Next Article in Journal
New Type of Halogen Bond: Multivalent Halogen Interacting with π- and σ-Electrons
Next Article in Special Issue
Frozen Microemulsions for MAPLE Immobilization of Lipase
Previous Article in Journal
Composition and Antibacterial Activity of the Essential Oils of Orthosiphon stamineus Benth and Ficus deltoidea Jack against Pathogenic Oral Bacteria
Previous Article in Special Issue
Monitoring the Activity of Immobilized Lipase with Quinizarin Diester Fluoro-Chromogenic Probe
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lipoprotein Lipase Expression in Chronic Lymphocytic Leukemia: New Insights into Leukemic Progression

1
Department of Developmental Neurobiology, Instituto de Investigaciones Biologicas Clemente Estable, 11400 Montevideo, Uruguay
2
Research Laboratory on Chronic Lymphocytic Leukemia, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
*
Author to whom correspondence should be addressed.
Molecules 2017, 22(12), 2083; https://doi.org/10.3390/molecules22122083
Submission received: 3 October 2017 / Revised: 24 November 2017 / Accepted: 24 November 2017 / Published: 5 December 2017
(This article belongs to the Special Issue Lipases and Lipases Modification)

Abstract

:
Lipoprotein lipase (LPL) is a central enzyme in lipid metabolism. Due to its catalytic activity, LPL is involved in metabolic pathways exploited by various solid and hematologic malignancies to provide an extra energy source to the tumor cell. We and others described a link between the expression of LPL in the tumor cell and a poor clinical outcome of patients suffering Chronic Lymphocytic Leukemia (CLL). This leukemia is characterized by a slow accumulation of mainly quiescent clonal CD5 positive B cells that infiltrates secondary lymphoid organs, bone marrow and peripheral blood. Despite LPL being found to be a reliable molecular marker for CLL prognosis, its functional role and the molecular mechanisms regulating its expression are still matter of debate. Herein we address some of these questions reviewing the current state of the art of LPL research in CLL and providing some insights into where currently unexplored questions may lead to.

1. Lipoprotein Lipase

Lipoprotein lipase (LPL, EC 3.1.1.34) is a N-glycosylated protein [1] that forms homodimers and is able to hydrolize triglycerides from chylomicrons [2] and very low-density lipoproteins [3]. The first evidence of its existence was serendipitously found when studying circulating red blood cell mass in dogs. In those experiments, it was found that the administration of heparin as an anticoagulant was able to counteract alimentary lipemia in five minutes or less [4]. LPL plays a central function in lipid metabolism and has been subject of intense and meticulous studies ever since. General aspects of LPL biology have already been reviewed elsewhere [5,6].

LPL Synthesis and Trafficking

LPL active dimer consists of two antiparallel subunits [7] whose formation and trafficking rely on a series of post-translational modifications. Interaction with calcium-dependent chaperones of the N-glycosylated polypeptide chain has been proven essential to the correct folding of LPL [8]. Furthermore, a lipase chaperon—Lipase-maturation factor 1 (Lmf1)—has been suggested to be required for dimer assembly and activity, as mutations in LMF1 cause lipase deficiency in mice [9]. A mouse model overexpressing LMF1 has increased LPL activity [10], and LPL has been co-immunoprecipitated with Lmf1 and Sortilin-related receptor (SorLA) [11]. It has been shown that LPL intracellular localization is regulated by SorLA, which directs its trafficking from the trans-Golgi network to endosomes [11]. LPL internalization by receptor-mediated endocytosis has been studied [12] either through LDL receptor-related protein [13] or by an LDL-receptor independent pathway [14].

2. Chronic Lymphocytic Leukemia

Chronic lymphocytic leukemia (CLL) is the most frequent form of leukemia among adult populations of Caucasian origin [15]. CLL is a malignancy of mature clonal B lymphocytes that accumulate in the blood, bone marrow and other lymphoid tissues, and is diagnosed upon the presence of ≥5000 clonal B lymphocytes per microliter of peripheral blood persisting for more than 3 months [16]. This leukemia is characterized by the accumulation of long-lived circulating clonal leukemic B-cells resulting from a complex balance between cell proliferation and apoptotic death. Increasing evidence suggests that CLL B-cells in lymph nodes (LN) and bone marrow (BM) that interact with stromal cells receive proliferative signals and are protected from cell death. These data led to the view that CLL is a dynamic process composed of cells that also proliferate and die, often at appreciable levels [17]. This crosstalk with accessory cells in specialized tissue microenvironments favors disease progression, by promoting malignant B-cell growth and the emergence of new genetic alterations which will lead to drug resistance [18]. Disease prognosis and the heterogeneous clinical evolution in CLL are probably related at least in part to these microenvironmental signaling, and although available treatments often induce remissions, CLL remains an incurable disease [19].
In CLL one third of the patients have an indolent disease with long survival and never require treatment, another third have an aggressive disease from onset and need to be immediately treated, and the last third have an indolent disease at onset which may last for years but then invariably progress to an aggressive disease [20]. It is because of this latter group that the search for strong prognostic markers in CLL predicting disease evolution has been of capital importance, and a number of them have been developed, the most reliable and universal still being the mutational status of the variable region of the heavy chain of immunoglobulin (IgHV) genes [21,22]. Patients carrying somatic hypermutation in their IgHV genes—mutated CLL (Mut)—display a better prognosis than patients with unmutated (Um) IgHV genes

3. LPL in Chronic Lymphocytic Leukemia

3.1. LPL As a Prognostic Marker of Disease Progression

Gene expression profiling analyses comparing Um and Mut patients were performed during the first decade of the century. We and others have performed these studies and described that LPL is differentially overexpressed in Um patients [23,24,25]. With these results in mind we selected and validated two genes, LPL for Um and ADAM29 for Mut CLLs, as candidates to propose a novel prognostic method. This methodology was tested in a cohort of 127 CLL patients, and correlated to clinical outcome and IgHV mutational status. Finally, we demonstrated that quantification of LPL and ADAM29 gene expression ratio is a strong prognostic indicator in CLL, providing better prognostic assessment than serologic markers in advanced stages of the disease [26]. A body of evidence has confirmed that the expression of LPL mRNA is associated to bad prognosis, and that it is the most robust of the molecular markers in CLL [27,28,29,30,31,32,33].
The elevated expression of LPL gene in Um CLL B-cells is a very remarkable observation, because there is no expression of LPL in normal B cells. This specific and ectopic expression constitutes not only a suitable prognostic marker to study disease evolution, but could also be helpful to understand the heterogeneous proliferative behavior in CLL. Despite the prognostic value of LPL expression is well established, the functional role of LPL overexpression in CLL pathogenesis as well as the molecular mechanisms regulating its expression are still open questions.
Concerning the functional role of LPL in CLL cells, increasing evidence supports the idea that LPL expression could help the leukemic clone to increase survival and proliferative signaling, leading to disease progression. We have also shown that microenvironmental signaling can induce LPL expression and proliferative phenotype in primary CLL B-cells [34,35]. Supporting this idea Rozovski, Grgurevic, et al. demonstrated that LPL confers CLL a survival advantage, since shRNA knockdown of LPL increases apoptotic death [36]. Accordingly, it has recently been reported that NOTCH1 gene mutations which are associated with disease progression and treatment refractoriness [37] are directly related to LPL expression in CLL [38].
Concerning the molecular mechanism that regulates LPL expression we previously demonstrated that abnormal expression of LPL gene in Um CLL patients results from the lack of methylation in the CpG island involving the whole exon 1 and the first nucleotides of intron 1 of LPL [34]. This epigenetic mechanism appears to be mainly triggered by proliferative T-cell-dependent signals and, in some patients, through the cross-linking of the B-cell receptor (BCR). By contrast, signaling through TLR9 or TLR1/2 pathways are unable to induce demethylation of the CpG island, LPL expression and B-cell proliferation [35]. Rozovski, Grgurevic, et al. have shown that LPL expression can also be transcriptionally regulated by STAT3 phosphorylation, and nuclear translocation where it can bind LPL promoter [36]. Additionally, it is necessary to mention that LPL expression can be regulated post-transcriptionally by miR-29 [39,40]. It has been reported that miR-29 expression is down-regulated in high-risk Um CLL patients [41,42,43,44]. In a more recent study of the microRNAome of a large patient cohort, down-regulation of miR-29c was the feature better related to IgHV Um profile [45]. In fact, Santanam et al. have developed a mouse model of early onset indolent CD5+ B-CLL by targeted overexpression of miR-29 in B-lymphocytes under control of the Eμ enhancer [46]. The authors focused on the effect on leukemogenesis by the interaction of miR-29 and TCL1 [44,47] and did not evaluate LPL expression, which would be expected to be low. Deregulation of miR-29 is known to have important effects in diverse hematological disorders (reviewed in [48]), to respond to cellular signaling processes such as BCR or CD40 stimulation, and to engage NF-κB activation through TCL1 [47]. Linking these microenvironmental signaling to the epigenetic changes described by us in Um patients as well as their correlation with miR-29 and LPL expression could be an interesting issue that is still awaiting to be studied in CLL progression.

3.2. LPL in CLL B-Cell Metabolism

LPL has been shown to mediate lipolysis and subsequent fatty acid (FA)-mediated fueling of cell proliferation in several solid tumors [49], and it has recently been shown that low-density lipoproteins may enhance proliferative responses of CLL cells to inflammatory signals [50]. PPARα protein levels in CLL B-cells have been shown to correlate with leukocytosis and clinical Rai stages, which suggests a metabolic switch to oxidation of fatty acids via PPARα [51] and PPARδ [50]. These findings are supported by the observation that CLL B-cells have lipid vacuoles in their cytoplasm, and that incubation with free fatty-acids (FFAs) increased their metabolic rate in terms of oxygen consumption [36]. Furthermore, the incidence of hyperlipidemia has been found to be higher in CLL patients, and treatment of hyperlipidemia with statins benefited them in terms of a delayed time to first treatment [52]. The same group expanded their initial study to a cohort of >2000 CLL patients in a retrospective analysis and found that both lipid-lowering drugs, as well as statin treatment prolonged overall survival by 3.7 years [53]. These findings suggest that a second mechanism mediated by LDL may be converging in STAT3 phosphorylation and generating an activated state in CLL B-cells [50].
Transcriptional profiling identified a metabolic shift into a muscle or adipose tissue-like strategy with lipid oxidation in poor prognosis Um IgHV and LPL expressing B CLL cells [54]. How this metabolic reprogramming ends up in a worse outcome for patients is only beginning to be understood. Long chain fatty acids, free cholesterol and vitamin E- increase STAT3 phosphorylation directed either by IL-10, IFNα or phorbol esters in CLL cells [50]. STAT3 phosphorylation in turn drives LPL expression directly, by binding to a GAS-like element 280 bp upstream of the LPL transcription start site and activating its transcription [36]. LPL expression favors FA oxidation, and this seems to result in higher cell survival as LPL knockdown or chemical inhibition reduced CLL cell viability [36,55], which might be explained in part by a transcriptional response [32]. Accordingly, microenvironmental induction of LPL expression stimulates CLL cell proliferation [35]. These findings indicate that LPL expression can be regulated by the microenvironment, either by autocrine or paracrine signaling and that it reflects a metabolic switch in CLL B-cells which confers an adaptive advantage. A positive feedback loop may maintain LPL expression and worsen the scenario for Um patients. In CLL, STAT3 is constitutively activated which also activates LPL transcription [36]. LPL breaks down very low-density lipoproteins (VLDL) and chylomicrons and liberates FFAs, generating a proinflammatory state which in turn activates STAT3 [51] and further activation of LPL transcription. This would further increase the levels of FFAs, thus exacerbating CLL cells responsiveness to cytokine signaling. More general aspects of metabolic pathways in CLL have been nicely reviewed recently [56].

3.3. Non-Metabolic Roles of LPL in CLL

Many studies have reported an increased expression of LPL in poor prognosis CLL, and several metabolic pathways could be involved in cancer progression as discussed above. However, attempts to determine metabolic activity of LPL directly have failed to correlate higher expression to higher metabolic activity. A seminal study with 33 CLL patients reported lower catalytic activity in Um patients than in their Mut counterpart [30]. Another report analyzing data from 42 patients did not find differences between CLL groups and reported that LPL activity was comparable to that of healthy individuals [32].
LPL can mediate lipoprotein uptake by cells [57], chylomicron attachment to cell surface through LDL-related receptor [58], and lipoprotein margination in small blood vessels, by binding on the one hand to the extracellular surface of endothelium via GPIHBP1, and on the other to triglyceride-rich lipoproteins [59]. Besides its canonical role in lipid metabolism, an interesting—yet quite unexplored—non-metabolic function of LPL has been known for 20 years. LPL can act as a bridging molecule between cells, as in the adhesion of monocytes to endothelial cells mediated by heparan sulfate proteoglycans (HSPGs) and LPL [60], whose interaction has recently been shown to be dynamic [61]. Provided that CLL cells display HSPGs on their surface [62] and that LPL forms homodimers, it could occur that a bridging between leukemic B-cells and other cells expressing surface HSPGs or GPIHBP1 such as endothelial cells would be mediated by LPL. Although several groups have already speculated about it, a cell–cell bridging role for LPL in CLL pathogenesis still has to be demonstrated [30,35,63]. If such a bridging actually occurred, LPL would be pivoting between surface HSPGs on the B-CLL cell side, and either HSPGs or GPIHBP1 on their counterpart.
Rombout et al. have found that two SNPs commonly found in LPL, rs328 (premature stop codon) and rs13702 were significantly associated with CLL outcome [63]. Although both SNPs are well-known gain-of-function mutations [64,65], the authors of the aforementioned study reported not to have been able to detect significant differences in LPL mRNA, protein levels, or enzymatic activity in patients carrying the SNPs [63]. How these mutations affect clinical outcome in CLL is still unclear, but whether these SNPs might have a role—if any—in LPL non-metabolic functions has not been explored yet. Furthermore, at least nine isoelectric point isoforms of LPL have been described in human blood of healthy individuals [66], thus opening a new dimension of studies to come for LPL in CLL and other pathologies.

4. Concluding Remarks

LPL is a protein located on the luminal side of the blood vessel wall, where it is anchored to heparan sulfate proteoglycans and contains binding sites for both heparan sulfate chains and apoproteins [67]. LPL is overexpressed in B-cells of unmutated IgHV CLL patients, and its expression can be used to predict their clinical outcome [23,24,25,26,27,28,29,30,31,32,33]. Accordingly, LPL could have a bridging function in the formation of a trimolecular complex including a lipoprotein particle, LPL and heparan sulfate proteoglycans from different cells [67]. The fact that CLL B-cells display heparan sulfate proteoglycans on their surface [62], invites to speculate about whether LPL localization on the cellular membrane could affect the biological behavior of CLL cells, by favoring cell spreading, migration and intracellular signaling following activation of the tumoral clone by an activated microenvironment. If it is the case, LPL might also act as a crosstalk factor facilitating specific interactions with accessory cells in tissue microenvironments. LPL might then be added to the list of proteins implicated in the activation of CLL proliferative pool together with integrins such as CD49d, metalloproteinases (MMP-9), antiapoptotic molecules (BCL2) as well as chemokines (CCL3, CCL4, CXCL12) [68,69]. Thus, LPL could be contributing to leukemic progression either per se through metabolic reprogramming, or through the synergistic contribution to an activating microenvironment in which the leukemic clone is continuously nourished (Figure 1).
The role that abnormal LPL expression could have in disease evolution, has also been addressed by previous work from Pallasch et al., demonstrating that lipase associated genes and triglyceride-specific lipase activity were significantly increased when comparing CLL B-cells to normal CD5+ B-cells [55]. The same authors reported that incubation of CLL tumoral cells with the lipase inhibitor orlistat resulted in increased apoptosis, which, could suggest that lipid metabolism and lipase activity could be functionally relevant in aggressive CLL [55]. Phenotypic analyses have shown that CLL B-cells expressing LPL are also enriched in FA degradation genes [54]. Recently, LPL has been shown to mediate lipolysis and subsequent FA-mediated fueling of cell proliferation in several solid tumors [49], and it has recently been shown that low-density lipoproteins may enhance proliferative responses of CLL cells to inflammatory signals [50].
A big amount of information is known nowadays about LPL some of which relates to CLL. Still, our understanding whether LPL overexpression in poor outcome CLL is a cause or consequence is poor. Many questions are still open and more answers will certainly come in next years.

Acknowledgments

The authors thank Uruguayan Agencia Nacional de Investigacion e Innovacion (ANII) for the graduate fellowship of DP and for the grants FMV_2_2011_1_7323; FMV_1_2014_1_104397 and FOCEM (MERCOSUR Structural Convergence Fund), COF 03/11. Open access publishing fees have been partially funded by Programa de Desarrollo de las Ciencias Básicas (PEDECIBA)—Uruguay.

Conflicts of Interest

The authors declare no conflict of interest. The founding sponsors had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish the results.

References

  1. Chajek-Shaul, T.; Friedman, G.; Knobler, H.; Stein, O.; Etienne, J.; Stein, Y. Importance of the different steps of glycosylation for the activity and secretion of lipoprotein lipase in rat preadipocytes studied with monensin and tunicamycin. Biochim. Biophys. Acta Lipids Lipid Metab. 1985, 837, 123–134. [Google Scholar] [CrossRef]
  2. Felts, J.M.; Itakura, H.; Crane, R.T. The mechanism of assimilation of constituents of chylomicrons, very low density lipoproteins and remnants—A new theory. Biochem. Biophys. Res. Commun. 1975, 66, 1467–1475. [Google Scholar] [CrossRef]
  3. Chajek, T.; Stein, O.; Stein, Y. Lipoprotein lipase of cultured mesenchymal rat heart cells. Biochim. Biophys. Acta Lipids Lipid Metab. 1978, 528, 466–474. [Google Scholar] [CrossRef]
  4. Hahn, P.F. Abolishment of Alimentary Lipemia Following Injection of Heparin. Science 1943, 98, 19–20. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, H.; Eckel, R.H. Lipoprotein lipase: From gene to obesity. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E271–E288. [Google Scholar] [CrossRef] [PubMed]
  6. Davies, B.S.J.; Beigneux, A.P.; Fong, L.G.; Young, S.G. New wrinkles in lipoprotein lipase biology. Curr. Opin. Lipidol. 2012, 23, 35–42. [Google Scholar] [CrossRef] [PubMed]
  7. Wong, H.; Yang, D.; Hill, J.S.; Davis, R.C.; Nikazy, J.; Schotz, M.C. A molecular biology-based approach to resolve the subunit orientation of lipoprotein lipase. Proc. Natl. Acad. Sci. USA 1997, 94, 5594–5598. [Google Scholar] [CrossRef] [PubMed]
  8. Zhang, L.; Wu, G.; Tate, C.G.; Lookene, A.; Olivecrona, G. Calreticulin promotes folding/dimerization of human lipoprotein lipase expressed in insect cells (Sf21). J. Biol. Chem. 2003, 278, 29344–29351. [Google Scholar] [CrossRef] [PubMed]
  9. Péterfy, M.; Ben-Zeev, O.; Mao, H.Z.; Weissglas-Volkov, D.; Aouizerat, B.E.; Pullinger, C.R.; Frost, P.H.; Kane, J.P.; Malloy, M.J.; Reue, K.; et al. Mutations in LMF1 cause combined lipase deficiency and severe hypertriglyceridemia. Nat. Genet. 2007, 39, 1483–1487. [Google Scholar] [CrossRef] [PubMed]
  10. Péterfy, M. Lipase maturation factor 1: A lipase chaperone involved in lipid metabolism. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2012, 1821, 790–794. [Google Scholar] [CrossRef] [PubMed]
  11. Klinger, S.C.; Glerup, S.; Raarup, M.K.; Mari, M.C.; Nyegaard, M.; Koster, G.; Prabakaran, T.; Nilsson, S.K.; Kjaergaard, M.M.; Bakke, O.; et al. SorLA regulates the activity of lipoprotein lipase by intracellular trafficking. J. Cell Sci. 2011, 124, 1095–1105. [Google Scholar] [CrossRef] [PubMed]
  12. Casaroli-Marano, R.P.; García, R.; Vilella, E.; Olivecrona, G.; Reina, M.; Vilaró, S. Binding and intracellular trafficking of lipoprotein lipase and triacylglycerol-rich lipoproteins by liver cells. J. Lipid Res. 1998, 39, 789–806. [Google Scholar] [PubMed]
  13. Vergés, M.; Bensadoun, A.; Herz, J.; Belcher, J.D.; Havel, R.J. Endocytosis of Hepatic Lipase and Lipoprotein Lipase into Rat Liver Hepatocytes in Vivo Is Mediated by the Low Density Lipoprotein Receptor-related Protein. J. Biol. Chem. 2004, 279, 9030–9036. [Google Scholar] [CrossRef] [PubMed]
  14. Fernández-Borja, M.; Bellido, D.; Vilella, E.; Olivecrona, G.; Vilaró, S. Lipoprotein lipase-mediated uptake of lipoprotein in human fibroblasts: Evidence for an LDL receptor-independent internalization pathway. J. Lipid Res. 1996, 37, 464–481. [Google Scholar] [PubMed]
  15. Hallek, M. Chronic lymphocytic leukemia: 2015 Update on diagnosis, risk stratification, and treatment. Am. J. Hematol. 2015, 90, 446–460. [Google Scholar] [CrossRef] [PubMed]
  16. Fabbri, G.; Dalla-Favera, R. The molecular pathogenesis of chronic lymphocytic leukaemia. Nat. Rev. Cancer 2016, 16, 145–162. [Google Scholar] [CrossRef] [PubMed]
  17. Caligaris-Cappio, F.; Ghia, P. Novel insights in chronic lymphocytic leukemia: Are we getting closer to understanding the pathogenesis of the disease? J. Clin. Oncol. 2008, 26, 4497–4503. [Google Scholar] [CrossRef] [PubMed]
  18. Burger, J.A.; Ghia, P.; Rosenwald, A.; Caligaris-Cappio, F. The microenvironment in mature B-cell malignancies: A target for new treatment strategies. Blood 2009, 114, 3367–3375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Oppezzo, P.; Dighiero, G. Role of the B-cell receptor and the microenvironment in chronic lymphocytic leukemia. Blood Cancer J. 2013, 3, e149. [Google Scholar] [CrossRef] [PubMed]
  20. Dighiero, G. Unsolved issues in CLL biology and management. Leukemia 2003, 17, 2385–2391. [Google Scholar] [CrossRef] [PubMed]
  21. Damle, R.N.; Wasil, T.; Fais, F.; Ghiotto, F.; Valetto, A.; Allen, S.L.; Buchbinder, A.; Budman, D.; Dittmar, K.; Kolitz, J.; et al. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood 1999, 94, 1840–1847. [Google Scholar] [PubMed]
  22. Hamblin, T.J.; Davis, Z.; Gardiner, A.; Oscier, D.G.; Stevenson, F.K. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 1999, 94, 1848–1854. [Google Scholar] [PubMed]
  23. Klein, U.; Tu, Y.; Stolovitzky, G.A.; Mattioli, M.; Cattoretti, G.; Husson, H.; Freedman, A.; Inghirami, G.; Cro, L.; Baldini, L.; et al. Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J. Exp. Med. 2001, 194, 1625–1638. [Google Scholar] [CrossRef] [PubMed]
  24. Rosenwald, A.; Alizadeh, A.A.; Widhopf, G.; Simon, R.; Davis, R.E.; Yu, X.; Yang, L.; Pickeral, O.K.; Rassenti, L.Z.; Powell, J.; et al. Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J. Exp. Med. 2001, 194, 1639–1647. [Google Scholar] [CrossRef] [PubMed]
  25. Vasconcelos, Y.; De Vos, J.; Vallat, L.; Rème, T.; Lalanne, A.I.; Wanherdrick, K.; Michel, A.; Nguyen-Khac, F.; Oppezzo, P.; Magnac, C.; et al. Gene expression profiling of chronic lymphocytic leukemia can discriminate cases with stable disease and mutated Ig genes from those with progressive disease and unmutated Ig genes. Leukemia 2005, 19, 2002–2005. [Google Scholar] [CrossRef] [PubMed]
  26. Oppezzo, P.; Vasconcelos, Y.; Settegrana, C.; Jeannel, D.; Vuillier, F.; Legarff-Tavernier, M.; Kimura, E.Y.; Bechet, S.; Dumas, G.; Brissard, M.; et al. The LPL/ADAM29 expression ratio is a novel prognosis indicator in chronic lymphocytic leukemia. Blood 2005, 106, 650–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Heintel, D.; Kienle, D.; Shehata, M.; Kröber, A.; Kroemer, E.; Schwarzinger, I.; Mitteregger, D.; Le, T.; Gleiss, A.; Mannhalter, C.; et al. High expression of lipoprotein lipase in poor risk B-cell chronic lymphocytic leukemia. Leukemia 2005, 19, 1216–1223. [Google Scholar] [CrossRef] [PubMed]
  28. Van’t Veer, M.B.; Brooijmans, A.M.; Langerak, A.W.; Verhaaf, B.; Goudswaard, C.S.; Graveland, W.J.; van Lom, K.; Valk, P.J.M. The predictive value of lipoprotein lipase for survival in chronic lymphocytic leukemia. Haematologica 2006, 91, 56–63. [Google Scholar] [PubMed]
  29. Nückel, H.; Hüttmann, A.; Klein-Hitpass, L.; Schroers, R.; Führer, A.; Sellmann, L.; Dührsen, U.; Dürig, J. Lipoprotein lipase expression is a novel prognostic factor in B-cell chronic lymphocytic leukemia. Leuk. Lymphoma 2006, 47, 1053–1061. [Google Scholar] [CrossRef] [PubMed]
  30. Mansouri, M.; Sevov, M.; Fahlgren, E.; Tobin, G.; Jondal, M.; Osorio, L.; Roos, G.; Olivecrona, G.; Rosenquist, R. Lipoprotein lipase is differentially expressed in prognostic subsets of chronic lymphocytic leukemia but displays invariably low catalytical activity. Leuk. Res. 2010, 34, 301–306. [Google Scholar] [CrossRef] [PubMed]
  31. Kaderi, M.A.; Kanduri, M.; Buhl, A.M.; Sevov, M.; Cahill, N.; Gunnarsson, R.; Jansson, M.; Smedby, K.E.; Hjalgrim, H.; Jurlander, J.; et al. LPL is the strongest prognostic factor in a comparative analysis of RNA-based markers in early chronic lymphocytic leukemia. Haematologica 2011, 96, 1153–1160. [Google Scholar] [CrossRef] [PubMed]
  32. Porpaczy, E.; Tauber, S.; Bilban, M.; Kostner, G.; Gruber, M.; Eder, S.; Heintel, D.; Le, T.; Fleiss, K.; Skrabs, C.; et al. Lipoprotein lipase in chronic lymphocytic leukaemia—Strong biomarker with lack of functional significance. Leuk. Res. 2013, 37, 631–636. [Google Scholar] [CrossRef] [PubMed]
  33. Mátrai, Z.; Andrikovics, H.; Szilvási, A.; Bors, A.; Kozma, A.; Ádám, E.; Halm, G.; Karászi, É.; Tordai, A.; Masszi, T. Lipoprotein Lipase as a Prognostic Marker in Chronic Lymphocytic Leukemia. Pathol. Oncol. Res. 2017, 23, 165–171. [Google Scholar] [CrossRef] [PubMed]
  34. Abreu, C.; Moreno, P.; Palacios, F.; Borge, M.; Morande, P.; Landoni, A.I.; Gabus, R.; Dighiero, G.; Giordano, M.; Gamberale, R.; et al. Methylation status regulates lipoprotein lipase expression in chronic lymphocytic leukemia. Leuk. Lymphoma 2013, 54, 1844–1888. [Google Scholar] [CrossRef] [PubMed]
  35. Moreno, P.; Abreu, C.; Borge, M.; Palacios, F.; Morande, P.; Pegazzano, M.; Bianchi, S.; Landoni, A.I.; Agrelo, R.; Giordano, M.; et al. Lipoprotein lipase expression in unmutated CLL patients is the consequence of a demethylation process induced by the microenvironment. Leukemia 2013, 27, 721–725. [Google Scholar] [CrossRef] [PubMed]
  36. Rozovski, U.; Grgurevic, S.; Bueso-Ramos, C.; Harris, D.M.; Li, P.; Liu, Z.; Wu, J.Y.; Jain, P.; Wierda, W.; Burger, J.; et al. Aberrant LPL Expression, Driven by STAT3, Mediates Free Fatty Acid Metabolism in CLL Cells. Mol. Cancer Res. 2015, 13, 944–953. [Google Scholar] [CrossRef] [PubMed]
  37. Sportoletti, P.; Baldoni, S.; Cavalli, L.; Del Papa, B.; Bonifacio, E.; Ciurnelli, R.; Bell, A.S.; Di Tommaso, A.; Rosati, E.; Crescenzi, B.; et al. NOTCH1 PEST domain mutation is an adverse prognostic factor in B-CLL. Br. J. Haematol. 2010, 151, 404–406. [Google Scholar] [CrossRef] [PubMed]
  38. Kristensen, L.; Kristensen, T.; Abildgaard, N.; Royo, C.; Frederiksen, M.; Mourits-Andersen, T.; Campo, E.; Møller, M.B. LPL gene expression is associated with poor prognosis in CLL and closely related to NOTCH1 mutations. Eur. J. Haematol. 2016, 97, 175–182. [Google Scholar] [CrossRef] [PubMed]
  39. Chen, T.; Li, Z.; Tu, J.; Zhu, W.; Ge, J.; Zheng, X.; Yang, L.; Pan, X.; Yan, H.; Zhu, J. MicroRNA-29a regulates pro-inflammatory cytokine secretion and scavenger receptor expression by targeting LPL in oxLDL-stimulated dendritic cells. FEBS Lett. 2011, 585, 657–663. [Google Scholar] [CrossRef] [PubMed]
  40. Bouvy-Liivrand, M.; Heinäniemi, M.; John, E.; Schneider, J.G.; Sauter, T.; Sinkkonen, L. Combinatorial regulation of lipoprotein lipase by microRNAs during mouse adipogenesis. RNA Biol. 2014, 11, 76–91. [Google Scholar] [CrossRef] [PubMed]
  41. Calin, G.A.; Ferracin, M.; Cimmino, A.; Di Leva, G.; Shimizu, M.; Wojcik, S.E.; Iorio, M.V.; Visone, R.; Sever, N.I.; Fabbri, M.; et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N. Engl. J. Med. 2005, 353, 1793–1801. [Google Scholar] [CrossRef] [PubMed]
  42. Fulci, V.; Chiaretti, S.; Goldoni, M.; Azzalin, G.; Carucci, N.; Tavolaro, S.; Castellano, L.; Magrelli, A.; Citarella, F.; Messina, M.; et al. Quantitative technologies establish a novel microRNA profile of chronic lymphocytic leukemia. Blood 2007, 109, 4944–4951. [Google Scholar] [CrossRef] [PubMed]
  43. Marton, S.; Garcia, M.R.; Robello, C.; Persson, H.; Trajtenberg, F.; Pritsch, O.; Rovira, C.; Naya, H.; Dighiero, G.; Cayota, A. Small RNAs analysis in CLL reveals a deregulation of miRNA expression and novel miRNA candidates of putative relevance in CLL pathogenesis. Leukemia 2008, 22, 330–338. [Google Scholar] [CrossRef] [PubMed]
  44. Stamatopoulos, B.; Meuleman, N.; Haibe-kains, B.; Saussoy, P.; Van Den Neste, E.; Michaux, L.; Heimann, P.; Martiat, P.; Bron, D.; Lagneaux, L. microRNA-29c and microRNA-223 down-regulation has in vivo significance in chronic lymphocytic leukemia and improves disease risk stratification sion levels decreased significantly with progression from Binet stage A to C were significantly lower in poor p. Blood 2009, 113, 5237–5245. [Google Scholar] [PubMed]
  45. Negrini, M.; Cutrona, G.; Bassi, C.; Fabris, S.; Zagatti, B.; Colombo, M.; Ferracin, M.; D’Abundo, L.; Saccenti, E.; Matis, S.; et al. microRNAome expression in chronic lymphocytic leukemia: Comparison with normal B cell subsets and correlations with prognostic and clinical parameters. Clin. Cancer Res. 2014. [Google Scholar] [CrossRef] [PubMed]
  46. Santanam, U.; Zanesi, N.; Efanov, A.; Costinean, S.; Palamarchuk, A.; Hagan, J.P.; Volinia, S.; Alder, H.; Rassenti, L.; Kipps, T.; et al. Chronic lymphocytic leukemia modeled in mouse by targeted miR-29 expression. Proc. Natl. Acad. Sci. USA 2010, 107. [Google Scholar] [CrossRef] [PubMed]
  47. Pekarsky, Y.; Santanam, U.; Cimmino, A.; Palamarchuk, A.; Efanov, A.; Maximov, V.; Volinia, S.; Alder, H.; Liu, C.G.; Rassenti, L.; et al. Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res. 2006, 66, 11590–11593. [Google Scholar] [CrossRef] [PubMed]
  48. Kluiver, J.L.; Chen, C.-Z. MicroRNAs regulate B-cell receptor signaling-induced apoptosis. Genes Immun. 2012, 13, 239–244. [Google Scholar] [CrossRef] [PubMed]
  49. Kuemmerle, N.B.; Rysman, E.; Lombardo, P.S.; Flanagan, A.J.; Lipe, B.C.; Wells, W.A.; Pettus, J.R.; Froehlich, H.M.; Memoli, V.A.; Morganelli, P.M.; et al. Lipoprotein lipase links dietary fat to solid tumor cell proliferation. Mol. Cancer Ther. 2011, 10, 427–436. [Google Scholar] [CrossRef] [PubMed]
  50. McCaw, L.; Shi, Y.; Wang, G.; Li, Y.-J.; Spaner, D.E. Low Density Lipoproteins Amplify Cytokine-signaling in Chronic Lymphocytic Leukemia Cells. EBioMedicine 2017, 15, 24–35. [Google Scholar] [CrossRef] [PubMed]
  51. Spaner, D.E.; Lee, E.; Shi, Y.; Wen, F.; Li, Y.; Tung, S.; McCaw, L.; Wong, K.; Gary-Gouy, H.; Dalloul, A.; et al. PPAR-alpha is a therapeutic target for chronic lymphocytic leukemia. Leukemia 2013, 27, 1090–1099. [Google Scholar] [CrossRef] [PubMed]
  52. Chow, S.; Buckstein, R.; Spaner, D.E. A link between hypercholesterolemia and chronic lymphocytic leukemia. Leuk. Lymphoma 2015, 1–6. [Google Scholar] [CrossRef] [PubMed]
  53. Mozessohn, L.; Earle, C.; Spaner, D.; Cheng, S.Y.; Kumar, M.; Buckstein, R. The association of dyslipidemia with chronic lymphocytic leukemia: A population-based study. J. Natl. Cancer Inst. 2017, 109, 1–9. [Google Scholar] [CrossRef] [PubMed]
  54. Bilban, M.; Heintel, D.; Scharl, T.; Woelfel, T.; Auer, M.M.; Porpaczy, E.; Kainz, B.; Kröber, A.; Carey, V.J.; Shehata, M.; et al. Deregulated expression of fat and muscle genes in B-cell chronic lymphocytic leukemia with high lipoprotein lipase expression. Leukemia 2006, 20, 1080–1088. [Google Scholar] [CrossRef] [PubMed]
  55. Pallasch, C.P.; Schwamb, J.; Königs, S.; Schulz, A.; Debey, S.; Kofler, D.; Schultze, J.L.; Hallek, M.; Ultsch, A.; Wendtner, C.-M. Targeting lipid metabolism by the lipoprotein lipase inhibitor orlistat results in apoptosis of B-cell chronic lymphocytic leukemia cells. Leukemia 2008, 22, 585–592. [Google Scholar] [CrossRef] [PubMed]
  56. Rozovski, U.; Hazan-Halevy, I.; Barzilai, M.; Keating, M.J.; Estrov, Z. Metabolism pathways in chronic lymphocytic leukemia. Leuk. Lymphoma 2016. [Google Scholar] [CrossRef] [PubMed]
  57. Merkel, M.; Kako, Y.; Radner, H.; Cho, I.S.; Ramasamy, R.; Brunzell, J.D.; Goldberg, I.J.; Breslow, J.L. Catalytically inactive lipoprotein lipase expression in muscle of transgenic mice increases very low density lipoprotein uptake: Direct evidence that lipoprotein lipase bridging occurs in vivo. Proc. Natl. Acad. Sci. USA 1998, 95, 13841–13846. [Google Scholar] [CrossRef] [PubMed]
  58. Beisiegel, U.; Weber, W.; Bengtsson-Olivecrona, G. Lipoprotein lipase enhances the binding of chylomicrons to low density lipoprotein receptor-related protein. Proc. Natl. Acad. Sci. USA 1991, 88, 8342–8346. [Google Scholar] [CrossRef] [PubMed]
  59. Goulbourne, C.N.; Gin, P.; Tatar, A.; Nobumori, C.; Hoenger, A.; Jiang, H.; Grovenor, C.R.M.; Adeyo, O.; Esko, J.D.; Goldberg, I.J.; et al. The GPIHBP1-LPL Complex Is Responsible for the Margination of Triglyceride-Rich Lipoproteins in Capillaries. Cell Metab. 2014, 19, 849–860. [Google Scholar] [CrossRef] [PubMed]
  60. Mamputu, J.C.; Desfaits, A.C.; Renier, G. Lipoprotein lipase enhances human monocyte adhesion to aortic endothelial cells. J. Lipid Res. 1997, 38, 1722–1729. [Google Scholar] [PubMed]
  61. Allan, C.M.; Larsson, M.; Jung, R.S.; Ploug, M.; Bensadoun, A.; Beigneux, A.P.; Fong, L.G.; Young, S.G. Mobility of “HSPG-bound” LPL explains how LPL is able to reach GPIHBP1 on capillaries. J. Lipid Res. 2017, 58, 216–225. [Google Scholar] [CrossRef] [PubMed]
  62. Van Bockstaele, F.; Pede, V.; Janssens, A.; Callewaert, F.; Offner, F.; Verhasselt, B.; Philippé, J. Lipoprotein lipase mRNA expression in whole blood is a prognostic marker in B cell chronic lymphocytic leukemia. Clin. Chem. 2007, 53, 204–212. [Google Scholar] [CrossRef] [PubMed]
  63. Rombout, A.; Verhasselt, B.; Philippé, J. Lipoprotein lipase in chronic lymphocytic leukemia: Function and prognostic implications. Eur. J. Haematol. 2016, 97, 409–415. [Google Scholar] [CrossRef] [PubMed]
  64. Deo, R.C.; Reich, D.; Tandon, A.; Akylbekova, E.; Patterson, N.; Waliszewska, A.; Kathiresan, S.; Sarpong, D.; Taylor, H.A.; Wilson, J.G. Genetic differences between the determinants of lipid profile phenotypes in African and European Americans: The Jackson Heart Study. PLoS Genet. 2009, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Richardson, K.; Nettleton, J.A.; Rotllan, N.; Tanaka, T.; Smith, C.E.; Lai, C.Q.; Parnell, L.D.; Lee, Y.C.; Lahti, J.; Lemaitre, R.N.; et al. Gain-of-function lipoprotein lipase variant rs13702 modulates lipid traits through disruption of a MicroRNA-410 seed site. Am. J. Hum. Genet. 2013, 92, 5–14. [Google Scholar] [CrossRef] [PubMed]
  66. Badia-Villanueva, M.; Carulla, P.; Carrascal, M.; Abián, J.; Llobera, M.; Casanovas, A.; Dolores López-Tejero, M. Lipoprotein lipase isoelectric point isoforms in humans. Biochem. Biophys. Res. Commun. 2014, 445, 480–485. [Google Scholar] [CrossRef] [PubMed]
  67. Kolset, S.O.; Salmivirta, M. Cell surface heparan sulfate proteoglycans and lipoprotein metabolism. Cell. Mol. Life Sci. 1999, 56, 857–870. [Google Scholar] [CrossRef] [PubMed]
  68. Borge, M.; Nannini, P.R.; Galletti, J.G.; Morande, P.E.; Sanchez Avalos, J.; Bezares, R.F.; Giordano, M.; Gamberale, R. CXCL12-induced chemotaxis is impaired in T cells from ZAP-70-chronic lymphocytic leukemia patients. Haematologica 2010, 95, 768–775. [Google Scholar] [CrossRef] [PubMed]
  69. Palacios, F.; Moreno, P.; Morande, P.; Abreu, C.; Correa, A.; Porro, V.; Landoni, A.I.; Gabus, R.; Giordano, M.; Dighiero, G.; et al. High expression of AID and active class switch recombination might account for a more aggressive disease in unmutated CLL patients: Link with an activated microenvironment in CLL disease. Blood 2010, 115, 4488–4496. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Hypothetical model of LPL function in CLL B-cells in secondary lymphoid organs (SLO, left) and peripheral blood (PB, right). HSPG-attached LPL molecules at the surface of B-CLL cells can bind very low-density lipoproteins and chylomicrons thus contributing to oxidative metabolism and fatty-acid signaling. LPL has been proposed to play a similar role in the intracellular compartment by releasing FFAs from cytosolic lipid droplets [36,56]. A non-canonical role for LPL in CLL B-cell surface would contribute to microenvironmental crosstalk. LPL would act as a bridging molecule between cells able to bind LPL either by heparan sulfate proteoglycans or GPIHBP1, thus facilitating modulatory interactions, exemplified here by a T-cell dependent activation through CD40/CD40L interaction.
Figure 1. Hypothetical model of LPL function in CLL B-cells in secondary lymphoid organs (SLO, left) and peripheral blood (PB, right). HSPG-attached LPL molecules at the surface of B-CLL cells can bind very low-density lipoproteins and chylomicrons thus contributing to oxidative metabolism and fatty-acid signaling. LPL has been proposed to play a similar role in the intracellular compartment by releasing FFAs from cytosolic lipid droplets [36,56]. A non-canonical role for LPL in CLL B-cell surface would contribute to microenvironmental crosstalk. LPL would act as a bridging molecule between cells able to bind LPL either by heparan sulfate proteoglycans or GPIHBP1, thus facilitating modulatory interactions, exemplified here by a T-cell dependent activation through CD40/CD40L interaction.
Molecules 22 02083 g001

Share and Cite

MDPI and ACS Style

Prieto, D.; Oppezzo, P. Lipoprotein Lipase Expression in Chronic Lymphocytic Leukemia: New Insights into Leukemic Progression. Molecules 2017, 22, 2083. https://doi.org/10.3390/molecules22122083

AMA Style

Prieto D, Oppezzo P. Lipoprotein Lipase Expression in Chronic Lymphocytic Leukemia: New Insights into Leukemic Progression. Molecules. 2017; 22(12):2083. https://doi.org/10.3390/molecules22122083

Chicago/Turabian Style

Prieto, Daniel, and Pablo Oppezzo. 2017. "Lipoprotein Lipase Expression in Chronic Lymphocytic Leukemia: New Insights into Leukemic Progression" Molecules 22, no. 12: 2083. https://doi.org/10.3390/molecules22122083

Article Metrics

Back to TopTop