- Adult Stem Cells cytology, Adult Stem Cells transplantation, Animals, Antigens, Differentiation genetics, Hematopoietic Stem Cells cytology, Humans, Mice, Mice, Transgenic, Transplantation, Homologous, Adult Stem Cells metabolism, Aldehyde Dehydrogenase metabolism, Antigens, Differentiation metabolism, G1 Phase physiology, Graft Survival physiology, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells metabolism
- Abstract
Aldehyde dehydrogenase (ALDH) activity is a widely used marker for human hematopoietic stem cells (HSCs), yet its relevance and role in murine HSCs remain unclear. We found that murine marrow cells with a high level of ALDH activity as measured by Aldefluor staining (ALDH(br) cells) do not contain known HSCs or progenitors. In contrast, highly enriched murine HSCs defined by the CD48(-)EPCR(+) and other phenotypes contain two subpopulations, one that stains dimly with Aldefluor (ALDH(dim)) and one that stains at intermediate levels (ALDH(int)). The CD48(-)EPCR(+)ALDH(dim) cells are virtually all in G(0) and yield high levels of engraftment via both intravenous and intrabone routes. In contrast the CD48(-)EPCR(+)ALDH(int) cells are virtually all in G(1), have little intravenous engraftment potential, and yet can engraft long-term after intrabone transplantation. These data demonstrate that Aldefluor staining of unfractionated murine marrow does not identify known HSCs or progenitors. However, varying levels of Aldefluor staining when combined with CD48 and EPCR detection can identify novel populations in murine marrow including a highly enriched population of resting HSCs and a previously unknown HSC population in G(1) with an intravenous engraftment defect., (Copyright © 2012 ISEH - Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
41. Aldehyde dehydrogenases are regulators of hematopoietic stem cell numbers and B-cell development.
- Author
-
Gasparetto M, Sekulovic S, Brocker C, Tang P, Zakaryan A, Xiang P, Kuchenbauer F, Wen M, Kasaian K, Witty MF, Rosten P, Chen Y, Imren S, Duester G, Thompson DC, Humphries RK, Vasiliou V, and Smith C
- Subjects
- Aldehyde Dehydrogenase biosynthesis, Aldehyde Dehydrogenase deficiency, Aldehyde Dehydrogenase genetics, Aldehyde Dehydrogenase 1 Family, Aldehydes metabolism, Animals, Animals, Congenic, B-Lymphocytes cytology, Bone Marrow Transplantation, Cell Count, Cell Cycle physiology, Cell Lineage, Cells, Cultured cytology, Cells, Cultured metabolism, Colony-Forming Units Assay, DNA Damage, Enzyme Induction, Gene Expression Regulation physiology, Hematopoietic Stem Cells cytology, Lymphopenia genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Radiation Chimera, Reactive Oxygen Species metabolism, Retinal Dehydrogenase, Signal Transduction physiology, p38 Mitogen-Activated Protein Kinases metabolism, Aldehyde Dehydrogenase physiology, B-Lymphocytes enzymology, Hematopoiesis physiology, Hematopoietic Stem Cells enzymology
- Abstract
High levels of the aldehyde dehydrogenase isoform ALDH1A1 are expressed in hematopoietic stem cells (HSCs); however, its importance in these cells remains unclear. Consistent with an earlier report, we find that loss of ALDH1A1 does not affect HSCs. Intriguingly, however, we find that ALDH1A1 deficiency is associated with increased expression of the ALDH3A1 isoform, suggesting its potential to compensate for ALDH1A1. Mice deficient in ALDH3A1 have a block in B-cell development as well as abnormalities in cell cycling, intracellular signaling, and gene expression. Early B cells from these mice exhibit excess reactive oxygen species and reduced metabolism of reactive aldehydes. Mice deficient in both ALDH3A1 and ALDH1A1 have reduced numbers of HSCs as well as aberrant cell cycle distribution, increased reactive oxygen species levels, p38 mitogen-activated protein kinase activity and sensitivity to DNA damage. These findings demonstrate that ALDH3A1 can compensate for ALDH1A1 in bone marrow and is important in B-cell development, both ALDH1A1 and 3A1 are important in HSC biology; and these effects may be due, in part, to changes in metabolism of reactive oxygen species and reactive aldehydes., (Copyright © 2012 ISEH - Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
42. Functional regulation of pre-B-cell leukemia homeobox interacting protein 1 (PBXIP1/HPIP) in erythroid differentiation.
- Author
-
Manavathi B, Lo D, Bugide S, Dey O, Imren S, Weiss MJ, and Humphries RK
- Subjects
- Antigens, CD34 metabolism, CCAAT-Enhancer-Binding Protein-alpha metabolism, CCCTC-Binding Factor, Chromatin drug effects, Chromatin genetics, Chromatin metabolism, Co-Repressor Proteins, Dimethyl Sulfoxide pharmacology, Erythroid Cells drug effects, Erythropoietin pharmacology, GATA1 Transcription Factor metabolism, Gene Expression Regulation drug effects, Gene Knockdown Techniques, HL-60 Cells, Hematopoiesis drug effects, Humans, K562 Cells, Myeloid Cells cytology, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Repressor Proteins metabolism, Signal Transduction drug effects, Transcription Factors deficiency, Transcription Factors genetics, Transcription, Genetic drug effects, Cell Differentiation drug effects, Erythroid Cells cytology, Erythroid Cells metabolism, Transcription Factors metabolism
- Abstract
Pre-B-cell leukemia homeobox interacting protein 1 or human PBX1 interacting protein (PBXIP1/HPIP) is a co-repressor of pre-B-cell leukemia homeobox 1 (PBX1) and is also known to regulate estrogen receptor functions by associating with the microtubule network. Despite its initial discovery in the context of hematopoietic cells, little is yet known about the role of HPIP in hematopoiesis. Here, we show that lentivirus-mediated overexpression of HPIP in human CD34(+) cells enhances hematopoietic colony formation in vitro, whereas HPIP knockdown leads to a reduction in the number of such colonies. Interestingly, erythroid colony number was significantly higher in HPIP-overexpressing cells. In addition, forced expression of HPIP in K562 cells, a multipotent erythro-megakaryoblastic leukemia cell line, led to an induction of erythroid differentiation. HPIP overexpression in both CD34(+) and K562 cells was associated with increased activation of the PI3K/AKT pathway, and corresponding treatment with a PI3K-specific inhibitor, LY-294002, caused a reduction in clonogenic progenitor number in HPIP-expressing CD34(+) cells and decreased K562 cell differentiation. Combined, these findings point to an important role of the PI3K/AKT pathway in mediating HPIP-induced effects on the growth and differentiation of hematopoietic cells. Interestingly, HPIP gene expression was found to be induced in K562 cells in response to erythroid differentiation signals such as DMSO and erythropoietin. The erythroid lineage-specific transcription factor GATA1 binds to the HPIP promoter and activates HPIP gene transcription in a CCCTC-binding factor (CTCF)-dependent manner. Co-immunoprecipitation and co-localization experiments revealed the association of CTCF with GATA1 indicating the recruitment of CTCF/GATA1 transcription factor complex onto the HPIP promoter. Together, this study provides evidence that HPIP is a target of GATA1 and CTCF in erythroid cells and plays an important role in erythroid differentiation by modulating the PI3K/AKT pathway.
- Published
- 2012
- Full Text
- View/download PDF
43. Insights into leukemia-initiating cell frequency and self-renewal from a novel canine model of leukemia.
- Author
-
Imren S, Zhang XB, Humphries RK, and Kiem HP
- Subjects
- Animals, Cell Line, Tumor, Dogs, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Transplantation, Disease Models, Animal, Leukemia pathology, Neoplastic Stem Cells pathology
- Abstract
Objective: Leukemia-initiating cells (LICs) have been the subject of considerable investigation because of their ability to self-renew and maintain leukemia. Thus, selective targeting and killing of LICs would provide highly efficient and novel therapeutic strategies. Here we explored whether we could use a canine leukemia cell line (G374) derived from a dog that received HOXB4-transduced repopulating cells to study leukemia in the murine xenograft model and the dog., Materials and Methods: G374 cells were infused in dogs intravenously and in nonobese diabetic/severe combined immunodeficient and nonobese diabetic/severe combined immunodeficient/IL2Rγ(null) mice either intravenously or directly into the bone cavity. Animals were bled to track engraftment and proliferation of G374 cells, and were sacrificed when they appeared ill., Results: We found that canine LICs are capable of sustained in vitro self-renewal while maintaining their ability to induce acute myeloid leukemia, which resembles human disease in both dogs and mice. Furthermore, we developed a novel strategy for the quantification of LIC frequency in large animals and showed that this frequency was highly comparable to that determined by limited dilution in mouse xenotransplants. We also demonstrated, using single-cell analysis, that LICs are heterogeneous in their self-renewal capacity and regenerate a leukemic cell population consistent with a hierarchical leukemia model., Conclusions: The availability of this novel framework should accelerate the characterization of LICs and the translation of animal studies into clinical trials., (Copyright © 2011. Published by Elsevier Inc.)
- Published
- 2011
- Full Text
- View/download PDF
44. Identification of E74-like factor 1 (ELF1) as a transcriptional regulator of the Hox cofactor MEIS1.
- Author
-
Xiang P, Lo C, Argiropoulos B, Lai CB, Rouhi A, Imren S, Jiang X, Mager D, and Humphries RK
- Subjects
- Gene Knockdown Techniques, HL-60 Cells, Humans, K562 Cells, Myeloid Ecotropic Viral Integration Site 1 Protein, Nuclear Proteins genetics, Transcription Factors genetics, Fetal Blood metabolism, Gene Expression Regulation, Leukemic, Homeodomain Proteins biosynthesis, Leukemia metabolism, Neoplasm Proteins biosynthesis, Nuclear Proteins metabolism, Response Elements, Transcription Factors metabolism
- Abstract
Objective: Myeloid ectropic viral integration site 1 (MEIS1) is a Hox cofactor known for its role in development and is strongly linked to normal and leukemic hematopoiesis. Although previous studies have focused on identifying protein partners of MEIS1 and its transcriptionally regulated targets, little is known about the upstream transcriptional regulators of this tightly regulated gene. Understanding the regulation of MEIS1 is important to understanding normal hematopoiesis and leukemogenesis., Materials and Methods: Here we describe our studies focusing on the evolutionary conserved putative MEIS1 promoter region. Phylogenetic sequence analysis and reporter assays in MEIS1-expressing (K562) and nonexpressing (HL60) leukemic cell line models were used to identify key regulatory regions and potential transcription factor binding sites within the candidate promoter region followed by functional and expression studies of one identified regulator in both cell lines and primary human cord blood and leukemia samples., Results: Chromatin status of MEIS1 promoter region is associated with MEIS1 expression. Truncation and mutation studies coupled with reporter assays revealed that a conserved ETS family member binding site located 289 bp upstream of the annotated human MEIS1 transcription start site is required for promoter activity. Of the three ETS family members tested, only ELF1 was enriched on the MEIS1 promoter as assessed by both electrophoretic mobility shift assay and chromatin immunoprecipitation experiments in K562. This finding was confirmed in MEIS1-expressing primary human samples. Moreover, small interfering RNA-mediated knockdown of ELF1 in K562 cells was associated with a decreased MEIS1 expression., Conclusions: We conclude that the ETS transcription factor ELF1 is an important positive regulator of MEIS1 expression.
- Published
- 2010
- Full Text
- View/download PDF
45. High level in vitro expansion of murine hematopoietic stem cells.
- Author
-
Sekulovic S, Imren S, and Humphries K
- Subjects
- Animals, Cell Proliferation, Cells, Cultured, Mice, Mice, Inbred C57BL, Nuclear Pore Complex Proteins metabolism, Time Factors, Transduction, Genetic, Cell Culture Techniques methods, Hematopoietic Stem Cells cytology
- Abstract
Development of strategies to extensively expand hematopoietic stem cells (HSCs) in vitro will be a major factor in enhancing the success of a range of transplant-based therapies for malignant and genetic disorders. In addition to potential clinical applications, the ability to increase the number of HSCs in culture will facilitate investigations into the mechanisms underlying self-renewal. In this unit, we describe a robust strategy for consistently achieving over 1000-fold net expansion of HSCs in short-term in vitro culture by using novel engineered fusions of the N-terminal domain of nucleoporin 98 (NUP98) and the homeodomain of the hox transcription factor, HOXA10 (so called NUP98-HOXA10hd fusion). We also provide a detailed protocol for monitoring the magnitude of HSC expansion in culture by limiting dilution assay of competitive lympho-myeloid repopulating units (CRU Assay). These procedures provide new possibilities for achieving significant numbers of HSCs in culture, as well as for studying HSCs biochemically and genetically.
- Published
- 2008
- Full Text
- View/download PDF
46. High-level beta-globin expression and preferred intragenic integration after lentiviral transduction of human cord blood stem cells.
- Author
-
Imren S, Fabry ME, Westerman KA, Pawliuk R, Tang P, Rosten PM, Nagel RL, Leboulch P, Eaves CJ, and Humphries RK
- Subjects
- Animals, Cell Lineage, Cell Transplantation, Cells, Cultured, Chromosomes, Human, Gene Transfer Techniques, Genetic Therapy methods, Genetic Vectors genetics, Globins genetics, Hematopoietic Stem Cells cytology, Humans, Lentivirus genetics, Mice, Mice, Inbred NOD, Mice, SCID, Transgenes, Fetal Blood cytology, Genetic Vectors metabolism, Globins metabolism, Hematopoietic Stem Cells physiology, Lentivirus metabolism, Transduction, Genetic
- Abstract
Transplantation of genetically corrected autologous hematopoietic stem cells is an attractive approach for the cure of sickle-cell disease and beta-thalassemia. Here, we infected human cord blood cells with a self-inactivating lentiviral vector encoding an anti-sickling betaA-T87Q-globin transgene and analyzed the transduced progeny produced over a 6-month period after transplantation of the infected cells directly into sublethally irradiated NOD/LtSz-scid/scid mice. Approximately half of the human erythroid and myeloid progenitors regenerated in the mice containing the transgene, and erythroid cells derived in vitro from these in vivo-regenerated cells produced high levels of betaA-T87Q-globin protein. Linker-mediated PCR analysis identified multiple transgene-positive clones in all mice analyzed with 2.1 +/- 0.1 integrated proviral copies per cell. Genomic sequencing of vector-containing fragments showed that 86% of the proviral inserts had occurred within genes, including several genes implicated in human leukemia. These findings indicate effective transduction of very primitive human cord blood cells with a candidate therapeutic lentiviral vector resulting in the long-term and robust, erythroid-specific production of therapeutically relevant levels of beta-globin protein. However, the frequency of proviral integration within genes that regulate hematopoiesis points to a need for additional safety modifications.
- Published
- 2004
- Full Text
- View/download PDF
47. Differential effects of granulocyte colony-stimulating factor on marrow- and blood-derived hematopoietic and immune cell populations in healthy human donors.
- Author
-
Shier LR, Schultz KR, Imren S, Regan J, Issekutz A, Sadek I, Gilman A, Luo Z, Panzarella T, Eaves CJ, and Couban S
- Subjects
- Adult, Antigens, CD blood, Antigens, CD34 blood, Bone Marrow Cells drug effects, Hematopoietic Stem Cells drug effects, Humans, Reference Values, Bone Marrow Cells cytology, Granulocyte Colony-Stimulating Factor pharmacology, Hematopoietic Stem Cell Mobilization methods, Hematopoietic Stem Cells cytology, Lymphocytes immunology
- Abstract
A recent phase III trial comparing granulocyte colony-stimulating factor (G-CSF)-stimulated bone marrow (G-BM) and G-CSF-mobilized peripheral blood (G-PB) in matched sibling allograft recipients showed that G-BM produced a similar hematologic recovery but a reduced incidence of extensive chronic graft-versus-host disease, indicating differences in the cell populations infused. As a first step toward identifying these differences, we treated a group of healthy adult humans with 4 daily doses of G-CSF 10 microg/kg and monitored the effects on various hematopoietic and immune cell types in the PB and BM over 12 days. G-CSF treatment caused rapid and large but transient increases in the number of circulating CD34+ cells, colony-forming cells, and long-term culture-initiating cells and in the short-term repopulating activity detectable in nonobese diabetic/severe combined immunodeficiency/beta2-microglobulin-null mice. Similar but generally less marked changes occurred in the same cell populations in the BM. G-CSF also caused transient perturbations in some immune cell types in both PB and BM: these included a greater increase in the frequency of naive B cells and CD123+ dendritic cells in the BM. The rapidity of the effects of G-CSF on the early progenitor activity of the BM provides a rationale for the apparent equivalence in rates of hematologic recovery obtained with G-BM and G-PB allotransplants. Accompanying effects on immune cell populations are consistent with a greater ability of G-BM to promote tolerance in allogeneic recipients, and this could contribute to a lower rate of chronic graft-versus-host disease.
- Published
- 2004
- Full Text
- View/download PDF
48. Permanent and panerythroid correction of murine beta thalassemia by multiple lentiviral integration in hematopoietic stem cells.
- Author
-
Imren S, Payen E, Westerman KA, Pawliuk R, Fabry ME, Eaves CJ, Cavilla B, Wadsworth LD, Beuzard Y, Bouhassira EE, Russell R, London IM, Nagel RL, Leboulch P, and Humphries RK
- Subjects
- 3T3 Cells, Animals, COS Cells, Chlorocebus aethiops, Disease Models, Animal, Erythrocytes cytology, Gene Expression, Humans, Liver pathology, Mice, Mice, Inbred C57BL, Spleen pathology, Erythroid Precursor Cells metabolism, Genetic Vectors genetics, Globins genetics, Lentivirus genetics, Virus Integration, beta-Thalassemia therapy
- Abstract
Achieving long-term pancellular expression of a transferred gene at therapeutic level in a given hematopoietic lineage remains an important goal of gene therapy. Advances have recently been made in the genetic correction of the hemoglobinopathies by means of lentiviral vectors and large locus control region (LCR) derivatives. However, panerythroid beta globin gene expression has not yet been achieved in beta thalassemic mice because of incomplete transduction of the hematopoietic stem cell compartment and position effect variegation of proviruses integrated at a single copy per genome. Here, we report the permanent, panerythroid correction of severe beta thalassemia in mice, resulting from a homozygous deletion of the beta major globin gene, by transplantation of syngeneic bone marrow transduced with an HIV-1-derived [beta globin gene/LCR] lentiviral vector also containing the Rev responsive element and the central polypurine tract/DNA flap. The viral titers produced were high enough to achieve transduction of virtually all of the hematopoietic stem cells in the graft with an average of three integrated proviral copies per genome in all transplanted mice; the transduction was sustained for >7 months in both primary and secondary transplants, at which time approximately 95% of the red blood cells in all mice contained human beta globin contributing to 32 +/- 4% of all beta-like globin chains. Hematological parameters approached complete phenotypic correction, as assessed by hemoglobin levels and reticulocyte and red blood cell counts. All circulating red blood cells became and remained normocytic and normochromic, and their density was normalized. Free alpha globin chains were completely cleared from red blood cell membranes, splenomegaly abated, and iron deposit was almost eliminated in liver sections. These findings indicate that virtually complete transduction of the hematopoietic stem cell compartment can be achieved by high-titer lentiviral vectors and that position effect variegation can be mitigated by multiple events of proviral integration to yield balanced, panerythroid expression. These results provide a solid foundation for the initiation of human clinical trials in beta thalassemia patients.
- Published
- 2002
- Full Text
- View/download PDF
49. Feasibility of using autologous transplantation to evaluate hematopoietic stem cell-based gene therapy strategies in transgenic mouse models of human disease.
- Author
-
Miller CL, Imren S, Antonchuk J, Kalberer C, Fabry ME, Nagel RL, Humphries RK, and Eaves CJ
- Subjects
- Anemia, Sickle Cell blood, Anemia, Sickle Cell genetics, Anemia, Sickle Cell therapy, Animals, Blood Cell Count, Disease Models, Animal, Erythrocytes, Hematopoietic Stem Cells, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Genetic Diseases, Inborn therapy, Genetic Therapy, Hematopoietic Stem Cell Transplantation, Transplantation, Autologous
- Abstract
Histoincompatibility between murine donors and recipients of bone marrow (BM) transplants reduces engraftment, and this compromises assessment of hematopoietic stem cells (HSCs) in certain transgenic mice. To study HSCs in the S+S-Antilles mouse model of human sickle cell disease (SCD), we developed an autotransplant protocol. Initial experiments showed no differences between S+S-Antilles mice and normal C57BL/6 (+/+) mice in their radiosensitivity or baseline hematopoietic progenitor numbers. The kinetics of red blood cell (RBC) replacement post-transplant in +/+ recipients of mixtures of transgenic and +/+ BM cells also showed no competitive advantage of the +/+ cells. BM cells were then aspirated from mice 4 days after 5-fluorouracil treatment, transduced with a green fluorescent protein (GFP)-encoding retrovirus, and transplanted into the same recipients that, just before transplant, were irradiated with 800 cGy. We subsequently detected high levels of GFP(+) RBCs (21-79%) and white blood cells (WBCs; 35-88%) in the blood for 11 months and showed that transduced HSCs regenerated in the primary mice also repopulated secondary mice. These findings provide a generally applicable protocol for performing autotransplants in mice and forecast the potential utility of this approach in assessing HSC-based gene therapy protocols in transgenic mouse models of many human diseases.
- Published
- 2002
- Full Text
- View/download PDF
50. Expression of a human beta-globin transgene in erythroid cells derived from retrovirally transduced transplantable human fetal liver and cord blood cells.
- Author
-
Nicolini FE, Imren S, Oh IH, Humphries RK, Leboulch P, Fabry ME, Nagel RL, and Eaves CJ
- Subjects
- Animals, Antigens, CD34 analysis, Cell Differentiation, Cells, Cultured, Erythrocytes metabolism, Genetic Therapy, Genetic Vectors, Green Fluorescent Proteins, Hematopoietic Stem Cell Transplantation, Humans, Liver cytology, Luminescent Proteins genetics, Mice, Retroviridae genetics, Reverse Transcriptase Polymerase Chain Reaction, Erythroid Precursor Cells metabolism, Fetal Blood cytology, Gene Expression, Globins genetics, Liver embryology, Transfection, Transgenes genetics
- Abstract
Transfer of therapeutic genes to human hematopoietic stem cells (HSCs) using complex vectors at clinically relevant efficiencies remains a major challenge. Recently we described a stable retroviral vector that sustains long-term expression of green fluorescent protein (GFP) and a human beta-globin gene in the erythroid progeny of transduced murine HSCs. We now report the efficient transduction of primitive human CD34(+) fetal liver or cord blood cells with this vector and expression of the beta-globin transgene in the erythroid progeny of these human cells for at least 2 months. After growth factor prestimulation and then a 2- to 3-day exposure to the virus, 35% to 55% GFP(+) progeny were seen in assays of transduced colony-forming cells, primitive erythroid precursors that generate large numbers of glycophorin A(+) cells in 3-week suspension cultures, and 6-week long-term culture-initiating cells. In immunodeficient mice injected with unselected infected cells, 5% to 15% of the human cells regenerated in the marrow (including the erythroid cells) were GFP(+) 3 and 6 weeks after transplantation. Importantly, the numbers of GFP(+) human lymphoid and either granulopoietic or erythroid cells in individual mice 6 weeks after transplantation were significantly correlated, indicative of the initial transduction of human multipotent cells with in vivo repopulating activity. Expression of the transduced beta-globin gene in human cells obtained directly from the mice or after their differentiation into erythroid cells in vitro was demonstrated by reverse transcriptase-polymerase chain reaction using specific primers. These experiments represent a significant step toward the realization of a gene therapy approach for human beta-globin gene disorders.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.