212 results on '"Ly P"'
Search Results
2. A Novel Bi-Specific T-Cell Engager Targeting ILT3 Is Potently Effective in Multiple Myeloma
- Author
-
Di Meo, Francesco, primary, Iyer, Anjushree, additional, Akama, Keith, additional, Yu, Christina, additional, Cheng, Rujin, additional, Cesarano, Annamaria, additional, Marino, Silvia, additional, Aljoufi, Arafat, additional, Soni, Rajesh, additional, Roda, Julie M, additional, Sissons, James, additional, Vu, Ly P, additional, Guzman, Monica L., additional, Huang, Kun, additional, Roodman, David G., additional, and Perna, Fabiana, additional
- Published
- 2022
- Full Text
- View/download PDF
3. RNA Deadenylation Subunit CNOT3 Promotes Myeloid Leukemia By Driving Translation of Oncogenic Targets
- Author
-
Ghashghaei, Maryam, primary, Yue, Marty, additional, Arsalan, Aaremish, additional, Bombaci, Giuseppe, additional, Spencer Miko, Sandra, additional, Shaalan, Haya, additional, Edin, Glenn, additional, Morin, Gregg, additional, Perna, Fabiana, additional, and Vu, Ly P, additional
- Published
- 2022
- Full Text
- View/download PDF
4. RNA Deadenylation Subunit CNOT3 Promotes Myeloid Leukemia By Driving Translation of Oncogenic Targets
- Author
-
Maryam Ghashghaei, Marty Yue, Aaremish Arsalan, Giuseppe Bombaci, Sandra Spencer Miko, Haya Shaalan, Glenn Edin, Gregg Morin, Fabiana Perna, and Ly P Vu
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
5. A Novel Bi-Specific T-Cell Engager Targeting ILT3 Is Potently Effective in Multiple Myeloma
- Author
-
Francesco Di Meo, Anjushree Iyer, Keith Akama, Christina Yu, Rujin Cheng, Annamaria Cesarano, Silvia Marino, Arafat Aljoufi, Rajesh Soni, Julie M Roda, James Sissons, Ly P Vu, Monica L. Guzman, Kun Huang, David G. Roodman, and Fabiana Perna
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
6. RNA Binding Protein Syncrip Is Required for the Low-Output HSC By Sustaining Proteome Quality
- Author
-
Chavez, Florisela Herrejon, primary, Cifani, Paolo, additional, Pine, Alli, additional, Chu, Karen L, additional, Barin, Ersilia, additional, Schurer, Alexandra, additional, Luo, Hanzhi, additional, Nguyen, Diu T.T., additional, Kentsis, Alex, additional, Leslie, Christina, additional, Kharas, Michael G, additional, and Vu, Ly P, additional
- Published
- 2021
- Full Text
- View/download PDF
7. Structural variation cooperates with permissive chromatin to control enhancer hijacking–mediated oncogenic transcription
- Author
-
Botten, Giovanni A., Zhang, Yuannyu, Dudnyk, Kseniia, Kim, Yoon Jung, Liu, Xin, Sanders, Jacob T., Imanci, Aygun, Droin, Nathalie, Cao, Hui, Kaphle, Pranita, Dickerson, Kathryn E., Kumar, Kirthi R., Chen, Mingyi, Chen, Weina, Solary, Eric, Ly, Peter, Zhou, Jian, and Xu, Jian
- Abstract
•We describe a multimodal approach for quantitative analysis of transcriptional and structural impacts imposed by structural variants.•Enhancer hijacking cooperates with permissive chromatin to activate oncogenic transcription in leukemic genomes.
- Published
- 2023
- Full Text
- View/download PDF
8. RNA Binding Protein Syncrip Is Required for the Low-Output HSC By Sustaining Proteome Quality
- Author
-
Hanzhi Luo, Diu T.T. Nguyen, Paolo Cifani, Karen L Chu, Alex Kentsis, Ersilia Barin, Florisela Herrejon Chavez, Christina S. Leslie, Alexandra Schurer, Ly P. Vu, Alli Pine, and Michael G. Kharas
- Subjects
Chemistry ,media_common.quotation_subject ,Immunology ,Proteome ,RNA-binding protein ,Quality (business) ,Cell Biology ,Hematology ,Biochemistry ,Cell biology ,media_common - Abstract
RNA binding proteins (RBPs) have been increasingly recognized as an important class of regulators of normal and malignant hematopoiesis. However, the exact function and underpinning mechanisms of the RBPs that govern hematopoietic stem cells (HSCs) remains poorly characterized. We had previously identified SYNCRIP as a critical RBP that controls leukemia stem cell program in myeloid leukemia. Here, using the novel murine genetic conditional knockout (cKO) model, we delineated the role of SYNCRIP in regulating the low-output HSC. We developed a Syncrip cKO allele and crossed Syncripf/f mice to the interferon (IFN) -a-inducible Mx-1-Cre mice to create Syncripf/f Mx-1-Cre+. We consistently obtained near complete depletion of SYNCRIP 3 weeks after two consecutive Poly(I:C) injections. We observed that SYNCRIP is dispensable for static hematopoiesis and Syncrip KO animals showed equivalent number and frequencies of stem and progenitor cells (Lin-Sca+cKit+ (LSK)- LT-HSC (CD48-CD150+); MPP1 (CD48-CD150-); MPP2 (CD48+CD150+); MPP4 (CD48-CD150-)). However, KO SyncripD/D deficient cells were outcompeted by WT Syncripf/f cells in the transplantation setting (bone marrow (BM) chimerism WT (n=9) 38% ± 7.8% vs. KO (n=9) 2.7% ± 0.8%, p Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
- Full Text
- View/download PDF
9. Evidence supporting a role for the immune checkpoint protein B7-H3 in NK cell-mediated cytotoxicity against AML
- Author
-
Tyagi, Anudishi, Ly, Stanley, El-Dana, Fouad, Yuan, Bin, Jaggupilli, Appalaraju, Grimm, Sabrina, Konopleva, Marina, Bühring, Hans-Jörg, and Battula, V. Lokesh
- Abstract
We observed that the immune checkpoint protein B7-H3 is overexpressed in acute myeloid leukemia (AML) patients with poor treatment outcomes. Inhibition of B7-H3 expression or blocking of its activity using a novel monoclonal antibody (T-1A5) in AML cells significantly enhanced natural killer (NK) cell-mediated cytotoxicity in AML cells in vitro and in vivo. Moreover, a human-mouse chimera of this antibody (ChT-1A5) induced antibody-dependent cell-mediated cytotoxicity (ADCC) in B7-H3+ primary AML cells, but not in normal hematopoietic cells, suggesting the specify of this antibody for AML cells. Epitope mapping studies identified that both T-1A5 and ChT-1A5 antibodies bind to the FG-loop region of B7-H3, which is known to regulate the immunosuppressive function of B7-H3. Furthermore, treatment with ChT-1A5 in combination with human NK cells significantly prolonged survival in AML patient-derived xenograft (PDX) models. Our results suggest that the ChT-1A5 antibody can inhibit the immunosuppressive function of B7-H3 protein as well as induce ADCC in B7-H3+ AML.
- Published
- 2022
- Full Text
- View/download PDF
10. Transcriptional Control of HP1a By the RNA Binding Proteins Rbmx/L1 Maintain Chromatin State in Myeloid Leukemia
- Author
-
Alexendar R. Perez, Ly P. Vu, James Taggart, Michael G. Kharas, Trevor Stephen Barlowe, Arthur Chow, Diu T.T. Nguyen, Zhaoqi Liu, Raul Rabadan, Alexandra Schurer, Timothy Chou, Justin C. Wheat, Ersilia Barin, Ulrich Steidl, Camila Prieto, Meera Dhodapkar, Katerina Hoskova, Saroj Gourkanti, and Christina S. Leslie
- Subjects
Myeloid ,Immunology ,Myeloid leukemia ,RNA-binding protein ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Chromatin ,Cell biology ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Conditional gene knockout ,medicine ,K562 cells - Abstract
Mutations and aberrant expression of RNA binding proteins (RBPs) have recently been found to contribute to leukemia development (Prieto and Kharas, CSH, 2020). Previously we have identified the RBP RBMX (RNA binding motif protein, X-linked) in our shRNA in vivo screen using murine MLL-AF9 driven leukemia (Prieto et al, ASH abstract 2018). Here we uncover the role of RBMX and its retrogene RBMXL1 in normal hematopoiesis and leukemogenesis in mouse and human systems. To investigate RBMX function in normal hematopoiesis, we created an Rbmx Mx1-Cre conditional knockout model to specifically delete Rbmx in the hematopoietic system. We gender stratified our studies since Rbmx is sex-linked. Deletion of Rbmx in both female and male primary mice as well as in non-competitive transplant animals did not affect hematopoiesis. Additionally, Rbmx knockout (KO) leads to a mild reduction in multipotent progenitors (MPP2 and MPP4) in female competitive transplanted mice although no defects in long-term hematopoiesis was observed in male competitive transplanted mice. These data suggest that Rbmx is dispensable for normal hematopoiesis. To identify the role of RBMX in leukemogenesis we knockout Rbmx in MLL-AF9 murine leukemia cells and found it significantly reduced colony formation in vitro and delayed leukemogenesis in vivo, indicating that Rbmx is required for leukemia maintenance. We observed, however, that MLL-AF9 transformed cells from Rbmx KO donor mice showed no delay in leukemia initiation versus cells from wildtype. We then determined that while Rbmx deletion is effective with complete depletion of mRNA, the retrogene RBMXL1 expression is maintained in Rbmx deficient cells, which may compensate for Rbmx deletion in leukemia initiation. Indeed, depletion of RBMXL1 by shRNAs (KD) in Rbmx deficient leukemia cells results in a drastic reduction in colony formation, increased colony myeloid differentiation, and induced apoptosis in cells deficient for both RBMXL1 and RBMX compared to those only depleted of RBMX. Correspondingly, RBMXL1 KD in Rbmx deficient leukemia cells resulted in further delayed leukemogenesis in vivo, indicating that RBMXL1 is functionally redundant to RBMX and both genes are required for leukemia development and maintenance. We next investigated the role of RBMX/L1 in normal human hematopoietic and leukemia cells. We found that RBMX/L1 expression were higher in AML cell lines (n=10/11) and primary AML patient samples (n=2/4) compared to healthy individuals. RBMX/L1 KD by shRNAs led to a dramatic decrease in cell proliferation, induction of apoptosis and myeloid differentiation in several human myeloid leukemia cell lines (MOLM-13, THP-1, K562, and KCL-22). Additionally, RBMX/L1 depletion significantly delayed leukemogenesis in vivo of AML cell lines (median survival of 51.5 days in control vs. median 'not reached' in shRNA1 and shRNA2), and of primary leukemia cells derived from an AML patient (median 50 days in control vs. "not reached" in shRNA2). We next depleted RBMX/L1 in human CD34+ stem and progenitor cells and observed reduced colony formation but no increase in apoptosis. Taken together, our data suggest that RBMX/L1 are differentially required in leukemia cells versus normal cells in both human and mouse systems. To uncover the mechanism of RBMX/L1 function, we performed complex chromosomal karyotyping analysis of RBMX/L1 depleted MOLM13 cells and revealed increased metaphases with breaks and gaps. In addition, ATAC-seq analysis showed profound genome-wide changes in chromatin accessibility and compaction upon RBMX/L1 depletion. Analyses of RNA-seq and transcriptome-wide RBMX/L1 binding targets (PAR-CLIP; Liu et al. 2017) revealed that RBMX/L1 directly binds to mRNAs and affects transcription of multiple loci including the heterochromatin protein 1 alpha (HP1a) mRNA. Using single molecule RNA FISH, we uncovered that RBMX/L1 controls the nascent transcription of the HP1a locus. Forced HP1a expression rescued the RBMX/L1 depletion effects on cell growth and apoptosis. Overall, we determine that RBMX/L1 control leukemia cell survival by regulating chromatin state through its downstream target HP1a. This study describes a novel genetic compensation phenomenon in leukemia and illuminate a mechanism for RBPs directly promoting transcription. Our results suggest RBMX/L1 as well as HP1a as potential novel therapeutic targets in myeloid malignancies. Disclosures Steidl: Aileron Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Stelexis Therapeutics: Consultancy, Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Bayer Healthcare: Research Funding; Pieris Pharmaceuticals: Consultancy. Kharas:Accent Therapeutics: Consultancy; 28-7: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
11. Disrupting the adult globin promoter alleviates promoter competition and reactivates fetal globin gene expression
- Author
-
Topfer, Sarah K., Feng, Ruopeng, Huang, Peng, Ly, Lana C., Martyn, Gabriella E., Blobel, Gerd A., Weiss, Mitchell J., Quinlan, Kate G. R., and Crossley, Merlin
- Abstract
The benign condition hereditary persistence of fetal hemoglobin (HPFH) is known to ameliorate symptoms of co-inherited β-hemoglobinopathies, such as sickle cell disease and β-thalassemia. The condition is sometimes associated with point mutations in the fetal globin promoters that disrupt the binding of the repressors BCL11A or ZBTB7A/LRF, which have been extensively studied. HPFH is also associated with a range of deletions within the β-globin locus that all reside downstream of the fetal HBG2 gene. These deletional forms of HPFH are poorly understood and are the focus of this study. Numerous different mechanisms have been proposed to explain how downstream deletions can boost the expression of the fetal globin genes, including the deletion of silencer elements, of genes encoding noncoding RNA, and bringing downstream enhancer elements into proximity with the fetal globin gene promoters. Here we systematically analyze the deletions associated with both HPFH and a related condition known as δβ-thalassemia and propose a unifying mechanism. In all cases where fetal globin is upregulated, the proximal adult β-globin (HBB) promoter is deleted. We use clustered regularly interspaced short palindromic repeats-mediated gene editing to delete or disrupt elements within the promoter and find that virtually all mutations that reduce ΗΒΒ promoter activity result in elevated fetal globin expression. These results fit with previous models where the fetal and adult globin genes compete for the distal locus control region and suggest that targeting the ΗΒΒ promoter might be explored to elevate fetal globin and reduce sickle globin expression as a treatment of β-hemoglobinopathies.
- Published
- 2022
- Full Text
- View/download PDF
12. Updated Outcomes of the University of Southern California (USC) ALL Frontline Regimen (based on CCG-1882) after Protocol Modification in the Era of Novel Agents in Ph-Negative ALL Patients
- Author
-
Mendiola, Vincent Louie Ramos, Ly, Catherine, Wang, Jennifer, Bui, Thuy, Rodman, Jack, Ali, Amir, Vergara-Lluri, Maria E., Woan, Karrune, Tam, Eric, Chaudhary, Preet M., Douer, Dan, Ladha, Abdullah, and Yaghmour, George
- Published
- 2022
- Full Text
- View/download PDF
13. Updated Comparison of Outcomes in Hispanic and Non-Hispanic Patients with Ph-Negative Acute Lymphoblastic Leukemia Using the Modified Pediatric-Based University of Southern California Acute Lymphoblastic Leukemia (USC ALL) Regimen in the Era of Novel Agents
- Author
-
Mendiola, Vincent Louie, Ly, Catherine, Wang, Jennifer, Bui, Thuy, Rodman, Jack, Ali, Amir, Woan, Karrune, Tam, Eric, Vergara-Lluri, Maria E., Chaudhary, Preet M., Douer, Dan, Ladha, Abdullah, and Yaghmour, George
- Published
- 2022
- Full Text
- View/download PDF
14. Updated Outcomes in Adult Philadelphia-Positive Acute Lymphoblastic Leukemia Patients Using Tyrosine Kinase Inhibitors Combined with the Modified University of Southern California (USC ALL) Regimen without Peg-Asparaginase in the Era of Novel Agents
- Author
-
Mendiola, Vincent Louie Louie, Ly, Catherine, Wang, Jennifer, Bui, Thuy, Rodman, Jack, Ali, Amir, Woan, Karrune, Tam, Eric, Vergara-Lluri, Maria E., Chaudhary, Preet M., Douer, Dan, Ladha, Abdullah, and Yaghmour, George
- Published
- 2022
- Full Text
- View/download PDF
15. m6A Maintains Hematopoietic Stem and Progenitor Cell Identity
- Author
-
Cheng, Yuanming, primary, Luo, Hanzhi, additional, Izzo, Franco, additional, Pickering, Brian F, additional, Vu, Ly P, additional, Schurer, Alexandra, additional, Gourkanti, Saroj, additional, Jaffrey, Samie R., additional, Landau, Dan A., additional, and Kharas, Michael G., additional
- Published
- 2018
- Full Text
- View/download PDF
16. RNA Binding Protein Rbmx Is Required in Acute Myeloid Leukemia By Regulating the Transcriptional Activity of the Heterochromatin Protein HP1α
- Author
-
Prieto, Camila, primary, Nguyen, Diu, additional, Vu, Ly P, additional, Perez, Alexendar, additional, Gourkanti, Saroj, additional, Schurer, Alexandra, additional, Chou, Timothy, additional, Chow, Arthur, additional, Taggart, James, additional, Barlowe, Trevor Stephen, additional, Leslie, Christina, additional, and Kharas, Michael G., additional
- Published
- 2018
- Full Text
- View/download PDF
17. CRISPR screen identifies genes that sensitize AML cells to double-negative T-cell therapy
- Author
-
Soares, Fraser, Chen, Branson, Lee, Jong Bok, Ahmed, Musaddeque, Ly, Dalam, Tin, Enoch, Kang, Hyeonjeong, Zeng, Yong, Akhtar, Nayeema, Minden, Mark D., He, Housheng Hansen, and Zhang, Li
- Abstract
Acute myeloid leukemia (AML) remains a devastating disease in need of new therapies to improve patient survival. Targeted adoptive T-cell therapies have achieved impressive clinical outcomes in some B-cell leukemias and lymphomas but not in AML. Double-negative T cells (DNTs) effectively kill blast cells from the majority of AML patients and are now being tested in clinical trials. However, AML blasts obtained from ∼30% of patients show resistance to DNT-mediated cytotoxicity; the markers or mechanisms underlying this resistance have not been elucidated. Here, we used a targeted clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) screen to identify genes that cause susceptibility of AML cells to DNT therapy. Inactivation of the Spt-Ada-Gcn5-acetyltransferase (SAGA) deubiquitinating complex components sensitized AML cells to DNT-mediated cytotoxicity. In contrast, CD64 inactivation resulted in resistance to DNT-mediated cytotoxicity. Importantly, the level of CD64 expression correlated strongly with the sensitivity of AML cells to DNT treatment. Furthermore, the ectopic expression of CD64 overcame AML resistance to DNTs in vitro and in vivo. Altogether, our data demonstrate the utility of CRISPR/Cas9 screens to uncover mechanisms underlying the sensitivity to DNT therapy and suggest CD64 as a predictive marker for response in AML patients.
- Published
- 2021
- Full Text
- View/download PDF
18. m6A Maintains Hematopoietic Stem and Progenitor Cell Identity
- Author
-
Alexandra Schurer, Dan A. Landau, Brian F. Pickering, Ly P. Vu, Franco Izzo, Michael G. Kharas, Saroj Gourkanti, Samie R. Jaffrey, Hanzhi Luo, and Yuanming Cheng
- Subjects
Myeloid ,Cell division ,Immunology ,Cell Biology ,Hematology ,Biology ,Cell cycle ,Biochemistry ,Embryonic stem cell ,Cell biology ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Stem cell ,Progenitor cell - Abstract
N-6-methyladenosine (m6A) is one of the most abundant posttranscriptional modifications in eukaryotic mRNAs and long noncoding RNAs. We previously found a critical role for m6A in promoting human myeloid leukemia (Vu et al. Nature Medicine 2017). Targeting the RNA methylation program in leukemias has been suggested as a potential novel therapeutic strategy. However, it is unknown whether the m6A modification controls normal adult hematopoiesis and hematopoietic stem cells (HSC) function. To investigate the role of m6A in adult hematopoiesis, we crossed with the Mettl3 conditional knockout (cKO) mouse model with the interferon inducible Mx1-Cre system to abolish m6A in the hematopoietic compartment. Deletion of Mettl3 (3 weeks post pIpC injections) resulted in pancytopenia (white blood count 9.44 k/ul in WT versus 4.35 k/ul cKO) and a 55% reduction in red blood cell counts and nearly 70% loss in platelet counts. Most remarkably, METTL3 depletion resulted in a 5-fold increase in the number of overall hematopoietic stem and progenitors HSPCs (LSKs; Lin-c-kit+Sca1+). Within the HSPC compartment there was a about a 10-fold expansion in immunophenotypic long term hematopoietic stem cells LT-HSCs (Lin-c-kit+Sca1+CD150+CD48-), multipotent progenitors (MPP-2s), (Lin-c-kit+Sca1+CD150+CD48+) and MPP4s (Lin-c-kit+Sca1+CD150-CD48+). In contrast to this general increase in HSPCs, we observed a decrease in common myeloid progenitor (CMP), granulocyte-monocyte progenitor (GMP) populations by 70% and 60% respectively. Altogether, these results suggest that loss of METTL3 results in a partial blockage in hematopoietic stem and progenitor cell differentiation, and an accumulation of LT-HSC and MPPs. Despite this phenotypic expansion of LT-HSCs and MPPs, their function was impaired as demonstrated by a reduction in long-term chimerism in non-competitive transplants into congenic mice compared to the remaining wildtype host cells (66% in WT vs 26% in cKO). Interestingly, the relative differentiation block and accumulation of HSCs remained after transplantation. Furthermore, Mettl3 cKO HSCs are less quiescent (79% WT vs. 44% cKO) and more proliferative based on cell cycle profiling with pyronin Y/Hoechst or Ki67/Hoechst. RNA-seq in sorted LT-HSC, MPP1, MPP2 and MPP4s from WT and Mettl3 cKO mice demonstrated that Mettl3 cKO HSCs lose the HSC signature while MPP1 and MPP2 cells exhibited gene signatures resembling WT HSCs. Interestingly, genes uniquely upregulated in Mettl3 cKO LT-HSC showed a significant enrichment of the Mettl3 KO mouse embryonic stem cells (mESCs) expression signature. To decipher METTL3 regulation of transcriptional states in individual cells along the hematopoietic hierarchy, we performed single-cell RNA-seq using sorted WT and Mettl3 cKO cKit+ cells. Surprisingly, tSNE analysis of scRNA-seq data uncovered the loss of the normal HSC cluster and the emergence of three unique HSC-like populations in Mettl3 cKO mice. Single cell analysis identified a reduction in c-MYC levels in the HSPCs that was then confirmed by immunofluorescence in sorted HSCs. More importantly, when forced into division, Mettl3 cKO HSCs exhibited increased symmetric c-MYC low division pattern compared to control (60% to 48%) based on c-MYC IF staining in paired daughter cells. We further confirmed that sorted HSCs underwent increased symmetric renewal divisions in the Mettl3 cKO compared to controls (65% to 49%) without any effect on asymmetric division based on NUMB IF staining in paired daughter cells. These data suggest that without METTL3, HSCs are partially blocked in a self-renewing state albeit with reduced fitness compared to wildtype HSCs. Overall, our studies uncovered a novel role for METTL3 and RNA methylation to maintain normal HSC identity and progenitor differentiation. Also, these studies suggest that inhibiting METTL3 could result in significant hematopoietic defects. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
19. RNA Binding Protein Rbmx Is Required in Acute Myeloid Leukemia By Regulating the Transcriptional Activity of the Heterochromatin Protein HP1α
- Author
-
Arthur Chow, Timothy Chou, Saroj Gourkanti, James Taggart, Ly P. Vu, Alexendar R. Perez, Camila Prieto, Michael G. Kharas, Christina S. Leslie, Trevor Stephen Barlowe, Alexandra Schurer, and Diu T.T. Nguyen
- Subjects
biology ,DNA damage ,Heterochromatin ,Immunology ,Myeloid leukemia ,RNA-binding protein ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Chromatin ,Cell biology ,Leukemia ,Histone ,hemic and lymphatic diseases ,Nucleic acid ,biology.protein ,medicine - Abstract
Acute myeloid leukemia (AML) is characterized by a block in the development of myeloid cells, often due to dysregulation of genes involved in key processes including self-renewal, proliferation, and differentiation. Somatic mutations and aberrant expression of RNA binding proteins (RBPs) have recently been found to be important in hematological malignancies. For example, our group and others have recently determined that increased expression of MUSASHI-2 and SYNCRIP drives aggressive leukemia. To discover novel RBP regulators of leukemia, we performed an in vivo pooled shRNA screen of 127 MSI2 direct protein interactors and associated genes (Vu et al. Nat Gen. 2017). In this screen, shRNAs specific to the RBP RBMX (RNA binding motif protein, X-linked) were selectively depleted in murine MLL-AF9 driven leukemia. RBMX has been implicated in regulating alternative splicing, chromatin cohesion, and DNA-damage response, but its function in hematopoiesis and leukemia is not known. We confirmed that depletion of RBMX with shRNAs in murine MLL-AF9 leukemia cells resulted in reduced myeloid colony formation, increased apoptosis, and increased differentiation as determined by flow analysis of myeloid cell surface markers Gr-1 and Mac-1 (mean of 61-65% shRNA versus mean of 12.95% control). Furthermore, RBMX is highly expressed among human myeloid leukemia cell lines (n=10/11) and primary AML patient samples (n=2/4). Depletion of RBMX with shRNAs led to a dramatic decrease in cell proliferation and 3-fold induction of apoptosis in several human myeloid leukemia cell lines (MOLM-13, THP-1, K562, and KCL-22). Additionally, RBMX depletion in AML cells induced myeloid differentiation and significantly delayed leukemogenesis cells in vivo (median survival of 51.5 days in control versus median 'not reached' in shRNA1 and shRNA2). To determine if there is a differential requirement of RBMX in survival of leukemia cells compared to normal hematopoietic stem and progenitor cells (HSPCs), we depleted RBMX with shRNAs in normal murine bone marrow c-Kit+ cells and found no significant changes in colony formation. Depleting RBMX with shRNAs in human cord blood derived CD34+ HSPCs resulted in reduced colony formation but no increase in apoptosis. Thus, these data suggest that there is a differential requirement for RBMX in myeloid leukemia cells compared to normal cells. To uncover the mechanism of RBMX function, we performed RNA-sequencing of human AML cells (MOLM-13) depleted for RBMX. Gene set enrichment analysis demonstrated a loss of cell cycle and DNA repair associated programs in RBMX depleted cells. Complex chromosomal karyotyping analysis of these cells revealed increased metaphases with breaks and gaps (mean of 30.67% shRNA versus mean of 13.33% control) and irregular chromatin compaction (mean of 47.67%shRNA versus mean of 20% control), while cell cycle analysis showed significantly increased S-phase arrest as determined by flow analysis of Hoechst stained cells (mean of 37-40% shRNA versus of 24.18% control). Reanalysis of RBMX transcriptome-wide binding sites in 293T cells revealed that RBMX directly binds to heterochromatin protein HP1α transcripts (Liu et al. Nucleic Acids Res. 2017). HP1α, also called CBX5, is a key heterochromatin protein that binds to histone H3-K9 tri-methylation marks to promote heterochromatin formation, which is critical in chromatin condensation and chromosome segregation. HP1α has also been determined to be required for MLL leukemia stem cell maintenance. We demonstrated that RBMX depletion resulted in a significant decrease of HP1α mRNA expression without affecting its mRNA stability in AML cells. We confirmed that RBMX depletion reduced the protein abundance of HP1α. Moreover, overexpression of HP1α rescued the effect of RBMX depletion on cell growth and apoptosis. Our study finds that RBMX binds to HP1α mRNA and regulates the transcriptional activity of the HP1α locus, which then maintains proper chromatin compaction in leukemia cells. Overall, we determine that RBMX function is critical for myeloid leukemia survival and has potential as a novel therapeutic target in AML. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
20. m6a Regulates Differentiation State and mRNA Translation in Myeloid Leukemia
- Author
-
Vu, Ly P, primary, Pickering, Brian F, additional, Cheng, Yuanming, additional, Sara, Zaccara, additional, Nguyen, Diu, additional, Minuesa, Gerard, additional, Chou, Timothy, additional, Chow, Arthur, additional, MacKay, Matthew, additional, Famulare, Christopher, additional, Patel, Minal, additional, Garret-Bakelman, Francine, additional, Melnick, Ari, additional, Carroll, Martin, additional, Mason, Christopher, additional, Jaffrey, Samie R., additional, and Kharas, Michael G., additional
- Published
- 2017
- Full Text
- View/download PDF
21. m6a Regulates Differentiation State and mRNA Translation in Myeloid Leukemia
- Author
-
Michael G. Kharas, Diu T.T. Nguyen, Matthew MacKay, Arthur Chow, Francine Garret-Bakelman, Timothy Chou, Gerard Minuesa, Minal Patel, Ari Melnick, Ly P. Vu, Yuanming Cheng, Christopher E. Mason, Samie R. Jaffrey, Christopher Famulare, Brian F. Pickering, Martin Carroll, and Zaccara Sara
- Subjects
Gene knockdown ,Myeloid ,Immunology ,CD34 ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Cancer research ,medicine ,Progenitor cell ,Stem cell - Abstract
N 6-methyladenosine (m6A) is a nucleotide modification in mRNA that is required for the acquisition of cell fate in embryonic stem cells. Recent studies have indicated that methylation writers can act as both oncogenes and tumor suppressor genes. Here we show that m6A is a critical regulator of myeloid differentiation of human hematopoietic stem and progenitors and myeloid leukemia cells. Depletion of the m6A-forming enzyme METTL3 with shRNAs in human cord blood derived CD34+ hematopoietic stem/progenitor cells (HSPCs) decreased global m6A levels, promoted differentiation, and reduced cell proliferation and colony formation. Conversely, overexpression of wild type METTL3-WT, but not the catalytic dead form METTL3-CD, in HSPCs increased global m6A levels, inhibited differentiation, and promoted cell growth. We surveyed METTL3 expression across different cancers and found that METTL3 is expressed at the highest level in acute myeloid leukemia (AML) compared to a broad range of solid tumors. METTL3 was highly expressed in wide variety of human AML cell lines (11/11) and in primary AML patients (3/3). Utilizing 2D-Thin layer chromatography (TLC), we found that m6A levels in poly(A) mRNAs were significantly increased in the MOLM13 leukemia cells and primary AML patient cells compared to normal HSPCs. To directly address the role of m6A in human myeloid leukemia cells, we demonstrated that METTL3 depletion with shRNAs resulted in reduced cell proliferation and the induction of apoptosis in human AML cell lines (MOLM13, NOMO-1 and KASUMI-1), and delayed leukemia in vivo (MOLM13 cells - median survival of 23 days control versus 35 and 36 days shRNA#9 and shRNA#12 respectively - in vivo knockdown was negatively selected as determined by immunoblotting). Validating an on-target effect, expression of shRNA-resistant form of METTL3 rescued both the apoptotic and differentiation phenotypes in METTL3-depleted cells and CRISPR/Cas9 mediate depletion of METTL3 (using two independent guide RNAs) resulted in similar phenotypes. Additionally, we looked into the rank of m6A "writers" and "erasers" in the genome-wide CRISPR-based screen (Wang et al. Cell 2017) for genes essential for survival in 14 AML cell lines. Interestingly, while all members of the "writer" complexes, METTL3, METTL14, WTAP and KIAA1429, were scored highly (at the top 10%), the eraser ALKBH5 showed no essentiality and FTO is only important for survival of EOL-1 cells. This data suggests that the m6A writer complex is required for leukemia. To gain insights into the mechanism of why m6A and METTL3 are required in leukemia, we performed single-nucleotide resolution mapping of m6A in MOLM13 cells using miCLIP, gene expression analysis, ribosome profiling and reverse phase protein arrays in MOLM13 cells depleted for METTL3. We profiled mRNAs targeted for m6A modifications and found that transcripts with m6A were more stable but were translated with less efficiency. Gene Set Enrichment Analysis revealed negative enrichment of MYC and ESCs gene sets. We further validated c-MYC, BCL2 and PTEN as targets for m6A modifications. We performed meRIP-qPCR and showed that knockdown of METTL3 specifically reduced enrichment of m6A at mapped sites of these transcripts. We demonstrated that c-MYC, BCL2 and PTEN protein expression were reduced despite a 2-5 log2 fold increase in mRNA expression after METTL3 depletion. The effects were reversed in cells overexpressing METTL3-WT, but not the METTL3-CD. Importantly, we observed a robust activation of p-AKT upon METTL3 knockdown, and treatment with inhibitors of PI3K and AKT partially rescued the differentiation effects in METTL3 depleted cells. Overall, m6A is critical for maintaining the differentiation program in the hematopoietic system and that this process is dysregulated in myeloid leukemia. Our data provides a rationale for targeting the mRNA methylation program in myeloid leukemia. Disclosures Carroll: Astellas Pharmaceuticals: Research Funding; Incyte Pharmaceuticals: Research Funding.
- Published
- 2017
- Full Text
- View/download PDF
22. RNA Binding Protein Syncrip Regulates the Leukemia Stem Cell Program
- Author
-
Vu, Ly P., primary, Prieto, Camila, additional, Amin, Elianna, additional, Minuesa, Gerard, additional, Chhangawala, Sagar, additional, Vidal, Maria J, additional, Krivtsov, Andrei V., additional, Chou, Tim, additional, Barlowe, Trevor, additional, Taggart, James, additional, Tivnan, Patrick, additional, Deering, Raquel P., additional, Gonen, Mithat, additional, Figueroa, Maria E, additional, Paietta, Elisabeth, additional, Tallman, Martin S., additional, Melnick, Ari, additional, Levine, Ross L., additional, Al-Shahrour, Fatima, additional, Järås, Marcus, additional, Chu, Lisa, additional, Garippa, Ralph, additional, Lengner, Christopher, additional, Armstrong, Scott A., additional, Cowley, Glenn, additional, Root, David, additional, Doench, John G, additional, Cerchietti, Leandro, additional, Leslie, Christina, additional, Ebert, Benjamin, additional, and Kharas, Michael G., additional
- Published
- 2016
- Full Text
- View/download PDF
23. RNA Binding Protein Syncrip Regulates the Leukemia Stem Cell Program
- Author
-
Patrick Tivnan, Ross L. Levine, Andrei V. Krivtsov, Elianna M. Amin, Scott A. Armstrong, Leandro Cerchietti, Raquel P. Deering, Gerard Minuesa, Camila Prieto, Elisabeth Paietta, Mithat Gonen, Michael G. Kharas, David E. Root, Tim Chou, Trevor Stephen Barlowe, Christina S. Leslie, Maria E. Figueroa, Sagar Chhangawala, Christopher J. Lengner, John G. Doench, Martin S. Tallman, Fatima Al-Shahrour, Ralph Garippa, Marcus Järås, Maria Jesus Vidal, James Taggart, Ly P. Vu, Ari Melnick, Lisa Chu, Glenn S. Cowley, and Benjamin L. Ebert
- Subjects
Immunology ,Myeloid leukemia ,RNA-binding protein ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Cell biology ,Small hairpin RNA ,Haematopoiesis ,Leukemia ,Myeloid stem cell ,Cancer stem cell ,medicine ,Stem cell - Abstract
RNA binding proteins (RBPs) tightly control mRNA abundance, stability and translation while mutations or altered expression of specific factors can drive malignancy. Yet, the identity of the RBPs that govern myeloid stem cells remains poorly characterized. We and others have recently demonstrated that MUSASHI-2 (MSI2) is a central regulator of the cancer stem cell program in myeloid leukemia. Therefore, we curated a list of 127 MSI2 direct protein interactors and associated genes to perform an in vivo shRNA screen using MLL-AF9 leukemia cells. We identified shRNAs corresponding to 24 genes that were significantly depleted in vivo after sequencing and comparing their representation from day 16 to day 0. We confirmed knockdown and demonstrated marked reduction in myeloid colony formation in vitro after depleting 7 hits identified in our screen. Additionally, we tested these genes in normal bone marrow c-Kit positive cells and found that the most differentially required gene in leukemia cells compared to normal cells was SYNCRIP (Synaptotagmin-binding, cytoplasmic RNA-interacting protein). SYNCRIP is an RNA binding protein that has been implicated in various RNA regulatory processes but its role in the hematopoietic system is virtually unknown. Depletion of SYNCRIP with shRNAs in murine MLL-AF9 leukemia cells resulted in an increase in myeloid differentiation, apoptosis and delayed leukemogenesis in vivo (median survival of 35 days; control versus 61 days shRNA#1 knockdown was selected against, and "not reached" shRNA#2). To further assess SYNCRIP function in vivo, we developed a germline Syncrip knockout (KO) by injecting Cas9-DNA and Syncrip - guides RNAs into embryos and harvested E13 fetal liver cells. After Syncrip deletion was verified by immunoblotting, we observed normal numbers of HSCs and equivalent engraftment in lethally irradiated animals in both primary and secondary transplants. In contrast, we observed a delay in leukemeogenesis (median survival of 87.5 days; WT versus 118 days KO) in recipient mice after transplantation of MLL-AF9 transformed LSKs. Notably, non-deleted leukemia cells outcompeted the SYNCRIP deleted cells based on a reemergence of SYNCRIP expression. These data suggest that SYNCRIP is differentially required in myeloid leukemia cells compared to normal cells. Furthermore, we found that SYNCRIP was highly expressed in wide variety of human AML cell lines and in primary AML patients (n=4/5). SYNCRIP depletion with shRNAs resulted in reduced cell proliferation and the induction of apoptosis in human AML cell lines (MOLM13, NOMO-1, KASUMI-1 and NB4) and a marked decrease in engraftment of primary AML patient cells. To gain insights into SYNCRIP function, we performed RNA-sequencing of leukemia cells depleted for SYNCRIP. Gene set enrichment analysis (GSEA) negatively enriched for the MLL-AF9, HOXA9 and stem cell programs in SYNCRIP-KD cells and positively enriched for MSI2's direct mRNA binding targets and a MSI2 deficient LSC signature. Reciprocal immunoblotting in the presence or absence of RNAse demonstrated that SYNCRIP and MSI2 interaction is RNA dependent. We validated their shared targets by performing SYNCRIP RNA-immunoprecipitation (RIP) for previously identified MSI2's direct mRNAs targets (HOXA9 and c-MYC). SYNCRIP depletion resulted in reduced protein abundance of HOXA9 and c-MYC. Forced MSI2 expression partially rescued the colony formation and HOXA9 expression in SYNCRIP-KD cells. To assess the functional downstream targets of SYNCRIP in leukemia, we overexpressed HOXA9 and c-MYC in SYNCRIP-KD cells and observed that HOXA9 expression but not c-MYC partially rescues the effect of SYNCRIP depletion on myeloid colony formation. Mechanistically, we showed that SYNCRIP regulates translation of HOXA9 without affecting HoxA9 mRNA stability. Overall, we provide a strategy for interrogating the functional RNA binding network in leukemia using shRNA screening. Additionally, we validated SYNCRIP as a novel RBP that controls the leukemia stem cell program and propose that targeting these functional complexes might provide a novel therapeutic strategy in myeloid leukemia. Disclosures Melnick: Janssen: Research Funding. Levine:Novartis: Consultancy; Qiagen: Membership on an entity's Board of Directors or advisory committees. Järås:Cantargia AB: Equity Ownership.
- Published
- 2016
- Full Text
- View/download PDF
24. CD38-Directed, Single-Chain T Cell-Engager Targets Leukemia Stem Cells through IFNγ-Induced CD38 Expression
- Author
-
Murtadha, Mariam, Park, Miso, Zhu, Yinghui, Caserta, Enrico, Napolitano, Ottavio, Tandoh, Theophilus, Moloudizargari, Milad, Pozhitkov, Alexander, Singer, Mahmoud, Dona, Ada Alice, Vahed, Hawa, Gonzalez, Asaul, Ly, Kevin, Ouyang, Ching, Sanchez, James, Nigam, Lokesh, Chowdhury, Arnab, Ghoda, Lucy Y., Li, Ling, Zhang, Bin, Krishnan, Amrita, Marcucci, Guido, Williams, John C., and Pichiorri, Flavia
- Abstract
Leukemia stem cells (LSCs), enriched in the CD34+CD38- AML population, are considered the main drivers of relapse in acute myeloid leukemia (AML). While more differentiated progenitors and bulk AML blasts reside in the CD34+CD38+ fraction, targeting CD38 in AML is less pursued, as LSCs are mainly CD38-. We show IFNγ (10 ng/ml) induced an increase in CD38 mRNA (mean fold change IFNγ over vehicle = 7.7, p=0.03, n=3) and surface CD38 (mean CD38 High% population IFNγ 56%±17 vs vehicle 16%±10, p<0.0001) in both CD34+ and CD34- AML primary blasts, and decreased the clonogenic activities of primary AML cells (colony forming unit IFNγ 50±36 vs vehicle 216±117, p=0.002, n=4) regardless of cytogenetics. scRNA and total RNA sequencing data in CD34+ AML primary blasts showed that upon IFNγ treatment, CD38 and Interferon Regulatory Factor 1 (IRF-1) were consistently the most upregulated genes compared to vehicle (>2.5 Log 2Fold Change; p=0; FDR=0). Furthermore, gene set enrichment analysis (FDR=0) indicated IFNγ induced an upregulation of genes involved in apoptosis and DNA repair, and a suppression of genes in the pro-oncogenic Wnt beta catenin pathways. CD38 luciferase-based reporter assays, chromatin immunoprecipitation, and IRF-1 knockdown experiments in AML cells showed that, upon IFNγ treatment, IRF1 is pivotal in CD38 transcriptional regulation.
- Published
- 2023
- Full Text
- View/download PDF
25. IFNγ Signaling in Acute Myeloid Leukemia Mediates Immunomodulation and Reveals Therapeutic Strategies
- Author
-
Wang, Bofei, Reville, Patrick K, Jelloul, Fatima Z, Desai, Poonam N, Borges, Pamella, Ly, Christopher, Veletic, Ivo, Dasdemir, Enes, Burks, Jared K., Tang, Guilin, Guo, Shengnan, Vaugh, Nicole, Baran, Natalia, Deng, Qing, Matthews, Jairo, Gunaratne, Preethi, Antunes, Dinler A, Ekmekcioglu, Suhendan, Sasaki, Koji, Garcia, Miriam B., Cuglievan, Branko, Hao, Dapeng, Daver, Naval, Green, Michael R., Konopleva, Marina Y., Futreal, Andy, Post, Sean M., and Abbas, Hussein A
- Abstract
Background: Interferon gamma (IFNγ) is a critical cytokine known for its diverse roles in immune regulation, inflammation, and tumor surveillance. Studies have reported the dichotomous nature of IFNγ signaling in both the pathogenesis of cancer and immunotherapy response. However, its complex interplay with acute myeloid leukemia (AML) remains insufficiently understood.
- Published
- 2023
- Full Text
- View/download PDF
26. Characterizing Inferred Functional and Phenotypical States of Newly Diagnosed Acute Myeloid Leukemia from Single Cell Transcriptomic Profiling
- Author
-
Wang, Bofei, Ly, Christopher, Dasdemir, Enes, Jelloul, Fatima Z, Desai, Poonam N, Borges, Pamella, Root, Jessica Lynn, Tang, Guilin, Gunaratne, Preethi, Basu, Sreyashi, Jindal, Sonali, Chen, Yulong, Wani, Khalida M, Lazar, Alexander J, Sharma, Padmanee, Reville, Patrick K, and Abbas, Hussein A
- Abstract
Background:Acute myeloid leukemia (AML) is a heterogeneous disease, marked by leukemia stem cells (LSCs), which significantly contribute to treatment resistance and dismal prognosis. While appreciable efforts have been made to study the diversity of functional and phenotypic states of LSCs, integrating these states from inferred single cell profiling has not been explored. Moreover, the spatial distribution and variability of hierarchical leukemic states within diverse cytogenetic groups, and their potential connection to transcriptional profiles, remain understudied, highlighting a gap in our understanding of this complex disease.
- Published
- 2023
- Full Text
- View/download PDF
27. Phase 1 Dose Escalation Study of Pembrolizumab in Combination with Rituximab and Lenalidomide for Relapsed/Refractory Large B-Cell or Follicular Lymphoma Following Progression after Anti-CD19 Chimeric Antigen Receptor (CAR) T-Cell Therapy
- Author
-
McCurry, Dustin, Westin, Jason, Feng, Lei, Ahmed, Sairah, Nair, Ranjit, Steiner, Raphael Eric, Wynn, Lauren, Thomas, Jennifer, Dsouza, Ly Huynh, Samaniego, Felipe, Neelapu, Sattva S., and Nastoupil, Loretta J.
- Abstract
Introduction:While autologous CD19-targeting chimeric antigen receptor T-cells (CAR T) can induce high complete response (CR) rates in relapsed and refractory (r/r) large B-cell lymphoma (LBCL) and follicular lymphoma (FL), over 50% of patients experience progressive disease. Survival following CAR T failure is dismal, with only 1 in 5 patients living >2 years. Multiple studies indicated that resistance to CAR T therapy may be driven by CAR T cell intrinsic and/or tumor intrinsic mechanisms. Here, we evaluated the combination of PD-1 blockade with rituximab (R) and lenalidomide (len) with the rationale that it would restore the function of exhausted T cells, enhance CAR T function with len and target the tumor and its immunosuppressive microenvironment.
- Published
- 2023
- Full Text
- View/download PDF
28. Prospective Molecular Characterization of Multiple Myeloma Patient Samples Identifies High-Risk Patients and Informs Treatment Sequences through Resistance Mechanisms to Immunotherapies
- Author
-
Pham, Phillip, Sudha, Parvathi, Wang, Lin, Niu, Wen, Morgan, Cameron, Ligocki, Cameron, Al-Azzawi, Ramzi, Ly, Reynold, Vetrini, Francesco, Czader, Magdalena, Tayeh, Marwan, Abu Zaid, Mohammad, Lee, Kelvin P., Suvannasankha, Attaya, Abonour, Rafat, and Walker, Brian A.
- Abstract
Introduction.Multiple myeloma (MM) genetic diagnostics has traditionally been performed using fluorescence in situ hybridization (FISH), giving prognostic information based on immunoglobulin (Ig) translocations and key copy number abnormalities such as 1p, 1q, and 17p. However, in recent years it has become clear that more mutations, biallelic events, and focal deletions are also prognostically important and can be captured better through molecular techniques such as DNA sequencing. In addition, as targeted and immunotherapies become standard, monitoring the molecular targets will be important in determining the optimal sequence of therapies.
- Published
- 2023
- Full Text
- View/download PDF
29. Mitochondrial Pyruvate Carrier Inhibition Mitigates Murine Chronic Graft Versus Host Disease By Attenuating the Germinal Center Reaction
- Author
-
Mohamed, Fathima A, Rhee, Stephanie Y, Ly, Joanna, Aguilar, Ethan G, Melin, Haley, Sage, Peter T, Schumacher, Tanner, Thangavelu, Govindarajan, Zaiken, Michael C, Liu, Juan, Mereddy, Venkatram, Locasale, Jason W, and Blazar, Bruce R
- Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potent therapy for many hematopoietic diseases and malignancies. However, the long-term efficacy of allo-HSCT is limited by the development of chronic graft versus host disease (cGVHD). During cGVHD, interactions between donor-derived T follicular helper cells (TFH) and germinal center B cells (GCB) result in the production of alloreactive class-switched affinity-matured antibodies that attack host tissues, causing multi-organ damage and tissue fibrosis. Novel treatment approaches are warranted to treat cGVHD since it is often refractory to current broad immune suppressive drugs. In this study we sought to delineate the metabolic dependencies of germinal center (GC) immune cells to discover new strategies to treat active murine cGVHD.
- Published
- 2023
- Full Text
- View/download PDF
30. Targeting Aberrantly Activated Glycolysis in Natural Killer T Cell Lymphomas
- Author
-
Espinoza, Luis, Nguyen, Viet Hoang, Quoc Trung, Ly, and Takami, Akiyoshi
- Abstract
Background and aims:With advances in the diagnosis and treatment of malignant lymphoma, improvements in long-term survival have been achieved for most common lymphoma types. However, natural killer/T-cell lymphoma (NKTL), which is rare in Europe and North America (0.3%) but relatively common in Asia and Latin America, has been left behind as a subtype of lymphoma with a high relapse rate and a poor prognosis. In particular, the outcome of patients with advanced or recurrent NKTL is dismal, with an overall survival of only a few months. This is mainly attributed to the intrinsic resistance of NKTL cells to anthracycline due to the high expression of P-glycoprotein. Molecular-targeted drugs used for other hematological malignancies, such as anti-CD30 and anti-CD52 monoclonal antibodies, have only limited effects on NKTL, and conversely, these drugs can cause severe immunodeficiency and severe autoimmune diseases, often with a fatal course. The development of NKTL-specific and effective molecular-target therapies is expected to improve the prognosis of NKTL. Methods:We performed a gene expression array targeting genes involved in glucose cell metabolism to compare the gene signatures of NK cell lines (NKL, NK92, YT, and KYHG1) and freshly isolated NK cells from three healthy donors (FNK). Results:The expression of various glucose metabolism genes was higher in NK cell lines than in FNK cells, and in particular, the PKM gene was highly enriched in NK cell lines. The PKM gene encodes the pyruvate kinase muscle isozyme 2 (PKM2), which is responsible for ATP production within the glycolytic sequence and plays an essential role in aerobic glycolysis (the Warburg effect), a dominant metabolic pathway used by cancer cells. Immunohistochemistry studies revealed high PKM2 protein expression in 17 out of 20 tumor tissues derived from patients with extranodal NK/T cell lymphoma. Treatment of NKL, NK92, YT, and KYHG1 cells with Benserazide or Shikonin, two known PKM2 inhibitors, robustly induced cell apoptosis in a time- and dose-dependent manner and inhibited colony formation in in vitroclonogenic assays. Western blotting studies showed a dose-dependent inhibition of STAT1 and STAT3 in NK cell lines exposed to PKM2 inhibitors. A colorimetric assay showed that PKM2 glycolytic activity correlated with the PKM2 gene expression in NK cells, with significantly higher glycolytic activity in malignant NK cell lines than in normal FNK cells. Further mechanistic studies showed that in normal FNK cells, PKM2 is mainly in the tetrameric form, where glucose is converted to pyruvate to generate ATP. In contrast, in malignant NK cell lines, PKM2 is mainly expressed in its dimeric form, with a low affinity for pyruvate and a higher affinity for glycolytic intermediates that promote the synthetic processes and thereby lead to persistent cell proliferation. Conclusions:These results substantiate the importance of aerobic glycolysis in malignat NK cells and indicate that targeting this pathway by blocking PKM2 activity has therapeutic potential for NKTL.
- Published
- 2023
- Full Text
- View/download PDF
31. Eosinophils to Basophils Rate, Mean Platelet Volume, and Neutrophils to Lymphocyte Rate for the Diagnosis of Systemic Amyloidosis
- Author
-
Quoc Trung, Ly, Nguyen, Viet Hoang, Takami, Akiyoshi, and Espinoza, Luis
- Abstract
Methods: CBC, performed by an automated hematology analyzer, included hemoglobin
- Published
- 2023
- Full Text
- View/download PDF
32. Single-Cell Polyfunctional Proteomic Profiling Reveals Temporal and Niche Differences in CD4 and CD8 T Cells in Acute Myeloid Leukemia Following PD-1 Blockade Therapy
- Author
-
Root, Jessica Lynn, Desai, Poonam N, Reville, Patrick K, Ly, Christopher, Wang, Bofei, Jelloul, Fatima Z, Zhou, Jing, Mackay, Sean Gregory, Alfayez, Mansour, Matthews, Jairo, Pierce, Sherry, Daver, Naval, and Abbas, Hussein A
- Abstract
Immune checkpoint blockade (ICB) has revolutionized solid tumor treatment by unleashing T cell cytotoxic potential, however, its effectiveness in leukemias such acute myeloid leukemia (AML) has been limited. Understanding the functional states of T cells in AML within the context of ICB is crucial to understanding its limited therapeutic potential. Here, we leveraged the IsoPlexis single cell secretome platform to assess T cell polyfunctional activity in relapsed/refractory (RelRef) AML patients treated on the phase 2 clinical trial with combination azacitidine and nivolumab (Aza/Nivo) (NCT02397720). We isolated CD4 and CD8 T cells by magnetic bead sorting from the peripheral blood (PB) and bone marrows (BMs) of 21 AML patients before (pretreatment) and after ICB (post-ICB). Isolated CD4 and CD8 cells were stimulated with CD3/CD28 agonism and functionally characterized by IsoPlexis, which measures T cell secreted cytokines, chemokines, and effector molecules at the single-cell level. In total, we characterized 52 patient samples (30 from PB and 22 from BMs). Functional CD4 and CD8 profiles were correlated with clinical and demographic characteristics of the patients. This enabled us to explore the temporal and niche differences in T cell functional states, specifically in the context of PD-1 blockade therapy, in a unique cohort of AML patients at both a pseudobulk and single-cell level.
- Published
- 2023
- Full Text
- View/download PDF
33. IKZF2, a Novel Target of MSI2 RNA-Binding Protein Plays an Oncogenic Role in Myeloid Leukemia
- Author
-
Park, Sun Mi, primary, Thornton, Angela, additional, Vu, Ly P., additional, Chhangawala, Sagar, additional, Minuesa, Gerard, additional, Barlowe, Trevor, additional, Taggart, James, additional, Tivnan, Patrick, additional, Kim, Theodore, additional, Shevach, Ethan, additional, Leslie, Christina S, additional, and Kharas, Michael G., additional
- Published
- 2015
- Full Text
- View/download PDF
34. IKZF2, a Novel Target of MSI2 RNA-Binding Protein Plays an Oncogenic Role in Myeloid Leukemia
- Author
-
Ly P. Vu, Angela M. Thornton, Theodore Kim, Sun Mi Park, Ethan M. Shevach, Trevor S. Barlowe, Sagar Chhangawala, James Taggart, Gerard Minuesa, Michael G. Kharas, Patrick Tivnan, and Christina S. Leslie
- Subjects
Myeloid ,Cell growth ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Transplantation ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Bone marrow ,Stem cell - Abstract
Deregulated epigenetic program is found in many cancers, and genetic aberrations of histone methyltransferases contribute to transformation in myeloid leukemias. Post-transcriptional regulation in leukemia has recently been highlighted as a novel way for maintaining the leukemia stem cell (LSC) program. We have recently demonstrated that Msi2 is required for LSC function in a murine MLL-AF9 leukemia model. We determined that MSI2 maintains the mixed-lineage leukemia (MLL) self-renewal program by interacting and retaining efficient translation of critical MLL regulated transcription factors including Hoxa9, Myc and Ikzf2. Despite extensive studies implicating Myc and Hoxa9 in leukemia, the role for Ikzf2 in myeloid leukemia is not known. Ikzf2 is a member of the Ikaros transcription factor family and regulates lymphocyte development by controlling regulatory T-cell function. Ikzf2 is highly expressed in Hematopoietic Stem Cells (HSC) and to investigate if Ikzf2 is involved in HSC function, we utilized mice that have a specific deletion of Ikzf2 in the hematopoietic system through the Vav-cre system. We found similar frequencies for different populations in the stem, progenitor and mature cells in the bone marrow of Ikzf2f/f and Ikzf2D/D mice. Colony assays of isolated Lin- Sca1+ c-Kit+ (LSK) cells from Ikzf2f/f and Ikzf2D/D mice resulted in a comparable number of myeloid progenitor colonies. Furthermore, noncompetitive transplant of Ikzf2f/f and Ikzf2D/D bone marrow cells showed similar chimerism after 34 months indicating that I kzf2D/D mice have normal HSC function and hematopoiesis. To interrogate the role of Ikzf2 in acute myeloid leukemia we utilized the MLL-AF9 retroviral transduction model. Intracellular flow cytometry showed that IKZF2 is highly expressed in the LSC population compared to the non-LSCs. We then transduced Ikzf2f/f and Ikzf2D/D LSK cells with MLL-AF9 and found that Ikzf2 deletion results in a ten-fold reduction in colony formation compared to Ikzf2f/f cells. Transplantation of transduced cells results in delayed leukemia progression with reduced disease burden. Secondary transplantation of the initiation experiment exhibited a significant delay in leukemogenesis in the Ikzf2D/D compared to the Ikzf2f/f mice (median survival of 32 and 19.5 days, respectively). The role for Ikzf2 in maintenance was assessed with an inducible puro-creER system, which resulted in 80% decrease in viable cell number within 24hrs of 4-hydroxytamoxifen (4-OHT) treatment. Flow cytometric analysis showed that the Ikzf2-deficient cells had increased apoptosis and differentiation, shown by AnnexinV/7-AAD and Mac1 expression respectively. Furthermore, inducible deletion of Ikzf2 using puro-creER system in vivo revealed that Ikzf2 deletion leads to a delay in leukemia after tamoxifen administration in mice. These results indicate that Ikzf2 is required for both leukemia initiation and maintenance. To determine a role for IKZF2 in human leukemia cells, we performed Ikzf2 knockdown experiments with shRNAs in Kasumi-1, KG1, KCL22 and MOLM13 cells. Ikzf2 depletion resulted in decreased cell growth and increased apoptosis compared to cells infected with scramble shRNA. To determine the mechanism for how IKZF2 controls leukemia cell survival and self-renewal, we performed gene expression profiling of the Ikzf2-deficient Vav-cre LSCs and demonstrated enrichment in signatures for self-renewal loss, increased differentiation, loss of Myc-regulated genes and loss for targets of Hoxa9 and Meis1. Further analysis overlapping our MSI2 HITS-CLIP data and our differentially regulated genes revealed a strong enrichment suggesting that the MSI2 bound targets are transcriptionally regulated by IKZF2. Lastly, ATAC-sequencing of Ikzf2f/f and Ikzf2D/D LSCs revealed alterations in chromatin accessibility that correlated closely with differentially expressed genes. Utilizing the ATAC-seq data we predicted that HOXA9 and MYC sites were significantly altered. We validated that MYC RNA and protein levels were reduced in both murine and human AML cell lines. In contrast to its known tumor suppressor role in hypodiploid B-ALL and T-ALL, these results suggest that Ikzf2 contributes to MLL leukemia cell initiation and maintenance. Thus, we provide evidence that Ikzf2 can regulate c-MYC expression helping in maintaining the stem cell self-renewal program in LSCs. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
- Full Text
- View/download PDF
35. Msi2 Maintains the MLL Leukemia Stem Cell Regulatory Program
- Author
-
Park, Sun Mi, primary, Gönen, Mithat, additional, Vu, Ly P., additional, Minuesa, Gerard, additional, Tivnan, Patrick, additional, Barlowe, Trevor, additional, Taggart, James, additional, Lu, Yuheng, additional, Deering, Raquel P, additional, Hacohen, Nir, additional, Figueroa, Maria, additional, Paietta, Elisabeth M., additional, Tallman, Martin S., additional, Melnick, Ari M., additional, Levine, Ross L., additional, Leslie, Christina, additional, Lengner, Christopher J, additional, and Kharas, Michael G, additional
- Published
- 2014
- Full Text
- View/download PDF
36. Msi2 Maintains the MLL Leukemia Stem Cell Regulatory Program
- Author
-
Christina S. Leslie, Trevor S. Barlowe, Elisabeth Paietta, Nir Hacohen, Mithat Gonen, Michael G. Kharas, Ly P. Vu, Ari Melnick, Christopher J. Lengner, Gerard Minuesa, Patrick Tivnan, Ross L. Levine, Raquel P. Deering, Sun Mi Park, James Taggart, Martin S. Tallman, Maria E. Figueroa, and Yuheng Lu
- Subjects
Myeloid ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Gene expression profiling ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,Conditional gene knockout ,medicine ,Cancer research ,Stem cell ,Progenitor cell - Abstract
Leukemia stem cells (LSCs) are found in most aggressive myeloid diseases and contribute to therapeutic resistance. LSCs are characterized by their gain of a self-renewal program that is normally associated with hematopoietic stem cells (HSCs). Previously we have shown that the RNA binding protein, Msi2 contributes to both HSC and myeloid leukemia function. Elevated MSI2 expression predicts a poor prognosis in a variety of leukemias and shRNA-mediated depletion in human AML cell lines reduces proliferation, increases differentiation and induces apoptosis. Despite these in vitroand correlative studies, MSI2’s molecular mechanism is not known and its role in LSC function has not been assessed. To elucidate MSI2’s role in LSC function, we utilized the MLL-AF9 leukemia mouse model. Initially we found MSI2 was elevated in the LSC enriched compartment (c-KitHigh cells) compared to non-LSCs (c-KitLow cells) based on flow cytometric intracellular staining. Therefore, to establish a model to study Msi2 and its contribution to myeloid LSCs, we have utilized the Msi2 conditional knockout mice that we previously crossed (Msi2f/f) into an Mx1-Cre background to generate the Msi2Δ/Δallele (injection of polyinositol-polycytosine; pIpC). In order to test if Msi2 is critical for MLL-AF9 mediated initiation, we transduced control Msi2f/f and Msi2Δ/ΔLin- Sca1+ c-Kit+cells (LSKs) with MLL-AF9 expressing retroviruses co-expressing GFP. Msi2 deleted LSKs or granulocyte-monocyte progenitors (GMPs) transduced with MLL-AF9 demonstrated delayed leukemogenesis with dramatically reduced diseased burden. Msi2 deficient leukemias were found to have a 4-fold reduced phenotypic LSC population and were more differentiated based on cellular morphology. Msi2 deficient leukemias failed to transplant into secondary recipients demonstrating that Msi2 is required for maintaining LSCs. Deletion of Msi2after leukemia engraftment led to a delay in leukemogenesis indicating that Msi2 is also important for leukemia maintenance. Gene expression profiling of the Msi2 ablated LSCs resulted in a loss of the HSC/LSC program and an increase in differentiation gene sets. The gene signature from the Msi2 deleted murine LSCs (121 genes) was overlapped and subjected to unsupervised clustering with the gene expression profiles from 336 AML patients (ECOG1900 dataset). This analysis resulted in distinct clusters that had differential MSI2 expression and the MSI2“high” cluster predicted a worse clinical outcome when compared to the other clusters. Overlapping of the differential transcriptional analysis of the Msi2 deleted murine LSCs with our global MSI2 direct mRNA targets (HITS-CLIP) led us to identify that MSI2 binds to transcripts that are associated with the downstream MLL self-renewal program, including Myc and Ikzf2. Ikzf2 is a member of the Ikaros transcription factor family and is known to regulate lymphocyte development by controlling regulatory T-cell function and chromatin remodeling. Ikzf2 shRNA mediated depletion resulted in reduced colony formation, decreased proliferation and increased apoptosis. The MLL associated targets were also reduced, which included Bcl-2 and Hoxa9. In contrast to its tumor suppressor role in hypodiploid B-ALL, these results suggest that Ikzf2 contributes to MLL leukemia cell maintenance. Thus, we provide evidence that MSI2 maintains the oncogenic LSC epigenetic program and the rationale for clinically targeting MSI2 in myeloid leukemia. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
37. Activin-like Kinase 2 (ALK2/ACVR1) Is a Resistance Factor and Therapeutic Vulnerability to FLT3 Inhibition in Acute Myeloid Leukemia
- Author
-
Tyagi, Anudishi, Ly, Stanley, Yuan, Bin, El-Dana, Fouad, Anand, Vivek, Jaggupilli, Appalaraju, Borthakur, Gautam, Foulks, Jason M, Warner, Steven L., and Battula, Venkata Lokesh
- Published
- 2022
- Full Text
- View/download PDF
38. Interim Phase I Clinical Data of FT576 As Monotherapy and in Combination with Daratumumab in Subjects with Relapsed/Refractory Multiple Myeloma
- Author
-
Dhakal, Binod, Berdeja, Jesus G., Gregory, Tara, Ly, Thomas, Bickers, Cara, Zong, Xingyue, Wong, Lilly, Goodridge, Jode P., Cooley, Sarah, Valamehr, Bahram, Chu, Yu-Waye, Byon, John, and Abonour, Rafat
- Published
- 2022
- Full Text
- View/download PDF
39. Interim Phase I Clinical Data of FT819-101, a Study of the First-Ever, Off-the-Shelf, iPSC-Derived TCR-Less CD19 CAR T-Cell Therapy for Patients with Relapsed/Refractory B-Cell Malignancies
- Author
-
Mehta, Amitkumar, Farooq, Umar, Chen, Andy, McGuirk, Joseph P., Ly, Thomas, Wong, Lilly, Cooley, Sarah, Valamehr, Bahram, Elstrom, Rebecca, Chu, Yu-Waye, and Park, Jae H.
- Published
- 2022
- Full Text
- View/download PDF
40. A First-in-Human Phase 1, Multicenter, Open-Label Study of CB-010, a Next-Generation CRISPR-Edited Allogeneic Anti-CD19 CAR-T Cell Therapy with a PD-1 Knockout, in Patients with Relapsed/Refractory B Cell Non-Hodgkin Lymphoma (ANTLER Study)
- Author
-
O'Brien, Susan, Nastoupil, Loretta J., Essell, James, Dsouza, Ly, Hart, Doug, Matsuda, Emiri, Satterfield, Tarah, Nesheiwat, Tonia, Hammad, Ashley, Davi, Franco, Chung, Shally, Shamsi, S. Asim, Bryan, Mara, Skoble, Justin, Garner, Elizabeth, Kanner, Steven, Rizvi, Syed, and Holmes, Houston
- Published
- 2022
- Full Text
- View/download PDF
41. A Phase I Study of FT538, an Off-the-Shelf, Multiplexed-Engineered, iPSC-Derived NK Cell Therapy in Combination with Daratumumab in Relapsed/Refractory Multiple Myeloma
- Author
-
Juckett, Mark, Mailankody, Sham, Eghtedar, Alireza, Bickers, Cara, Zong, Xingyue, Wong, Lilly, Ly, Thomas, Byon, John, Cooley, Sarah, Valamehr, Bahram, Chu, Yu-Waye, and Vij, Ravi
- Published
- 2022
- Full Text
- View/download PDF
42. Structural Variation Cooperates with Permissive Chromatin to Control Enhancer Hijacking-Mediated Oncogenic Transcription
- Author
-
Botten, Giovanni A., Zhang, Yuannyu, Dudnyk, Kseniia, Liu, Xin, Sanders, Jacob T., Imanci, Aygun, Droin, Nathalie, Cao, Hui, Kaphle, Pranita, Dickerson, Kathryn E., Chen, Mingyi, Chen, Weina, Solary, Eric, Ly, Peter, Zhou, Jian, and Xu, Jian
- Published
- 2022
- Full Text
- View/download PDF
43. L3MBTL1: A Polycomb Protein At the Node of Crosstalk Between the BMP4 and Hippo Signaling Pathways in Erythropoiesis.
- Author
-
Perna, Fabiana, primary, Vu, Ly P., additional, Themeli, Maria, additional, Hoya-Arias, Ruben, additional, Liu, Fan, additional, Wang, Lan, additional, Gurvich, Nadia, additional, Hricik, Todd, additional, Papapetrou, Eirini P., additional, Moore, Malcolm A.S. AS, additional, Levine, Ross L., additional, Sadelain, Michel, additional, and Nimer, Stephen, additional
- Published
- 2012
- Full Text
- View/download PDF
44. Degree of anemia, indirect markers of hemolysis, and vascular complications of sickle cell disease in Africa
- Author
-
Dubert, Marie, Elion, Jacques, Tolo, Aissata, Diallo, Dapa Aly, Diop, Saliou, Diagne, Ibrahima, Sanogo, Ibrahima, Belinga, Suzanne, Guifo, Odette, Wamba, Guillaume, Ngo Sack, Françoise, Boidy, Kouakou, Kamara, Ismael, Traore, Youssouf, Diakite, Cheick Oumar, Gbonon, Valérie, Faye, Blaise Felix, Seck, Moussa, Deme Ly, Indou, Chelo, David, N'Guetta, Roland, Diop, Ibrahima Bara, Gaye, Bamba, Jouven, Xavier, and Ranque, Brigitte
- Abstract
The hyperhemolysis paradigm that describes overlapping “hyperhemolytic-endothelial dysfunction” and “high hemoglobin-hyperviscous” subphenotypes of sickle cell disease (SCD) patients is based on North American studies. We performed a transversal study nested in the CADRE cohort to analyze the association between steady-state hemolysis and vascular complications of SCD among sub-Saharan African patients. In Mali, Cameroon, and Ivory Coast, 2407 SCD patients (1751 SS or sickle β-zero-thalassemia [Sβ0], 495 SC, and 161 sickle β+-thalassemia [Sβ+]), aged 3 years old and over, were included at steady state. Relative hemolytic intensity was estimated from a composite index derived from principal component analysis, which included bilirubin levels or clinical icterus, and lactate dehydrogenase levels. We assessed vascular complications (elevated tricuspid regurgitant jet velocity [TRV], microalbuminuria, leg ulcers, priapism, stroke, and osteonecrosis) by clinical examination, laboratory tests, and echocardiography. After adjustment for age, sex, country, and SCD phenotype, a low hemoglobin level was significantly associated with TRV and microalbuminuria in the whole population and with leg ulcers in SS-Sβ0adults. A high hemolysis index was associated with microalbuminuria in the whole population and with elevated TRV, microalbuminuria, and leg ulcers in SS-Sβ0adults, but these associations were no longer significant after adjustment for hemoglobin level. In conclusion, severe anemia at steady state in SCD patients living in West and Central Africa is associated with elevated TRV, microalbuminuria, and leg ulcers, but these vascular complications are not independently associated with indirect markers of increased hemolysis. Other mechanisms leading to anemia, including malnutrition and infectious diseases, may also play a role in the development of SCD vasculopathy.
- Published
- 2017
- Full Text
- View/download PDF
45. Methylation of RUNX1 by the Nuclear PRMT5/COPR5 Complex Regulates Its Cellular Location in Leukemia Cells,
- Author
-
Zhao, Xinyang, primary, Vu, Ly P., additional, Perna, Fabiana, additional, Liu, Fan, additional, Xu, Hao, additional, and Nimer, Stephen, additional
- Published
- 2011
- Full Text
- View/download PDF
46. Regulation of AML1/RUNX1 Function by Protein Arginine Methyltransferase 4 (PRMT4) in Myeloid Differentiation
- Author
-
Vu, Ly P., primary, Zhao, Xinyang, additional, Perna, Fabiana, additional, and Nimer, Stephen D., additional
- Published
- 2011
- Full Text
- View/download PDF
47. Targeting a Novel Epigenetic Silencing Mechanism to Efficiently Upregulate Fetal Globin Gene Expression
- Author
-
Perna, Fabiana, primary, Vu, Ly P., additional, Themeli, Maria, additional, Hoya-Arias, Ruben, additional, Zhao, Xinyang, additional, Papapetrou, Eirini P., additional, Sadelain, Michel, additional, and Nimer, Stephen D., additional
- Published
- 2011
- Full Text
- View/download PDF
48. L3MBTL1: A Polycomb Protein At the Node of Crosstalk Between the BMP4 and Hippo Signaling Pathways in Erythropoiesis
- Author
-
Todd Hricik, Eirini P. Papapetrou, Nadia Gurvich, Malcolm A.S. Moore, Ruben Hoya-Arias, Ly P. Vu, Michel Sadelain, Fabiana Perna, Ross L. Levine, Maria Themeli, Stephen D. Nimer, Lan Wang, and Fan Liu
- Subjects
Hippo signaling pathway ,Cellular differentiation ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Haematopoiesis ,Hippo signaling ,Erythropoiesis ,Signal transduction ,Progenitor cell ,Stem cell - Abstract
Abstract 2296 The Hippo signaling pathway, first discovered in Drosophila, is emerging as an important regulator of stem cell behavior. Upon still-unclear upstream stimuli, the hippo pathway kinase cascade phosphorylates and inhibits the function of YAP, a transcription coactivator, by inducing its cytoplasmic retention. While recent evidences indicate that inhibition of YAP affects cell fate decisions, and proliferation, in many tissues, little is known about the relevance of this pathway in hematopoiesis. However, the interaction of YAP with Smad1, identified in flies and human cells (Alarcon C. et al. Cell 2009), prevents smurf-mediated Smad1 degradation, potentially enhancing BMP signaling. Our ongoing studies have indentified crosstalk between the BMP4 and the Hippo pathways in hematopoietic cells, and in induced-pluripotent stem (iPS) cells that we differentiated towards the erythroid lineage. This crosstalk involves the chromatin-binding, Polycomb protein L3MBTL1, which clearly regulate the effects of BMP on the erythroid differentiation of hematopoietic stem/progenitor cells and on fetal globin gene expression. We find that the Lats2 kinase, a core component of the Hippo pathway, physically interacts with L3MBTL1 and that treatment with BMP4 or Erythropoietin decreases the expression of both proteins in various hematopoietic cells, including primary human cord blood-derived CD34+ cells. By altering L3MBTL1 levels in K562 cells, we were able to show that the L3MBTL1-Lats2 interaction enhances Lats-mediated phosphorylation and the cytoplasmic retention of YAP. Furthermore, L3MBTL1-depleted iPS cells have an enhanced smad-mediated transcriptional response; by analyzing the gene expression profile of these cells, we found increased expression of several BMP target genes (such as HHEX and ID genes), suggesting that L3MBTL1 negatively titrates the BMP4 signaling pathway at least in part by affecting YAP phosphorylation and localization. Gene Set Enrichment Analysis confirmed enrichment of many smad-related genes, and yet, these cells presented enhanced smad1/5/8 phosphorylation by WB analysis, indicating that BMP4 signaling is triggered by L3MBTL1 depletion. We also found that hematopoietic differentiation of L3MBTL1-KD iPS cells generates high-fetal globin gene expressing erythroid progeny, suggesting a role for the BMP4 signaling pathway and the targeting of L3MBTL1 in the treatment of hemoglobinopathies. To further evaluate the effect of BMP4 signaling on hematopoietic cells that lack L3MBTL1, we analyzed the stress erythroid response of L3MBTL1 KO mice: while no difference was observed at baseline in the null mice compared to wt littermates, the L3mbtl1 null mice had a more severe anemia, with increased leukocytosis, and thrombocytosis post-hydrazine (PHZ) or Epo. We found a significant increase in the colony-forming ability of the l3mbtl1 null spleen and bone marrow cells, compared to controls, as well as increased spleen size and an expansion of the spleen erythroid compartment. Thus, l3mbtl1 null hematopoietic stem cells are more sensitive to the PHZ-mediated cytokine storm, which includes BMP4. Interestingly, the L3mbtl1 null BM and spleen cells showed diminished expression of Lats2 and phospho-YAP, consistent with our in vitro findings. In conclusion, these investigations have shown that L3MBTL1 not only negatively titrates the BMP4 signaling pathway, but also provides a nodal point for crosstalk between the BMP4 and Hippo signaling pathways in erythropoiesis. Thus, these data provide insights into possible novel treatments for genetic red cell disorders (such as β-thalassemia) and for acquired bone marrow failure syndromes such as Epo-resistant anemia. Disclosures: Levine: Agios Pharmaceuticals: Research Funding.
- Published
- 2012
- Full Text
- View/download PDF
49. Downregulation of Protein Arginine Methyltransferase – 4 (PRMT4) Promotes MiR-223 Expression and Myeloid Differentiation.
- Author
-
Vu, Ly P, primary, Zhao, Xinyang, additional, Perna, Fabiana, additional, and Nimer, Stephen D, additional
- Published
- 2010
- Full Text
- View/download PDF
50. Targeting a Novel Epigenetic Silencing Mechanism to Efficiently Upregulate Fetal Globin Gene Expression
- Author
-
Eirini P. Papapetrou, Maria Themeli, Ly P. Vu, Michel Sadelain, Xinyang Zhao, Fabiana Perna, Ruben Hoya-Arias, and Stephen D. Nimer
- Subjects
Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Embryonic stem cell ,Chromatin ,Cell biology ,Haematopoiesis ,Erythropoiesis ,Stem cell ,Induced pluripotent stem cell ,Chromatin immunoprecipitation ,Locus control region - Abstract
Abstract 352 L3MBTL1 is a Polycomb group protein, commonly deleted in patients with myeloid disorders associated with the 20q- chromosomal abnormality. After crystallizing the MBT repeat domain, we demonstrated that L3MBTL1 compacts chromatin by binding mono- and di-methylated lysine residues in histones H1 (H1K26) and H4 (H4K20), ultimately leading to gene repression. Despite its role in affecting the chromatin structure, the role of L3MBTL1 in hematopoiesis has remained largely unknown. We recently demonstrated that lack of L3MBTL1 accelerates the erythroid differentiation of human hematopoietic stem cells and here we reveal that L3MBTL1 represses the expression of the fetal gamma globin gene. We lentivirally expressed shRNAs targeting L3MBTL1 in human cord blood (CB) CD34+ cells and in K562 erythroleukemia cells, and consistently observed upregulation of gamma globin gene expression, while beta globin gene expression decreased. Remarkably, we observed similar findings in human embryonic stem (hES) cells, where knock-down of L3MBTL1 triggered a BMP4-like spontaneous differentiation. Given the potential impact of therapeutically increasing fetal hemoglobin expression in patients with hemoglobinopathies, we targeted L3MBTL1 in induced pluripotent stem (iPS) cells derived from patients with β-thalassemia. The gene expression profile of L3MBTL1-KD normal and thalassemic iPS cells indicated clear activation of fetal hemoglobin (HbF) expression, activation of BMP4 signaling and upregulation of specific smad5 target genes (e.g. EKLF, HHEX, ID2/3). We generated and utilized a model of “stress erythropoiesis” in L3MBTL1 KO mice and observed in vivo BMP4-mediated expansion of spleen immature erythroid progenitors, as indicated by increased spleen weight and splenic BFU-E colonies in KO mice compared to controls. We also examined K562 cells, human CB CD34+ cells and hES cells, using chromatin immunoprecipitation assays, and found that L3MBTL1 directly associates with the human β-globin locus, occupying discrete regions within the human β-globin cluster. Furthermore, L3MBTL1 colocalized with H4K20me within the Locus Control Region (LCR), a primary attachment site for chromatin modifiers. We observed clearance of L3MBTL1 and its associated histone marks (H4K20me1/2) from the LCR upon treatment with hemin, erythropoietin or TGFβ, three agents that potently induce erythroid differentiation. This suggests that this polycomb repressor complex responds to cytokine signaling. In summary, we have identified a novel epigenetic regulatory mechanism to control fetal globin gene expression; the Polycomb protein L3MBTL1 regulates BMP4 signaling and the chromatin structure of globin genes. Targeting this regulatory system represents a means to efficiently increase HbF in a human model of β-thalassemia (i.e. with the use of patient-derived iPS cells) and to potentially ameliorate hematological and clinical symptoms of patients with red cell disorders. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.