scholarly journals Sphingosine kinase-1 and sphingosine 1-phosphate receptor 2 mediate Bcr-Abl1 stability and drug resistance by modulation of protein phosphatase 2A

Blood ◽  
2011 ◽  
Vol 117 (22) ◽  
pp. 5941-5952 ◽  
Author(s):  
Arelis Salas ◽  
Suriyan Ponnusamy ◽  
Can E. Senkal ◽  
Marisa Meyers-Needham ◽  
Shanmugam Panneer Selvam ◽  
...  

Abstract The mechanisms by which sphingosine kinase-1 (SK-1)/sphingosine 1-phosphate (S1P) activation contributes to imatinib resistance in chronic myeloid leukemia (CML) are unknown. We show herein that increased SK-1/S1P enhances Bcr-Abl1 protein stability, through inhibition of its proteasomal degradation in imatinib-resistant K562/IMA-3 and LAMA-4/IMA human CML cells. In fact, Bcr-Abl1 stability was enhanced by ectopic SK-1 expression. Conversely, siRNA-mediated SK-1 knockdown in K562/IMA-3 cells, or its genetic loss in SK-1−/− MEFs, significantly reduced Bcr-Abl1 stability. Regulation of Bcr-Abl1 by SK-1/S1P was dependent on S1P receptor 2 (S1P2) signaling, which prevented Bcr-Abl1 dephosphorylation, and degradation via inhibition of PP2A. Molecular or pharmacologic interference with SK-1/S1P2 restored PP2A-dependent Bcr-Abl1 dephosphorylation, and enhanced imatinib- or nilotinib-induced growth inhibition in primary CD34+ mononuclear cells obtained from chronic phase and blast crisis CML patients, K562/IMA-3 or LAMA4/IMA cells, and 32Dcl3 murine progenitor cells, expressing the wild-type or mutant (Y253H or T315I) Bcr-Abl1 in situ. Accordingly, impaired SK-1/S1P2 signaling enhanced the growth-inhibitory effects of nilotinib against 32D/T315I-Bcr-Abl1–derived mouse allografts. Since SK-1/S1P/S1P2 signaling regulates Bcr-Abl1 stability via modulation of PP2A, inhibition of SK-1/S1P2 axis represents a novel approach to target wild-type- or mutant-Bcr–Abl1 thereby overcoming drug resistance.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 571-571
Author(s):  
Lars Klemm ◽  
Cihangir Duy ◽  
Niklas Feldhahn ◽  
John Groffen ◽  
Yong-mi Kim ◽  
...  

Abstract Chronic myeloid leukemia (CML) in chronic phase has a disease-free survival of 87% (DFS; 5 years) and long-term treatment with Imatinib is effective. In a large subgroup of patients with CML, however, the disease ultimately progresses into B lymphoid blast crisis (LBC) with only 6% DFS and resistance to Imatinib develops in virtually all cases. In most cases, acquired resistance to Imatinib can be attributed to somatic mutations within the BCR-ABL1 kinase domain. Whereas BCR-ABL1 kinase mutations are rare in chronic phase CML, such mutations are found in >80% of patients with B cell lineage LBC. Likewise, deletions of the ARF and INK4B genes are rare in chronic phase CML but found in ~50% of B cell lineage LBC. In a search for a B cell lineage-specific mutation mechanism responsible for BCR-ABL1 kinase mutations, we tested the hypothesis that aberrant activation of somatic hypermutation may give rise to drug-resistance and progression of chronic phase CML into LBC. Somatic hypermutation drives affinity maturation of immunoglobulins expressed by germinal center B cells and requires the cytidine deaminase AID. Expression of AID depends on PAX5, a transcription factor that determines B cell lineage commitment of hematopoietic progenitor cells. The dependence of AID expression on PAX5 limits somatic hypermutation to the B cell lineage. Consistent with aberrant activation of somatic hypermutation in B cell lineage LBC, we found both PAX5 and AID expression at the mRNA and protein level in B lymphoid but not myeloid subclones from patient-derived blast crisis CML. However, AID protein levels in LBC clones were 5–10-fold lower than in germinal center B cells. To confirm lineage-specific activation of AID-expression in BCR-ABL1 driven leukemia, we isolated bone marrow from Aid-GFP reporter transgenic mice and transformed the bone marrow cells with BCR-ABL1 under either myeloid (IL3, IL6, SCF) or B lymphoid (IL7) culture conditions. The Aid-GFP reporter drives GFP expression under control of upstream and downstream regulatory elements of the Aid locus (Crouch et al., 2007). BCR-ABL1-induced Aid-expression was only observed under B lymphoid culture conditions and was very heterogeneous among the leukemia cell population: Only about 5–10% of CD19+ B lymphoid leukemia clones express Aid-GFP. In these cells, however, Aid mRNA levels are 240-fold higher than in Aid-GFP-negative cells and even 1.5-fold higher than in normal germinal center B cells. Consistent with these findings, we found aberrant somatic hypermutation of the IGHM, BCL6 and MYC loci as well as evidence of ongoing DNA single-strand breaks at the ARF and INK4B loci in B cell lineage LBC but not myeloid CML clones. Ectopic expression of AID in seven otherwise AID-negative CML cell lines cells leads to the acquisition of Imatinib-resistance and sequence analysis of the Imatinib-resistant clones revealed accumulation of mutations within the BCR-ABL1 kinase domain that cause Imatinib-resistance in patients (e.g. L248V, E225K, T315I). Aberrant expression of AID also caused Imatinib-resistance of CML cells in vivo: NOD/SCID mice were injected with CML cells that were either transduced with AID/GFP or GFP alone. Whereas more than the half of the mice injected with GFP+ CML cells were still alive after 170 days, all mice in the AID/GFP+ CML group died within 54 days after injection despite Imatinib-treatment. Forced expression of the B cell-specific transcription factor PAX5 in otherwise PAX5-negative CML cells resulted in a partial B lymphoid lineage conversion similar to LBC. Of note, ectopic expression of PAX5 also resulted in aberrant AID expression, subsequent acquisition of BCR-ABL1 kinase mutations and development of drug-resistance. We conclude that B cell-specific activation of PAX5/AID-induced aberrant somatic hypermutation provides a genetic basis for the strikingly different outcome of myeloid lineage CML as compared to LBC.


2015 ◽  
Vol 83 (7) ◽  
pp. 2705-2713 ◽  
Author(s):  
Amir M. Farnoud ◽  
Arielle M. Bryan ◽  
Talar Kechichian ◽  
Chiara Luberto ◽  
Maurizio Del Poeta

Cryptococcus neoformansis a fungal pathogen that causes pulmonary infections, which may progress into life-threatening meningitis. In commonly used mouse models ofC. neoformansinfections, fungal cells are not contained in the lungs, resulting in dissemination to the brain. We have previously reported the generation of an engineeredC. neoformansstrain (C. neoformansΔgcs1) which can be contained in lung granulomas in the mouse model and have shown that granuloma formation is dependent upon the enzyme sphingosine kinase 1 (SK1) and its product, sphingosine 1-phosphate (S1P). In this study, we have used four mouse models, CBA/J and C57BL6/J (both immunocompetent), Tgε26 (an isogenic strain of strain CBA/J lacking T and NK cells), and SK−/−(an isogenic strain of strain C57BL6/J lacking SK1), to investigate how the granulomatous response and SK1-S1P pathway are interrelated duringC. neoformansinfections. S1P and monocyte chemotactic protein-1 (MCP-1) levels were significantly elevated in the bronchoalveolar lavage fluid of all mice infected withC. neoformansΔgcs1but not in mice infected with theC. neoformanswild type. SK1−/−mice did not show elevated levels of S1P or MCP-1. Primary neutrophils isolated from SK1−/−mice showed impaired antifungal activity that could be restored by the addition of extracellular S1P. In addition, high levels of tumor necrosis factor alpha were found in the mice infected withC. neoformansΔgcs1in comparison to the levels found in mice infected with theC. neoformanswild type, and their levels were also dependent on the SK1-S1P pathway. Taken together, these results suggest that the SK1-S1P pathway promotes host defense againstC. neoformansinfections by regulating cytokine levels, promoting extracellular killing by phagocytes, and generating a granulomatous response.


Blood ◽  
2008 ◽  
Vol 111 (7) ◽  
pp. 3735-3741 ◽  
Author(s):  
Catherine Roche-Lestienne ◽  
Lauréline Deluche ◽  
Sélim Corm ◽  
Isabelle Tigaud ◽  
Sami Joha ◽  
...  

Abstract Acquired molecular abnormalities (mutations or chromosomal translocations) of the RUNX1 transcription factor gene are frequent in acute myeloblastic leukemias (AMLs) and in therapy-related myelodysplastic syndromes, but rarely in acute lymphoblastic leukemias (ALLs) and chronic myelogenous leukemias (CMLs). Among 18 BCR-ABL+ leukemias presenting acquired trisomy of chromosome 21, we report a high frequency (33%) of recurrent point mutations (4 in myeloid blast crisis [BC] CML and one in chronic phase CML) within the DNA-binding region of RUNX1. We did not found any mutation in de novo BCR-ABL+ ALLs or lymphoid BC CML. Emergence of the RUNX1 mutations was detected at diagnosis or before the acquisition of trisomy 21 during disease progression. In addition, we also report a high frequency of cryptic chromosomal RUNX1 translocation to a novel recently described gene partner, PRDM16 on chromosome 1p36, for 3 (21.4%) of 14 investigated patients: 2 myeloid BC CMLs and, for the first time, 1 therapy-related BCR-ABL+ ALL. Two patients presented both RUNX1 mutations and RUNX1-PRDM16 fusion. These events are associated with a short survival and support the concept of a cooperative effect of BCR-ABL with molecular RUNX1 abnormalities on the differentiation arrest phenotype observed during progression of CML and in BCR-ABL+ ALL.


2004 ◽  
Vol 279 (50) ◽  
pp. 52487-52492 ◽  
Author(s):  
Maria L. Allende ◽  
Teiji Sasaki ◽  
Hiromichi Kawai ◽  
Ana Olivera ◽  
Yide Mi ◽  
...  

Sphingosine-1-phosphate (S1P), a lipid signaling molecule that regulates many cellular functions, is synthesized from sphingosine and ATP by the action of sphingosine kinase. Two such kinases have been identified, SPHK1 and SPHK2. To begin to investigate the physiological functions of sphingosine kinase and S1P signaling, we generated mice deficient in SPHK1.Sphk1null mice were viable, fertile, and without any obvious abnormalities. Total SPHK activity in mostSphk1-/-tissues was substantially, but not completely, reduced indicating the presence of multiple sphingosine kinases. S1P levels in most tissues from theSphk1-/- mice were not markedly decreased. In serum, however, there was a significant decrease in the S1P level. Although S1P signaling regulates lymphocyte trafficking, lymphocyte distribution was unaffected in lymphoid organs ofSphk1-/- mice. The immunosuppressant FTY720 was phosphorylated and elicited lymphopenia in theSphk1null mice showing that SPHK1 is not required for the functional activation of this sphingosine analogue prodrug. The results with theseSphk1null mice reveal that some key physiologic processes that require S1P receptor signaling, such as vascular development and proper lymphocyte distribution, can occur in the absence of SPHK1.


Blood ◽  
2003 ◽  
Vol 101 (11) ◽  
pp. 4611-4614 ◽  
Author(s):  
Amie S. Corbin ◽  
Paul La Rosée ◽  
Eric P. Stoffregen ◽  
Brian J. Druker ◽  
Michael W. Deininger

Abstract Imatinib mesylate is a selective Bcr-Abl kinase inhibitor, effective in the treatment of chronic myelogenous leukemia. Most patients in chronic phase maintain durable responses; however, many in blast crisis fail to respond, or relapse quickly. Kinase domain mutations are the most commonly identified mechanism associated with relapse. Many of these mutations decrease the sensitivity of the Abl kinase to imatinib, thus accounting for resistance to imatinib. The role of other mutations in the emergence of resistance has not been established. Using biochemical and cellular assays, we analyzed the sensitivity of several mutants (Met244Val, Phe311Leu, Phe317Leu, Glu355Gly, Phe359Val, Val379Ile, Leu387Met, and His396Pro/Arg) to imatinib mesylate to better understand their role in mediating resistance.While some Abl mutations lead to imatinib resistance, many others are significantly, and some fully, inhibited. This study highlights the need for biochemical and biologic characterization, before a resistant phenotype can be ascribed to a mutant.


Author(s):  
Jiujiang Liao ◽  
Yangxi Zheng ◽  
Mingyu Hu ◽  
Ping Xu ◽  
Li Lin ◽  
...  

Incomplete spiral artery remodeling, caused by impaired extravillous trophoblast invasion, is a fundamental pathogenic process associated with malplacentation and the development of preeclampsia. Nevertheless, the mechanisms controlling this regulation of trophoblast invasion are largely unknown. We report that sphingosine-1-phosphate synthesis and expression is abundant in healthy trophoblast, whereas in pregnancies complicated by preeclampsia the placentae are associated with reduced sphingosine-1-phosphate and lower SPHK1 (sphingosine kinase 1) expression and activity. In vivo inhibition of sphingosine kinase 1 activity during placentation in pregnant mice led to decreased placental sphingosine-1-phosphate production and defective placentation, resulting in a preeclampsia phenotype. Moreover, sphingosine-1-phosphate increased HTR8/SVneo (immortalized trophoblast cells) cell invasion in a Hippo-signaling–dependent transcriptional coactivator YAP (Yes-associated protein) dependent manner, which is activated by S1PR2 (sphingosine-1-phosphate receptor-2) and downstream RhoA/ROCK induced actin polymerization. Mutation-based YAP-5SA demonstrated that sphingosine-1-phosphate activation of YAP could be either dependent or independent of Hippo signaling. Together, these findings suggest a novel pathogenic pathway of preeclampsia via disrupted sphingosine-1-phosphate metabolism and signaling-induced, interrupted actin dynamics and YAP deactivation; this may lead to potential novel intervention targets for the prevention and management of preeclampsia.


2020 ◽  
Vol 2020 ◽  
pp. 1-7 ◽  
Author(s):  
Parnpen Viriyavejakul ◽  
Chuchard Punsawad

Pulmonary edema (PE) is a major cause of pulmonary manifestations of severe Plasmodium falciparum malaria and is usually associated with acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). The sphingosine kinase-1 (SphK-1)/sphingosine-1-phosphate receptor-3 (S1PR-3) pathway has recently been reported to affect the pathogenesis of lung injury, but the expression of these proteins in the lungs of severe P. falciparum malaria patients has not been investigated. The cellular expression of SphK-1 and S1PR-3 in lung tissues from autopsied patients with P. falciparum malaria was investigated using immunohistochemistry (IHC). Lung tissues from patients who died of severe P. falciparum malaria were classified into two groups based on histopathological findings: those with PE (18 patients) and those without PE (non-PE, 19 patients). Ten samples of normal lung tissues were used as the control group. The protein expression levels of SphK-1 and S1PR-3 were significantly upregulated in endothelial cells (ECs), alveolar epithelial cells, and alveolar macrophages (AMs) in the lungs of severe P. falciparum malaria patients with PE compared to those in the non-PE and control groups (all p<0.001). In addition, the SphK-1 and S1PR-3 expression levels were significantly positively correlated in pulmonary ECs (rs=0.922, p<0.001), alveolar epithelial cells (rs=0.995, p<0.001), and AMs (rs=0.969, p<0.001). In conclusion, both the SphK-1 and S1PR-3 proteins were overexpressed in the lung tissues of severe P. falciparum malaria patients with PE, suggesting that SphK-1 and S1PR-3 mediate the pathogenesis of PE in severe malaria. Targeting the regulation of SphK-1 and/or S1PR-3 may be an approach to treat pulmonary complications in severe P. falciparum patients.


Sign in / Sign up

Export Citation Format

Share Document