scholarly journals The Tumor Suppressor Roles of MYBBP1A, a Major Contributor to Metabolism Plasticity and Stemness

Cancers ◽  
2020 ◽  
Vol 12 (1) ◽  
pp. 254 ◽  
Author(s):  
Blanca Felipe-Abrio ◽  
Amancio Carnero

The MYB binding protein 1A (MYBBP1A, also known as p160) acts as a co-repressor of multiple transcription factors involved in many physiological processes. Therefore, MYBBP1A acts as a tumor suppressor in multiple aspects related to cell physiology, most of them very relevant for tumorigenesis. We explored the different roles of MYBBP1A in different aspects of cancer, such as mitosis, cellular senescence, epigenetic regulation, cell cycle, metabolism plasticity and stemness. We especially reviewed the relationships between MYBBP1A, the inhibitory role it plays by binding and inactivating c-MYB and its regulation of PGC-1α, leading to an increase in the stemness and the tumor stem cell population. In addition, MYBBP1A causes the activation of PGC-1α directly and indirectly through c-MYB, inducing the metabolic change from glycolysis to oxidative phosphorylation (OXPHOS). Therefore, the combination of these two effects caused by the decreased expression of MYBBP1A provides a selective advantage to tumor cells. Interestingly, this only occurs in cells lacking pVHL. Finally, the loss of MYBBP1A occurs in 8%–9% of renal tumors. tumors, and this subpopulation could be studied as a possible target of therapies using inhibitors of mitochondrial respiration.

BMC Cancer ◽  
2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Dominique M. O. Higgins ◽  
Maisel Caliva ◽  
Mark Schroeder ◽  
Brett Carlson ◽  
Pavan S. Upadhyayula ◽  
...  

Abstract Background Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults, with a median survival of approximately 15 months. Semaphorin 3A (Sema3A), known for its axon guidance and antiangiogenic properties, has been implicated in GBM growth. We hypothesized that Sema3A directly inhibits brain tumor stem cell (BTSC) proliferation and drives invasion via Neuropilin 1 (Nrp1) and Plexin A1 (PlxnA1) receptors. Methods GBM BTSC cell lines were assayed by immunostaining and PCR for levels of Semaphorin 3A (Sema3A) and its receptors Nrp1 and PlxnA1. Quantitative BrdU, cell cycle and propidium iodide labeling assays were performed following exogenous Sema3A treatment. Quantitative functional 2-D and 3-D invasion assays along with shRNA lentiviral knockdown of Nrp1 and PlxnA1 are also shown. In vivo flank studies comparing tumor growth of knockdown versus control BTSCs were performed. Statistics were performed using GraphPad Prism v7. Results Immunostaining and PCR analysis revealed that BTSCs highly express Sema3A and its receptors Nrp1 and PlxnA1, with expression of Nrp1 in the CD133 positive BTSCs, and absence in differentiated tumor cells. Treatment with exogenous Sema3A in quantitative BrdU, cell cycle, and propidium iodide labeling assays demonstrated that Sema3A significantly inhibited BTSC proliferation without inducing cell death. Quantitative functional 2-D and 3-D invasion assays showed that treatment with Sema3A resulted in increased invasion. Using shRNA lentiviruses, knockdown of either NRP1 or PlxnA1 receptors abrogated Sema3A antiproliferative and pro-invasive effects. Interestingly, loss of the receptors mimicked Sema3A effects, inhibiting BTSC proliferation and driving invasion. Furthermore, in vivo studies comparing tumor growth of knockdown and control infected BTSCs implanted into the flanks of nude mice confirmed the decrease in proliferation with receptor KD. Conclusions These findings demonstrate the importance of Sema3A signaling in GBM BTSC proliferation and invasion, and its potential as a therapeutic target.


2015 ◽  
Author(s):  
david miguez

The understanding of the regulatory processes that orchestrate stem cell maintenance is a cornerstone in developmental biology. Here, we present a mathematical model based on a branching process formalism that predicts average rates of proliferative and differentiative divisions in a given stem cell population. In the context of vertebrate spinal neurogenesis, the model predicts complex non-monotonic variations in the rates of pp, pd and dd modes of division as well as in cell cycle length, in agreement with experimental results. Moreover, the model shows that the differentiation probability follows a binomial distribution, allowing us to develop equations to predict the rates of each mode of division. A phenomenological simulation of the developing spinal cord informed with the average cell cycle length and division rates predicted by the mathematical model reproduces the correct dynamics of proliferation and differentiation in terms of average numbers of progenitors and differentiated cells. Overall, the present mathematical framework represents a powerful tool to unveil the changes in the rate and mode of division of a given stem cell pool by simply quantifying numbers of cells at different times.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1805-1805
Author(s):  
Ying Liang ◽  
Gary Van Zant

Abstract We recently found that latexin is a negative regulator of the size of the hematopoietic stem cell population in mice. It acts by increasing apoptosis and decreasing cell proliferation. This 29 kD protein bears a strong structural resemblance to tazarotene-induced gene 1 (TIG1), a tumor suppressor down-regulated in a variety of cancers. The structural similarity and close genetic linkage led us to hypothesize that latexin also may have tumor suppressor properties. We found that latexin was down-regulated in a variety of human leukemias and lymphomas as determined by a survey of malignant cell lines and by analysis of CD34+ cells isolated from the blood and marrow of patients diagnosed with these malignancies and presenting with very high white cell counts. Bisulfite sequencing revealed that methylation of CpG dinucleotides in the latexin promoter at least partially accounted for latexin down-regulation. 5-aza-deoxycytidine treatment reinitiated or significantly increased latexin expression in K562, Molt4, CRF-CEM, Jurkat, U937, HL60, KG-1, and Sup B15 cell lines. To test the hypothesis that ectopic latexin expression in tumor cells would inhibit their growth, we developed a retrovirus-based expression vector with which we infected the murine lymphoma cell lines, WEHI231 and A20, neither of which contained significant latexin levels by Western blot. A vector containing GFP, but not latexin, was used to infect control cells. In triplicate experiments, the growth of both cell lines in vitro was inhibited an average 48% by infection with the latexin vector. Western blots revealed that latexin was durably expressed throughout the 2-week culture period at 2- to 4-fold the level expressed in normal T and B cells. As we found in our normal stem cell studies, latexin caused growth inhibition of lymphoma cells by significantly increasing apoptosis by 6-fold, and by suppressing cell proliferation by 2-fold. To test whether tumor inhibition extended to lymphomas in vivo, we injected either control or latexin vector-infected A20 cells subcutaneously in the flanks of BALB/c mice. Three weeks following adoptive transfer of identical numbers of cells, in duplicate experiments the latexin-expressing cells developed tumors only half the volume of those caused by the control cells. These results are consistent with a tumor suppressor role for latexin and suggest that latexin, or molecular mimics thereof, may have clinical efficacy in the treatment of malignancies.


2021 ◽  
Vol 11 ◽  
Author(s):  
Jiaxin Liang ◽  
Linda Oyang ◽  
Shan Rao ◽  
Yaqian Han ◽  
Xia Luo ◽  
...  

RAS-related C3 botulinum toxin substrate 1 (Rac.1) is one of the important members of Rho GTPases. It is well known that Rac1 is a cytoskeleton regulation protein that regulates cell adhesion, morphology, and movement. Rac1 is highly expressed in different types of tumors, which is related to poor prognosis. Studies have shown that Rac1 not only participates in the tumor cell cycle, apoptosis, proliferation, invasion, migration and angiogenesis, but also participates in the regulation of tumor stem cell, thus promoting the occurrence of tumors. Rac1 also plays a key role in anti-tumor therapy and participates in immune escape mediated by the tumor microenvironment. In addition, the good prospects of Rac1 inhibitors in cancer prevention and treatment are exciting. Therefore, Rac1 is considered as a potential target for the prevention and treatment of cancer. The necessity and importance of Rac1 are obvious, but it still needs further study.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1244-1244
Author(s):  
Kathrin Krowiorz ◽  
Razan Jammal ◽  
Stephan Emmrich ◽  
Arefeh Rouhi ◽  
Michael Heuser ◽  
...  

Abstract MicroRNAs (miRNAs) are essential for maintenance and differentiation of normal hematopoietic cells and their dysregulation is strongly implicated in leukemias. In order to identify tumor suppressor miRNAs in the context of hematological malignancies, we performed two complementary miRNA expression screenings in normal hematopoiesis as well as in childhood and adult acute myeloid leukemias (AML). We reasoned that tumor suppressor miRNAs are upregulated in mature myeloid cells, as compared to normal hematopoietic stem and progenitor cells (HSPCs), and downregulated in AML. Based on this screening strategy, we identified the miR-193 family members as potent suppressors of HSPC activity and AML growth. During normal hematopoiesis mmu-miR-193a-3p is exclusively expressed in mature myeloid cells and absent in normal HSPCs. Accordingly, in a cohort of 165 pediatric AML patients hsa-miR-193b-3p was broadly repressed throughout the cytogenetically characterized subgroups. In addition, in a cohort of 43 adult AML patients, its homolog hsa-miR-193a-3p was significantly upregulated in APL cases (p=0.0025, n=7) compared to bone marrow from healthy donors (n=5). To assess the impact of the miR-193 family members on AML maintenance and development, we lentivirally expressed miR-193a/b in the MLL-rearranged cell lines ML2 and THP1, which induced monocytic differentiation in concert with calcitriol treatment, measured by CD11b/CD14 expression (p=0.024). Consistently, enforced miR-193-expression led to a significant growth disadvantage in ML2 and THP1 cells (p=<0.001 and p=0.02, respectively) as well as to reduced colony formation (p=0.008) in methylcellulose-based colony-forming unit (CFU) assays. Noteworthy, these effects were not restricted to MLL-rearranged AML cell lines only, but were also evident in six other AML cell lines representing the most common AML subgroups, such as t(8;21) and t(15;17). Beyond the growth-suppressive and differentiation-inductive effect of miR-193 in human AML cell lines, overexpression of miR-193a caused a significant decrease of proliferation in murine bone marrow cells immortalized in vitro by retroviral expression of Hoxa9 or Hoxa9 and Meis1 (p=0.019 and p=0.008, respectively). Based on these findings in AML, we further investigated the impact of the miR-193 family on normal hematopoiesis. We retrovirally expressed miR-193a in 32D cells treated with granulocyte-colony stimulating factor (G-CSF), which resulted in a strong induction of myeloid differentiation already after day 2 (p=0.006) as assessed by CD11b/Gr-1 surface marker expression. We lentivirally transduced mouse lineage negative (Lin-) HSPCs and transplanted them into irradiated isogenic recipients. Bleedings performed on weeks 4, 8 and 11, as well as the examination of the bone marrow on week 11, showed a severe competitive disadvantage of miR-193-transduced cells (week 11: 2% GFP+ miR-193- vs. 25% GFP+ miR-NSC-transduced cells). These results were further refined using highly purified ESLAM (CD45+ EPCR+ CD48− CD150+) HSCs which failed to reconstitute hematopoiesis when overexpressing miR-193a, indicated by the absence of miR-193a/GFP+ cells at week 8 post transplantation. These observations might be explained by a potent G1 cell cycle arrest in HSPCs when overexpressing miR-193a/b (4-fold decrease in the S phase population) and induction of apoptosis. Our results in normal and malignant hematopoiesis suggested that the miR-193 family acts globally through targeting relevant stem cell pathways. To validate this hypothesis we quantified the knockdown of ten predicted miR-193 target genes. qRT-PCR analysis confirmed the down regulation of KIT, KRAS, SOS2 (key components of the MAPK signaling pathway) and CCND1, a CDK regulator of G1/S phase transition. We propose a dual regulatory platform where firstly, miR-193 targets CCND1 and controls the cell cycle kinetics of stem cells. Secondly, miR-193 interferes with the KIT proto-oncogene and the RAS pathway thereby inhibiting crucial pro-proliferation and anti-apoptotic signaling cascades. Taken together, we identified the miR-193 family as a pan-tumor suppressor in childhood and adult AML. Its anti-leukemic effect is mediated by targeting the stem cell KIT/SOS2/RAS/RAF axis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1997 ◽  
Vol 89 (10) ◽  
pp. 3596-3606 ◽  
Author(s):  
Troy D. Randall ◽  
Irving L. Weissman

Abstract A significant fraction of hematopoietic stem cells (HSCs) have been shown to be resistant to the effects of cytotoxic agents such as 5-fluorouracil (5-FU), which is thought to eliminate many of the rapidly dividing, more committed progenitors in the bone marrow and to provide a relatively enriched population of the most primitive hematopoietic progenitor cells. Although differences between 5-FU–enriched progenitor populations and those from normal bone marrow have been described, it remained unclear if these differences reflected characteristics of the most primitive stem cells that were revealed by 5-FU, or if there were changes in the stem-cell population itself. Here, we have examined some of the properties of the stem cells in the bone marrow before and after 5-FU treatment and have defined several activation-related changes in the stem-cell population. We found that long-term reconstituting stem cells decrease their expression of the growth factor receptor c-kit by 10-fold and increase their expression of the integrin Mac-1 (CD11b). These changes begin as early as 24 hours after 5-FU treatment and are most pronounced within 2 to 3 days. This activated phenotype of HSCs isolated from 5-FU–treated mice is similar to the phenotype of stem cells found in the fetal liver and to the phenotype of transiently repopulating progenitors in normal bone marrow. We found that cell cycle is induced concomitantly with these physical changes, and within 2 days as many as 29% of the stem-cell population is in the S/G2/M phases of the cell cycle. Furthermore, when examined at a clonal level, we found that 5-FU did not appear to eliminate many of the transient, multipotent progenitors from the bone marrow that were found to be copurified with long-term repopulating, activated stem cells. These results demonstrate the sensitivity of the hematopoietic system to changes in its homeostasis and correlate the expression of several important surface molecules with the activation state of HSCs.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 700-700
Author(s):  
Michihiro Kobayashi ◽  
Edward F. Srour

Abstract Abstract 700 Cell cycle status of hematopoietic stem cells (HSC) is tightly regulated most likely to balance quiescence and proliferation/differentiation of these cells which fulfil the immediate and continuous demands for maintenance of the size of the stem cell pool as well as the production of all the formed elements of the blood, respectively. Tumor suppressor genes, which negatively influence cell cycle regulation and control cell survival have different important roles in HSC function. Recently, Dmtf1 (Cyclin D-binding Myb-like Transcription Factor 1) was recognized as a tumor suppressor gene that is deleted in lung cancer and leukemic cells. Dmtf1 has been shown to regulate cell cycle progression by mainly an Arf-Mdm2-p53 dependent mechanism. Since the role of Dmtf1 in the hematopoietic system has not been explored, we investigated weather Dmtf1 plays a role in regulating HSC quiescence by analyzing Dmtf1 KO mice. Dmtf1 was expressed in many stages and lineages of hematopoietic cells, including Kit+Sca-1+Lineage- (KSL) cells, Kit+/lin- cells, T-cells, B-cells, and granulocytes. As previously reported, KO mice were smaller than WT mice (25.1 vs 28.0 g at 13-14 weeks old male, p<0.01), however, bone marrow (BM) of KO mice contained a higher number of total nucleated cells than that of WT mice (6.8 vs 5.1 × 10e7 in 2 femurs, p<0.01). Population of myeloid and T/B-cell were similar in both genotype. Absolute KSL count in KO BM was more than 2-fold higher than that of WT marrow (6.1 vs 2.2 × 10e4/femur, p<0.03). Although the frequency of ST-HSC (CD34+CD135+ KSL) was higher in KO mice (0.17 vs 0.09%, p<0.05), the frequency of LT-HSC (CD34-CD135- KSL) was similar in both genotypes (0.03 vs 0.03%). Sorted KSL from KO mice contained higher numbers of clonogenic cells (62.3 vs 32.3 CFU/1000KSL, p<0.01) and exhibited a higher proliferative potential in liquid culture. Competitively transplanted low-density BM cells from KO mice sustained a higher level of chimerism in recipient mice than their WT counterparts at 16 weeks post-transplantation (83 vs 48%, p<0.05). To evaluate differences in the repopulation potential of LT-HSC, we transplanted 100 sorted CD34-CD135- KSL (CD45.2) with 5×10e5 competitor BM low-density cells (CD45.1). KO LT-HSC supported markedly higher chimerism than WT cells at 16 weeks (45.6 vs 3.8%, p<0.01). Results of secondary transplantation are pending. Because it has been shown that Dmtf1 is induced by oncogenic or proliferative RAS/ERK signals as an initiation of negative feed back regulation, we investigated the relationship between Dmtf1 expression and cell cycle status of KSL cells in the early phases of proliferation. At steady state (0 hr), sorted KO-KSL showed a higher percentage of cycling cells (S+G2/M) by Hst/Pyronin staining (16.8 vs 11.2%) and the same percentage of G0 cells (51.2 vs 52.6%). Percentage of cycling KO- & WT-KSL cells after 24hr in culture was 37.8% and 24.2%, respectively, and at 48hr, the fraction of KO-KSL in G0 was lower than that among WT cells (1.6 vs 7.8%). Apoptosis was not increased among KO BM cells. Accordingly, expression of Dmtf1 was increased at 24 & 48 hr compared to 0 hr by real-time qPCR analysis. Interestingly, a marked suppression of CDKN1a (p21) expression in KO-KSL cells was observed at both 0 and 48hr. Given that Arf expression is induced by Dmtf1 in epithelial cells, we examined the relationship between Arf and Dmtf1 in KSL cells. Arf was not detected at 0hr in WT and KO KSL cells. While cultured KO-KSL cells failed to express Arf after 48 & 72hr in culture, WT cells expressed Arf after 72hr suggesting that induction of Arf may be partially responsible for the dysregulation of cell cycle progression in cultured KO-KSL cells and Arf does not have important role for regulating the cell cycle of steady state HSCs. Taken together, our data suggest that loss of Dmtf1 make LT-HSC acquire a higher long-term repopulating potential compared to WT cells and that in the hematopoietic system that might be involved in development of leukemic stem cell. Dmtf1 regulates HSC quiescence by the induction of CDKN1a via an Arf independent mechanism. Disclosures: No relevant conflicts of interest to declare.


Oncogene ◽  
2015 ◽  
Vol 35 (21) ◽  
pp. 2777-2788 ◽  
Author(s):  
I Ferrer ◽  
E M Verdugo-Sivianes ◽  
M A Castilla ◽  
R Melendez ◽  
J J Marin ◽  
...  

Blood ◽  
1997 ◽  
Vol 89 (10) ◽  
pp. 3596-3606 ◽  
Author(s):  
Troy D. Randall ◽  
Irving L. Weissman

A significant fraction of hematopoietic stem cells (HSCs) have been shown to be resistant to the effects of cytotoxic agents such as 5-fluorouracil (5-FU), which is thought to eliminate many of the rapidly dividing, more committed progenitors in the bone marrow and to provide a relatively enriched population of the most primitive hematopoietic progenitor cells. Although differences between 5-FU–enriched progenitor populations and those from normal bone marrow have been described, it remained unclear if these differences reflected characteristics of the most primitive stem cells that were revealed by 5-FU, or if there were changes in the stem-cell population itself. Here, we have examined some of the properties of the stem cells in the bone marrow before and after 5-FU treatment and have defined several activation-related changes in the stem-cell population. We found that long-term reconstituting stem cells decrease their expression of the growth factor receptor c-kit by 10-fold and increase their expression of the integrin Mac-1 (CD11b). These changes begin as early as 24 hours after 5-FU treatment and are most pronounced within 2 to 3 days. This activated phenotype of HSCs isolated from 5-FU–treated mice is similar to the phenotype of stem cells found in the fetal liver and to the phenotype of transiently repopulating progenitors in normal bone marrow. We found that cell cycle is induced concomitantly with these physical changes, and within 2 days as many as 29% of the stem-cell population is in the S/G2/M phases of the cell cycle. Furthermore, when examined at a clonal level, we found that 5-FU did not appear to eliminate many of the transient, multipotent progenitors from the bone marrow that were found to be copurified with long-term repopulating, activated stem cells. These results demonstrate the sensitivity of the hematopoietic system to changes in its homeostasis and correlate the expression of several important surface molecules with the activation state of HSCs.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4794-4794
Author(s):  
Peter J Quesenberry ◽  
Mark S Dooner ◽  
Laura R Goldberg

Abstract Abstract 4794 We propose that the long-term multipotent marrow renewal stem cell is on a continuum of change in potential tied to cell cycle stage. Furthermore our data indicate that marrow stem cells cannot be defined on a clonal basis and that purification of stem cells, as is currently carried out, leads to a biased population which provides only limited information with regard to marrow stem cells. Previous data have clearly demonstrated continuous and reversible phenotype change when murine marrow stem cells are driven through cell cycle in vitro by cytokines (Quesenberry et al, Exp Hematol, 2010). The relevance of this to normal hematopoiesis is placed into question by numerous observations indicating that highly purified long-term hematopoietic stem cells are in the G0 phase of cell cycle. However, no studies have addressed the cell cycle status of long-term multilineage stem cells in vivo from normal unseparated marrow. Given the fact that marrow stem cell separations are not random and that over 90% of stem cells are lost in these separations (Nilsson et al, Blood, 1997), the possibility exists that the purified stem cells are not representative of stem cells in the whole population. Accordingly, we have investigated the cell cycle status of murine whole marrow stem cells employing Hoechst 33342 alone or pyronin/Hoechst 33342 staining with FACS separation into G0, G1 and S/G2/M fractions, followed by competitive engraftment of each sub fraction into lethally irradiated mice and analysis of multilineage engraftment from 28 to 52 weeks. In five separate experiments we have found that 50 ± 4 % of whole marrow long-term engrafting cells were in S/G2/M. Engraftment in every instance was multilineage as determined by analysis of B220, Gr-1. CD11b and CD3 engraftment. Given that this is an instantaneous look at cell cycle status, it implies that virtually all long-term engrafting cells are in active cell cycle. Similar studies on LT-HSC (lin-Sca+c-kit+ Flk2-) essentially confirmed previous studies (with one stochastic exception) that LT-HSC are a dormant, noncycling population in G0. In order to further confirm these studies, donor whole marrow was subjected to high-specific activity tritiated thymidine suicide for thirty minutes and long-term multilineage engraftment was determined in lethally irradiated mice. In these on-going studies we find that 52 ± 8.6 % of engrafting cells were in S/G2/M out to 6 months (5 experiments). Given the short exposure to tritiated thymidine, these data also indicate that virtually all marrow stem cells are in cell cycle. With progression through cell cycle there is a continuous change in all aspects of cellular phenotype and thus the characteristics of the stem cell will vary from moment to moment. In this setting, purification of individual cells can not define the population. Thus the marrow long-term repopulating stem cell is on a continuum of change in potential which will be realized if interrogated with appropriate stimuli. These potentials continuously change with progression through cell cycle with different fate outcomes at different points in cell cycle. The present challenge for the field is to appropriately define the stem cell population, not the “stem cell”. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document