#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Impact of Nestin Analysis in Multiple Myeloma


Authors: H. Šváchová 1,2;  L. Kovářová 1,4;  J. Štossová 1;  A. Potáčová 1;  L. Pour 1,3;  R. Hájek 1,3,4
Authors‘ workplace: Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic 1;  Department of Experimental Biology, Faculty of Science, Masaryk University 2;  Department of Internal Medicine – Hematooncology, University Hospital Brno, Czech Republic 3;  Laboratory of Experimental Hematology and Cell Immunotherapy, Department of Clinical Haematology, University Hospital Brno 4
Published in: Klin Onkol 2011; 24(Supplementum 1): 53-57

Overview

Nestin, a marker of multipotent precursor cells, is an important dynamic structure; its polymerization/depolymerization influences intracellular signaling and participates in key cell pro­cesses such as proliferation, migration and cell survival. It is presumed that nestin plays a central role in carcinogenesis. It is suggested that nestin might be a suitable diagnostic and prognostic indicator of malignancy and a potential marker of cancer stem cells. Unexpectedly, nestin has been identified in mature CD138+CD38+ plasma cells (PC) of multiple myeloma patients (MM). Expression of nestin, a marker of stem/progenitor cells, in malignant PC, that are considered to be terminally differentiated, indicates that nestin might play a unique role in pathology of MM.

Key words:
nestin – plasma cells – multiple myeloma – myeloma stem cells – myeloma-initiating cells – flow cytometry

This work was supported by research grants of The Ministry of Education, Youth and Sports: LC06027, MSM0021622434; research projects of IGA of The Ministry of Health: NS10406, NS10408, NT11154 and grants of The Czech Science Foundation GAP304/10/1395.

The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study.

The Editorial Board declares that the manuscript met the ICMJE “uniform requirements” for biomedical papers.

Introduction

Multiple myeloma (MM) is a hematological malignancy characterized by clonal expansion of malignant plasma cells (PC) in bone marrow, production of monoclonal immunoglobulin and osteolytic lesions [1]. Despite of vast advances in myeloma treatment, the disease is still incurable [2–4]. Short median survival of patients treated with conventional therapy (4–5 years) has led to initiate a new trend in prediction and prevention of progression to malignant stage of MM [5]. Benign stage of MM is represented by asymptomatic monoclonal gammopathy of undetermined significance (MGUS) that is associated with a 1% per year risk of progression to multiple myeloma (MM) or related disease [6]. However, current MGUS risk-stratification model for accurate and early assessment of malignant transformation risk is still insufficient, as described by Klincová et al in this supplementum of Klinická onkologie. New diagnostic and prognostic parameters and detailed characterization of population responsible for the origin, progression and sustainability of the disease are required.

Myeloma progenitors/myeloma--initiating cells (MIC) are supposed to be a minor population of slightly differentiated cells reminiscent of memory B-lymphocytes with surface markers CD19+CD27+CD138 [7]. However, Yaccoby & Epstein [8] proved that also dominant population of human CD45lowCD38highCD138+ PC is able to proliferate and produce tumor mass. More­over, myeloma PC show high phenotypic plasticity and are able to dedifferentiate and acquire autonomous survival properties under specific cultivation conditions [9]. The real origin of MIC is still unclear, although these data indicate that myeloma PC have ability to reprogram, reverse senescence and induce stem cell properties. This hypothesis is supported by our current results indicating that potential marker of MIC responsible for plasticity of mature PC and clinical relevant factor for MM might be nestin, a marker of stem/progenitor cells [10]. Furthermore, expression of stem/germ line cell markers, such as MAGE, KLF4, SOX2, CD117, has been already reported in monoclonal gammopathies [11–14]. Detailed characterization of MIC would enable identification of new prognostic markers for progression rate monitoring but also lead to targeted eradication of this population and delay progression to symptomatic disease. In the present work, we discuss the importance of nestin analysis for MM and describe methodological approaches for study of intracellular proteins in rare populations.

Neural Stem Cell Marker Nestin

Nestin, a class VI intermediate filament protein, was originally described as a neuronal stem cell marker during central nervous system (CNS) development [15] but currently shows a wider range of expression that has been previously thought [16–20]. Human nestin gene is located on the long arm of chromosome 1. Time- and site-specific expression of nestin is driven from promoter activated by Sp-1 transcription factor [21]. The protein has a high molecular weight (about 240 kDa), which differs among organs because of protein modifications [22]. Nestin is characterized by an α-helical central “rod” domain, typical for all intermediate filaments, short N-terminus and a very long C-terminus which could function as a linker or cross-bridge between intermediate filaments, microfilaments and microtubules [23]. Nestin is unable to self-assemble; therefore, it requires the presence of other IF proteins, such as vimentin, desmin or α-internexin [16,24,25]. Phosphorylation/dephosphorylation of nestin may modulate disassembly and assembly of intermediate filaments [26]. These processes might play a role during increased cytoplasmic trafficking in progenitor cells undergoing division or in migrating interphase cells [27–29]. Nestin was shown to participate in asymmetric redistribution of cytoskeletal proteins and other factors to daughter neuroepithelial cells [30]. Moreover, cytoprotective effect of nestin expression has been demonstrated in neuronal progenitor cells undergoing oxidative stress [31,32].

Nestin is expressed predominantly in rapidly dividing progenitor cells of embryonal and fetal tissue [17–18,33,34]. Upon differentiation, nestin becomes downregulated and is replaced by tissue-specific intermediate filament proteins but reappears transiently after injury of muscle or the CNS [34,35]. Nestin-positive cells were also found in adult tissues, such as CNS and skin where they are restricted to defined locations and may function as cellular “reserve” capable of proliferation, differentiation and migration [36,37,20]. Furthermore, increased nestin expression has been reported in various tumor cells, including CNS tumors, gastrointestinal stromal tumors, pancreatic cancer, prostate cancer, breast cancer, malignant melanoma, osteosarcomas, thyroid tumors and currently in PC of MM [10,33,38–42]. Nestin expression correlates with aggressive growth, metastasis, and poor prognosis in some tumors; it is considered to be a suitable diagnostic and prognostic indicator of malignant grade of tumors. Besides being a possible clinical marker, it has been recently shown that co-expression of nestin and CD133 might be a putative marker of cancer stem cells in neuroepithelial tumors [42–44]. Liu et al [42] first demonstrated gene expression of nestin in 5 MM patients and MM cell lines. Results referred to the exi­stence of CD56+ primary MM cells expressing neuronal markers, such as nestin, neuron-specific enolase and β-tubulin III. Despite of wide spectrum of nestin occurrence under normal and pathological conditions, its true biological role in cells is still unknown. Increasing importance of nestin analyses in solid tumors and missing information about expression of nestin in MM led us to evaluate nestin levels in PC ex vivo.

Methodology

Immunophenotypic studies were performed on bone marrow mononuclear cells (BMMNC) which were analyzed within 7 hours after sampling. BMMNC of MM patients and individuals without hemato­logical malignancy were isolated by Histopaque® 1077-1 (Sigma-Aldrich, Chemie GmbH, Steinheim, Germany), and stained with two-color combination of monoclonal antibodies for identification of PC – CD138-PE (clone: B-A38, EXBIO, Praha, Czech Rep.)/CD38-PE-Cy7 (Beckman Coulter, Marseille, France), based on manufacturer’s instructions. After surface staining, cells were fixed with 3% paraformaldehyde (Sigma-Aldrich) for 20 min at RT, washed with PBS and followed by intracellular staining with anti-human nestin-allophycocyanin (APC) conjugated monoclonal antibody (clone: 196908, RD Systems, Minneapolis, USA) in 0.1% Triton X-100 (USB, Cleveland, OH, USA) for 1h at RT. An isotypic control IgG1-APC (RD Systems) was used each time and applied as described above for anti-human nestin antibody. Data acquisition was performed on FACSCanto II flow cytometer (BD Biosciences, San Jose, CA, USA) using the BD FACSDiva 4.0 software (BD Biosciences); 104 events/tube were collected. Nestin expression was assessed as the percentage of PC showing positive nestin staining (Nes+PC), ratio of median fluorescence intensity (MFINes) and median fluorescence intensity of isotypic control (MFIIC) (Tab. 1).

1. Formula for calculating of relative levels of intracellular proteins in rare population.
Formula for calculating of relative levels of intracellular proteins in rare population.

Methodological Pitfalls in Multiple Myeloma

Proteomic analyses in MM are generally limited because of low amounts of cells. Therefore, analyses of intracellular protein levels of nestin needed to be optimized. As a suitable approach for analysis of nestin levels in PC of MM, we selected flow cytometry. This choice was made for three reasons:

  1. It is a well standardized method for analyses of cell surface markers and detection of intracellular light chains (kappa, lambda) in PC [45,46].
  2. It does not require high amounts of separated PC as other proteomic methods.
  3. Flow cytometry has already been used for nestin detection in human gliomas [47].

In this study, the intensity of nestin expression corresponded to the mean fluorescence intensity expressed as arbitrary relative linear fluorescence channel units scaled from 0 to 104 of the stained cells after subtracting the mean fluorescence intensity obtained for control unstained cells. However, this method did not consider non-specific binding of nestin antibody. Van Stijn et al [48] developed an accurate analysis of intracellular proteins in combination with extracellular antigens. The method enables detection and quantification of proteins in very small cell populations using isotypic control (IgGx-fluorochrom-conjugated monoclonal antibody intended for determination of non-specific staining in flow cytometric analysis; x – IgG subclass) (Tab. 1). According this method, we showed that nestin was heterogeneously expressed among MM patients (Fig. 1). Based on percentage of nestin-positive PC and relative nestin levels, significant differences were confirmed between MM patients and the control group without hematological malignancy but we did not find any statistically significant differences between newly diagnosed and relapsed patients based on our flow cytometry data [10].

Fig. 1. Heterogeneous nestin levels in CD138+38+ PC of MM patients. Representative results demonstrate diff erent shifts of fl uorescent intensity of entire PC population compared to isotypic control. Equivalent amounts of isotype control mouse IgG1-APC (tinted gray histograms) were assessed in parallel with anti-human nestin-APC (solid blue histograms).
Fig. 1. Heterogeneous nestin levels in CD138+38+ PC of MM patients. Representative results demonstrate diff erent shifts of fl uorescent intensity of entire PC population compared to isotypic control. Equivalent amounts of isotype control mouse IgG1-APC (tinted gray histograms) were assessed in parallel with anti-human nestin-APC (solid blue histograms).

The presence of nestin, a marker of multipotent proliferative cells, in malignant PC which are considered to be mature and terminally differentiated cells, is highly controversial. There are a few proposals for the explanation of nestin role in MM:

  1. Specific stimulus of damaged microenvironment may lead to activation of “stem cell” characteristics in more matured cell than was initially supposed [49]. Induction of stem cell gene expression might support survival or give some growth advantages to the PC subset [50,9].
  2. Dynamic character of nestin network plays a role in metastatic and migratory potential of solid tumors [51]. It is possible that nestin expression might be responsible for migration of PC to the extramedullary site.
  3. Considering the exceptional role of PC in the immune system, nestin might have another unknown function in malignant PC that might be associated with overproduction of abnormal proteins [41].

Conclusion

Nestin may become a useful diagnostic and prognostic marker for MM; therefore, it deserves further research. Our limited information about the role of nestin in MM requires clarifying its biological implication in the pathology of MM. As our data showed, flow cytometry might be a suitable tool for analyses of relative levels of nestin and simplify proteomic analysis of intracellular proteins. Clinical application of flow cytometry is currently increasing in research of monoclonal gammopathies and has become almost routine instrument for fast and easy screening of B-cell populations in monoclonal gammopathies.

Prof. MUDr. Roman Hájek, CSc.
Babak Myeloma Group
Department of Pathological Physiology
Faculty of Medicine
Masaryk University
Kamenice 5
625 00 Brno
Czech Republic
e-mail: r.hajek@fnbrno.cz


Sources

1. Billadeau D, Ahmann G, Greipp P et al. The bone marrow of multiple myeloma patients contains B cell populations at different stages of differentiation that are clonally related to the malignant plasma cell. J Exp Med 1993; 178(3): 1023–1031.

2. Hájek R, Maisnar V, Krejčí M. Thalidomid. Klin Farmakol Farm 2005; 19(1): 43–46.

3. Hájek R, Holánek M. Lenalidomid v léčbě mnohočetného myelomu. Farmakoterapie 2009; 2(5): 159–163.

4. Berenson JR, Yang HH, Sadler K et al. Phase I/II trial assessing bortezomib and melphalan combination therapy for the treatment of patients with relapsed or refractory multiple myeloma. J Clin Oncol 2006; 24(6): 937–944.

5. Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med 2004; 351(18): 1860–1873.

6. Kyle RA. New strategies for MGUS and smoldering multiple myeloma. Clin Adv Hematol Oncol 2004; 2(8): 507–509.

7. Matsui W, Huff CA, Wang Q et al. Characterization of clonogenic multiple myeloma cells. Blood 2004; 103(6): 2332–2336.

8. Yaccoby S, Epstein J. The proliferative potential of myeloma plasma cells manifest in the SCID-hu host. Blood 1999; 94(10): 3576–3582.

9. Yaccoby S. The phenotypic plasticity of myeloma plasma cells as expressed by dedifferentiation into an immature, resilient, and apoptosis-resistant phenotype. Clin Cancer Res 2005; 11(21): 7599–7606.

10. Svachova H, Pour L, Sana J et al. Stem cell marker nestin is expressed in plasma cells of multiple myeloma patients. Leuk Res 2011. Epub ahead of print.

11. Spisek R, Kukreja A, Chen LC et al. Frequent and specific immunity to the embryonal stem cell-associated antigen SOX2 in patients with monoclonal gammopathy. J Exp Med 2007; 204(4): 831–840.

12. Jungbluth AA, Ely S, DiLiberto M et al. The cancer-testis antigens CT7 (MAGE-C1) and MAGE-A3/6 are commonly expressed in multiple myeloma and correlate with plasma-cell proliferation. Blood 2005; 106(1): 167–174.

13. Zhu L, Somlo G, Zhou B et al. Fibroblast growth factor receptor 3 inhibition by short hairpin RNAs leads to apoptosis in multiple myeloma. Mol Cancer Ther 2005; 4(5): 787–798.

14. Nečasová J, Kadlecová J, Spěšná R et al. Význam sledování exprese nádorových testikulárních antigenů u mnohočetného myelomu.Klin Onkol 2008; 21 (Suppl 4): 223–225.

15. Hockfield S, McKay RD. Identification of major cell classes in the developing mammalian nervous system. J Neurosci 1985; 5(12): 3310–3328.

16. Sjöberg G, Jiang WQ, Ringertz NR et al. Colocalization of nestin and vimentin/desmin in skeletal muscle cells demonstrated by three-dimensional fluorescence digital imaging microscopy. Exp Cell Res 1994; 214(2): 447–458.

17. Walcott JC, Provis JM. Müller cells express the neuronal progenitor cell marker nestin in both differentiated and undifferentiated human foetal retina. Clin Experiment Ophthalmol 2003; 31(3): 246–249.

18. Sun XY, An J. Expression of nestin, an intermediate filament protein, in human fetal hepatic stem cells. Di Yi Jun Yi Da Xue Xue Bao 2004; 24(2): 207–209.

19. Vanderwinden JM, Gillard K, de Laet MH et al. Distribution of the intermediate filament nestin in the muscularis propria of the human gastrointestinal tract. Cell Tissue Res 2002; 309(2): 261–268.

20. Mayer EJ, Hughes EH, Carter DA et al. Nestin positive cells in adult human retina and in epiretinal membranes. Br J Ophthalmol 2003; 87(9): 1154–1158.

21. Lothian C, Prakash N, Lendahl U et al. Identification of both general and region-specific embryonic CNS enhancer elements in the nestin promoter. Exp Cell Res 1999; 248(2): 509–519.

22. Lobo MV, Arenas MI, Alonso FJ et al. Nestin, a neuroectodermal stem cell marker molecule, is expressed in Leydig cells of the human testis and in some specific cell types from human testicular tumours. Cell Tissue Res 2004; 316(3): 369–376.

23. Marvin MJ, Dahlstrand J, Lendahl U et al. A rod end deletion in the intermediate filament protein nestin alters its subcellular localization in neuroepithelial cells of transgenic mice. J Cell Sci 1998; 111 (Pt 14): 1951–1961.

24. Michalczyk K, Ziman M. Nestin structure and predicted function in cellular cytoskeletal organization. Histol Histopathol 2005; 20(2): 665–671.

25. Coulombe PA, Wong P. Cytoplasmic intermediate filaments revealed as dynamic and multipurpose scaffolds. Nat Cell Biol 2004; 6(8): 699–706.

26. Steinert PM, Chou YH, Prahlad V et al. A high molecular weight intermediate filament-associated protein in BHK-21 cells is nestin, a type VI intermediate filament protein. Limited co-assembly in vitro to form heteropolymers with type III vimentin and type IV alpha-internexin. J Biol Chem 1999; 274(14): 9881–9890.

27. Lendahl U, Zimmerman LB, McKay RD. CNS stem cells express a new class of intermediate filament protein. Cell 1990; 60(4): 585–595.

28. Kachinsky AM, Dominov JA, Miller JB. Intermediate filaments in cardiac myogenesis: nestin in the developing mouse heart. J Histochem Cytochem 1995; 43(8): 843–847.

29. Vaittinen S, Lukka R, Sahlgren C et al. The expression of intermediate filament protein nestin as related to vimentin and desmin in regenerating skeletal muscle. J Neuropathol Exp Neurol 2001; 60(6): 588–597.

30. Chou YH, Khuon S, Herrmann H et al. Nestin promotes the phosphorylation-dependent disassembly of vimentin intermediate filaments during mitosis. Mol Biol Cell 2003; 14(4): 1468–1478.

31. Sahlgren CM, Mikhailov A, Hellman J et al. Mitotic reorganization of the intermediate filament protein nestin involves phosphorylation by cdc2 kinase. J Biol Chem 2001; 276(19): 16456–16463.

32. Sahlgren CM, Pallari HM, He T et al. A nestin scaffold links Cdk5/p35 signaling to oxidant-induced cell death. EMBO J 2006; 25(20): 4808–4819.

33. Wiese C, Rolletschek A, Kania G et al. Nestin expression – a property of multi-lineage progenitor cells? Cell Mol Life Sci 2004; 61(19–20): 2510–2522.

34. About I, Laurent-Maquin D, Lendahl U et al. Nestin expression in embryonic and adult human teeth under normal and pathological conditions. Am J Pathol 2000; 157(1): 287–295.

35. Ha Y, Kim T, Yoon DH et al. Reinduced expression of developmental proteins (nestin, small heat shock protein) in and around cerebral arteriovenous malformations. Clin Neuropathol 2003; 22(5): 252–261.

36. Klein T, Ling Z, Heimberg H et al. Nestin is expressed in vascular endothelial cells in the adult human pancreas. J Histochem Cytochem 2003; 51(6): 697–706.

37. Gu H, Wang S, Messam CA et al. Distribution of nestin immunoreactivity in the normal adult human forebrain. Brain Res 2002; 943(2): 174–180.

38. Qiang L, Yang Y, Ma YJ et al. Isolation and characterization of cancer stem like cells in human glioblastoma cell lines. Cancer Lett 2009; 279(1): 13–21.

39. Veselska R, Kuglik P, Cejpek P et al. Nestin expression in the cell lines derived from glioblastoma multiforme. BMC Cancer 2006; 6: 32.

40. Krupkova O Jr, Loja T, Zambo I et al. Nestin expression in human tumors and tumor cell lines. Neoplasma 2010; 57(4): 291–298.

41. Yamada H, Takano T, Ito Y et al. Expression of nestin mRNA is a differentiation marker in thyroid tumors. Cancer Lett 2009; 280(1): 61–64.

42. Liu S, Otsuyama K, Ma Z e al. Induction of multilineage markers in human myeloma cells and their down-regulation by interleukin 6. Int J Hematol 2007; 85(1): 49–58.

43. Ignatova TN, Kukekov VG, Laywell ED et al. Human cortical glial tumors contain neural stem-like cells expressing astroglial and neuronal markers in vitro. Glia 2002; 39(3): 193–206.

44. Fang D, Nguyen TK, Leishear K et al. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res 2005; 65(20): 9328–9337.

45. Kovářová L, Buršová I, Suská R et al. Rozlišení nádorových klonálních a fyziologických polyklonálch plazmatických buněk pomocí flocytometrie. Klin Onkol 2008; 21 (Suppl 1): 254–257.

46. Kovarova L, Buresova I, Buchler T et al. Phenotype of plasma cells in multiple myeloma and monoclonal gammopathy of undetermined significance. Neoplasma 2009; 56(6): 526–532.

47. Balik V, Mirossay P, Bohus P et al. Flow cytometry analysis of neural differentiation markers expression in human glioblastomas may predict their response to chemotherapy. Cell Mol Neurobiol 2009; 29(6–7): 845–858.

48. van Stijn A, Kok A, van der Pol MA et al. Multiparameter flow cytometric quantification of apoptosis-related protein expression. Leukemia 2003; 17(4): 787–788.

49. Means AL, Meszoely IM, Suzuki K et al. Pancreatic epithelial plasticity mediated by acinar cell transdifferentiation and generation of nestin-positive intermediates. Development 2005; 132(16): 3767–3776.

50. Blau HM, Brazelton TR, Weimann JM. The evolving concept of a stem cell: entity or function? Cell 2001; 105(7): 829–841.

51. Kleeberger W, Bova GS, Nielsen ME et al. Roles for the stem cell associated intermediate filament Nestin in prostate cancer migration and metastasis. Cancer Res 2007; 67(19): 9199–1206.

Labels
Paediatric clinical oncology Surgery Clinical oncology
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#