1401
|
Borrello MG, Alberti L, Fischer A, Degl'innocenti D, Ferrario C, Gariboldi M, Marchesi F, Allavena P, Greco A, Collini P, Pilotti S, Cassinelli G, Bressan P, Fugazzola L, Mantovani A, Pierotti MA. Induction of a proinflammatory program in normal human thyrocytes by the RET/PTC1 oncogene. Proc Natl Acad Sci U S A 2005; 102:14825-30. [PMID: 16203990 PMCID: PMC1253545 DOI: 10.1073/pnas.0503039102] [Citation(s) in RCA: 246] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rearrangements of the RET receptor tyrosine kinase gene generating RET/PTC oncogenes are specific to papillary thyroid carcinoma (PTC), the most frequent thyroid tumor. Here, we show that the RET/PTC1 oncogene, when exogenously expressed in primary normal human thyrocytes, induces the expression of a large set of genes involved in inflammation and tumor invasion, including those encoding chemokines (CCL2, CCL20, CXCL8, and CXCL12), chemokine receptors (CXCR4), cytokines (IL1B, CSF-1, GM-CSF, and G-CSF), matrix-degrading enzymes (metalloproteases and urokinase-type plasminogen activator and its receptor), and adhesion molecules (L-selectin). This effect is strictly dependent on the presence of the RET/PTC1 Tyr-451 (corresponding to RET Tyr-1062 multidocking site). Selected relevant genes (CCL20, CCL2, CXCL8, CXCR4, L-selectin, GM-CSF, IL1B, MMP9, UPA, and SPP1/OPN) were found up-regulated also in clinical samples of PTC, particularly those characterized by RET/PTC activation, local extrathyroid spread, and lymph node metastases, when compared with normal thyroid tissue or follicular thyroid carcinoma. These results, demonstrating that the RET/PTC1 oncogene activates a proinflammatory program, provide a direct link between a transforming human oncogene, inflammation, and malignant behavior.
Collapse
Affiliation(s)
- Maria Grazia Borrello
- Departments of Experimental Oncology, Research Units 3 and 14, and Pathology, Istituto Nazionale Tumori, 20133 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1402
|
Hagemann T, Wilson J, Kulbe H, Li NF, Leinster DA, Charles K, Klemm F, Pukrop T, Binder C, Balkwill FR. Macrophages induce invasiveness of epithelial cancer cells via NF-kappa B and JNK. THE JOURNAL OF IMMUNOLOGY 2005; 175:1197-205. [PMID: 16002723 DOI: 10.4049/jimmunol.175.2.1197] [Citation(s) in RCA: 333] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages may influence tumor progression, angiogenesis and invasion. To investigate mechanisms by which macrophages interact with tumor cells, we developed an in vitro coculture model. Previously we reported that coculture enhanced invasiveness of the tumor cells in a TNF-alpha- and matrix metalloprotease-dependent manner. In this report, we studied intracellular signaling pathways and induction of inflammatory genes in malignant cells under the influence of macrophage coculture. We report that coculture of macrophages with ovarian or breast cancer cell lines led to TNF-alpha-dependent activation of JNK and NF-kappaB pathways in tumor cells, but not in benign immortalized epithelial cells. Tumor cells with increased JNK and NF-kappaB activity exhibited enhanced invasiveness. Inhibition of the NF-kappaB pathway by TNF-alpha neutralizing Abs, an NF-kappaB inhibitor, RNAi to RelA, or overexpression of IkappaB inhibited tumor cell invasiveness. Blockade of JNK also significantly reduced invasiveness, but blockade of p38 MAPK or p42 MAPK had no effect. Cocultured tumor cells were screened for the expression of 22 genes associated with inflammation and invasion that also contained an AP-1 and NF-kappaB binding site. EMMPRIN and MIF were up-regulated in cocultured tumor cells in a JNK- and NF-kappaB-dependent manner. Knocking down either MIF or EMMPRIN by RNAi in the tumor cells significantly reduced tumor cell invasiveness and matrix metalloprotease activity in the coculture supernatant. We conclude that TNF-alpha, via NF-kappaB, and JNK induces MIF and EMMPRIN in macrophage to tumor cell cocultures and this leads to increased invasive capacity of the tumor cells.
Collapse
Affiliation(s)
- Thorsten Hagemann
- Cancer Research U.K., Translational Oncology Laboratory, Barts and The London, Queen Mary's School of Medicine and Dentistry, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1403
|
An J, Rettig MB. Mechanism of von Hippel-Lindau protein-mediated suppression of nuclear factor kappa B activity. Mol Cell Biol 2005; 25:7546-56. [PMID: 16107702 PMCID: PMC1190288 DOI: 10.1128/mcb.25.17.7546-7556.2005] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Biallelic inactivating mutations of the von Hippel-Lindau tumor suppressor gene (VHL) are a hallmark of clear cell renal cell carcinoma (CCRCC), the most common histologic subtype of RCC. Biallelic VHL loss results in accumulation of hypoxia-inducible factor alpha (HIFalpha). Restoring expression of the wild-type protein encoded by VHL (pVHL) in tumors with biallelic VHL inactivation (VHL(-)(/)(-)) suppresses tumorigenesis, and pVHL-mediated degradation of HIFalpha is necessary and sufficient for VHL-mediated tumor suppression. The downstream targets of HIFalpha that promote renal carcinogenesis have not been completely elucidated. Recently, VHL loss was shown to activate nuclear factor kappa B (NF-kappaB), a family of transcription factors that promotes tumor growth. Here we show that VHL loss drives NF-kappaB activation by resulting in HIFalpha accumulation, which induces expression of transforming growth factor alpha, with consequent activation of an epidermal growth factor receptor/phosphatidylinositol-3-OH kinase/protein kinase B (AKT)/IkappaB-kinase alpha/NF-kappaB signaling cascade. We also show that components of this signaling pathway promote the growth of VHL(-)(/)(-) tumor cells. Members of this pathway represent viable drug targets in VHL(-)(/)(-) tumors, such as those associated with CCRCC.
Collapse
Affiliation(s)
- Jiabin An
- Division of Hematology/Oncology, Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System-West Los Angeles, CA 90073, USA
| | | |
Collapse
|
1404
|
Zijlstra A, Seandel M, Kupriyanova TA, Partridge JJ, Madsen MA, Hahn-Dantona EA, Quigley JP, Deryugina EI. Proangiogenic role of neutrophil-like inflammatory heterophils during neovascularization induced by growth factors and human tumor cells. Blood 2005; 107:317-27. [PMID: 16174763 PMCID: PMC1895349 DOI: 10.1182/blood-2005-04-1458] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A quantitative in vivo angiogenesis model employing collagen onplants placed on the chick embryo chorioallantoic membrane (CAM) has been used in this study to assess the spatial and temporal associations between neutrophil-like inflammatory cells, namely chicken heterophils, and the development of new blood vessels. Previously we have demonstrated that monocytes/macrophages infiltrating the onplants were associated with extracellular matrix remodeling and angiogenesis, in particular by delivering MMP-13 collagenase. By introducing chicken gelatinase B (chMMP-9) as a specific marker for heterophils, we now show that the onset and extent of angiogenesis induced by purified growth factors or by human HT-1080 fibrosarcoma cells correlated with the initial influx of chMMP-9-positive heterophils. This early heterophil arrival was followed by the infiltration of monocytes/macrophages and appeared to sustain further blood vessel formation. The disruption of inflammatory cell influx by 2 mechanistically distinct anti-inflammatory drugs, cortisone and ibuprofen, significantly inhibited angiogenesis, indicating a functional involvement of these inflammatory cells in new blood vessel development. A direct addition of isolated heterophils or purified chMMP-9 into the HT-1080 onplants engrafted into cortisone- or ibuprofen-treated embryos reversed the antiangiogenic effects of the drugs. The exogenously added heterophils induced in vivo a further infiltration of endogenous heterophils and monocytes and dramatically rescued the impaired angiogenesis, highlighting the importance of early inflammatory leukocytes in tumor-induced angiogenesis. Moreover, purified heterophils incorporated into onplants lacking growth factors or tumor cells induced angiogenesis in nontreated embryos, further indicating a direct proangiogenic role for neutrophil-like leukocytes.
Collapse
Affiliation(s)
- Andries Zijlstra
- Department of Cell Biology, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
1405
|
Farinha P, Masoudi H, Skinnider BF, Shumansky K, Spinelli JJ, Gill K, Klasa R, Voss N, Connors JM, Gascoyne RD. Analysis of multiple biomarkers shows that lymphoma-associated macrophage (LAM) content is an independent predictor of survival in follicular lymphoma (FL). Blood 2005; 106:2169-74. [PMID: 15933054 DOI: 10.1182/blood-2005-04-1565] [Citation(s) in RCA: 375] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
We studied the role of multiple biomarkers in determining outcome in follicular lymphoma (FL), concentrating in particular on the role of benign macrophages. The study group consisted of uniformly staged and treated patients with FL enrolled in a phase 2 trial between 1987 and 1993. All patients were younger than 61 years of age, had advanced-stage FL, and were treated with a multiagent chemotherapy regimen, BP-VACOP (bleomycin, cisplatin, etoposide, doxorubicin, cyclophosphamide, vincristine, and prednisone), followed by involved region radiation. The median follow-up of living patients was 12.5 years, and the median survival was 16.3 years. The International Prognostic Index (IPI) was predictive of overall survival (OS) (P = .003). Biopsy specimens from all cases were stained with an anti-CD68 antibody. Of the 99 evaluable patients with FL, 87 had less than 15 CD68+ macrophages/high-power field (hpf) (median, 7; range, 1-14) and 12 had more than 15 CD68+ macrophages/hpf (median, 20; range, 16-25) with a median OS of 16.3 vs 5.0 years, respectively (P < .001). A multivariate Cox model that included the IPI score, the histologic grade, and the lymphoma-associated macrophage (LAM) score, showed IPI and LAM to be independent predictors of OS (P = .009 and P = .004, respectively). The LAM content of FL predicts survival, and these data support a prominent role for nonneoplastic immune cells in the biology of FL. (Blood. 2005;106:2169-2174)
Collapse
Affiliation(s)
- Pedro Farinha
- Department of Pathology, BC Cancer Agency, 600 W 10th Ave, Vancouver, BC V5Z 4E6, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1406
|
Lewis C, Murdoch C. Macrophage responses to hypoxia: implications for tumor progression and anti-cancer therapies. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 167:627-35. [PMID: 16127144 PMCID: PMC1698733 DOI: 10.1016/s0002-9440(10)62038-x] [Citation(s) in RCA: 314] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/19/2005] [Indexed: 01/22/2023]
Abstract
The presence of multiple areas of hypoxia (low oxygen tension) is a hallmark feature of human and experimental tumors. Monocytes are continually recruited into tumors, differentiate into tumor-associated macrophages (TAMs), and then accumulate in these hypoxic areas. A number of recent studies have shown that macrophages respond to the levels of hypoxia found in tumors by up-regulating such transcription factors as hypoxia-inducible factors 1 and 2, which in turn activate a broad array of mitogenic, pro-invasive, pro-angiogenic, and pro-metastatic genes. This could explain why high numbers of TAMs correlate with poor prognosis in various forms of cancer. In this review, we assess the evidence for hypoxia activating a distinct, pro-tumor phenotype in macrophages and the possible effect of this on the growth, invasion, angiogenesis, and immune evasion of tumors. We also discuss current attempts to selectively target TAMs for destruction or to use them to deliver gene therapy specifically to hypoxic tumor sites.
Collapse
Affiliation(s)
- Claire Lewis
- Tumor Targeting Group, Academic Unit of Pathology, Division of Genomic Medicine, Sir Henry Wellcome Laboratories for Medical Research, University of Sheffield Medical School, Sheffield S10 2RX, UK.
| | | |
Collapse
|
1407
|
Stout RD, Jiang C, Matta B, Tietzel I, Watkins SK, Suttles J. Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. THE JOURNAL OF IMMUNOLOGY 2005; 175:342-9. [PMID: 15972667 DOI: 10.4049/jimmunol.175.1.342] [Citation(s) in RCA: 703] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Recent studies have described the development of distinct functional subsets of macrophages in association with cancer, autoimmune disease, and chronic infections. Based on the ability of Th1 vs Th2 cytokines to promote opposing activities in macrophages, it has been proposed that macrophages develop into either type 1 inflammatory or type 2 anti-inflammatory subsets. As an alternative to the concept of subset development, we propose that macrophages, in response to changes in their tissue environment, can reversibly and progressively change the pattern of functions that they express. As demonstrated herein, macrophages can reversibly shift their functional phenotype through a multitude of patterns in response to changes in cytokine environment. Macrophages display distinct functional patterns after treatment with IFN-gamma, IL-12, IL-4, or IL-10 and additional functional patterns are displayed depending on whether the cytokine is present alone or with other cytokines and whether the cytokines are added before or concomitantly with the activating stimulus (LPS). Sequential treatment of macrophages with multiple cytokines results in a progression through multiple functional phenotypes. This ability to adapt to changing cytokine environments has significant in vivo relevance, as evidenced by the demonstration that macrophage functional phenotypes established in vivo in aged or tumor-bearing mice can be altered by changing their microenvironment. A concept of functional adaptivity is proposed that has important implications for therapeutic targeting of macrophages in chronic diseases that result in the dominance of particular functional phenotypes of macrophages that play a significant role in disease pathology.
Collapse
Affiliation(s)
- Robert D Stout
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Health Sciences Center, Louisville, KY 40292, USA.
| | | | | | | | | | | |
Collapse
|
1408
|
Hamada K, Sasaki T, Koni PA, Natsui M, Kishimoto H, Sasaki J, Yajima N, Horie Y, Hasegawa G, Naito M, Miyazaki JI, Suda T, Itoh H, Nakao K, Mak TW, Nakano T, Suzuki A. The PTEN/PI3K pathway governs normal vascular development and tumor angiogenesis. Genes Dev 2005; 19:2054-65. [PMID: 16107612 PMCID: PMC1199575 DOI: 10.1101/gad.1308805] [Citation(s) in RCA: 231] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PTEN is an important tumor suppressor gene. Hereditary mutation of PTEN causes tumor-susceptibility diseases such as Cowden disease. We used the Cre-loxP system to generate an endothelial cell-specific mutation of Pten (Tie2CrePten) in mice. Tie2CrePten(flox/+) mice displayed enhanced tumorigenesis due to an increase in angiogenesis driven by vascular growth factors. This effect was partially dependent on the PI3K subunits p85alpha and p110gamma. In vitro, Tie2CrePten(flox/+) endothelial cells showed enhanced proliferation/migration. Tie2CrePten(flox/flox) mice died before embryonic day 11.5 (E11.5) due to bleeding and cardiac failure caused by impaired recruitment of pericytes and vascular smooth muscle cells to blood vessels, and of cardiomyocytes to the endocardium. These phenotypes depend strongly on p110gamma rather than on p85alpha and were associated with decreased expression of Ang-1, VCAM-1, connexin 40, and ephrinB2 but increased expression of Ang-2, VEGF-A, VEGFR1, and VEGFR2. Pten is thus indispensable for normal cardiovascular morphogenesis and post-natal angiogenesis, including tumor angiogenesis.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cells, Cultured
- Female
- Fetal Heart/embryology
- Fetal Heart/metabolism
- Genes, Tumor Suppressor
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Mutation
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/genetics
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- PTEN Phosphohydrolase
- Phosphatidylinositol 3-Kinases/deficiency
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/physiology
- Protein Tyrosine Phosphatases/deficiency
- Protein Tyrosine Phosphatases/genetics
- Protein Tyrosine Phosphatases/physiology
- RNA, Small Interfering/genetics
- Tumor Suppressor Proteins/deficiency
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/physiology
Collapse
Affiliation(s)
- Koichi Hamada
- Department of Molecular Biology, Department of Microbiology, Department of Gastroenterology, Akita University School of Medicine, Akita 010-8543, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1409
|
Abstract
Tissues maintain homeostasis by monitoring and responding to varied physical interactions between cells and their microenvironment. In situations where acute tissue damage occurs, such as wounding, pathogenic assault, or toxic exposure, regulatory circuits that monitor tissue homeostasis are rapidly engaged to initiate tissue repair by regulating cell polarity, proliferation and death, matrix metabolism, inflammation, and vascular and lymphatic function. The critical feature of regulating these acute responses is the innate ability to discriminate between homeostatic versus damaged tissue states and engage or disengage regulatory machinery as appropriate; thus, a major distinction between acute versus chronic disease is the altered ability to appropriately activate and?or inactivate reparative regulatory programs. Since cancer is a chronic disease characterized by altered cell polarity, enhanced cell survival, inflammation, increased matrix metabolism, and enhanced vascular and lymphatic function, considerable attention is now focused on understanding the cellular and molecular mechanisms regulating these responsive pathways. Since chemoattractant cytokines are important mediators of leukocyte recruitment following acute tissue stress, and demonstrate altered characteristics of expression and activation in chronically inflamed tissue, they have been implicated as key regulators of inflammation and angiogenesis during cancer development. This chapter focuses on the clinical and experimental data implicating proinflammatory cytokines and chemokines as important potentiators of carcinogenesis.
Collapse
Affiliation(s)
- Stephen C Robinson
- Cancer Research Institute, University of California, San Francisco, 94143, USA
| | | |
Collapse
|
1410
|
Toulza F, Eliaou JF, Pinet V. Breast tumor cell soluble factors induce monocytes to produce angiogenic but not angiostatic CXC chemokines. Int J Cancer 2005; 115:429-36. [PMID: 15688373 DOI: 10.1002/ijc.20705] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Tumor cells are known to interact closely with nontumoral infiltrating cells in order to grow and proliferate. Monocyte-derived cells constitute a major component of the tumoral infiltrate and a high level of these cells has been associated with increased tumor growth and poor prognosis in patients with breast cancer. For their growth and metastatic propagation, solid tumors are dependant on angiogenesis and accumulated evidences suggest that monocyte-derived cells could also play an important role in this phenomenon. However, the precise nature of proangiogenic factors secreted by these cells in breast carcinomas, and their direct influence on vessel formation, has not been determined. In the present study, we show that soluble factors secreted by breast tumor cells induce monocytes to produce a variety of proangiogenic CXC chemokines without secretion of angiostatic CXC chemokines. Using in vitro tubule formation in Matrigel, we demonstrated that the CXC chemokines secreted by MTSs (monocytes cultured with tumor cell supernatants) were able to induce microvessel formation. The profile of secreted CXC chemokines was characteristic for each tumor cell line or fresh tumor cells. This last result points out that a precise profiling of secreted proangiogenic factors inside the tumor, by tumor cells themselves or tumor-infiltrating monocyte-derived cells, is important for a precise targeting of therapeutic agents against neovascularization.
Collapse
|
1411
|
van Deventer HW, O'Connor W, Brickey WJ, Aris RM, Ting JPY, Serody JS. C-C chemokine receptor 5 on stromal cells promotes pulmonary metastasis. Cancer Res 2005; 65:3374-9. [PMID: 15833871 DOI: 10.1158/0008-5472.can-04-2616] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have shown that mice that express the C-C chemokine receptor 5 (CCR5) have enhanced local tumor growth and an impaired response to vaccine therapy compared with CCR5 knockout (CCR5(-/-)) mice. Here, we extend these observations to evaluate the function of CCR5 in pulmonary metastasis and the mechanism underlying the diminished tumor growth in CCR5(-/-) mice. Lung metastases were counted in wild-type (WT) and CCR5(-/-) mice following the injection of 1 x 10(6) B16-F10 melanoma cells. These results were compared with those from syngeneic bone marrow chimeric mice formed by the transfer of WT bone marrow into irradiated CCR5(-/-) and CCR5(-/-) marrow into irradiated WT mice. Intact CCR5(-/-) mice developed fewer metastases than WT mice (40.2 versus 70.6; P < 0.05). Bone marrow chimeras formed by the transfer of WT bone marrow into CCR5(-/-) hosts had fewer metastases than WT hosts injected with knockout marrow (46.6 versus 98.6; P < 0.01). Adoptive transfer of CCR5-expressing leukocytes also failed to promote metastasis in CCR5(-/-) mice. However, the i.v. transfer of WT pulmonary stromal cells into CCR5(-/-) mice increased the number of metastases compared with transfer of CCR5(-/-) stromal cells (102.8 versus 26.0; P < 0.05). These results show for the first time that CCR5 expression on stromal and not hematopoietic cells contributes to tumor metastasis. Therefore, recently developed CCR5 inhibitors may have a novel benefit in cancer therapy.
Collapse
MESH Headings
- Animals
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
- Immunotherapy, Adoptive
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/secondary
- Male
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Melanoma, Experimental/secondary
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, CCR5/biosynthesis
- Receptors, CCR5/deficiency
- Receptors, CCR5/physiology
- Stromal Cells/metabolism
Collapse
Affiliation(s)
- Hendrik W van Deventer
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Room 3009, Old Clinic Building, NC 27599, USA.
| | | | | | | | | | | |
Collapse
|
1412
|
Stout RD, Suttles J. Immunosenescence and macrophage functional plasticity: dysregulation of macrophage function by age-associated microenvironmental changes. Immunol Rev 2005; 205:60-71. [PMID: 15882345 PMCID: PMC1201508 DOI: 10.1111/j.0105-2896.2005.00260.x] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The macrophage lineage displays extreme functional and phenotypic heterogeneity, which appears to be because, in large part, of the ability of macrophages to functionally adapt to changes in their tissue microenvironment. This functional plasticity of macrophages plays a critical role in their ability to respond to tissue damage and/or infection and to contribute to clearance of damaged tissue and invading microorganisms, to recruitment of the adaptive immune system, and to resolution of the wound and of the immune response. Evidence has accumulated that environmental influences, such as stromal function and imbalances in hormones and cytokines, contribute significantly to the dysfunction of the adaptive immune system. The innate immune system also appears to be dysfunctional in aged animals and humans. In this review, the hypothesis is presented and discussed that the observed age-associated 'dysfunction' of macrophages is the result of their functional adaptation to the age-associated changes in tissue environments. The resultant loss of orchestration of the manifold functional capabilities of macrophages would undermine the efficacy of both the innate and adaptive immune systems. The macrophages appear to maintain functional plasticity during this dysregulation, making them a prime target of cytokine therapy that could enhance both innate and adaptive immune systems.
Collapse
Affiliation(s)
- Robert D Stout
- Department of Microbiology and Immunology, University of Louisville School of Medicine, KY 40292, USA.
| | | |
Collapse
|
1413
|
Valković T, Fuckar D, Stifter S, Matusan K, Hasan M, Dobrila F, Jonjić N. Macrophage level is not affected by monocyte chemotactic protein-1 in invasive ductal breast carcinoma. J Cancer Res Clin Oncol 2005; 131:453-8. [PMID: 15883814 DOI: 10.1007/s00432-004-0667-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2004] [Accepted: 12/12/2004] [Indexed: 10/25/2022]
Abstract
PURPOSE AND METHOD Monocyte chemotactic protein-1 (MCP-1) is a chemokine involved in the macrophage infiltration of tumor tissue. Tumor-associated macrophages (TAMs) are a population of mononuclear phagocytic cells that can have a complex function in tumor biology. The aim of this study was to determine the possible correlation between parenchymal MCP-1 expression and TAM level by immunohistochemical analysis of 97 invasive ductal breast carcinomas, not otherwise specified (NOS), and to investigate their relation with tumor size, histological grade, mitotic activity index (MAI) and lymph node status. Secondly, the MCP-1 mRNA was determined by reverse transcriptase-polymerase chain reaction (RT-PCR) in eight samples of normal breast tissue and 27 samples of invasive breast carcinomas and compared with TAMs. RESULTS MCP-1 immunoreactivity was present in tumor cells (17/97), but also in TAMs, fibroblasts and endothelial cells. The statistical analysis did not show a significant correlation between MCP-1 expression in tumoral epithelium and tumor size, histological grade, MAI, lymph node status or TAMs. The results of RT-PCR showed that, in all cases of breast carcinomas (27/27) and the majority of normal breast tissues (7/8), the number of detected MCP-1 cDNA copies was above the detection limit. However, carcinomas showed higher levels of MCP-1 mRNA than normal breast tissue. Nevertheless, the statistical analysis did not find a significant correlation between MCP-1 expression and macrophage infiltrations. CONCLUSION These results indicate that MCP-1 is probably not the only and/or crucial factor involved in macrophage attraction to tumor locus in breast carcinoma.
Collapse
MESH Headings
- Breast/metabolism
- Breast/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- DNA/genetics
- DNA/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Humans
- Macrophages/metabolism
- Macrophages/pathology
- Neoplasm Invasiveness/pathology
- Prognosis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- T Valković
- Department of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | | | | | | | | | | | | |
Collapse
|
1414
|
Villeneuve J, Tremblay P, Vallières L. Tumor Necrosis Factor Reduces Brain Tumor Growth by Enhancing Macrophage Recruitment and Microcyst Formation. Cancer Res 2005; 65:3928-36. [PMID: 15867393 DOI: 10.1158/0008-5472.can-04-3612] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent findings implicate macrophages and some of their secreted products, especially tumor necrosis factor (TNF), as tumor promoters. Inhibitors of these inflammatory components are currently regarded as potential therapeutic tools to block tumor progression. Here, we show that infiltrating macrophages represented a significant population of nonneoplastic cells within malignant gliomas, in which they were the exclusive producers of TNF. Contrary to the reported pro-oncogenic effects of TNF in other types of solid tumors, glioma-bearing mice deficient in TNF developed larger tumors and had reduced survival compared with their wild-type controls. Histologic examinations revealed that glioma volume was negatively correlated with the number of macrophages and small cavities called microcysts. Overall, our results support the concept that macrophages alter brain tumor development through a TNF-dependent process that culminates in the formation of microcysts. This raises the question of whether anti-inflammatory drugs, such as those commonly administrated to patients with brain cancer, could interfere with antitumor mechanisms.
Collapse
Affiliation(s)
- Jérôme Villeneuve
- Department of Oncology and Molecular Endocrinology, Laval University Hospital Research Center, Québec City, Québec, Canada.
| | | | | |
Collapse
|
1415
|
de Moreno de LeBlanc A, Matar C, LeBlanc N, Perdigón G. Effects of milk fermented by Lactobacillus helveticus R389 on a murine breast cancer model. Breast Cancer Res 2005; 7:R477-86. [PMID: 15987453 PMCID: PMC1175061 DOI: 10.1186/bcr1032] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Revised: 03/21/2005] [Accepted: 03/31/2005] [Indexed: 12/20/2022] Open
Abstract
Introduction Antitumour activity is one of the health-promoting effects attributed to the lactic acid bacteria and their products of fermentation. Previous studies in mice demonstrated that bioactive compounds released in milk fermented by Lactobacillus helveticus R389 contribute to its immunoenhancing and antitumour properties. The aim of the present work was to study the effects of the consumption of milk fermented by L. helveticus R389 or its proteolytic-deficient variant, L. helveticus L89, on a murine hormone-dependent breast cancer model. Methods Mice were fed with milk fermented by L. helveticus R389 or L. helveticus L89, during 2 or 7 days. The tumour control group received no special feeding. At the end of the feeding period, the mice were challenged by a subcutaneous injection of tumour cells in the mammary gland. Four days post-injection, the mice received fermented milk on a cyclical basis. The rate of tumour development and the cytokines in serum, mammary gland tissue and tumour-isolated cells were monitored. Bcl-2-positive cells in mammary glands and cellular apoptosis in tumour tissue were also studied. Results Seven days of cyclical administration of milk fermented by either bacterial strain delayed or stopped the tumour development. Cytokines demonstrated that L. helveticus R389 modulated the immune response challenged by the tumour. IL-10 and IL-4 were increased in all the samples from this group. In comparison with the tumour control, all test groups showed a decrease of IL-6, a cytokine involved in oestrogen synthesis. Seven days of cyclical feeding with milk fermented by L. helveticus R389 produced an increase in the number of apoptotic cells, compared with all other groups. Conclusion This study demonstrated that 7 days of cyclical administration of milk fermented by both strains of L. helveticus diminishes tumour growth, stimulating an antitumour immune response. Compounds released during milk fermentation with L. helveticus R389 would be implicated in its immunoregulatory capacity on the immune response in mammary glands and tumour, which were correlated with the cytokines found at the systemic level. The milk fermented by L. helveticus R389 was able to modulate the relationship between immune and endocrine systems (by IL-6 diminution), which is very important in oestrogen-dependent tumour and induced cellular apoptosis.
Collapse
Affiliation(s)
- Alejandra de Moreno de LeBlanc
- Départment de Chimie-Biochimie, Université de Moncton, NB, Canada
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Tucumán, Argentina
- Cátedra de Inmunología, Facultad de Bioquimíca, Química y Farmacia, Universidad Nacional de Tucumán, Argentina
| | - Chantal Matar
- Départment de Chimie-Biochimie, Université de Moncton, NB, Canada
| | - Nicole LeBlanc
- Départment de Chimie-Biochimie, Université de Moncton, NB, Canada
| | - Gabriela Perdigón
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Tucumán, Argentina
- Cátedra de Inmunología, Facultad de Bioquimíca, Química y Farmacia, Universidad Nacional de Tucumán, Argentina
| |
Collapse
|
1416
|
Ribatti D, Crivellato E, Roccaro AM, Ria R, Vacca A. Mast cell contribution to angiogenesis related to tumour progression. Clin Exp Allergy 2005; 34:1660-4. [PMID: 15544587 DOI: 10.1111/j.1365-2222.2004.02104.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The current wisdom is that tumours are endowed with an angiogenic capability and that their growth, invasion and metastasis are angiogenesis dependent. It is now well documented that neoplastic cells are influenced by their microenvironment and vice versa. The specific organ microenvironment determines the extent of cancer cell proliferation, angiogenesis, invasion and survival. Tumour cells are surrounded by an infiltrate of inflammatory cells, namely lymphocytes, neutrophils, macrophages and mast cells (MCs), which communicate via a complex network of intercellular signalling pathways, mediated by surface adhesion molecules, cytokines and their receptors. This review article summarizes: (i) the MC mediators involved in angiogenesis; (ii) the experimental evidence concerning the role played by MCs in angiogenesis; (iii) the list of solid and haematological tumours in which a close relationship between angiogenesis, tumour progression and MCs has been demonstrated; (iv) the circumstances in which MCs are a critical source of angiogenic factors in vivo, and in such cases, the signals that regulate their production and secretion that need to be determined as a prelude to the elaboration of new therapeutic strategies associated with MC presence and activation.
Collapse
Affiliation(s)
- D Ribatti
- Department of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy.
| | | | | | | | | |
Collapse
|
1417
|
Allavena P, Signorelli M, Chieppa M, Erba E, Bianchi G, Marchesi F, Olimpio CO, Bonardi C, Garbi A, Lissoni A, de Braud F, Jimeno J, D'Incalci M. Anti-inflammatory Properties of the Novel Antitumor Agent Yondelis (Trabectedin): Inhibition of Macrophage Differentiation and Cytokine Production. Cancer Res 2005; 65:2964-71. [PMID: 15805300 DOI: 10.1158/0008-5472.can-04-4037] [Citation(s) in RCA: 234] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Yondelis (Trabectedin) is a novel antitumor agent of marine origin extracted from the tunicate Ecteinascidia turbinata. This original compound is active against several human tumors including sarcoma and ovarian and breast adenocarcinoma, as evidenced in phase II clinical trials in advanced multitreated patients. Yondelis is a DNA minor groove binder that blocks cell cycle and interferes with inducible gene transcription in a selective manner. In this study, we investigated the immunomodulatory properties of Yondelis on leukocytes. Human blood monocytes were highly susceptible in vitro to its cytotoxic effect and underwent apoptosis at pharmacologically relevant concentrations (5 nmol/L), whereas lymphocytes were up to 5-fold less sensitive. Macrophages differentiated in vitro with macrophage colony-stimulating factor and tumor-associated macrophages (TAM), isolated from patients with ovarian cancer, were also susceptible. At subcytotoxic concentrations, Yondelis inhibited the in vitro differentiation of monocytes to macrophages. In tumor-treated patients, drug infusion caused a selective decrease of monocyte counts and of ex vivo macrophage differentiation. The in vitro production of two proinflammatory mediators, CCL2 and IL-6, was markedly reduced by Yondelis in monocytes, macrophages, TAM, and freshly isolated ovarian tumor cells. The chemokine CCL2 is the major determinant of monocyte recruitment at tumor sites, whereas IL-6 is a growth factor for ovarian tumors. In view of the protumor activity of TAM and of the strong association between chronic inflammation and cancer progression, the inhibitory effect of Yondelis on macrophage viability, differentiation, and cytokine production is likely to contribute to the antitumor activity of this agent in inflammation-associated human tumors.
Collapse
Affiliation(s)
- Paola Allavena
- Department of Immunology, Mario Negri Institute, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1418
|
Kluk MJ, Grant-Kels JM, Kerr P, Hoss D, Berke A, Claffey KP, Murphy M. Melanoma on the move: the progression of melanoma: novel concepts with histologic correlates. Am J Dermatopathol 2005; 26:504-10. [PMID: 15618934 DOI: 10.1097/00000372-200412000-00011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Michael J Kluk
- Division of Dermatopathology, Department of Dermatology, University of Connecticut School of Medicine, Farmington, Connecticut 06030-6230, USA
| | | | | | | | | | | | | |
Collapse
|
1419
|
Abstract
Cancer is a difficult target for any therapeutic strategy; therefore, there is a continuous search for new therapeutic modalities, for application either alone or in combination. In this regard, gene-based therapy is a new approach that offers hope of improved control of tumors. Intensive research to apply gene therapy for cancer treatment has led to identification of the most important technical and theoretical barriers that need to be overcome for clinical success. One of the central unresolved challenges remains the issue of specific and efficient delivery of genes to target cells or tissues, emphasizing the importance of the gene carrier. Along with different viral and non-viral vector systems, mammalian cells have also been considered as vehicles for delivery of anti-cancer therapeutics. The cell-based delivery approach was introduced as the first attempt to apply gene therapy to cancer treatment, and in general, has followed most of the ups and downs of gene therapy applications, progressing alongside new knowledge gained in this field. As a result, significant progress has been made in some aspects of the cell-based approach, while the development of other essential issues is only just gaining speed. It appears that the initial phase of development of cell-based protocols - the achievement of efficient ex vivo cell loading with therapeutics - has largely been fulfilled. However, the desired efficacy of cell-based strategies in general has not yet been reached, and specificity of tumor homing needs to be improved considerably. There is hope that advances in related scientific fields will promote the utilization of cells as powerful and versatile vehicles for cancer gene therapy.
Collapse
Affiliation(s)
- Larisa Pereboeva
- Division of Human Gene Therapy, Department of Medicine, The Gene Therapy Center, BMRII-572, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL 35294, USA.
| | | |
Collapse
|
1420
|
Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 2005; 7:211-7. [PMID: 15766659 DOI: 10.1016/j.ccr.2005.02.013] [Citation(s) in RCA: 1315] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Frances Balkwill
- Cancer Research UK, Translational Oncology Laboratory, Barts and The London, Queen Mary's Medical School, EC1M 6BQ London, United Kingdom.
| | | | | |
Collapse
|
1421
|
Corthay A, Skovseth DK, Lundin KU, Røsjø E, Omholt H, Hofgaard PO, Haraldsen G, Bogen B. Primary Antitumor Immune Response Mediated by CD4+ T Cells. Immunity 2005; 22:371-83. [PMID: 15780993 DOI: 10.1016/j.immuni.2005.02.003] [Citation(s) in RCA: 345] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2004] [Revised: 01/27/2005] [Accepted: 02/01/2005] [Indexed: 12/01/2022]
Abstract
Gene-targeted mice have recently revealed a role for lymphocytes and interferon-gamma (IFNgamma) in conferring protection against cancer, but the mechanisms remain unclear. Here, we have characterized a successful primary antitumor immune response initiated by naive CD4+ T cells. Major histocompatibility complex class II (MHC-II)-negative myeloma cells injected subcutaneously into syngeneic mice were surrounded within 3 days by macrophages that captured tumor antigens. Within 6 days, naive myeloma-specific CD4+ T cells became activated in draining lymph nodes and subsequently migrated to the incipient tumor site. Upon recognition of tumor-derived antigenic peptides presented on MHC-II by macrophages, the myeloma-specific CD4+ T cells were reactivated and started to secrete cytokines. T cell-derived IFNgamma activated macrophages in close proximity to the tumor cells. Tumor cell growth was completely inhibited by such locally activated macrophages. These data indicate a mechanism for immunosurveillance of MHC-II-negative cancer cells by tumor-specific CD4+ T cells through collaboration with macrophages.
Collapse
Affiliation(s)
- Alexandre Corthay
- Institute of Immunology, University of Oslo, Rikshospitalet and Rikshospitalet University Hospital, 0027 Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
1422
|
Marchesi F, Monti P, Leone BE, Zerbi A, Vecchi A, Piemonti L, Mantovani A, Allavena P. Increased survival, proliferation, and migration in metastatic human pancreatic tumor cells expressing functional CXCR4. Cancer Res 2005; 64:8420-7. [PMID: 15548713 DOI: 10.1158/0008-5472.can-04-1343] [Citation(s) in RCA: 254] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, we have evaluated 11 pancreatic tumor cell lines and tumor cells from surgical samples of patients with pancreatic adenocarcinoma for expression of the chemokine receptor CXCR4. Six of 11 cell lines expressed detectable mRNA of CXCR4, with three cell lines (AsPC1, Capan1, and Hs766T) having substantial amounts of transcripts. Expression was higher in lines derived from metastatic lesions compared with those derived from primary tumors. Different inflammatory cytokines did not modify expression, whereas IFN-gamma down-regulated and hypoxia up-regulated CXCR4 transcripts. Transcript expression was associated with surface expression in pancreatic carcinoma cell lines. All surgical carcinoma samples tested expressed higher levels of CXCR4 than normal pancreatic ducts, which were used as reference tissue. The chemokine CXCL12 induced chemotaxis in CXCR4-positive pancreatic carcinoma cell lines, which was inhibited by anti-CXCR4 monoclonal antibody and by the antagonist AMD3100. Transendothelial migration, Matrigel invasion, and activation of matrix metalloproteases were also enhanced by CXCL12. In CXCR4-positive cell lines, CXCL12 stimulated cell proliferation. The cell line Hs766T produces high levels of CXCL12, and addition of the CXCR4 antagonist AMD3100 partially inhibited proliferation, indicating an autocrine loop. Moreover, the addition of exogenous CXCL12 inhibited apoptosis induced by serum starvation. These results indicate that the CXCR4 receptor is frequently expressed in metastatic pancreatic tumor cells. CXCR4 not only stimulates cell motility and invasion but also promotes survival and proliferation. Strategies to target CXCR4 expressed on tumor cells may be of benefit in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Federica Marchesi
- Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
1423
|
Schiffelers RM, Metselaar JM, Fens MHAM, Janssen APCA, Molema G, Storm G. Liposome-encapsulated prednisolone phosphate inhibits growth of established tumors in mice. Neoplasia 2005; 7:118-27. [PMID: 15802017 PMCID: PMC1501128 DOI: 10.1593/neo.04340] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Revised: 06/21/2004] [Accepted: 06/21/2004] [Indexed: 01/01/2023]
Abstract
Glucocorticoids can inhibit solid tumor growth possibly due to an inhibitory effect on angiogenesis. The antitumor effects of the free drugs have only been observed using treatment schedules based on high and frequent dosing for prolonged periods of time. As long-circulating liposomes accumulate at sites of malignancy, we investigated the tumor-inhibiting potential of liposome-encapsulated prednisolone phosphate. Liposomal prednisolone phosphate could inhibit tumor growth dose-dependently, with 80% to 90% tumor growth inhibition of subcutaneous B16.F10 melanoma and C26 colon carcinoma murine tumor models at 20 mg/kg by single or weekly doses. Prednisolone phosphate in the free form was completely ineffective at this low-frequency treatment schedule, even when administered at a dose of 50 mg/kg. In vitro studies did not show an inhibitory effect of prednisolone (phosphate) on tumor cell, nor on endothelial cell proliferation. Histologic evaluation revealed that liposomal prednisolone phosphate-treated tumors contained a center with areas of picnotic/necrotic cells, which were not apparent in untreated tumors or tumors treated with the free drug. In conclusion, the present study shows potent antitumor effects of liposomal formulations of glucocorticoids in a low dose and low-frequency schedule, offering promise for liposomal glucocorticoids as novel antitumor agents.
Collapse
Affiliation(s)
- Raymond M Schiffelers
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
1424
|
Yamagishi SI, Abe R, Inagaki Y, Nakamura K, Sugawara H, Inokuma D, Nakamura H, Shimizu T, Takeuchi M, Yoshimura A, Bucala R, Shimizu H, Imaizumi T. Minodronate, a newly developed nitrogen-containing bisphosphonate, suppresses melanoma growth and improves survival in nude mice by blocking vascular endothelial growth factor signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 165:1865-74. [PMID: 15579431 PMCID: PMC1618714 DOI: 10.1016/s0002-9440(10)63239-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Angiogenesis, a process by which new vascular networks are formed from pre-existing capillaries, is required for tumors to grow, invade, and metastasize. Vascular endothelial growth factor (VEGF), a specific mitogen to endothelial cells, is a crucial factor for tumor angiogenesis. In this study, we investigated whether minodronate, a newly developed nitrogen-containing bisphosphonate, could inhibit melanoma growth and improve survival in nude mice by suppressing the VEGF signaling. We found here that minodronate inhibited melanoma growth and improved survival in nude mice by suppressing the tumor-associated angiogenesis and macrophage infiltration. Minodronate completely inhibited the VEGF-induced increase in DNA synthesis and tube formation in endothelial cells by suppressing NADPH oxidase-mediated reactive oxygen species generation and Ras activation. Furthermore, minodronate inhibited the VEGF-induced expression of intercellular adhesion molecule-1 and monocyte chemoattractant protein-1 in endothelial cells. Minodronate decreased DNA synthesis and increased apoptotic cell death of cultured melanoma cells as well. Our present study suggests that minodronate might suppress melanoma growth and improve survival in nude mice by two independent mechanisms; one is by blocking the VEGF signaling in endothelial cells, and the other is by inducing apoptotic cell death of melanoma. The present study provides a novel potential therapeutic strategy for the treatment of melanoma.
Collapse
Affiliation(s)
- Sho-ichi Yamagishi
- Department of Internal Medicine III, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1425
|
Junker N, Johansen JS, Andersen CB, Kristjansen PEG. Expression of YKL-40 by peritumoral macrophages in human small cell lung cancer. Lung Cancer 2005; 48:223-31. [PMID: 15829322 DOI: 10.1016/j.lungcan.2004.11.011] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Revised: 11/04/2004] [Accepted: 11/08/2004] [Indexed: 02/02/2023]
Abstract
YKL-40 is a 40 kDa protein with possible involvement in tissue remodeling, cell proliferation and angiogenesis. Elevated serum YKL-40 levels in patients with metastatic cancers (including small cell lung cancer (SCLC)) are associated with poor prognosis. The aim of this study was to identify the cellular source of YKL-40 in SCLC patient biopsies and in a panel of 20 human SCLC lines cultured in vitro and in vivo in nude mice. In general, the SCLC cell lines had no or very limited (human) YKL-40 expression, whereas, by RT-PCR a pronounced murine (i.e., stromal) YKL-40 expression was present in all tumors. YKL-40 mRNA transcripts were detected by in situ hybridization in 9 of 10 biopsies from SCLC patients, and in each case the signal was localized in the peritumoral stroma in cells of typical macrophage morphology (confirmed by a CD68 macrophage specific stain). No YKL-40 mRNA expression was found in the cancer cells, in macrophages infiltrating the solid tumor areas, or in non-malignant tissue. In conclusion, the predominant source of elevated serum YKL-40 in SCLC is peritumoral macrophages.
Collapse
Affiliation(s)
- Nanna Junker
- Laboratory of Experimental Oncology, Department of Molecular Pathology, University of Copenhagen, 11 Frederik V Vej, DK-2100 Copenhagen, Denmark
| | | | | | | |
Collapse
|
1426
|
Rehor A, Hubbell JA, Tirelli N. Oxidation-sensitive polymeric nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2005; 21:411-417. [PMID: 15620332 DOI: 10.1021/la0478043] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
We have recently demonstrated the possible use of organic polysulfides for the design of oxidation-sensitive colloidal carriers in the form of polymeric vesicles, which are particularly suitable for the encapsulation of hydrosoluble drugs. In the present research we extend our efforts to carriers specifically suitable for hydrophobic molecules. Exploiting the living emulsion polymerization of episulfides, we have produced new cross-linked polysulfide nanoparticles. Here we demonstrate how this process allows the production of stable nanoparticles with a good control over their size and functionality. The nanoparticles showed negligible cytotoxicity on a fibroblast model; furthermore, they exhibited sensitivity to oxidative conditions, which first produce swelling and then solubilize the material.
Collapse
Affiliation(s)
- Annemie Rehor
- Institute for Biomedical Engineering and Department of Materials, Swiss Federal Institute of Technology and University of Zurich, Moussonstrasse 18, CH-8044 Zurich, Switzerland
| | | | | |
Collapse
|
1427
|
Esposito I, Bergmann F, Büchler MW, Friess H. Role of Mast Cells in Pancreatic Carcinoma. HANDBOOK OF IMMUNOHISTOCHEMISTRY AND IN SITU HYBRIDIZATION OF HUMAN CARCINOMAS 2005:383-389. [DOI: 10.1016/s1874-5784(05)80046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
1428
|
Bissell MJ, Kenny PA, Radisky DC. Microenvironmental regulators of tissue structure and function also regulate tumor induction and progression: the role of extracellular matrix and its degrading enzymes. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2005; 70:343-56. [PMID: 16869771 PMCID: PMC3004779 DOI: 10.1101/sqb.2005.70.013] [Citation(s) in RCA: 211] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
It is now widely accepted that elements of the cellular and tissue microenvironment are crucial regulators of cell behavior in culture and homeostasis in vivo, and that many of the same factors influence the course of tumor progression. Less well established is the extent to which extracellular factors actually cause cancer, and the circumstances under which this may occur. Using physiologically relevant three-dimensional culture assays and transgenic animals, we have explored how the environmental and architectural context of cells, tissues, and organs controls mammary-specific gene expression, growth regulation, apoptosis, and drug resistance and have found that loss of tissue structure is a prerequisite for cancer progression. Here we summarize this evidence and highlight two of our recent studies. Using mouse mammary epithelial cells, we show that exposure to matrix metalloproteinase-3 (MMP-3) stimulates production of reactive oxygen species (ROS) that destabilize the genome and induce epithelial-mesenchymal transition, causing malignant transformation. Using a human breast cancer progression series, we find that ADAM-dependent growth factor shedding plays a crucial role in acquisition of the malignant phenotype. These findings illustrate how normal tissue structure controls the response to extracellular signals so as to preserve tissue specificity and growth status.
Collapse
Affiliation(s)
- M J Bissell
- Cancer Biology Department, Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, 94720, USA
| | | | | |
Collapse
|
1429
|
Chen JJW, Lin YC, Yao PL, Yuan A, Chen HY, Shun CT, Tsai MF, Chen CH, Yang PC. Tumor-associated macrophages: the double-edged sword in cancer progression. J Clin Oncol 2004; 23:953-64. [PMID: 15598976 DOI: 10.1200/jco.2005.12.172] [Citation(s) in RCA: 285] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Inflammation plays a critical role in cancer progression. In this study we investigate the pro-tumorigenic activities and gene expression profiles of lung cancer cells after interaction with macrophages. MATERIALS AND METHODS We measured intratumoral microvessel counts and macrophage density in 41 lung cancer tumor specimens and correlated these with the patients' clinical outcome. The interaction between macrophages and cancer cell lines was assessed using a transwell coculture system. The invasive potential was evaluated by in vitro invasion assay. The matrix-degrading activity was assayed by gelatin zymography. The microarray was applied to a large-scale analysis of the genes involved in the interaction, as well as to monitor the gene expression profiles of lung cancer cells responding to anti-inflammatory drugs in cocultures. RESULTS The macrophage density positively correlated with microvessel counts and negatively correlated with patient relapse-free survival (P < .05). After coculture with macrophages, lung cancer cell lines exhibited higher invasive potentials and matrix-degrading activities. We identified 50 genes by microarray that were upregulated more than two-fold in cancer cells after coculture. Northern blot analyses confirmed some gene expression such as interleukin-6, interleukin-8, and matrix metalloproteinase 9. The two-dimensional hierarchical clustering also demonstrated that the gene expression profiles of lung cancer cells responding to various anti-inflammatory drugs in cocultures are distinct. CONCLUSION The interaction of lung cancer cells and macrophages can promote the invasiveness and matrix-degrading activity of cancer cells. Our results also suggest that a great diversity of gene expression occurs in this interaction, which may assist us in understanding the process of cancer metastasis.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Anti-Inflammatory Agents/therapeutic use
- Carcinoma, Non-Small-Cell Lung/blood supply
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Count
- Cell Line, Tumor
- Coculture Techniques
- Disease Progression
- Disease-Free Survival
- Female
- Gelatinases/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Interleukin-6/genetics
- Interleukin-8/genetics
- Lung Neoplasms/blood supply
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/pathology
- Male
- Matrix Metalloproteinase 9/genetics
- Microcirculation/pathology
- Middle Aged
- Neoplasm Invasiveness
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Jeremy J W Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
1430
|
Abstract
Robustness is a ubiquitously observed property of biological systems. It is considered to be a fundamental feature of complex evolvable systems. It is attained by several underlying principles that are universal to both biological organisms and sophisticated engineering systems. Robustness facilitates evolvability and robust traits are often selected by evolution. Such a mutually beneficial process is made possible by specific architectural features observed in robust systems. But there are trade-offs between robustness, fragility, performance and resource demands, which explain system behaviour, including the patterns of failure. Insights into inherent properties of robust systems will provide us with a better understanding of complex diseases and a guiding principle for therapy design.
Collapse
Affiliation(s)
- Hiroaki Kitano
- Sony Computer Science Laboratories, Inc., 3-14-13 Higashi-Gotanda, Shinagawa, Tokyo 141-0022, Japan.
| |
Collapse
|
1431
|
Abstract
PURPOSE OF REVIEW Macrophage infiltration is a hallmark of all forms of inflammatory and non-inflammatory renal injury. However, the classical view of macrophages as cells that cause injury has been superseded with evidence of their heterogeneous role, i.e. with involvement in all stages of the inflammatory process including tissue repair and healing. This review summarizes the major advances in macrophage biology achieved in the last year, highlighting the different activation states, how these are regulated, and their relevance in renal disease. RECENT FINDINGS New concepts have emerged concerning the factors controlling monocyte recruitment into inflamed tissue and their subsequent differentiation into activated macrophages. There is now compelling evidence for the heterogeneity of macrophages in clinical disease, i.e. they appear to be able to both promote and downregulate inflammation. An increased understanding of the factors regulating the expression of pro-inflammatory or reparative characteristics by macrophages is establishing how their function can be manipulated to attenuate renal inflammation in experimental models. SUMMARY An understanding of the role of macrophages at different time-points in renal inflammation, and the development of techniques for modulating macrophage activation in vivo, will provide a powerful method for exploiting the reparative attributes of these cells in clinical settings, restoring regulation to the inflammatory process and promoting healing.
Collapse
Affiliation(s)
- Heather M Wilson
- Department of Medicine and Therapeutics, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK.
| | | | | |
Collapse
|
1432
|
Lundin KU, Screpanti V, Omholt H, Hofgaard PO, Yagita H, Grandien A, Bogen B. CD4+ T cells kill Id+ B-lymphoma cells: FasLigand-Fas interaction is dominant in vitro but is redundant in vivo. Cancer Immunol Immunother 2004; 53:1135-45. [PMID: 15696611 PMCID: PMC11032948 DOI: 10.1007/s00262-004-0538-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2004] [Accepted: 03/19/2004] [Indexed: 01/30/2023]
Abstract
B-lymphoma cells express a highly tumor-specific antigen, monoclonal Ig, which is a promising target for immunotherapy. Previous work has demonstrated that B-lymphoma cells spontaneously process their endogenous monoclonal Ig and present variable (V) region peptides (Id-peptides) on their MHC class II molecules to CD4+ T cells. Id-specific CD4+ T cells protect mice against B-lymphoma cells in the absence of antiidiotypic antibodies. The molecular mechanism by which Id-specific CD4+ T cells kill B-lymphoma cells is hitherto unknown. We here demonstrate in an Id-specific T-cell receptor (TCR)-transgenic mouse model that Id-specific CD4+ T cells induce apoptosis of Fas+ B-lymphoma cells in vitro by FasLigand (FasL)-Fas interaction. Moreover, the rare B lymphomas that had escaped rejection in TCR-transgenic mice had down-regulated their sensitivity to Fas-mediated apoptosis. Although these results suggest that FasL-Fas interaction is important, Id-specific CD4+ T cells could eliminate Id+ B-lymphoma cells in vivo by other mechanisms, since three independent ways of blocking FasL-Fas-mediated killing failed to abrogate tumor protection in TCR-transgenic mice. These results suggest that there are several redundant pathways by which Id-specific CD4+ T cells eliminate Id+ B-lymphoma cells in vivo, of which FasL-Fas interaction is only one.
Collapse
Affiliation(s)
- Katrin U Lundin
- Institute of Immunology, University of Oslo, Rikshospitalet, N-0027 Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|
1433
|
Okaji Y, Tsuno NH, Kitayama J, Saito S, Takahashi T, Kawai K, Yazawa K, Asakage M, Tsuchiya T, Sakurai D, Tsuchiya N, Tokunaga K, Takahashi K, Nagawa H. A novel method for isolation of endothelial cells and macrophages from murine tumors based on Ac-LDL uptake and CD16 expression. J Immunol Methods 2004; 295:183-93. [PMID: 15627623 DOI: 10.1016/j.jim.2004.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Revised: 09/25/2004] [Accepted: 10/18/2004] [Indexed: 11/25/2022]
Abstract
Analysis of specific properties of tumor endothelium should be useful for development of novel antiangiogenic strategies. However, the isolation of pure endothelial cells from tumor tissues is still a fundamental problem. In this study, we have attempted to develop a reliable method for the isolation of endothelial cells from murine tumors. We found that the labeling with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate-acetylated-low density lipoprotein (Dil-Ac-LDL), commonly used for this purpose, can result in the contamination of isolated endothelium by macrophages due to the overlapping staining patterns of these two distinct cell types. Therefore, we chose the CD16, which is expressed on macrophages but not endothelial cells, to better distinguish them when labeled with Dil-Ac-LDL. By using this method, we obtained pure populations of endothelial cells and macrophages from murine colorectal cancer tissues, showing characteristic morphological and functional properties of the either cell type. The endothelial cells were long spindle-shaped, spread on gelatin, formed tube-like structures on Matrigel and expressed MECA-32 but not CD68. In contrast, the macrophages were round-shaped, partially spread on gelatin, formed unorganized aggregates on Matrigel and expressed CD68 but not MECA-32. The additional analysis of normal and tumor tissues revealed a positive correlation between the relative numbers of tumor endothelial cells and macrophages, calculated as % total cells, as well as the respective relative number and tumor weight. The present method is hoped to be useful for the evaluation of tumor angiogenesis and antitumor immunity.
Collapse
Affiliation(s)
- Yurai Okaji
- Department of Surgical Oncology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1434
|
Singh N, Singh SM, Shrivastava P. Immunomodulatory and Antitumor Actions of Medicinal PlantTinospora cordifoliaAre Mediated Through Activation of Tumor‐Associated Macrophages. Immunopharmacol Immunotoxicol 2004; 26:145-62. [PMID: 15106739 DOI: 10.1081/iph-120029952] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The present investigations were under taken to study whether the tumor-associated macrophages (TAM) of Dalton's lymphoma (DL), a spontaneous transplantable T cell lymphoma, can be activated by the alcoholic extract of medicinal plant Tinospora cordifolia (ALTC). Intraperitoneal administration of ALTC in DL-bearing mice not only augments the basic function of macrophages such as Phagocytosis as well as their antigen presenting ability and secretion of IL-1, TNF and RNI. The results of the present investigation also indicate that the intraperitoneal administration of ALTC slow down the tumor growth and increases the life span of tumor bearing host, thus showing its anti tumor effect through destabilizing the membrane integrity of DL cells directly or indirectly. This is the first study of it's kind regarding the effect of alcoholic extract of Tinospora cordifolia on the activation of tumor associated macrophages and showing the antitumor effect on the spontaneous T-cell lymphoma (DL), thus may have clinical implications.
Collapse
Affiliation(s)
- Nisha Singh
- School of Biotechnology, Banaras Hindu University, Vanarasi, U.P., India
| | | | | |
Collapse
|
1435
|
Mühl H, Paulukat J, Höfler S, Hellmuth M, Franzen R, Pfeilschifter J. The HIV protease inhibitor ritonavir synergizes with butyrate for induction of apoptotic cell death and mediates expression of heme oxygenase-1 in DLD-1 colon carcinoma cells. Br J Pharmacol 2004; 143:890-8. [PMID: 15504750 PMCID: PMC1575947 DOI: 10.1038/sj.bjp.0706023] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The protease inhibitor ritonavir is an integral part of current antiretroviral therapy targeting human immunodeficiency virus. Recent studies demonstrate that ritonavir induces apoptotic cell death with high efficiency in lymphoblastoid cell lines. Moreover, ritonavir can suppress activation of the transcription factor nuclear factor-kappaB and is an inhibitor of interleukin-1beta and tumor necrosis factor-alpha production in peripheral blood mononuclear cells. Thus, ritonavir appears to have anti-inflammatory properties. In the present study, we investigated in DLD-1 colon carcinoma cell effects of ritonavir on apoptotic cell death and expression of heme oxygenase-1 (HO-1), an anti-inflammatory enzyme that may be critically involved in the modulation of colonic inflammation. Compared to unstimulated control, ritonavir resulted in a moderate increase in the rate of apoptotic cell death as observed after 20 h of incubation. Notably, ritonavir potently synergized with the short-chain fatty acid butyrate for induction of caspase-3-dependent apoptosis in DLD-1 cells. Ritonavir enhanced mRNA and protein expression of HO-1 in DLD-1 cells. Ritonavir-induced HO-1 protein was suppressed by SB203580 or SB202190 and preceded by immediate upregulation of cellular c-Fos and c-Jun protein levels. This process was associated with induction of activator protein-1 as detected by electrophoretic mobility shift analysis. The present data suggest that ritonavir has the potential to curb colon carcinogenesis by reducing cell growth via mechanisms that include apoptosis and by simultaneously modulating colonic inflammation via induction of anti-inflammatory HO-1.
Collapse
Affiliation(s)
- Heiko Mühl
- Pharmazentrum frankfurt (ZAFES), University Hospital, Johann Wolfgang Goethe-Universität Frankfurt am Main, Theodor-Stern Kai 7, Haus 75A, Frankfurt am Main D-60590, Germany.
| | | | | | | | | | | |
Collapse
|
1436
|
Shrivastava P, Singh SM, Singh N. Activation of tumor-associated macrophages by thymosin alpha 1. Int J Immunopathol Pharmacol 2004; 17:39-47. [PMID: 15000865 DOI: 10.1177/039463200401700106] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
It was shown earlier that the progressive growth of a transplantable T-cell lymphoma of spontaneous origin, designated as Dalton's lymphoma (DL), in a murine host is associated with an inhibition of macrophages (TAM) along with an involution of thymus. However, it remained unclear if a decline in the level of thymic peptides in DL-bearing host, due to thymic regression, has any implications in the inhibited responses of TAM. Therefore, the present investigation was under taken to study whether the TAM of DL-bearing host can be activated to tumoricidal state by peptides of thymic origin. It was observed that intraperitoneal administration of thymosin alpha 1 to DL-bearing mice resulted in activation of TAM. Such TAM were found to produce enhanced amount of interleukin-1 (IL-1), tumor necrosis factor (TNF), reactive oxygen intermediates (ROI), nitric oxide (NO) and showed an increased abilities of pinocytosis, phagocytosis, antigen presentation and tumor cytotoxicity. The TAM were found to be directly responsive to thymosin alpha1 as in vitro treatment with thymosin alpha 1 could activate TAM to tumoricidal state. Treatment of TAM with thymosin alpha 1 also enhanced their LPS responsiveness for an augmented state of activation. The findings of this study demonstrate for the first time that the TAM of a T cell lymphoma can be activated to tumoricidal state by thymosin alpha 1.
Collapse
Affiliation(s)
- P Shrivastava
- School of Biotechnology, Banaras Hindu University, Varanasi-221 005, U.P., India
| | | | | |
Collapse
|
1437
|
de Bruin M, Peters GJ, Oerlemans R, Assaraf YG, Masterson AJ, Adema AD, Dijkmans BAC, Pinedo HM, Jansen G. Sulfasalazine down-regulates the expression of the angiogenic factors platelet-derived endothelial cell growth factor/thymidine phosphorylase and interleukin-8 in human monocytic-macrophage THP1 and U937 cells. Mol Pharmacol 2004; 66:1054-1060. [PMID: 15269287 DOI: 10.1124/mol.104.000315] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Platelet-derived endothelial cell growth factor/thymidine phosphorylase (PD-ECGF/TP) and interleukin-8 (IL-8) are angiogenic factors produced by tumor infiltrating macrophages. Here, we show that prolonged exposure of human monocytic/macrophage THP1 and U937 cells to sulfasalazine, an anti-inflammatory drug and inhibitor of nuclear factor-kappaB (NF-kappaB), resulted in down-regulation of PD-ECGF/TP and IL-8 (mRNA, protein and activity) along with elimination of their induction by tumor necrosis factor-alpha and interferon-gamma. Concomitantly, sulfasalazine-exposed cells were markedly resistant to 5'-deoxyfluorouridine, the last intermediate of capecitabine requiring activation by PD-ECGF/TP. This is the first report suggesting that disruption of NF-kappaB-dependent signaling pathways can provoke a marked and sustained down-regulation of macrophage-related angiogenic factors. However, this may also negatively affect capecitabine efficacy.
Collapse
Affiliation(s)
- Michiel de Bruin
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
1438
|
De Ridder M, Verovski VN, Darville MI, Van Den Berge DL, Monsaert C, Eizirik DL, Storme GA. Macrophages enhance the radiosensitizing activity of lipid A: A novel role for immune cells in tumor cell radioresponse. Int J Radiat Oncol Biol Phys 2004; 60:598-606. [PMID: 15380597 DOI: 10.1016/j.ijrobp.2004.05.065] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2003] [Revised: 05/10/2004] [Accepted: 05/28/2004] [Indexed: 12/06/2022]
Abstract
PURPOSE This study examines whether activated macrophages may radiosensitize tumor cells through the release of proinflammatory mediators. METHODS AND MATERIALS RAW 264.7 macrophages were activated by lipid A, and the conditioned medium (CM) was analyzed for the secretion of cytokines and the production of nitric oxide (NO) through inducible nitric oxide synthase (iNOS). EMT-6 tumor cells were exposed to CM and analyzed for hypoxic cell radiosensitivity. The role of nuclear factor (NF)-kappaB in the transcriptional activation of iNOS was examined by luciferase reporter gene assay. RESULTS Clinical immunomodulator lipid A, at a plasma-relevant concentration of 3 microg/mL, stimulated RAW 264.7 macrophages to release NO, tumor necrosis factor (TNF)-alpha, and other cytokines. This in turn activated iNOS-mediated NO production in EMT-6 tumor cells and drastically enhanced their radiosensitivity. Radiosensitization was abrogated by the iNOS inhibitor aminoguanidine but not by a neutralizing anti-TNF-alpha antibody. The mechanism of iNOS induction was linked to NF-kappaB but not to JAK/STAT signaling. Interferon-gamma further increased the NO production by macrophages to a level that caused radiosensitization of EMT-6 cells through the bystanding effect of diffused NO. CONCLUSIONS We demonstrate for the first time that activated macrophages may radiosensitize tumor cells through the induction of NO synthesis, which occurs in both tumor and immune cells.
Collapse
Affiliation(s)
- Mark De Ridder
- Academic Hospital, Free University Brussels, Oncology Center, Cancer Research Unit, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
1439
|
Evans KN, Taylor H, Zehnder D, Kilby MD, Bulmer JN, Shah F, Adams JS, Hewison M. Increased expression of 25-hydroxyvitamin D-1alpha-hydroxylase in dysgerminomas: a novel form of humoral hypercalcemia of malignancy. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:807-13. [PMID: 15331405 PMCID: PMC1618616 DOI: 10.1016/s0002-9440(10)63343-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Humoral hypercalcemia of malignancy (HHM) is a common paraneoplastic disorder usually associated with increased synthesis of parathyroid hormone-related peptide (PTHrP). Unlike non-cancer forms of hypercalcemia, HHM does not routinely involve increased circulating levels of the active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3). Dysgerminomas are a notable exception to this rule, previous reports having described hypercalcemia with elevated serum 1,25(OH)2D3. To investigate the etiology of this form of HHM we have characterized expression and activity of the enzyme that catalyzes synthesis of 1,25(OH)2D3, 25-hydroxyvitamin D-1alpha-hydroxylase (1alpha-hydroxylase), in a collection of 12 dysgerminomas. RT-PCR analyses indicated that mRNA for 1alpha-hydroxylase was increased 222-fold in dysgerminomas compared to non-tumor ovarian tissue. Parallel enzyme assays in tissue homogenates showed that dysgerminomas produced fivefold higher levels of 1,25(OH)2D3 compared to normal ovarian tissue. Immunolocalization studies indicated that 1alpha-hydroxylase was expressed by both tumor cells and by macrophages within the inflammatory cell infiltrate associated with dysgerminomas. The immunological nature of the increased 1,25(OH)2D3 production observed in dysgerminomas was further emphasized by correlation between expression of 1alpha-hydroxylase and the endotoxin recognition factors CD14 and toll-like receptor 4 (TLR4). These data suggest that inflammatory mechanisms associated with dysgerminomas are the underlying cause of the increased expression and activity of 1alpha-hydroxylase associated with these tumors. We further postulate that this autocrine/paracrine action of 1alpha-hydroxylase may lead to increased circulating levels of 1,25(OH)2D3 and a form of HHM which is distinct from that seen with PTHrP-secreting tumors.
Collapse
Affiliation(s)
- Katie N Evans
- Division of Medical Sciences, Institute of Biomedical Research, The University of Birmingham, Birmingham, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
1440
|
Shunyakov L, Ryan CK, Sahasrabudhe DM, Khorana AA. The influence of host response on colorectal cancer prognosis. Clin Colorectal Cancer 2004; 4:38-45. [PMID: 15207019 DOI: 10.3816/ccc.2004.n.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death worldwide. Several tumor characteristics have been shown to be of prognostic significance, although stage at diagnosis continues to be the most important predictor of survival. Emerging new data suggest that the presence of a host response to CRC may also influence survival and other outcomes in CRC. This review summarizes recent evidence regarding the prognostic significance of the host response to CRC. In retrospective analyses, tumor-associated macrophages and tumor-infiltrating lymphocytes appear to be the elements most significantly associated with improved outcomes in CRC. The presence of other cells, including dendritic cells, natural killer cells, eosinophils, and mast cells, also appears to be associated with increased survival. The influence of the host response to CRC needs confirmation in prospective studies, but in the meantime should be part of risk stratification. Novel approaches to further augmenting this response merit study.
Collapse
Affiliation(s)
- Leo Shunyakov
- James P. Wilmot Cancer Center and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
1441
|
Tanaka Y, Kobayashi H, Suzuki M, Kanayama N, Suzuki M, Terao T. Upregulation of bikunin in tumor-infiltrating macrophages as a factor of favorable prognosis in ovarian cancer. Gynecol Oncol 2004; 94:725-34. [PMID: 15350365 DOI: 10.1016/j.ygyno.2004.06.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2003] [Indexed: 11/30/2022]
Abstract
OBJECTIVE This study was carried out to clarify the localization of bikunin, a Kunitz-type protease inhibitor, and relation between expression of individual bikunin protein and ovarian cancer progression. METHODS We performed a retrospective study on the immunohistochemical expression of bikunin, urokinase-type plasminogen activator (uPA) and macrophages (CD68) in surgical specimens derived from 89 ovarian cancer patients to investigate correlations between the expression of bikunin and the clinicopathologic features and the prognosis. Furthermore, bikunin and uPA levels were measured by immunoblot analysis. RESULTS Immunohistochemical staining revealed that the localization of bikunin was similar to that of CD68 for macrophages. We identified high expression of bikunin in 40 (45%) of 89 ovarian cancers. The results of Western blot analysis showed a significant correlation with immunohistochemical data. There was a significant inverse correlation between bikunin levels and uPA levels in ovarian cancer tissues. High bikunin expression was an independent predictor for disease-free survival (P = 0.040) and overall survival (P = 0.042). The 5-year survival rate of the 49 patients with low bikunin expression in ovarian cancers was 39%, whereas that of the other 40 patients with high bikunin expression was 63%. In addition, macrophage-derived bikunin protein was induced by exogenous IL-6. CONCLUSION Bikunin derived from tumor-infiltrating macrophages might be a prognostic indicator as an antiinvasive factor supplied from macrophages within and around the tumor possibly through down-regulation of tumor-associated uPA expression.
Collapse
Affiliation(s)
- Yoshiko Tanaka
- Department of Obstetrics and Gynecology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | | | | | | | | | | |
Collapse
|
1442
|
Stout RD, Suttles J. Functional plasticity of macrophages: reversible adaptation to changing microenvironments. J Leukoc Biol 2004; 76:509-13. [PMID: 15218057 PMCID: PMC1201486 DOI: 10.1189/jlb.0504272] [Citation(s) in RCA: 535] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
There has been substantial research activity in the past decade directed at phenotyping macrophage lineages and defining macrophage functional subsets or patterns of activity. The emphasis over the past 2-3 years has been to divide macrophage functional patterns into type 1 (Th1-driven) or type 2 (Th2-driven) functions. However, a huge array of environmental factors (including cytokines, chemokines, pattern recognition receptors, hormones) differentially regulates macrophage response patterns, resulting in the display of numerous distinct, functional phenotypes. Upon stimulation, a macrophage does not display just a single set of functions but rather displays a progression of functional changes in response to the progressive changes in its microenvironment. The remarkable ability of monocytes and tissue macrophages to adapt to changes in their microenvironment challenges the thesis that macrophages displaying unique tissue-specific or response-specific, functional patterns represent distinct lineages. With the exception of mature osteoclasts and mature dendritic cells, evidence supporting stable differentiation as the basis for macrophage functional heterogeneity is equivocal. The concept of whether macrophages develop into functional subsets as opposed to continuously adapting their functional pattern in response to the changing environment of a progressive inflammatory response is important to resolve from the perspectives of therapeutic targeting and understanding the role of macrophages in disease pathogenesis.
Collapse
Affiliation(s)
- Robert D Stout
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40292, USA.
| | | |
Collapse
|
1443
|
van Kempen LCLT, Ruiter DJ, van Muijen GNP, Coussens LM. The tumor microenvironment: a critical determinant of neoplastic evolution. Eur J Cell Biol 2004; 82:539-48. [PMID: 14703010 DOI: 10.1078/0171-9335-00346] [Citation(s) in RCA: 173] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Evolution of neoplastic cells has generally been regarded as a cumulative intrinsic process resulting in altered cell characteristics enabling enhanced growth properties, evasion of apoptotic signals, unlimited replicative potential and gain of properties enabling the ability to thrive in ectopic tissues and in some cases, ability to metastasize. Recently however, the role of the neoplastic microenvironment has become appreciated largely due to the realization that tumors are not merely masses of neoplastic cells, but instead, are complex tissues composed of both a non-cellular (matrix proteins) and a cellular 'diploid' component (tumor-associated fibroblasts, capillary-associated cells and inflammatory cells), in addition to the ever-evolving neoplastic cells. With these realizations, it has become evident that early and persistent inflammatory responses observed in or around many solid tumors, play important roles in establishing an environment suitable for neoplastic progression by providing diverse factors that alter tissue homeostasis. Using cutaneous melanoma and squamous cell carcinoma as tumor models, we review the current literature focussing on inflammatory and tumor-associated fibroblast responses as critical mediators of neoplastic progression for these malignancies.
Collapse
Affiliation(s)
- Léon C L T van Kempen
- University Medical Center Nijmegen, Department of Pathology, Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
1444
|
Affiliation(s)
- Fran Balkwill
- Cancer Research UK Translational Oncology Laboratory, Barts and The London, Queen Mary's Medical School, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
1445
|
Murdoch C, Giannoudis A, Lewis CE. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 2004; 104:2224-34. [PMID: 15231578 DOI: 10.1182/blood-2004-03-1109] [Citation(s) in RCA: 678] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The mechanisms responsible for recruiting monocytes from the bloodstream into solid tumors are now well characterized. However, recent evidence has shown that these cells then differentiate into macrophages and accumulate in large numbers in avascular and necrotic areas where they are exposed to hypoxia. This parallels their tendency to congregate in ischemic areas of other diseased tissues such as atherosclerotic plaques and arthritic joints. In tumors, macrophages appear to undergo marked phenotypic changes when exposed to hypoxia and to switch on their expression of a number of mitogenic and proangiogenic cytokines and enzymes. This then promotes tumor growth, angiogenesis, and metastasis. Here, we compare the various mechanisms responsible for monocyte recruitment into tumors with those regulating the accumulation of macrophages in hypoxic/necrotic areas. Because the latter are best characterized in human tumors, we focus mainly on these but also discuss their relevance to macrophage migration in ischemic areas of other diseased tissues. Finally, we discuss the relevance of these mechanisms to the development of novel cancer therapies, both in providing targets to reduce the proangiogenic contribution made by hypoxic macrophages in tumors and in developing the use of macrophages to deliver therapeutic gene constructs to hypoxic areas of diseased tissues.
Collapse
Affiliation(s)
- Craig Murdoch
- Tumor Targeting Group, Academic Unit of Pathology, Division of Genomic Medicine, University of Sheffield Medical School, Beech Hill Rd, Sheffield S10 2RX, United Kingdom
| | | | | |
Collapse
|
1446
|
Lin EY, Pollard JW. Role of infiltrated leucocytes in tumour growth and spread. Br J Cancer 2004; 90:2053-8. [PMID: 15164120 PMCID: PMC2410285 DOI: 10.1038/sj.bjc.6601705] [Citation(s) in RCA: 197] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2003] [Accepted: 01/19/2004] [Indexed: 12/11/2022] Open
Abstract
Leucocytes are a major component of the tumour microenvironment. Recent studies have indicated that the infiltration and activity of these host cells are regulated by the tumour to promote its survival and progression. Through the production of an array of growth factors, proteases and angiogenic mediators, leucocytes in the tumour microenvironment promote tumour growth, angiogenesis and metastasis.
Collapse
Affiliation(s)
- E Y Lin
- Departments of Developmental and Molecular Biology and Obstetrics and Gynecology and Women's Health, Center for the Study of Reproductive Biology and Women's Health and Albert Einstein Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | - J W Pollard
- Departments of Developmental and Molecular Biology and Obstetrics and Gynecology and Women's Health, Center for the Study of Reproductive Biology and Women's Health and Albert Einstein Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| |
Collapse
|
1447
|
Esposito I, Menicagli M, Funel N, Bergmann F, Boggi U, Mosca F, Bevilacqua G, Campani D. Inflammatory cells contribute to the generation of an angiogenic phenotype in pancreatic ductal adenocarcinoma. J Clin Pathol 2004; 57:630-636. [PMID: 15166270 PMCID: PMC1770337 DOI: 10.1136/jcp.2003.014498] [Citation(s) in RCA: 193] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2003] [Indexed: 01/07/2023]
Abstract
BACKGROUND Inflammatory cells contribute to the growth and spread of human malignancies by producing molecules that enhance tumour invasiveness. AIMS To characterise the inflammatory infiltrate in pancreatic ductal adenocarcinoma and to analyse its contribution to angiogenesis and its prognostic relevance. METHODS Immunohistochemistry was used to identify inflammatory cells and evaluate the expression of proangiogenic and prolymphangiogenic molecules (vascular endothelial growth factor A (VEGF-A), VEGF-C, and basic fibroblast growth factor (bFGF)) by inflammatory and cancer cells in 137 pancreatic cancers. Intratumorous microvessel density (IMD) was assessed using CD34 as an endothelial cell marker. RESULTS There were significantly more mast cells and macrophages in pancreatic cancers than in normal pancreas and the number of mast cells directly correlated with the presence of lymph node metastases. However, there was no relation between numbers of infiltrating inflammatory cells and the presence of chronic pancreatitis (CP)-like changes in the parenchyma surrounding the tumour. Double immunostaining revealed that both pancreatic mast cells and macrophages express VEGF-A, VEGF-C, and bFGF. These factors were also expressed in the tumour cells in many cases. The numbers of VEGF-A expressing tumour cells and bFGF expressing tumour and inflammatory cells significantly correlated with IMD. Moreover, tumours with higher IMD had higher numbers of infiltrating mast cells and macrophages. CONCLUSIONS Mononuclear inflammatory cells of the non-specific immune response are recruited to pancreatic cancer tissues independent of the presence of CP-like changes, may influence the metastatic capacity of the cancer cells, and may contribute to the development of tumours with high angiogenic activity.
Collapse
Affiliation(s)
- I Esposito
- Institute of Pathology, University of Heidelberg, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
1448
|
Gordillo GM, Onat D, Stockinger M, Roy S, Atalay M, Beck FM, Sen CK. A key angiogenic role of monocyte chemoattractant protein-1 in hemangioendothelioma proliferation. Am J Physiol Cell Physiol 2004; 287:C866-73. [PMID: 15163622 DOI: 10.1152/ajpcell.00238.2003] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Angiomatous lesions are common in infants and children. Hemangioendotheliomas (HE) represent one type of these lesions. Endothelial cell proliferation and the development of vascular/blood cell-filled spaces are inherent in the growth of HE. Therefore, understanding mechanisms that regulate the proliferation of these lesions should provide key insight into mechanisms regulating angiogenesis. A murine model was used to test the significance of monocyte chemoattractant protein (MCP)-1 in HE proliferation. EOMA cells, a cell line derived from a spontaneously arising murine HE, generate these lesions with 100% efficiency when injected subcutaneously into syngeneic mice. MCP-1 produced by EOMA cells recruit macrophages, which were shown to induce angiogenic behavior in EOMA cells by stimulating transwell migration and inducing sprout formation on type I collagen gels. When EOMA cells were injected into MCP-1(-/-) mice, only 50% of the mice developed tumors, presumably because the low levels of MCP-1 expressed by the injected EOMA cells were enough to overcome any host deficits of this chemokine. When EOMA cells were coinjected with a neutralizing antibody to MCP-1, tumors failed to develop in any of the treated mice, including syngeneic 129P3, C57Bl/6 (wild type), and MCP-1(-/-). These results present the first evidence that MCP-1 is required for HE proliferation and may promote the growth of these lesions by stimulating angiogenic behavior of endothelial cells. This study has produced the first in vivo evidence of a complete response for any neoplasm, specifically a vascular proliferative lesion, to anti-MCP-1 therapy in animals with intact immune systems.
Collapse
Affiliation(s)
- Gayle M Gordillo
- Division of Plastic Surgery, The Ohio State University Medical Center, 410 W. 10th Ave., Columbus, OH 43210, USA.
| | | | | | | | | | | | | |
Collapse
|
1449
|
Koga F, Kageyama Y, Kawakami S, Fujii Y, Hyochi N, Ando N, Takizawa T, Saito K, Iwai A, Masuda H, Kihara K. Prognostic significance of endothelial Per-Arnt-sim domain protein 1/hypoxia-inducible factor-2alpha expression in a subset of tumor associated macrophages in invasive bladder cancer. J Urol 2004; 171:1080-4. [PMID: 14767275 DOI: 10.1097/01.ju.0000110541.62972.08] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Endothelial Per-Arnt-Sim domain protein 1 (EPAS1) is induced under hypoxia and it transactivates a series of genes involved in angiogenesis and energy metabolism. Recent studies showed that EPAS1 is expressed in tumor associated macrophages (TAMs), which have multifaceted roles in tumor progression. We hypothesized that EPAS1 expressed in TAMs may contribute to bladder cancer progression. MATERIALS AND METHODS Clinicopathological and followup data on 69 patients undergoing radical cystectomy for T1-4N0-2M0 high grade bladder urothelial carcinoma were reviewed. Quantitative immunohistochemical analysis of TAMs and EPAS1 was performed separately in invasive front and in other superficial parts of carcinoma tissues. TAM counts and EPAS1 positive cell counts were compared with pathological variables and cancer specific survival (CSS). RESULTS The 5-year CSS rate in the 69 patients was 69% at a median followup of 58 months (range 2 to 196). EPAS1 expression was restricted to a small subset of TAMs. Although TAM counts were not associated with T stage or lymph node metastasis, EPAS1 expressing TAM counts were significantly associated with higher T stage. On univariate and multivariate analyses higher EPAS1 expressing TAM counts in invasive front along with higher T stage and positive lymph node metastasis were significantly associated with shorter CSS, while total TAM counts or EPAS1 expressing TAM counts in other superficial parts did not. CONCLUSIONS Despite limited prognostic effects of total TAMs EPAS1 expressing TAMs were associated with a poor prognosis of invasive bladder cancer, suggesting that EPAS1 expressed in a subset of TAMs mediates bladder cancer progression.
Collapse
Affiliation(s)
- Fumitaka Koga
- Department of Urology and Reproductive Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1450
|
Zielinski MR, Muenchow M, Wallig MA, Horn PL, Woods JA. Exercise delays allogeneic tumor growth and reduces intratumoral inflammation and vascularization. J Appl Physiol (1985) 2004; 96:2249-56. [PMID: 15020578 PMCID: PMC3645346 DOI: 10.1152/japplphysiol.01210.2003] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This investigation determined whether daily strenuous exercise would alter the progression and regression of an allogeneic lymphoid tumor in mice. We also determined whether exercise would alter the cellular composition and vascularity of the tumor. Female BALB/c mice (age 6-8 wk) were randomly assigned to sedentary control (Con) or daily exercised groups (EXH). EXH mice ran on a treadmill at incremental speeds (20-40 m/min) for 3 h or until fatigue. Each mouse was subcutaneously injected with 20 x 10(6) EL-4 lymphoma cells immediately after the first exercise bout (day 1) and run daily. Tumor volume was measured daily with calipers. In some experiments, mice were euthanized on days 5-10, 12, and 14. Tumors were excised and stained with hematoxylin and eosin or for Factor VIII-associated antigen using immunohistochemistry and analyzed in a blinded fashion under a light microscope. There was no significant treatment main effect found for tumor volumes. Interestingly, a significant treatment x time interaction was found, such that there was a 2-day delay in peak tumor volume and a more rapid tumor regression in EXH. Tumors isolated from Con exhibited significantly higher numbers of apoptotic bodies, blood vessels, macrophages, and neutrophils when compared with EXH. Intratumoral lymphocytes were higher in Con early in tumor growth but higher in EXH at peak tumor size. These data indicate that daily strenuous exercise may influence tumor growth by affecting the microenvironment of the tumor, resulting in a delay in tumor growth and a more rapid regression.
Collapse
Affiliation(s)
- Mark R Zielinski
- Department of Kinesiology, University of Illinois, Urbana, IL 61801, USA
| | | | | | | | | |
Collapse
|