scholarly journals The lysine-rich intracellular loop and cell type-specific co-factors are required for IFITM3 antiviral immunity in hematopoietic stem cells

2021 ◽  
Author(s):  
Giulia Unali ◽  
Anna Maria Sole Giordano ◽  
Ivan Cuccovillo ◽  
Monah Abou Alezz ◽  
Luis Apolonia ◽  
...  

The interferon-induced transmembrane protein 3 (IFITM3) inhibits lentiviral gene therapy vector entry into hematopoietic stem cells and can be overcome by Cyclosporine H (CsH), but underlying mechanisms remain unclear. Here, we show that mutating the evolutionarily conserved lysines of the IFITM3 intracellular loop abolishes its antiviral activity without affecting either its localization or its degradation by CsH through non-canonical lysosomal pathways. When confined to the plasma membrane, the lysine-competent IFITM3 lost restriction against VSV-G pseudotyped viral vectors but gained antiviral activity against vectors that fuse directly at the plasma membrane. Interestingly, altering the lysines did not alter IFITM3 homodimerization but impacted higher-order protein complex formation, suggesting loss of interaction with cellular co-factors. In agreement, IFITM3 expression was not sufficient to restrict viral vectors in myeloid K562 cells as opposed to promonocytic THP1 or primary HSC. We exclude the involvement of previously identified factors affecting IFITM3 biology and propose a novel model for IFITM3 restriction that depends on the presence of cellular co-factor(s) that may interact with IFITM3 through the intracellular loop lysine residues. Overall, our work provides significant insight into the mechanisms of action of IFITM3 and CsH that can be exploited for improved gene therapies and broadly acting antiviral strategies.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1778-1778
Author(s):  
Flavia O. Pinho ◽  
Dulcineia M. Albuquerque ◽  
Sara T.O Saad ◽  
Fernando F. Costa

Abstract Alpha Hemoglobin Stabilizing Protein (AHSP) binds alpha hemoglobin chain (αHb), avoiding its precipitation and its pro-oxidant activity. In the presence of beta hemoglobin chain (βHb), the αHb-AHSP complex is dismembered and βHb displaces AHSP to generate the quaternary structure of hemoglobin. These data have been obtained in vitro and in mouse cells, but strongly suggest the importance of AHSP for normal hemoglobin synthesis in humans. To the best of our knowledge, the relationship between hemoglobin formation and alterations in AHSP expression has not yet been described in human red cells. Hence, to investigate the consequences of a reduced AHSP synthesis in human red cells, we established the RNA interference-mediated knockdown of AHSP expression in human erythroleukemia cell line (K562 cells) and human hematopoietic stem cells (CD34+ cells) induced to erythroid differentiation, and analyzed the consequent cellular and molecular aspects of AHSP knockdown in these cells. shRNA expression vectors, aimed at the AHSP mRNA target sequence, were cloned and transfected into K562 and CD34+ cells using a non-liposomal lipid reagent. Following transfection, K562 cells that stably expressed AHSP-shRNA and CD34+ cells that transiently expressed AHSP-shRNA were selected. K562 and CD34+ cells were stimulated to erythroid differentiation by hemin and erythropoietin (EPO) respectively. The cells were examined in terms of gene expression using quantitative real-time PCR; production of reactive oxygen species (ROS), apoptosis and hemoglobin production through flow cytometry assays; and immunofluorescence assays for globin chains. AHSP-shRNA hemin-induced K562 cells and AHSP-shRNA EPO-induced CD34+ cells presented 71% and 75% decreases in AHSP expression levels, respectively. The RNAi-mediated knockdown of AHSP expression resulted in a considerable αHb precipitation, as well as in a significant decrease in fetal hemoglobin formation. In addition, AHSP-knockdown cells demonstrated an increased ROS production and increased rate of apoptosis. These findings strengthen the hypothesis that AHSP stabilizes the alpha hemoglobin chain, avoiding its precipitation and its ability to generate ROS which implicate in cell death. Moreover, data indicate that AHSP may be highly significant for human hemoglobin formation and suggest that AHSP is a key chaperone protein during human erythropoiesis.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3436-3436
Author(s):  
Tohru Fujiwara ◽  
Hisayuki Yokoyama ◽  
Yoko Okitsu ◽  
Mayumi Kamata ◽  
Shinichi Fujimaki ◽  
...  

Abstract Abstract 3436 Background: Aplastic anemia (AA) is characterized by a reduced number of hematopoietic stem cells (HSCs). It has been proposed that immunological injury in HSCs leads to reduced numbers of stem cells in the bone marrow. In addition, expression of the critical regulator of hematopoiesis GATA-2 is decreased in CD34-positive cells in AA (Fujimaki et al. Br J Haematol 2001). Despite the compelling results described above, only limited information has emerged regarding intrinsic abnormalities of hematopoietic stem cells in AA. It has been demonstrated that HOXB4 induces HSC expansion ex vivo (Antonchuk et al. Cell 2002), and restoring HOXB4 protein in HSCs from bone marrow failure patients promotes HSC expansion (Tang et al. Br J Haematol 2009). In conjunction with the evidence that recent genome-wide analysis of GATA factor chromatin occupancy identified GATA-2 peak at HOXB4 promoter (Fujiwara et al. Mol Cell 2009), we hypothesized that GATA-2 directly regulates HOXB4 expression in HSCs, which might contribute to the pathogenesis of AA. Here, we investigated possible link between GATA-2 and HOXB4, and also tested if HOXB4 is deregulated in CD34-positive cells from patients with AA. Method: For GATA-2 overexpression, human GATA-2 coding sequence was cloned into pcDNA3.1 expression vector as well as MSCV retroviral expression vector (Clontech). For GATA-2 knockdown, siRNA specific for human GATA-2 was transfected into CD34-positive cells or K562 cells by Amaxa Nucleofector kit (Amaxa Inc.). For promoter assay, DNA fragment of the HOXB4 gene promoter region (up to −262 from 1st ATG) was cloned into pGL3-Basic (Promega), and the GATA deletion construct was subsequently created with QuickChange™Site-Directed Mutagenesis Kit (Stratagene). Quantitative chromatin immunoprecipitation (ChIP) analysis was performed using antibodies for GATA-2 (H-116, Santa Cruz). For analyzing clinical samples, informed consent was obtained in all cases and ethical considerations according to the declaration of Helsinki were followed. Results: To examine if GATA-2 and HOXB4 are functionally linked, we transfected a GATA-2 expression vector into K562 cells, and demonstrated that GATA-2 significantly upregulated endogeneous HOXB4 expression. Furthermore, siRNA-mediated GATA-2 knockdown in K562 cells significantly reduced HOXB4 expression, indicating that HOXB4 is a GATA-2 target gene. We overexpressed/reduced GATA-2 in cord blood-derived CD34+ cells, which also provided evidence for GATA-2 regulation of HOXB4 expression. Promoter analyses revealed that GATA sequence located at −160/-157 of the HOXB4 gene promoter region was required to confer luciferase activity in K562 cells. In vitro DNA binding studies and quantitative ChIP analysis revealed specific GATA-2 occupancy at a chromatin region containing this element. Finally, we demonstrated that HOXB4 gene expression was significantly decreased in CD34+ cells from patients with AA (n=10) compared to those with ITP (n=13). The expression levels of HOXB4 and GATA-2 also correlated in these populations (r=0.6573, p<0.01). Conclusion: Based on these findings, we propose that decreased expression of GATA-2 in hematopoietic stem cells of AA leads to reduced HOXB4 transcription, which may have an important role in the development and/or progression of the disease. Disclosures: No relevant conflicts of interest to declare.


2003 ◽  
Vol 12 (4) ◽  
pp. 365-377 ◽  
Author(s):  
Osamu Yamada ◽  
Masaharu Akiyama ◽  
Kiyotaka Kawauchi ◽  
Tomoko Adachi ◽  
Hisashi Yamada ◽  
...  

Leukemic stem cells that expressed endogenous telomerase activity were induced to show overexpression of exogenous hTERT and were analyzed for biological changes in order to assess the possible influence of telomerase gene therapy on the transplantation of normal hematopoietic stem cells. Introduction of hTERT into K562, a telomerase-positive immortal cell line, resulted in a 2.5-fold elevation of telomerase activity and the lengthening of telomeres by 6 kb to 23 kb. Real-time fluorescent PCR, which could perform quantitative analysis of transcripts, revealed a 175-fold increase in hTERT expression, suggesting the posttranscriptional regulation of telomerase. Ectopic expression of hTERT in K562 cells showed a survival advantage during culture in the absence of serum. Expression of mRNA for the telomeric-repeat binding factor 1 (TRF1) and caspase-3 activity were both decreased in hTERT-transfected K562 cells. Transduced cells retained their usual phenotypic characteristics, differentiation ability, and signal transduction response to TPA. These data suggest that ectopic expression of hTERT by normal hematopoietic stem cells may confer a survival advantage without changing their innate biological characteristics.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3722-3722 ◽  
Author(s):  
Erin H. Breese ◽  
Catherine Dawson ◽  
Corina Buechele ◽  
Marcus R. Breese ◽  
Michael L. Cleary ◽  
...  

Abstract Introduction Chromosomal rearrangements involving the MLL gene occur in both primary and treatment-related leukemias and convey a poor prognosis. Infants with acute lymphoblastic leukemia (ALL) containing an MLL translocation have a 5-year event free survival of 35% as compared to 69% for infants with germline MLL (Kang et al., Blood 2012). While animal models of MLL-AF9 translocations typically found in adult AML have improved our understanding of the role of MLL translocations in leukemia pathogenesis, modeling the MLL-AF4 translocation commonly found in infant ALL has proven difficult. We hypothesized that recent advances in genome engineering would facilitate induction of specific MLL translocations in primary hematopoietic stem cells to recapitulate the initiating event that leads to infant leukemia. A better understanding of the pathogenesis of the MLL-AF4 translocation will allow for the development of targeted therapies to improve outcome in MLL-rearranged infant leukemia. Methods Transcription activator-like effector nucleases (TALENs) were designed to recognize a patient-specific translocation site within the break point cluster region of the MLL gene and the AF4 gene, respectively. These TALEN pairs were then co-expressed in either K562 cells or primary human CD34+ cells isolated from umbilical cord blood to induce patient-specific double strand breaks. Traditional PCR, nested PCR, and high throughput next generation sequencing were used to detect MLL-AF4 translocations, reciprocal AF4-MLL translocations, as well as other MLL translocations. Clonal analysis was performed to determine the translocation efficiency resulting from two site-specific double strand breaks. Primary cells were maintained in long-term culture to assess for potential survival advantage conferred by the translocations. Finally, primary human CD34+ cells with an induced MLL-AF4translocation were transplanted into NSG mice to assess for leukemic potential. Results We successfully generated TALENs that induce a patient-specific double strand break within the breakpoint cluster regions of the endogenous MLL and AF4 genes. Co-expression of the MLL and AF4 TALENs in K562 cells as well as in primary human hematopoietic stem cells isolated from umbilical cord blood resulted in de novo MLL-AF4 translocations as well as the reciprocal AF4-MLL translocation. The MLL-AF4 translocation efficiency was appreciably higher in K562 cells (10-4) compared to primary hematopoietic stem cells (10-5). Long-term culture of the primary human hematopoietic cells showed an increase in the frequency of the MLL-AF4 translocation-positive cells, suggesting a survival advantage for the cells containing the translocation. These cells were subsequently transplanted into NSG mice to assess their ability to induce leukemia. These studies are currently ongoing. Conclusions Advances in genome engineering have provided the tools necessary to study leukemia pathogenesis in a prospective manner. We have designed an experimental system to recapitulate the process of leukemogenesis. TALENs are effective to induce a specific MLL translocation in primary human hematopoietic stem cells with minimal off target effects, under the control of the endogenous promoter, and in the presence of the reciprocal translocation. This system will allow us to prospectively investigate the downstream effects of this initiating event on gene expression, epigenetic regulation, and genome stability in order to understand the key steps critical for the pathogenesis of MLL-rearranged leukemias. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 87 (9) ◽  
pp. 3676-3687 ◽  
Author(s):  
AP Warren ◽  
K Patel ◽  
DJ McConkey ◽  
R Palacios

In our search for cell surface markers expressed on hematopoietic stem cells and/or very early progenitor cells we found that the Joro 177 monoclonal antibody (MoAb) bound to most hematopoietic cells in day 8/8.5 yolk sac, day 12 fetal liver, and day 13 fetal thymocytes; it stained hematopoietic stem cells and less immature lymphoid, myeloid, and erythroid-lineage cells, but not most thymocytes and splenic lymphocytes in adult mice. Joro 177 MoAb stimulated tyrosine phosphorylation of an integral of 124-kD protein and induced homotypic aggregation of lymphoid progenitor cells. Importantly, Joro 177 MoAb inhibited cell survival/growth and consequently the generation of lymphoid, myeloid, and erythroid lineage cells in vitro from early Lin- hematopoietic precursors. Joro 177 MoAb induced apoptosis of hematopoietic progenitor cells. Molecular cloning and expression indicated that Joro 177 MoAb recognizes a type II transmembrane protein, which is the mouse homologue of the human CD98 heavy chain gene. We suggest that CD98 is a cell membrane receptor involved in the control of cell survival/death of hematopoietic cells.


Blood ◽  
1999 ◽  
Vol 93 (10) ◽  
pp. 3276-3285 ◽  
Author(s):  
Alexis Grande ◽  
Bianca Piovani ◽  
Alessandro Aiuti ◽  
Sergio Ottolenghi ◽  
Fulvio Mavilio ◽  
...  

Targeted expression to specific tissues or cell lineages is a necessary feature of a gene therapy vector for many clinical applications, such as correction of hemoglobinopathies or thalassemias by transplantation of genetically modified hematopoietic stem cells. We developed retroviral vectors in which the constitutive viral enhancer in the U3 region of the 3′ LTR is replaced by an autoregulatory enhancer of the erythroid-specific GATA-1 transcription factor gene. The replaced enhancer is propagated to the 5′ LTR upon integration into the target cell genome. The modified vectors were used to transduce human hematopoietic cell lines, cord blood-derived CD34+ stem/progenitor cells, and murine bone marrow repopulating stem cells. The expression of appropriate reporter genes (▵LNGFR, EGFP) was analyzed in the differentiated progeny of transduced stem cells in vitro, in liquid culture as well as in clonogenic assay, and in vivo, after bone marrow transplantation in lethally irradiated mice. The GATA-1 autoregulatory enhancer effectively restricts the expression of the LTR-driven proviral transcription unit to the erythroblastic progeny of both human progenitors and mouse-repopulating stem cells. Packaging of viral particles, integration into the target genome, and stability of the integrated provirus are not affected by the LTR modification. Enhancer replacement is therefore an effective strategy to target expression of a retroviral transgene to a specific progeny of transduced hematopoietic stem cells.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4290-4290
Author(s):  
Naohito Fujishima ◽  
Makoto Hirokawa ◽  
Masumi Fujishima ◽  
Yoshikazu Ichikawa ◽  
Takahiro Horiuchi ◽  
...  

Abstract Background: We previously investigated the gene expression profile of drug-resistant human leukemia K562 cells by the serial analysis of gene expression (SAGE) (Ichikawa et al. Int J Hematol, 2004;79:276), and found that the HEI10 gene was overexpressed in K562 cells. With the same technology, we also observed that human CD34+ cells expressed high levels of HEI10 gene expression compared to erythroid progenitor cells (Fujishima et al. Int J Hematol, in press). HEI10 encodes a protein which belongs to a family of E3 ubiquitin ligase,and interacts with cyclin B1 leading to the regulation of cell cycle. In the present study, we examined the gene expression of HEI10 quantitatively in human leukemias in order to learn whether there would be any association between the expression levels and leukemia subtypes. We also investigated whether the expression levels of HEI10 would be related to differentiation of myeloid leukemia cell lines and normal hematopoietic stem cells. Methods: The quantitative real-time PCR method was employed to estimate the relative gene expression levels of HEI10. Total RNA was extracted from mononuclear cells of bone marrow that had been taken for diagnostic purposes. Patients include acute myeloid leukemia (AML) (n=21) and chronic myeloid leukemia (CML) (n=7; 2 in blastic crisis, 5 in chronic phase). Differentiation of human acute myeloid leukemia cell line HL60 cells was induced by PMA. Human normal CD34+ cells were purified from G-CSF mobilized blood stem cells and were induced to differentiate into erythroid, myeloid or dendritic cells by various cytokines. Results: Quantitative real-time PCR analysis revealed the high expression of HEI10 gene in patients with leukemia with undifferentiated phenotype (3 cases of AML M1 in FAB classification and 2 cases of CML in blast crisis) and low levels of expression in acute promyelocytic leukemia and CML in chronic phase. When HL60 cells were induced to differentiation with PMA, accumulation in the G0/G1 phase was observed and the expression of HEI10 was decreased. Differentiation of normal CD34+ cells also resulted in a decrease of HEI10 expression. Conclusions: The expression of HEI10 gene in hematopoietic cells appears to be upregulated when cells are immature. The role for HEI10 in hematopoietic stem cells remains to be elucidated. Biological effects of the HEI10 gene silencing in human hematopoietic cells are currently being investigated by RNAi technology. Figure Figure Figure Figure


2006 ◽  
Author(s):  
Hideyo Hirai ◽  
Pu Zhang ◽  
Tajhal Dayaram ◽  
Christopher Hetherington ◽  
Shin-ichi Mizuno ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document