Open access

Lymphocyte Commitment and Ikaros Transcription Factors

Written By

Mariastefania Antica

Submitted: 04 November 2010 Published: 22 December 2011

DOI: 10.5772/19794

From the Edited Volume

Acute Leukemia - The Scientist's Perspective and Challenge

Edited by Mariastefania Antica

Chapter metrics overview

2,141 Chapter Downloads

View Full Metrics

1. Introduction

T lymphocytes like all blood cells are progenies of a single multipotent hematopoietic stem cell (HSC). The existence of HSCs was proven by Till and McCulloch in 1961 when bone marrow cells injected into irradiated mice formed multilineage colonies in their spleens. These cells were called colony forming units (CFU-S) and they have potential for self-renewal and differentiation into all types of blood cells(Till and McCulloch 1961, Wu et al. 1968). For lymphocyte development HSCs migrate from the bone marrow, differentiate in the thymus into immunocompetent cells and seed the peripheral lymphatic organs. This process occurs during fetal development (for review see (Godin and Cumano 2002), but also during the adult life of an individual since T lymphocytes have to be continuously replenished. Transplantation experiments proved that thirty hematopoietic stem cells are sufficient to save 50 percent of lethally irradiated mice, and to reconstitute all blood cell types in vivo (Spangrude et al. 1988). Because it is extremely important to produce immunocompetent T cells the process of their maturation and development is strictly regulated and is succumb to very strict check points at several stages of differentiation. In this context we’ll discuss Ikaros family transcription factors as major regulators of T lymphocyte development.

Advertisement

2. Committed lymphocyte precursor

For a long time it has been postulated analogously to committed myeloid precursors that there is a committed lymphocyte precursor. However the first experiments that showed the existence of a committed lymphocyte precursor was described in the early 90tis when a new population of cells in the thymus was found and its features described (Wu 1991, Wu et al. 1991). These cells expressed most markers of the multipotent hematopoietic stem cells like Thy-1lowCD44+H2k+Sca-1+ but they expressed Sca-2 and low levels of CD4. These cells were negative for all mature blood cells markers lymphocytes (CD8-CD3-Ig-), macrophages (Mac-1-), granulocytes (Gr-1-) and erhytrocytes (TER119-), lineage negative, (Lin- cells). Their Ig and TCR receptor genes were in germline configuration. The described precursors represent 0.05% of the cells in an adult mouse thymus. If the cells were sorted and transferred by intrathymic injections (i.t.) into lethally irradiated congenic mice they developed into all thymic subpopulations, first CD4-CD8- (double negatives, DN) thymocytes, than into CD4+CD8+ double positives which give rise to mature CD4+ and CD8+ lymphocytes. Mature cells derived from the CD4low precursors seeded the spleen, lymph nodes and bone marrow. The reconstitution potential of the described precursors was lower and the time for development was shorter when compared to the multipotent hematopoietic stem cells from the bone marrow. These experiments proved that the CD4low precursors in the thymus can develop into T lymphocytes. Because of their resemblance to the multipotent progenitors from the bone marrow it was important to answer the question whether their potential was restricted only to develop into T lymphocytes. Therefore we transplanted these cells into the periphery by injecting them intravenously (i.v.) into lethally irradiated congenic animals. Their progenies were found to develop into T lymphocytes similarly to the results obtained by direct i.t. transplantation experiments. When injected intravenously they also developed into B lymphocytes but there were no myeloid progenies. During fetal development these cells were detected from day 14 in the fetal thymus but only after birth their function was comparable to the adult ones(Antica et al. 1993). In the bone marrow the CD4low precursor cells were described, but their function was not lymphocyte restricted (Antica et al. 1994a). Further studies showed that in the bone marrow the marker that could differentiate the lymphoid precursor cells from the multipotent precursors was IL7Rα (Kondo et al. 1997). This population, called common lymphoid precursor (CLP) is characterized by Lin Thy-1Sca-1loc-Kitlo IL-7R+ and possess rapid and prominent short-term lymphoid-restricted (T, B, and NK cells) reconstitution activity. However, this progenitor population clonally produces both B and T lymphocytes, but have little myeloid potential in vivo. A recently developed bioinformatics method, called Mining Developmentally Regulated Genes, which mines the publically available microarray data to identify genes that are up- or down-regulated within a developmental pathway was applied to identify surface proteins that distinguish functional CLPs from other progenitors (Inlay et al. 2009, Sahoo et al. 2010). A surface marker Ly6d dissects the CLP population in two and shows that it consists of a mixture of all lymphoid progenitor cells (ALP) which retain B and T lymphoid potential, and BLP (B cell biased lymphoid progenitors). This manuscript offers strong support for the validity of our earlier conclusions. However, ALPs still keep low myeloid potential indicating that ALPs are either a mixture of the CD4low precursor cells and myeloid progenitors or is a single population but at an earlier developmental stage and still multipotent(Wu et al. 1991, Antica et al. 1993, Antica et al. 1994b, Inlay et al. 2009). Here we also stress the necessity and importance of in vivo assays for the determination of physiologic lineage potentials since it has been recently shown that in vitro assays can misrepresent in vivo lineage potentials of murine lymphoid progenitors(Richie Ehrlich et al. 2011).

From the clinical point of view defining the earliest lymphoid precursor is important for a rapid engraftment and protection from infections after hematopoietic stem cell transplantation in chemotherapy or irradiation compromised patients (Arber et al. 2003, Holländer et al. 2010).

Advertisement

3. Ikaros transcription factors

Mechanisms and factors that regulate lymphocyte development from stem cells have to be very accurate since any alteration of this process may lead to serious diseases like leukemia.

Transcription factors from the Ikaros family play an essential role in the commitment of hematopoietic progenitors into the lymphoid lineage as well as in the choice of effector functions at later stages of development(Georgopoulos 1994, Sun et al. 1996, Wang et al. 1998, Cortes et al. 1999). Their role has been addressed by gene targeting and such gene inactivation studies have identified Ikaros, Aiolos and Helios as transcription factors required for the maturation of lymphocytes(Morgan 1997, Hahm 1998, Kelley 1998). It has been shown that mice homozygous for a deletion in these genes undergo remarkable changes in their lymphocyte populations and also those ageing animals with the same mutation develop lymphoproliferative disorders. A number of studies show that Ikaros genes in both mice and human malignancies might be deregulated (Winandy et al. 1995, Nichogiannopoulou et al. 1999, Nakase et al. 2000, Nakayama et al. 2000, Nakase et al. 2002, Rebollo and Schmitt 2003, Dovat et al. 2005; Mullighan et al. 2008, Matulic et al. 2009, Billot et al. 2010). Therefore we addressed the question whether a combination of transcription factor failures may contribute to the development of human lymphoma. We amplified human mRNA from formalin fixed paraffin embedded tissues from lymphoma patients in order to have consistent and well defined groups of patients. Hence, we were able to analyze Ikaros, Aiolos and Helios mRNA from archive tissue specimens from patients with Hodgkin’s and non- Hodgkin’s lymphoma and follicular hyperplasia (Antica et al. 2008, Antica et al. 2010). Further we and others show a deregulation in human leukemia. Acute lymphoblastic leukemia (ALL) is characterized by the Philadelphia chromosome (Ph) which encodes the BCR-ABL1 tyrosine kinase, the most frequent cytogenetic abnormality (∼25–30% of cases)(Mancini et al. 2005). Deletion of the IKAROS gene (IKZF1) was found in 83.7% cases of BCR-ABL1 ALL, but not in chronic-phase CML(Mullighan et al. 2008). Posttranscriptional regulation of alternative splicing of Ikaros was associated with resistance to tyrosine kinase inhibitors (TKIs) in Ph/positive acute lymphoblastic leukemia (ALL) patients (Iacobucci et al. 2008). Further, IKZF1 deletions are likely to be a genomic alteration that significantly affects the prognosis of Ph-positive ALL in adults (Martinelli et al. 2009). Further, when Ikaros expression was analyzed by real time RT-PCR the quantitative distribution of mRNA level in hematopoietic cells of patients with lymphocytic leukemia was similar but a clear difference among groups was due to Aiolos lower expression in all types of acute leukemia(Antica et al. 2007). The mechanisms involved have been tested in the mouse model. It has been found that pre-BCR induces Ikaros to inhibit the proliferation of Philadelphia chromosome-positive B-ALL cells (Trageser et al. 2009). Pre–B cell receptor/IKAROS-induced cell cycle arrest can be reversed by dominant-negative Ikaros splicing variant IK6(Trageser et al. 2009). A possible mechanism of Ikaros suppression has been described by Ma et al. Their experiments on mice show that Ikaros inhibits c-Myc as a direct target, resulting in inhibition of pre B-lymphocyte proliferation (Ma et al 2010).

In the last ten years a new system, besides fetal thymic organ cultures (FTOC) or reaggregation cultures, for T cell growth in vitro has been developed. It has been shown that OP9 stromal cells transfected with the Notch ligand delta like 1 DL1 (OP9-DL1) can support T lymphocyte differentiation in vitro ( Schmitt and Zúńiga-Pflücker 2002). This new technology provided a powerful tool for analyzing developmental phases from multipotent stem cells to mature T lymphocytes at single cell level in vitro and allowed a better understanding of the processes underlying development. However, there are still a lot of unanswered and exciting questions to be solved. Who is regulating Ikaros and Notch? How transcription factors regulate development with their partners and DNA where and how do they interfere with their partners, and DNA, how can we identify targets for new drugs and finally how can we produce T cells in vitro for practical applications and regenerative medicine?

References

  1. 1. Antica M. Cicin-Sain L. Kapitanovic S. Matulic M. Dzebro S. Dominis M. 2008 Aberrant Ikaros, Aiolos, and Helios expression in Hodgkin and non-Hodgkin lymphoma. Blood 111 6 3296 3297 .
  2. 2. Antica M. Dubravcic K. Weber I. Rasic L. Labar B. Batinic D. 2007 A search for a mutation of the Aiolos phosphorylation domain in lymphocytes from patients with leukemia. Haematologica 92 2 260 261 .
  3. 3. Antica M. Paradzik M. Novak S. Dzebro S. Dominis M. 2010 Gene expression in formalin-fixed paraffin-embedded lymph nodes. Journal of Immunological Methods 359(1-2): 42 EOF 46 EOF .
  4. 4. Antica M. Wu L. Shortman K. Scollay R. 1993 Intrathymic lymphoid precursor cells during fetal thymus development. Journal of Immunology 151 11 5887 5895 .
  5. 5. Antica M. Wu L. Shortman K. Scollay R. 1994 Thymic stem cells in mouse bone marrow. Blood 84 1 111 117 .
  6. 6. Arber C. Bit Mansour. A. Sparer T. E. Higgins J. P. Mocarski E. S. Weissman I. L. Shizuru J. A. Brown J. M. Y. 2003 Common lymphoid progenitors rapidly engraft and protect against lethal murine cytomegalovirus infection after hematopoietic stem cell transplantation. Blood 102 2 421 428 .
  7. 7. Billot K. Soeur J. Chereau F. Arrouss I. Merle-Beral H. Huang M. E. Mazier D. Baud V. Rebollo A. 2010 Deregulation of Aiolos expression in chronic lymphocytic leukemia is associated to epigenetic modifications. Blood 117 6 1917 1927
  8. 8. Cortes M. Wong E. Koipally J. Georgopoulos K. 1999 Control of lymphocyte development by the Ikaros gene family. Current Opinion in Immunology 11 2 167 171 .
  9. 9. Dovat S. Montecino-Rodriguez E. Schuman V. Teitell M. A. Dorshkind K. Smale S. T. 2005 Transgenic Expression of Helios in B Lineage Cells Alters B Cell Properties and Promotes Lymphomagenesis. J Immunol 175 6 3508 3515 .
  10. 10. Georgopoulos K. 1994 The Ikaros gene is required for the development of all lymphoid lineages. Cell 79 143 156 .
  11. 11. Godin I. Cumano A. 2002 The hare and the tortoise: an embryonic haematopoietic race. Nat Rev Immunol 2 8 593 604 .
  12. 12. Hahm K. 1998 Helios, a T-cell restricted Ikaros family member that quantitatively associates with Ikaros at centromeric heterochromatin. Genes Dev 12 782 796 .
  13. 13. Holländer G. A. Krenger W. Blazar B. R. 2010 Emerging strategies to boost thymic function. Current Opinion in Pharmacology 10 4 443 453 .
  14. 14. Iacobucci I. Lonetti A. Messa F. Cilloni D. Arruga F. Ottaviani E. Paolini S. Papayannidis C. Piccaluga P. P. Giannoulia P. et al. 2008 Expression of spliced oncogenic Ikaros isoforms in Philadelphia-positive acute lymphoblastic leukemia patients treated with tyrosine kinase inhibitors: implications for a new mechanism of resistance. Blood 112 9 3847 3855 .
  15. 15. Inlay M. A. Bhattacharya D. Sahoo D. Serwold T. Seita J. Karsunky H. Plevritis S. K. Dill D. L. Weissman I. L. 2009 Ly6d marks the earliest stage of B-cell specification and identifies the branchpoint between B-cell and T-cell development. Genes & Development 23 20 2376 2381 .
  16. 16. Kelley C. M. 1998 Helios, a novel dimerization partner of Ikaros expressed in the earliest hematopoietic progenitors. Curr Biol 8 508 515 .
  17. 17. Kondo M. Weissman I. L. Akashi K. 1997 Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 91 661 672 .
  18. 18. Ma S. Pathak S. Mandal M. Trinh L. Clark M. R. Lu R. 2010 Ikaros and Aiolos Inhibit Pre-B-Cell Proliferation by Directly Suppressing c-Myc Expression. Mol Cell Biol 30 17 4149 4158 .
  19. 19. Mancini M. Scappaticci D. Cimino G. Nanni M. Derme V. Elia L. Tafuri A. Vignetti M. Vitale A. Cuneo A. et al. 2005 A comprehensive genetic classification of adult acute lymphoblastic leukemia (ALL): analysis of the GIMEMA 0496 protocol. Blood 105 9 3434 3441 .
  20. 20. Martinelli G. Iacobucci I. Storlazzi C. T. Vignetti M. Paoloni F. Cilloni D. Soverini S. Vitale A. Chiaretti S. Cimino G. et al. 2009 IKZF1 (Ikaros) Deletions in BCR-ABL1 Positive Acute Lymphoblastic Leukemia Are Associated With Short Disease-Free Survival and High Rate of Cumulative Incidence of Relapse: A GIMEMA AL WP Report. Journal of Clinical Oncology 27 31 5202 5207 .
  21. 21. Matulic M. Paradzik M. Cicin-Sain L. Kapitanovic S. Dubravcic K. Batinic D. Antica M. 2009 Ikaros family transcription factors in chronic and acute leukemia. American Journal of Hematology 84 6 375 377 .
  22. 22. Morgan B. 1997 Aiolos, a lymphoid restricted transcription factor that interacts with Ikaros to regulate lymphocyte differentiation. EMBO J 16 2004 2013 .
  23. 23. Mullighan C. G. Miller C. B. Radtke I. Phillips L. A. Dalton J. Ma J. White D. Hughes T. P. Le Beau M. M. Pui C.H. etal 2008 BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature 453 7191 110 114 .
  24. 24. Nakase K. Ishimaru F. Avitahl N. Dansako H. Matsuo K. Fujii K. Sezaki N. Nakayama H. Yano T. Fukuda S. et al. 2000 Dominant Negative Isoform of the Ikaros Gene in Patients with Adult B-Cell Acute Lymphoblastic Leukemia. Cancer Res 60 15 4062 4065 .
  25. 25. Nakase K. Ishimaru F. Fujii K. Tabayashi T. Kozuka T. Sezaki N. Matsuo Y. Harada M. 2002 Overexpression of novel short isoforms of Helios in a patient with T-cell acute lymphoblastic leukemia. Experimental Hematology 30 4 313 317 .
  26. 26. Nakayama H. Ishimaru F. Katayama Y. Nakase K. Sezaki N. Takenaka K. Shinagawa K. Ikeda K. Niiya K. Harada M. 2000 Ikaros expression in human hematopoietic lineages. Experimental Hematology 28 11 1232 1238 .
  27. 27. Nichogiannopoulou N. Trevisan M. Naben S. Friedrich C. Georgopoulos K. 1999 Defects in the activity of hemopoietic stem cells in Ikaros mutant mice. J Exp Med 190 1201 1214 .
  28. 28. Rebollo A. Schmitt C. 2003 Ikaros, Aiolos and Helios: Transcription regulators and lymphoid malignancies. Immunology and Cell Biology 81 3 171 175 .
  29. 29. Richie Ehrlich. L. I. Serwold T. Weissman I. L. 2011 In vitro assays misrepresent in vivo lineage potentials of murine lymphoid progenitors. Blood 117 9 2618 2624 .
  30. 30. Sahoo D. Seita J. Bhattacharya D. Inlay M. A. Weissman I. L. Plevritis S. K. Dill D. L. 2010 A method of mining developmentally regulated genes using Boolean implications. Proceedings of the National Academy of Sciences 107 13 5732 5737 .
  31. 31. Schmitt T. M. Zúńiga-Pflücker J. C. 2002 Induction of T Cell Development from Hematopoietic Progenitor Cells by Delta-like-1 In Vitro. Immunity 17 6 749 756 .
  32. 32. Spangrude G. J. Heimfeld S. Weissman I. L. 1988 Purification and characterization of mouse hematopoietic stem cell. Science 241 58 92 .
  33. 33. Sun L. Liu A. Georgopoulos K. 1996 Zinc finger-mediated protein interactions modulate Ikaros activity, a molecular control of lymphocyte development. EMBO J 15 5358 5369 .
  34. 34. Till J. E. Mc Culloch C. E. 1961 A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiation Research 14 213 222 .
  35. 35. Trageser D. Iacobucci I. Nahar R. Duy C. von Levetzow. G. Klemm L. Park E. Schuh W. Gruber T. Herzog S. et al. 2009 Pre B cell receptor mediated cell cycle arrest in Philadelphia chromosome positive acute lymphoblastic leukemia requires IKAROS function. The Journal of Experimental Medicine 206 8 1739 1753 .
  36. 36. Wang J. H. Avitahl N. Cariappa A. Friedrich C. Ikeda T. Renold A. Andrikopoulos K. Liang L. Pillai S. Morgan B. A. et al. 1998 Aiolos Regulates B Cell Activation and Maturation to Effector State. Immunity 9 4 543 553 .
  37. 37. Winandy S. Wu P. Georgopoulos K. 1995 A dominant mutation in the Ikaros gene leads to rapid development of leukemia and lymphoma. Cell 83 2 289 299 .
  38. 38. Wu A. M. Siminovitch L. Till J. E. Mc Culloch E. A. 1968 Evidence for a relationship between mouse hemopoietic stem cells and cells forming colonies in culture. Proceedings of the National Academy of Sciences 59 1209 1215 .
  39. 39. Wu L. 1991 CD4 expressed on earliest T-lineage precursor cells in the adult murine thymus. Nature 349 71 74 .
  40. 40. Wu L. Antica M. Johnson G. R. Scollay R. Shortman K. 1991 Developmental potential of the earliest precursor cells from the adult mouse thymus. J Exp Med 174 6 1617 1627 .

Written By

Mariastefania Antica

Submitted: 04 November 2010 Published: 22 December 2011