全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Microenvironment-Centred Dynamics in Aggressive B-Cell Lymphomas

DOI: 10.1155/2012/138079

Full-Text   Cite this paper   Add to My Lib

Abstract:

Aggressive B-cell lymphomas share high proliferative and invasive attitudes and dismal prognosis despite heterogeneous biological features. In the interchained sequence of events leading to cancer progression, neoplastic clone-intrinsic molecular events play a major role. Nevertheless, microenvironment-related cues have progressively come into focus as true determinants for this process. The cancer-associated microenvironment is a complex network of nonneoplastic immune and stromal cells embedded in extracellular components, giving rise to a multifarious crosstalk with neoplastic cells towards the induction of a supportive milieu. The immunological and stromal microenvironments have been classically regarded as essential partners of indolent lymphomas, while considered mainly negligible in the setting of aggressive B-cell lymphomas that, by their nature, are less reliant on external stimuli. By this paper we try to delineate the cardinal microenvironment-centred dynamics exerting an influence over lymphoid clone progression in aggressive B-cell lymphomas. 1. Introduction B-cell malignancies represent a heterogeneous group of diseases characterized by different biological features and clinical behaviour, the latter ranging from indolent to highly aggressive. As for most neoplasms, the natural course of B-cell malignancies is characterized by tumour progression, featured by a flow of events leading to the enhancement of proliferative and invasive capabilities, towards the establishment of a more aggressive phenotype. Even if most of the processes involved in cancer progression are inherent to the neoplastic clone, this event is, actually, the result of an articulated mechanism, which seems to require the constant crosstalk between neoplastic cells and the faulty surrounding microenvironment. An ever-increasing amount of evidences suggest that this bijective relationship is a prime determinant of cancer natural history and evolution. Much has been so far discovered about the role of tumour intrinsic mechanisms of neoplastic progression, and the focus of research has been progressively shifting toward the study of microenvironment-centred dynamics. Cancer-associated microenvironment represents a multifaceted entity, which not only provides structural support to neoplastic cells (proper stroma) but also acts as a “fertile soil” that, through humoral factors (bioactive molecules such as cytokines, chemokines, and adhesion molecules), nonmalignant cellular elements of the stroma (fibroblasts and endothelial cells) and the immune system (macrophages, mast

References

[1]  A. Zucchetto, D. Benedetti, C. Tripodo et al., “CD38/CD31, the CCL3 and CCL4 chemokines, and CD49d/vascular cell adhesion molecule-1 are interchained bysequential events sustaining chronic lymphocytic leukemia cell survival,” Cancer Research, vol. 69, no. 9, pp. 4001–4009, 2009.
[2]  O. Tournilhac, D. D. Santos, L. Xu et al., “Mast cells in Waldenstrom's macroglobulinemia support lymphoplasmacytic cell growth through CD154/CD40 signaling,” Annals of Oncology, vol. 17, no. 8, pp. 1275–1282, 2006.
[3]  A. Carbone, A. Gloghini, A. Cabras, and G. Elia, “Differentiating germinal center-derived lymphomas through their cellular microenvironment,” American Journal of Hematology, vol. 84, no. 7, pp. 435–438, 2009.
[4]  J. Carreras, A. Lopez-Guillermo, B. C. Fox et al., “High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma,” Blood, vol. 108, no. 9, pp. 2957–2964, 2006.
[5]  C. Tripodo, S. Sangaletti, P. P. Piccaluga et al., “The bone marrow stroma in hematological neoplasms-a guilty bystander,” Nature Reviews Clinical Oncology, vol. 8, no. 8, pp. 456–466, 2011.
[6]  A. M. Florena, C. Tripodo, R. Porcasi et al., “Immunophenotypic profile and role of adhesion molecules in splenic marginal zone lymphoma with bone marrow involvement,” Leukemia and Lymphoma, vol. 47, no. 1, pp. 49–57, 2006.
[7]  L. Trentin, A. Cabrelle, M. Facco et al., “Homeostatic chemokines drive migration of malignant B cells in patients with non-Hodgkin lymphomas,” Blood, vol. 104, no. 2, pp. 502–508, 2004.
[8]  D. de Jong and G. Enblad, “Inflammatory cells and immune microenvironment in malignant lymphoma,” Journal of Internal Medicine, vol. 264, no. 6, pp. 528–536, 2008.
[9]  J. Delabie, E. Vandenberghe, C. Kennes et al., “Histiocyte-rich B-cell lymphoma: a distinct clinicopathologic entity possibly related to lymphocyte predominant Hodgkin's disease, paragranuloma subtype,” American Journal of Surgical Pathology, vol. 16, no. 1, pp. 37–48, 1992.
[10]  P. P. Piccaluga, C. Agostinelli, A. Gazzola et al., “Pathobiology of Hodgkin lymphoma,” Advances in Hematology, vol. 2011, Article ID 920898, 18 pages, 2011.
[11]  D. Aldinucci, A. Gloghini, A. Pinto, R. de Filippi, and A. Carbone, “The classical Hodgkin's lymphoma microenvironment and its role in promoting tumour growth and immune escape,” Journal of Pathology, vol. 221, no. 3, pp. 248–263, 2010.
[12]  C. Steidl, T. Lee, S. P. Shah et al., “Tumor-associated macrophages and survival in classic Hodgkin's lymphoma,” The New England Journal of Medicine, vol. 362, no. 10, pp. 875–885, 2010.
[13]  C. Brigati, D. M. Noonan, A. Albini, and R. Benelli, “Tumors and inflammatory infiltrates: friends or foes?” Clinical and Experimental Metastasis, vol. 19, no. 3, pp. 247–258, 2002.
[14]  P. van Loo, T. Tousseyn, V. Vanhentenrijk et al., “T-cell/histiocyte-rich large B-cell lymphoma shows transcriptional features suggestive of a tolerogenic host immune response,” Haematologica, vol. 95, no. 3, pp. 440–448, 2010.
[15]  C. A. Ogden, J. D. Pound, B. K. Batth et al., “Enhanced apoptotic cell clearance capacity and B cell survival factor production by IL-10-activated macrophages: implications for Burkitt's lymphoma,” Journal of Immunology, vol. 174, no. 5, pp. 3015–3023, 2005.
[16]  C. G. Mueller, C. Boix, W. H. Kwan et al., “Critical role of monocytes to support normal B cell and diffuse large B cell lymphoma survival and proliferation,” Journal of Leukocyte Biology, vol. 82, no. 3, pp. 567–575, 2007.
[17]  R. Küppers, “Mechanisms of B-cell lymphoma pathogenesis,” Nature Reviews Cancer, vol. 5, no. 4, pp. 251–262, 2005.
[18]  B. Treanor, D. Depoil, A. Gonzalez-Granja et al., “The membrane skeleton controls diffusion dynamics and signaling through the B cell receptor,” Immunity, vol. 32, no. 2, pp. 187–199, 2010.
[19]  L. Chen, S. Monti, P. Juszczynski et al., “SYK-dependent tonic B-cell receptor signaling is a rational treatment target in diffuse large B-cell lymphoma,” Blood, vol. 111, no. 4, pp. 2230–2237, 2008.
[20]  R. E. Davis, V. N. Ngo, G. Lenz et al., “Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma,” Nature, vol. 463, no. 7277, pp. 88–92, 2010.
[21]  M. Buchner, C. Baer, G. Prinz et al., “Spleen tyrosine kinase inhibition prevents chemokine- and integrin-mediated stromal protective effects in chronic lymphocytic leukemia,” Blood, vol. 115, no. 22, pp. 4497–4506, 2010.
[22]  W. W. Lin and M. Karin, “A cytokine-mediated link between innate immunity, inflammation, and cancer,” Journal of Clinical Investigation, vol. 117, no. 5, pp. 1175–1183, 2007.
[23]  P. Cheng, J. Zhou, and D. Gabrilovich, “Regulation of dendritic cell differentiation and function by Notch and Wnt pathways,” Immunological Reviews, vol. 234, no. 1, pp. 105–119, 2010.
[24]  P. Serafini, S. Mgebroff, K. Noonan, and I. Borrello, “Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells,” Cancer Research, vol. 68, no. 13, pp. 5439–5449, 2008.
[25]  G. Gallina, L. Dolcetti, P. Serafini et al., “Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells,” Journal of Clinical Investigation, vol. 116, no. 10, pp. 2777–2790, 2006.
[26]  K. C. Chang, G. C. Huang, D. Jones, and Y. H. Lin, “Distribution patterns of dendritic cells and T cells in diffuse large B-cell lymphomas correlate with prognoses,” Clinical Cancer Research, vol. 13, no. 22, part 1, pp. 6666–6672, 2007.
[27]  J. J. F. Muris, C. J. L. M. Meijer, S. A. G. M. Cillessen et al., “Prognostic significance of activated cytotoxic T-lymhocytes in primary nodal diffuse large B-cell lymphomas,” Leukemia, vol. 18, no. 3, pp. 589–596, 2004.
[28]  A. Tzankov, C. Meier, P. Hirschmann, P. Went, S. A. Pileri, and S. Dirnhofer, “Correlation of high numbers of intratumoral FOXP3+ regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin's lymphoma,” Haematologica, vol. 93, no. 2, pp. 193–200, 2008.
[29]  T. álvaro-Naranjo, M. Lejeune, M. T. Salvadó et al., “Immunohistochemical patterns of reactive microenvironment are associated with clinicobiologic behavior in follicular lymphoma patients,” Journal of Clinical Oncology, vol. 24, no. 34, pp. 5350–5357, 2006.
[30]  N. R. Lee, E. K. Song, K. Y. Jang et al., “Prognostic impact of tumor infiltrating FOXP3 positive regulatory T cells in diffuse large B-cell lymphoma at diagnosis,” Leukemia and Lymphoma, vol. 49, no. 2, pp. 247–256, 2008.
[31]  S. Hasselblom, M. Sigurdadottir, U. Hansson, H. Nilsson-Ehle, B. Ridell, and P. O. Andersson, “The number of tumour-infiltrating TIA-1+ cytotoxic T cells but not FOXP3+ regulatory T cells predicts outcome in diffuse large B-cell lymphoma,” British Journal of Haematology, vol. 137, no. 4, pp. 364–373, 2007.
[32]  S. Piconese, P. Pittoni, A. Burocchi et al., “A non-redundant role for OX40 in the competitive fitness of Treg in response to IL-2,” European Journal of Immunology, vol. 40, no. 10, pp. 2902–2913, 2010.
[33]  C. Tripodo, G. Gri, P. P. Piccaluga et al., “Mast cells and Th17 cells contribute to the lymphoma-associated pro-inflammatory microenvironment of angioimmunoblastic T-cell lymphoma,” American Journal of Pathology, vol. 177, no. 2, pp. 792–802, 2010.
[34]  A. Doganci, T. Eigenbrod, N. Krug et al., “The IL-6R α chain controls lung CD4+CD25+ Treg development and function during allergic airway inflammation in vivo,” Journal of Clinical Investigation, vol. 115, no. 2, pp. 313–325, 2005.
[35]  S. Wan, C. Xia, and L. Morel, “IL-6 produced by dendritic cells from lupus-prone mice inhibits CD4+CD25+ T cell regulatory functions,” Journal of Immunology, vol. 178, no. 1, pp. 271–279, 2007.
[36]  X. Valencia, G. Stephens, R. Goldbach-Mansky, M. Wilson, E. M. Shevach, and P. E. Lipsky, “TNF downmodulates the function of human CD4+CD25hi T-regulatory cells,” Blood, vol. 108, no. 1, pp. 253–261, 2006.
[37]  D. Gratzinger, S. Zhao, R. J. Marinelli et al., “Microvessel density and expression of vascular endothelial growth factor and its receptors in diffuse large B-cell lymphoma subtypes,” American Journal of Pathology, vol. 170, no. 4, pp. 1362–1369, 2007.
[38]  A. Potti, A. K. Ganti, S. Kargas, and M. Koch, “Immunohistochemical detection of C-kit (CD117) and vascular endothelial growth factor (VEGF) overexpression in mantle cell lymphoma,” Anticancer Research, vol. 22, no. 5, pp. 2899–2901, 2002.
[39]  S. M. Smith, J. Anastasi, K. S. Cohen, and L. A. Godley, “The impact of MYC expression in lymphoma biology: beyond Burkitt lymphoma,” Blood Cells, Molecules, and Diseases, vol. 45, no. 4, pp. 317–323, 2010.
[40]  A. Tzankov, S. Heiss, S. Ebner et al., “Angiogenesis in nodal B cell lymphomas: a high throughput study,” Journal of Clinical Pathology, vol. 60, no. 5, pp. 476–482, 2007.
[41]  J. Ruan, K. Hajjar, S. Rafii, and J. P. Leonard, “Angiogenesis and antiangiogenic therapy in non-Hodgkin's lymphoma,” Annals of Oncology, vol. 20, no. 3, pp. 413–424, 2009.
[42]  T. Takahashi, H. Ueno, and M. Shibuya, “VEGF activates protein kinase C-dependent, but Ras-independent Raf-MEK-MAP kinase pathway for DNA synthesis in primary endothelial cells,” Oncogene, vol. 18, no. 13, pp. 2221–2230, 1999.
[43]  B. Barleon, S. Sozzani, D. Zhou, H. A. Weich, A. Mantovani, and D. Marmé, “Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1,” Blood, vol. 87, no. 8, pp. 3336–3343, 1996.
[44]  B. L. Gruber, M. J. Marchese, and R. Kew, “Angiogenic factors stimulate mast-cell migration,” Blood, vol. 86, no. 7, pp. 2488–2493, 1995.
[45]  C. Sunderkotter, K. Steinbrink, M. Goebeler, R. Bhardwaj, and C. Sorg, “Macrophages and angiogenesis,” Journal of Leukocyte Biology, vol. 55, no. 3, pp. 410–422, 1994.
[46]  D. Ribatti, A. Vacca, A. Marzullo et al., “Angiogenesis and mast cell density with tryptase activity increase simultaneously with pathological progression in B-cell non-Hodgkin's lymphomas,” International Journal of Cancer, vol. 85, no. 2, pp. 171–175, 2000.
[47]  D. Ribatti, A. Vacca, B. Nico, E. Crivellato, L. Roncali, and F. Dammacco, “The role of mast cells in tumour angiogenesis,” British Journal of Haematology, vol. 115, no. 3, pp. 514–521, 2001.
[48]  S. Rafii, D. Lyden, R. Benezra, K. Hattori, and B. Heissig, “Vascular and haematopoietic stem cells: novel targets for anti-angiogenesis therapy?” Nature Reviews Cancer, vol. 2, no. 11, pp. 826–835, 2002.
[49]  M. Grunewald, I. Avraham, Y. Dor et al., “VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells,” Cell, vol. 124, no. 1, pp. 175–189, 2006.
[50]  D. Gratzinger, S. Zhao, R. J. Tibshirani et al., “Prognostic significance of VEGF, VEGF receptors, and microvessel density in diffuse large B cell lymphoma treated with anthracycline-based chemotherapy,” Laboratory Investigation, vol. 88, no. 1, pp. 38–47, 2008.
[51]  D. Ribatti, A. Vacca, B. Nico, M. Fanelli, L. Roncali, and F. Dammacco, “Angiogenesis spectrum in the stroma of B-cell non-Hodgkin's lymphomas. An immunohistochemical and ultrastructural study,” European Journal of Haematology, vol. 56, no. 1-2, pp. 45–53, 1996.
[52]  O. Bairey, Y. Zimra, E. Kaganovsky, M. Shaklai, E. Okon, and E. Rabizadeh, “Microvessel density in chemosensitive and chemoresistant diffuse large B-cell lymphomas,” Medical Oncology, vol. 17, no. 4, pp. 314–318, 2000.
[53]  K. N. Ganjoo, A. M. Moore, A. Orazi, J. A. Sen, C. S. Johnson, and C. S. An, “The importance of angiogenesis markers in the outcome of patients with diffuse large B cell lymphoma: a retrospective study of 97 patients,” Journal of Cancer Research and Clinical Oncology, vol. 134, no. 3, pp. 381–387, 2008.
[54]  T. M. Cardesa-Salzmann, L. Colomo, G. Gutierrez, et al., “High microvessel density determines a poor outcome in patients with diffuse large B-cell lymphoma treated with rituximab plus chemotherapy,” Haematologica, vol. 96, no. 7, pp. 996–1001, 2011.
[55]  T. A. Baudino, C. McKay, H. Pendeville-Samain et al., “c-Myc is essential for vasculogenesis and angiogenesis during development and tumor progression,” Genes and Development, vol. 16, no. 19, pp. 2530–2543, 2002.
[56]  A. Ruddell, P. Mezquita, K. A. Brandvold, A. Farr, and B. M. Iritani, “B lymphocyte-specific c-Myc expression stimulates early and functional expansion of the vasculature and lymphatics during lymphomagenesis,” American Journal of Pathology, vol. 163, no. 6, pp. 2233–2245, 2003.
[57]  U. E. Knies-Bamforth, S. B. Fox, R. Poulsom, G. I. Evan, and A. L. Harris, “c-Myc interacts with hypoxia to induce angiogenesis in vivo by a vascular endothelial growth factor-dependent mechanism,” Cancer Research, vol. 64, no. 18, pp. 6563–6570, 2004.
[58]  M. Baroudi, D. Corà, C. Bosia, M. Osella, and M. Caselle, “A curated database of miRNA mediated feed-forward loops involving MYC as master regulator,” PLoS ONE, vol. 6, no. 3, Article ID e14742, 2011.
[59]  J. Ruan, P. Martin, M. Coleman et al., “Durable responses with the metronomic rituximab and thalidomide plus prednisone, etoposide, procarbazine, and cyclophosphamide regimen in elderly patients with recurrent mantle cell lymphoma,” Cancer, vol. 116, no. 11, pp. 2655–2664, 2010.
[60]  S. Monestiroli, P. Mancuso, A. Burlini et al., “Kinetics and viability of circulating endothelial cells as surrogate angiogenesis marker in an animal model of human lymphoma,” Cancer Research, vol. 61, no. 11, pp. 4341–4344, 2001.
[61]  F. Bertolini, C. Dell'Agnola, P. Mancuso et al., “CXCR4 neutralization, a novel therapeutic approach for non-Hodgkin's lymphoma,” Cancer Research, vol. 62, no. 11, pp. 3106–3112, 2002.
[62]  B. Geiger, A. Bershadsky, R. Pankov, and K. M. Yamada, “Transmembrane extracellular matrix-cytoskeleton crosstalk,” Nature Reviews Molecular Cell Biology, vol. 2, no. 11, pp. 793–805, 2001.
[63]  A. Bergamaschi, E. Tagliabue, T. S?rlie et al., “Extracellular matrix signature identifies breast cancer subgroups with different clinical outcome,” Journal of Pathology, vol. 214, no. 3, pp. 357–367, 2008.
[64]  M. R. Ng and J. S. Brugge, “A stiff blow from the stroma: collagen crosslinking drives tumor progression,” Cancer Cell, vol. 16, no. 6, pp. 455–457, 2009.
[65]  G. Lenz, G. Wright, S. S. Dave et al., “Stromal gene signatures in large-B-cell lymphomas,” The New England Journal of Medicine, vol. 359, no. 22, pp. 2313–2323, 2008.
[66]  H. W. Tun, D. Personett, K. A. Baskerville et al., “Pathway analysis of primary central nervous system lymphoma,” Blood, vol. 111, no. 6, pp. 3200–3210, 2008.
[67]  A. Rosenwald, G. Wright, W. C. Chan et al., “The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma,” The New England Journal of Medicine, vol. 346, no. 25, pp. 1937–1947, 2002.
[68]  T. R. Kyriakides and P. Bornstein, “Matricellular proteins as modulators of wound healing and the foreign body response,” Thrombosis and Haemostasis, vol. 90, no. 6, pp. 986–992, 2003.
[69]  S. Sangaletti and M. P. Colombo, “Matricellular proteins at the crossroad of inflammation and cancer,” Cancer Letters, vol. 267, no. 2, pp. 245–253, 2008.
[70]  S. M. Pupa, S. Ménard, S. Forti, and E. Tagliabue, “New insights into the role of extracellular matrix during tumor onset and progression,” Journal of Cellular Physiology, vol. 192, no. 3, pp. 259–267, 2002.
[71]  P. Y. Wai and P. C. Kuo, “Osteopontin: regulation in tumor metastasis,” Cancer and Metastasis Reviews, vol. 27, no. 1, pp. 103–118, 2008.
[72]  A. F. Chambers, S. M. Wilson, N. Kerkvliet, F. P. O'Malley, J. F. Harris, and A. G. Casson, “Osteopontin expression in lung cancer,” Lung Cancer, vol. 15, no. 3, pp. 311–323, 1996.
[73]  K. A. Furger, R. K. Menon, A. B. Tuck, V. H. C. Bramwel, and A. F. Chambers, “The functional and clinical roles of osteopontin in cancer and metastasis,” Current Molecular Medicine, vol. 1, no. 5, pp. 621–632, 2001.
[74]  M. K. El-Tanani, “Role of osteopontin in cellular signaling and metastatic phenotype,” Frontiers in Bioscience, vol. 1, no. 13, pp. 4276–4284, 2008.
[75]  C. Chiodoni, M. P. Colombo, and S. Sangaletti, “Matricellular proteins: from homeostasis to inflammation, cancer, and metastasis,” Cancer and Metastasis Reviews, vol. 29, no. 2, pp. 295–307, 2010.
[76]  Y. H. Lin and H. F. Yang-Yen, “The osteopontin-CD44 survival signal involves activation of the phosphatidylinositol 3-kinase/Akt signaling pathway,” Journal of Biological Chemistry, vol. 276, no. 49, pp. 46024–46030, 2001.
[77]  R. S. Hauptschein, K. E. Sloan, C. Torella et al., “Functional proteomic screen identifies a modulating role for CD44 in death receptor-mediated apoptosis,” Cancer Research, vol. 65, no. 5, pp. 1887–1896, 2005.
[78]  S. A. Khan, A. C. Cook, M. Kappil et al., “Enhanced cell surface CD44 variant (v6, v9) expression by osteopontin in breast cancer epithelial cells facilitates tumor cell migration: novel post-transcriptional, post-translational regulation,” Clinical and Experimental Metastasis, vol. 22, no. 8, pp. 663–673, 2005.
[79]  A. Tzankov, A. C. Pehrs, A. Zimpfer et al., “Prognostic significance of CD44 expression in diffuse large B cell lymphoma of activated and germinal centre B cell-like types: a tissue microarray analysis of 90 cases,” Journal of Clinical Pathology, vol. 56, no. 10, pp. 747–752, 2003.
[80]  A. Mantovani, P. Allavena, A. Sica, and F. Balkwill, “Cancer-related inflammation,” Nature, vol. 454, no. 7203, pp. 436–444, 2008.
[81]  A. D. Bradshaw and E. H. Sage, “SPARC, a matricellular protein that functions in cellular differentiation and tissue response to injury,” Journal of Clinical Investigation, vol. 107, no. 9, pp. 1049–1054, 2001.
[82]  V. H. C. Bramwell, A. B. Tuck, S. M. Wilson et al., “Expression of osteopontin and HGF/met in adult soft tissue tumors,” Cancer Biology and Therapy, vol. 4, no. 12, pp. 1336–1341, 2005.
[83]  A. B. Tuck, C. Hota, S. M. Wilson, and A. F. Chambers, “Osteopontin-induced migration of human mammary epithelial cells involves activation of EGF receptor and multiple signal transduction pathways,” Oncogene, vol. 22, no. 8, pp. 1198–1205, 2003.
[84]  D. N. Haylock and S. K. Nilsson, “Osteopontin: a bridge between bone and blood,” British Journal of Haematology, vol. 134, no. 5, pp. 467–474, 2006.
[85]  A. D. Bradshaw, “The role of SPARC in extracellular matrix assembly,” Journal of Cell Communication and Signaling, vol. 3, no. 3-4, pp. 239–246, 2009.
[86]  J. R. Infante, H. Matsubayashi, N. Sato et al., “Peritumoral fibroblast SPARC expression and patient outcome with resectable pancreatic adenocarcinoma,” Journal of Clinical Oncology, vol. 25, no. 3, pp. 319–325, 2007.
[87]  O. L. Podhajcer, L. G. Benedetti, M. R. Girotti, F. Prada, E. Salvatierra, and A. S. Llera, “The role of the matricellular protein SPARC in the dynamic interaction between the tumor and the host,” Cancer and Metastasis Reviews, vol. 27, no. 4, pp. 691–705, 2008.
[88]  N. A. Said, A. A. Elmarakby, J. D. Imig, D. J. Fulton, and K. Motamed, “SPARC ameliorates ovarian cancer-associated inflammation,” Neoplasia, vol. 10, no. 10, pp. 1092–1104, 2008.
[89]  N. Said, I. Najwer, and K. Motamed, “Secreted protein acidic and rich in cysteine (SPARC) inhibits integrin-mediated adhesion and growth factor-dependent survival signaling in ovarian cancer,” American Journal of Pathology, vol. 170, no. 3, pp. 1054–1063, 2007.
[90]  P. P. Piccaluga, G. de Falco, M. Kustagi et al., “Gene expression analysis uncovers similarity and differences among Burkitt lymphoma subtypes,” Blood, vol. 117, no. 13, pp. 3596–3608, 2011.
[91]  S. Piconese, M. Costanza, C. Tripodo et al., “The matricellular protein SPARC supports follicular dendritic cell networking toward Th17 responses,” Journal of Autoimmunity, vol. 37, no. 4, pp. 300–310, 2011.
[92]  A. Vacca, R. Ria, M. Presta et al., “α(v)β(3) integrin engagement modulates cell adhesion, proliferation, and protease secretion in human lymphoid tumor cells,” Experimental Hematology, vol. 29, no. 8, pp. 993–1003, 2001.
[93]  U. Reuning, “Integrin αvβ3 promotes vitronectin gene expression in human ovarian cancer cells by implicating rel transcription factors,” Journal of Cellular Biochemistry, vol. 112, no. 7, pp. 1909–1919, 2011.
[94]  N. Ahmed, C. Riley, G. Rice, and M. Quinn, “Role of integrin receptors for fibronectin, collagen and laminin in the regulation of ovarian carcinoma functions in response to a matrix microenvironment,” Clinical and Experimental Metastasis, vol. 22, no. 5, pp. 391–402, 2005.
[95]  J. Wang, J. Wu, J. Hong et al., “PKC promotes the migration of colon cancer cells by regulating the internalization and recycling of integrin αvβ6,” Cancer Letters, vol. 311, no. 1, pp. 38–47, 2011.
[96]  J. J. Grzesiak, H. S. T. Cao, D. W. Burton et al., “Knockdown of the β(1) integrin subunit reduces primary tumor growth and inhibits pancreatic cancer metastasis,” International Journal of Cancer, vol. 129, no. 12, pp. 2905–2915, 2011.
[97]  I. Stamenkovic, “Extracellular matrix remodelling: the role of matrix metalloproteinases,” Journal of Pathology, vol. 200, no. 4, pp. 448–464, 2003.
[98]  S. Curran and G. I. Murray, “Matrix metalloproteinases in tumour invasion and metastasis,” Journal of Pathology, vol. 189, no. 3, pp. 300–308, 1999.
[99]  A. E. Kossakowska, A. Hinek, D. R. Edwards et al., “Proteolytic activity of human non-Hodgkin's lymphomas,” American Journal of Pathology, vol. 152, no. 2, pp. 565–576, 1998.
[100]  A. E. Kossakowska, D. R. Edwards, C. Prusinkiewicz et al., “Interleukin-6 regulation of matrix metalloproteinase (MMP-2 and MMP-9) and tissue inhibitor of metalloproteinase (TIMP-1) expression in malignant non-Hodgkin's lymphomas,” Blood, vol. 94, no. 6, pp. 2080–2089, 1999.
[101]  T. Sato, A. Ito, and Y. Mori, “Interleukin 6 enhances the production of tissue inhibitor of metalloproteinases (TIMP) but not that of matrix metalloproteinases by human fibroblasts,” Biochemical and Biophysical Research Communications, vol. 170, no. 2, pp. 824–829, 1990.
[102]  A. E. Kossakowska, S. J. Urbanski, and D. R. Edwards, “Tissue inhibitor of metalloproteinases-1 (TIMP-1) RNA is expressed at elevated levels in malignant non-Hodgkin's lymphomas,” Blood, vol. 77, no. 11, pp. 2475–2481, 1991.
[103]  L. Guedez, L. Courtemanch, and M. Stetler-Stevenson, “Tissue inhibitor of metalloproteinase (TIMP)-1 induces differentiation and an antiapoptotic phenotype in germinal center B cells,” Blood, vol. 92, no. 4, pp. 1342–1349, 1998.
[104]  J. W. Choi, J. S. An, J. H. Lee, E. S. Lee, K. H. Kim, and Y. S. Kim, “Clinicopathologic implications of tissue inhibitor of metalloproteinase-1- positive diffuse large B-cell lymphoma,” Modern Pathology, vol. 19, no. 7, pp. 963–973, 2006.
[105]  C. Fang, D. X. Zhu, H. J. Dong et al., “Serum microRNAs are promising novel biomarkers for diffuse large B cell lymphoma,” Annals of Hematology. In press.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413