Browsing by Subject "Hematopoiesis"
Results Per Page
Sort Options
Item Open Access Early hematopoiesis inhibition under chronic radiation exposure in humans.(Radiat Environ Biophys, 2010-05) Akleyev, Alexander V; Akushevich, Igor V; Dimov, Georgy P; Veremeyeva, Galina A; Varfolomeyeva, Tatyana A; Ukraintseva, Svetlana V; Yashin, Anatoly IThe major goal of this study was to identify and quantitatively describe the association between the characteristics of chronic (low-dose rate) exposure to (low LET) ionizing radiation and cellularity of peripheral blood cell lines. About 3,200 hemograms (i.e., spectra of blood counts) obtained over the years of maximal exposure to ionizing radiation (1950-1956) for inhabitants of the Techa River were used in analyses. The mean cumulative red bone marrow dose (with standard errors), calculated using Techa River Dosimetry System-2000, was 333.6 +/- 4.6 mGy (SD = 259.9 mGy, max = 1151 mGy) to the year 1956. The statistical approach included both empirical methods for estimating frequencies of cytopenic states of the investigated blood cell lines (e.g. neutrophile, platelets, erythrocyte, etc.), and regression methods, including generalized linear models and logistic regressions which allowed taking into account confounding factors (e.g., attained age, age at maximal exposure, presence of concomitant diseases, and demographic characteristics). The results of the analyses demonstrated hematopoiesis inhibition manifested by a decrease in peripheral blood cellularity and an increase in the frequency of cytopenia in all blood cell lines (leukocytes, including lymphocytes, monocytes, neutrophiles, as well as platelets and erythrocytes). The intensity of hematopoiesis inhibition in the period of maximal exposures is determined by the combined influence of the dose rate and cumulative dose. The contribution of specific confounding factors was quantified and shown to be much less important than dose characteristics. The best predictor among dose characteristics was identified for each blood cell line. A 2-fold increase in dose rate is assumed to be a characteristic of radiosensitivity and a quantitative characteristic of the effect.Item Open Access Early hematopoietic effects of chronic radiation exposure in humans.(Health Phys, 2010-09) Akleyev, Alexander V; Akushevich, Igor V; Dimov, Georgy P; Veremeyeva, Galina A; Varfolomeyeva, Tatyana A; Ukraintseva, Svetlana V; Yashin, Anatoly IThe major goal of this study is to investigate and quantitatively describe the nature of the relationship between the characteristics of chronic exposure to ionizing radiation and specific patterns of hematopoiesis reduction. The study is based on about 3,200 hemograms taken for inhabitants of the Techa riverside villages over the years 1951-1956, i.e., the period characterized by a gradual decrease in dose rates. The mean cumulative red bone marrow dose was 333.6 + or - 4.6 mGy. The approach to statistical analyses involved both empirical methods and modeling (generalized linear models and logistic regressions). The results of the analyses highlighted a gradual increase in the frequency of cytopenias with dose rate. The impact of exposure on hematopoiesis reduction patterns was found to be more substantial than that of age and health status. Dose rates resulting in a two-fold increase in the frequency of cytopenias have been estimated.Item Open Access Epidermal growth factor regulates hematopoietic regeneration after radiation injury.(Nat Med, 2013-03) Doan, Phuong L; Himburg, Heather A; Helms, Katherine; Russell, J Lauren; Fixsen, Emma; Quarmyne, Mamle; Harris, Jeffrey R; Deoliviera, Divino; Sullivan, Julie M; Chao, Nelson J; Kirsch, David G; Chute, John PThe mechanisms that regulate hematopoietic stem cell (HSC) regeneration after myelosuppressive injury are not well understood. We identified epidermal growth factor (EGF) to be highly enriched in the bone marrow serum of mice bearing deletion of Bak and Bax in TIE2-expressing cells in Tie2Cre; Bak1(-/-); Bax(flox/-) mice. These mice showed radioprotection of the HSC pool and 100% survival after a lethal dose of total-body irradiation (TBI). Bone marrow HSCs from wild-type mice expressed functional EGF receptor (EGFR), and systemic administration of EGF promoted the recovery of the HSC pool in vivo and improved the survival of mice after TBI. Conversely, administration of erlotinib, an EGFR antagonist, decreased both HSC regeneration and the survival of mice after TBI. Mice with EGFR deficiency in VAV-expressing hematopoietic cells also had delayed recovery of bone marrow stem and progenitor cells after TBI. Mechanistically, EGF reduced radiation-induced apoptosis of HSCs and mediated this effect through repression of the proapoptotic protein PUMA. Our findings show that EGFR signaling regulates HSC regeneration after myelosuppressive injury.Item Open Access Essential cell-extrinsic requirement for PDIA6 in lymphoid and myeloid development.(The Journal of experimental medicine, 2020-04) Choi, Jin Huk; Zhong, Xue; Zhang, Zhao; Su, Lijing; McAlpine, William; Misawa, Takuma; Liao, Tzu-Chieh; Zhan, Xiaoming; Russell, Jamie; Ludwig, Sara; Li, Xiaohong; Tang, Miao; Anderton, Priscilla; Moresco, Eva Marie Y; Beutler, BruceIn a forward genetic screen of N-ethyl-N-nitrosourea (ENU)-induced mutant mice for aberrant immune function, we identified mice with a syndromic disorder marked by growth retardation, diabetes, premature death, and severe lymphoid and myeloid hypoplasia together with diminished T cell-independent (TI) antibody responses. The causative mutation was in Pdia6, an essential gene encoding protein disulfide isomerase A6 (PDIA6), an oxidoreductase that functions in nascent protein folding in the endoplasmic reticulum. The immune deficiency caused by the Pdia6 mutation was, with the exception of a residual T cell developmental defect, completely rescued in irradiated wild-type recipients of PDIA6-deficient bone marrow cells, both in the absence or presence of competition. The viable hypomorphic allele uncovered in these studies reveals an essential role for PDIA6 in hematopoiesis, but one extrinsic to cells of the hematopoietic lineage. We show evidence that this role is in the proper folding of Wnt3a, BAFF, IL-7, and perhaps other factors produced by the extra-hematopoietic compartment that contribute to the development and lineage commitment of hematopoietic cells.Item Open Access Intercellular Protein Transfer from Thymocytes to Thymic Epithelial Cells.(PLoS One, 2016) Wang, Hong-Xia; Qiu, Yu-Rong; Zhong, Xiao-PingPromiscuous expression of tissue restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs) is crucial for negative selection of self-reactive T cells to establish central tolerance. Intercellular transfer of self-peptide-MHC complexes from mTECs to thymic dendritic cells (DCs) allows DCs to acquire TRAs, which in turn contributes to negative selection and regulatory T cell generation. However, mTECs are unlikely to express all TRAs, such as immunoglobulins generated only in B cells after somatic recombination, hyper-mutation, or class-switches. We report here that both mTECs and cortical TECs can efficiently acquire not only cell surface but also intracellular proteins from thymocytes. This reveals a previously unappreciated intercellular sharing of molecules from thymocytes to TECs, which may broaden the TRA inventory in mTECs for establishing a full spectrum of central tolerance.Item Open Access Modeling hematopoietic system response caused by chronic exposure to ionizing radiation.(Radiat Environ Biophys, 2011-05) Akushevich, Igor V; Veremeyeva, Galina A; Dimov, Georgy P; Ukraintseva, Svetlana V; Arbeev, Konstantin G; Akleyev, Alexander V; Yashin, Anatoly IA new model of the hematopoietic system response in humans chronically exposed to ionizing radiation describes the dynamics of the hematopoietic stem cell compartment as well as the dynamics of each of the four blood cell types (lymphocytes, neutrophiles, erythrocytes, and platelets). The required model parameters were estimated based on available results of human and experimental animal studies. They include the steady-state number of hematopoietic stem cells and peripheral blood cell lines in an unexposed organism, amplification parameters for each blood line, parameters describing proliferation and apoptosis, parameters of feedback functions regulating the steady-state numbers, and characteristics of radiosensitivity related to cell death and non-lethal cell damage. The model predictions were tested using data on hematological measurements (e.g., blood counts) performed in 1950-1956 in the Techa River residents chronically exposed to ionizing radiation since 1949. The suggested model of hematopoiesis is capable of describing experimental findings in the Techa River Cohort, including: (1) slopes of the dose-effect curves reflecting the inhibition of hematopoiesis due to chronic ionizing radiation, (2) delay in effect of chronic exposure and accumulated character of the effect, and (3) dose-rate patterns for different cytopenic states (e.g., leukopenia, thrombocytopenia).Item Open Access Plerixafor (a CXCR4 antagonist) following myeloablative allogeneic hematopoietic stem cell transplantation enhances hematopoietic recovery.(J Hematol Oncol, 2018-03-04) Green, Michael MB; Chao, Nelson; Chhabra, Saurabh; Corbet, Kelly; Gasparetto, Cristina; Horwitz, Ari; Li, Zhiguo; Venkata, Jagadish Kummetha; Long, Gwynn; Mims, Alice; Rizzieri, David; Sarantopoulos, Stefanie; Stuart, Robert; Sung, Anthony D; Sullivan, Keith M; Costa, Luciano; Horwitz, Mitchell; Kang, YubinBACKGROUND: The binding of CXCR4 with its ligand (stromal-derived factor-1) maintains hematopoietic stem/progenitor cells (HSPCs) in a quiescent state. We hypothesized that blocking CXCR4/SDF-1 interaction after hematopoietic stem cell transplantation (HSCT) promotes hematopoiesis by inducing HSC proliferation. METHODS: We conducted a phase I/II trial of plerixafor on hematopoietic cell recovery following myeloablative allogeneic HSCT. Patients with hematologic malignancies receiving myeloablative conditioning were enrolled. Plerixafor 240 μg/kg was administered subcutaneously every other day beginning day +2 until day +21 or until neutrophil recovery. The primary efficacy endpoints of the study were time to absolute neutrophil count >500/μl and platelet count >20,000/μl. The cumulative incidence of neutrophil and platelet engraftment of the study cohort was compared to that of a cohort of 95 allogeneic peripheral blood stem cell transplant recipients treated during the same period of time and who received similar conditioning and graft-versus-host disease prophylaxis. RESULTS: Thirty patients received plerixafor following peripheral blood stem cell (n = 28) (PBSC) or bone marrow (n = 2) transplantation. Adverse events attributable to plerixafor were mild and indistinguishable from effects of conditioning. The kinetics of neutrophil and platelet engraftment, as demonstrated by cumulative incidence, from the 28 study subjects receiving PBSC showed faster neutrophil (p = 0.04) and platelet recovery >20 K (p = 0.04) compared to the controls. CONCLUSIONS: Our study demonstrated that plerixafor can be given safely following myeloablative HSCT. It provides proof of principle that blocking CXCR4 after HSCT enhances hematopoietic recovery. Larger, confirmatory studies in other settings are warranted. TRIAL REGISTRATION: ClinicalTrials.gov NCT01280955.Item Open Access Umbilical cord blood expansion with nicotinamide provides long-term multilineage engraftment.(The Journal of clinical investigation, 2014-07) Horwitz, Mitchell E; Chao, Nelson J; Rizzieri, David A; Long, Gwynn D; Sullivan, Keith M; Gasparetto, Cristina; Chute, John P; Morris, Ashley; McDonald, Carolyn; Waters-Pick, Barbara; Stiff, Patrick; Wease, Steven; Peled, Amnon; Snyder, David; Cohen, Einat Galamidi; Shoham, Hadas; Landau, Efrat; Friend, Etty; Peleg, Iddo; Aschengrau, Dorit; Yackoubov, Dima; Kurtzberg, Joanne; Peled, TonyBackground
Delayed hematopoietic recovery is a major drawback of umbilical cord blood (UCB) transplantation. Transplantation of ex vivo-expanded UCB shortens time to hematopoietic recovery, but long-term, robust engraftment by the expanded unit has yet to be demonstrated. We tested the hypothesis that a UCB-derived cell product consisting of stem cells expanded for 21 days in the presence of nicotinamide and a noncultured T cell fraction (NiCord) can accelerate hematopoietic recovery and provide long-term engraftment.Methods
In a phase I trial, 11 adults with hematologic malignancies received myeloablative bone marrow conditioning followed by transplantation with NiCord and a second unmanipulated UCB unit. Safety, hematopoietic recovery, and donor engraftment were assessed and compared with historical controls.Results
No adverse events were attributable to the infusion of NiCord. Complete or partial neutrophil and T cell engraftment derived from NiCord was observed in 8 patients, and NiCord engraftment remained stable in all patients, with a median follow-up of 21 months. Two patients achieved long-term engraftment with the unmanipulated unit. Patients transplanted with NiCord achieved earlier median neutrophil recovery (13 vs. 25 days, P < 0.001) compared with that seen in historical controls. The 1-year overall and progression-free survival rates were 82% and 73%, respectively.Conclusion
UCB-derived hematopoietic stem and progenitor cells expanded in the presence of nicotinamide and transplanted with a T cell-containing fraction contain both short-term and long-term repopulating cells. The results justify further study of NiCord transplantation as a single UCB graft. If long-term safety is confirmed, NiCord has the potential to broaden accessibility and reduce the toxicity of UCB transplantation.Trial registration
Clinicaltrials.gov NCT01221857.Funding
Gamida Cell Ltd.Item Embargo ZFP36L2 in Development and Adulthood: A Critical Regulator of Hematopoietic Stem Cell Homeostasis(2023) Huang, RuiThe tristetraprolin (TTP) protein family of RNA-binding proteins contains three widely expressed mammalian protein members: TTP (ZFP36), ZFP36L1, and ZFP36L2, all of which can regulate gene expression by binding to specific AU-rich sequences located in the 3'-untranslated regions (3’-UTR) of mRNAs and accelerating their decay. Unique among the three, ZFP36L2 plays a pivotal role in maintaining hematopoietic stem cells (HSC) during development. ZFP36L2-deficient mice exhibit severely impaired definitive hematopoiesis and die approximately two weeks after birth due to severe anemia, thrombocytopenia, and internal hemorrhage. Recent single-cell RNA sequencing (scRNA-seq) studies have demonstrated widespread Zfp36l2 expression in HSC and the hematopoietic system during both development and adulthood.
Despite the recognized importance of ZFP36L2 in maintaining HSC, there are still numerous aspects of its role that are not fully understood, hindering our understanding of hematopoietic regulation. While prior studies have provided valuable insights into the physiological function of ZFP36L2, its molecular mechanisms in HSC development and hematopoietic system maintenance remain poorly defined. To decipher its involvement in hematopoiesis, it is crucial to identify the mRNA targets and pathways regulated by ZFP36L2 and determine whether this regulation is intrinsic to HSC. Moreover, the premature death of ZFP36L2-deficient mice makes it unclear to what extent this protein governs adult HSC function. Lastly, considering the pivotal position of HSC in the hematopoietic hierarchy, it is important to investigate how ZFP36L2's activity in HSC affects the subsequent differentiation of hematopoietic lineages. Clarifying this relationship could yield valuable insights into the post-transcriptional mechanisms that govern HSC biology and potentially lead to the identification of new therapeutic targets for hematological disorders.
To address the knowledge gap, we employed a detailed analysis combining flow cytometry and scRNA-seq to examine HSC and hematopoietic progenitor cells (HSPC) at several critical developmental stages. Our studies revealed that the absence of ZFP36L2 resulted in significant reductions in both HSC and immature progenitors during mouse development, primarily due to HSC-autonomous dysregulation. In addition, scRNA-seq analysis of HSC and progenitors revealed that ZFP36L2 deficiency caused abnormal upregulation of transcripts related to cell cycle regulation and lymphoid specification, leading to aberrant cell cycle progression and premature lymphoid lineage commitment. This ultimately resulted in cellular damage and HSC exhaustion at birth. These findings demonstrate that ZFP36L2 is essential for maintaining the homeostasis of HSC, and emphasizes the significance of restraining lineage commitment and excessive self-renewal during HSC development.
In a related study, we investigated possible functional overlap of ZFP36L2 and TTP. We developed mice (L2KO/TTP∆ARE) that lacked Zfp36l2 but modestly overexpressed TTP throughout the body. L2KO/TTP∆ARE mice not only survived but also exhibited normal peripheral blood counts, except for residual moderate thrombocytopenia. We took advantage of this rescued ZFP36L2-deficient model and investigated the role of ZFP36L2 in adult hematopoiesis. We discovered that megakaryocyte (MK) progenitors and MK-biased HSC were decreased in bone marrow from L2KO/TTP∆ARE mice and exhibited enriched erythroid and decreased MK gene signatures. In addition, L2KO/TTP∆ARE HSC failed to reconstitute hematopoiesis upon non-competitive transplantation, and showed molecular features of stress and reduced cycling. Thus, TTP can assume some functions of ZFP36L2 in a genetic dose-dependent manner, but ZFP36L2 may be specifically required for the maintenance of megakaryopoiesis and HSC function.In summary, our studies provide novel insights into the essential role of ZFP36L2 in the maintenance of HSC throughout both developmental stages and adulthood. We demonstrate that ZFP36L2 is essential for restraining abnormal cell cycling and lymphoid commitment during HSC development, thereby ensuring proper HSC maintenance. Interestingly, our results also suggest that TTP may partially compensate for ZFP36L2 deficiency during the development of the hematopoietic system, but that ZFP36L2 may have specific functions in maintaining megakaryopoiesis and HSC function in adulthood. Overall, our research provides a strong foundation for future studies aimed at elucidating the underlying mechanisms that govern ZFP36L2's function in hematopoiesis, further advancing our understanding of the intricate regulatory paradigm of HSC biology.