Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yves A. DeClerck is active.

Publication


Featured researches published by Yves A. DeClerck.


Journal of Biological Chemistry | 1998

Tissue Inhibitor of Metalloproteinase-2 (TIMP-2) Binds to the Catalytic Domain of the Cell Surface Receptor, Membrane Type 1-Matrix Metalloproteinase 1 (MT1-MMP)

Stanley Zucker; Michelle Drews; Cathleen Conner; Hussein D. Foda; Yves A. DeClerck; Keith E. Langley; Wadie F. Bahou; Andrew J. P. Docherty; Jian Cao

It has been proposed that tissue inhibitor of metalloproteinase-2 (TIMP-2), in stoichiometric concentrations, serves as an intermediate in progelatinase A activation by binding to activated membrane type 1-matrix metalloproteinase 1 (MT1-MMP) on the plasma membrane. An MT1-MMP-independent cell surface receptor for TIMP-2 has also been postulated. To clarify TIMP-2 binding, we have performed 125I-TIMP-2 binding studies on transfected COS-1 cells and endothelial cells. Specific receptors for TIMP-2 were identified on COS-1 cells transfected with MT1-MMP cDNA, but not on vector-transfected cells. Treatment of MT1-MMP transfected COS-1 cells with a hydroxamic acid inhibitor of MMPs, CT-1746, but not an inactive stereoisomer, CT-1915, produced dose-dependent inhibition of specific TIMP-2 binding comparable with that noted with excess unlabeled TIMP-2. This result suggests that TIMP-2 binds to the zinc catalytic site of MT1-MMP. As demonstrated by the limited competition for binding of C-terminal deleted TIMP-2, the C-terminal domain of TIMP-2 participates in binding to MT1-MMP. Cross-linking studies followed by immunoprecipitation using antibodies to MT1-MMP were employed to identify 125I-TIMP-2·MT1-MMP complexes in MT1-MMP-transfected COS-1 cell membrane extracts. TIMP-2 receptors were also identified on concanavalin A-treated human umbilical vein endothelial cells; inhibition of TIMP-2 binding with CT-1746 was demonstrated.


Cancer Research | 2012

Tumor Microenvironment Complexity: Emerging Roles in Cancer Therapy

Melody A. Swartz; Noriho Iida; Edward Roberts; Sabina Sangaletti; Melissa H. Wong; Fiona E. Yull; Lisa M. Coussens; Yves A. DeClerck

The tumor microenvironment (TME) consists of cells, soluble factors, signaling molecules, extracellular matrix, and mechanical cues that can promote neoplastic transformation, support tumor growth and invasion, protect the tumor from host immunity, foster therapeutic resistance, and provide niches for dormant metastases to thrive. An American Association for Cancer Research (AACR) special conference held on November 3-6, 2011, addressed five emerging concepts in our understanding of the TME: its dynamic evolution, how it is educated by tumor cells, pathways of communication between stromal and tumor cells, immunomodulatory roles of the lymphatic system, and contribution of the intestinal microbiota. These discussions raised critical questions on how to include the analysis of the TME in personalized cancer diagnosis and treatment.


European Journal of Cancer | 2010

Interleukin-6 in Bone Metastasis and Cancer Progression

Tasnim Ara; Yves A. DeClerck

The bone and bone marrow are among the most frequent sites of cancer metastasis. It is estimated that 350,000 patients die with bone metastases annually in the United States. The ability of tumor cells to colonize the bone marrow and invade the bone is the result of close interactions between tumor cells and the bone marrow microenvironment. In this article, we review the contribution of interleukin-6 (IL-6) produced in the bone marrow microenvironment to bone metastasis. This cytokine has a strong pro-tumorigenic activity due to its multiple effects on bone metabolism, tumor cell proliferation and survival, angiogenesis, and inflammation. These effects are mediated by several signaling pathways, in particular the Janus kinase/signal transducer and transcription activator (JAK/STAT-3), Ras/mitogen activated protein kinase (MAPK), and phosphoinositol-3 kinase (PI3K)-protein kinase B/Akt (PkB/Akt), which are activated by IL-6 and amplified in the presence of soluble IL-6 receptor (sIL-6R). Supporting the role of IL-6 in human cancer is the observation of elevated serum levels of IL-6 and sIL-6R in patients with bone metastasis and their association with a poor clinical outcome. Over the last decade several large (monoclonal antibodies) and small (inhibitors of IL-6 mediated signaling) molecules that inhibit IL-6 activity in preclinical models have been developed. Several of these inhibitors are now undergoing phases I and II clinical trials, which will determine their inclusion in the list of effective targeted agents in the fight against cancer.


Cancer and Metastasis Reviews | 2010

Bone marrow-derived mesenchymal stem cells and the tumor microenvironment

Scott A. Bergfeld; Yves A. DeClerck

Over the last decade, there has been a growing interest in the role of mesenchymal stem cells (MSC) in cancer progression. These cells have the potential to give rise to a variety of mesenchymal cells like osteoblasts, chondrocytes, adipocytes, fibroblasts, and muscle cells. In contrast to their hematopoetic counterparts, MSC are not as clearly defined, which makes the interpretation of their role in cancer progression more complex. However, the nature of the relationship between MSC and tumor cells appears dual. Primary and metastatic tumors attract MSC in their microenvironment where they become tumor-associated fibroblasts, affect tumor cell survival and angiogenesis, and have an immunomodulatory function, and vice versa in the bone marrow MSC attract tumor cells and contribute to a microenvironment that promotes osteolysis, tumor growth, survival, and drug resistance. Whether MSC are pro- or anti-tumorigenic is a subject of controversial reports that is in part explained by the complexity of their interaction with tumor cells and the large range of cytokines and growth factors they produce. The study of these interactions is a fertile ground of investigation that—as already demonstrated in the case of myeloma—should lead to novel therapeutic approaches in cancer. In this article, the biology and role of MSC in cancer is reviewed with a primary focus on bone marrow-derived MSC.


American Journal of Pathology | 2004

Proteases, Extracellular Matrix, and Cancer: A Workshop of the Path B Study Section

Yves A. DeClerck; Arthur M. Mercurio; M. Sharon Stack; Harold A. Chapman; Mary M. Zutter; Ruth J. Muschel; Avraham Raz; Lynn M. Matrisian; Bonnie F. Sloane; Agnès Noël; Mary J.C. Hendrix; Lisa M. Coussens; Martin Padarathsingh

The role of the extracellular matrix (ECM) in the tumor microenvironment is not limited to being a barrier against tumor invasion. The ECM is a reservoir of cell binding proteins and growth factors that affect tumor cell behavior. It is also substantially modified by proteases produced by tumor cells or stroma cells. As a result of the activity of these proteases, cell-cell and cell-ECM interactions are altered, new biologically active ECM molecules are generated, and the bioavailability and activity of many growth factors, growth factor receptors, and cytokines are modified. ECM-degrading proteases also play a critical role in angiogenesis, where they can act as positive as well as negative regulators of endothelial cell proliferation and vascular morphogenesis. This review article summarizes some of the most relevant findings made over the recent years that were discussed at a workshop organized by the Path B Study Section of the National Institutes of Health in October 2002.


Cancer Research | 2004

Stromal Matrix Metalloproteinase-9 Regulates the Vascular Architecture in Neuroblastoma by Promoting Pericyte Recruitment

Christophe Chantrain; Hiroyuki Shimada; Sonata Jodele; Susan Groshen; Wei Ye; David R. Shalinsky; Zena Werb; Lisa M. Coussens; Yves A. DeClerck

Advanced stages of neuroblastoma show increased expression of matrix metalloproteinases MMP-2 and MMP-9 (Y. Sugiura et al., Cancer Res., 58: 2209–2216, 1998) that have been implicated in many steps of tumor progression, suggesting that they play a contributory role. Using pharmacological and genetic approaches, we have examined the role of these MMPs in progression of SK-N-BE (2).10 human neuroblastoma tumors orthotopically xenotransplanted into immunodeficient mice. Mice treated with Prinomastat, a synthetic inhibitor of MMPs, showed an inhibition of tumor cell proliferation in implanted tumors and a prolonged survival (50 versus 39 days in control group, P < 0.035). Treatment with Prinomastat did not affect formation of liver metastases (P = 0.52) but inhibited intravascular colonization by the tumor cells in the lung by 73.8% (P = 0.03) and angiogenesis in both primary tumors and experimental liver metastases. The primary tumors from Prinomastat-treated mice showed a 39.3% reduction in endothelial area detected by PECAM/CD31 staining in tumor sections (P < 0.001), primarily due to the presence of smaller vessels (P = 0.004). MMP-2 is expressed by neuroblastoma tumor cells and stromal cells, whereas MMP-9 is exclusively expressed by stromal cells, particularly vascular cells. To examine the contribution of MMP-9 to tumor angiogenesis, we generated RAG1/MMP-9 double-deficient mice. We observed a significant inhibition of angiogenesis in the immunodeficient RAG1/MMP-9 double-deficient mice orthotopically implanted with tumor cells (P = 0.043) or implanted s.c. with a mixture of tumor cells and Matrigel (P < 0.001). Using an FITC-labeled lectin, we demonstrated an inhibition in the architecture of the tumor vasculature in MMP-9-deficient mice, resulting in fewer and smaller blood vessels. These changes were associated with a 48% decrease in pericytes present along microvessels. Taken together, the data demonstrate that in neuroblastoma, stromally derived MMP-9 contributes to angiogenesis by promoting blood vessel morphogenesis and pericyte recruitment.


Cancer and Metastasis Reviews | 2006

Modifying the soil to affect the seed: role of stromal-derived matrix metalloproteinases in cancer progression

Sonata Jodele; Laurence Blavier; Janet M. Yoon; Yves A. DeClerck

In the 1980s, as the importance of matrix metalloproteinases (MMPs) in cancer progression was discovered, it was recognized that in most tumors these proteases were abundantly and sometimes exclusively expressed not by tumor cells, but by normal host-derived cells like fibroblasts, vascular endothelial cells, myofibroblasts, pericytes or inflammatory cells that contribute to the tumor microenvironment. Later experiments in mice deficient in specific MMPs revealed that host-derived MMPs play a critical role not only in tumor cell invasion, but also in carcinogenesis, angiogenesis, vasculogenesis and metastasis. Tumor cells secrete many factors, cytokines and chemokines that directly or indirectly increase the expression of these MMPs in the tumor microenvironment where they exert extracellular matrix (ECM) degrading and sheddase activities. The knowledge of the complex role that stromal-derived MMPs play in the interaction between tumor cells and stromal cells should allow us to consider specific windows in cancer treatment when MMP inhibition could have a valuable therapeutic effect.


Journal of Clinical Investigation | 2009

Vα24-invariant NKT cells mediate antitumor activity via killing of tumor-associated macrophages

Liping Song; Shahab Asgharzadeh; Jill Salo; Kelly Engell; Hong-Wei Wu; Richard Sposto; Tasnim Ara; Ayaka M. Silverman; Yves A. DeClerck; Robert C. Seeger; Leonid S. Metelitsa

Tumor infiltration with Valpha24-invariant NKT cells (NKTs) associates with favorable outcome in neuroblastoma and other cancers. Although NKTs can be directly cytotoxic against CD1d+ cells, the majority of human tumors are CD1d-. Therefore, the role of NKTs in cancer remains largely unknown. Here, we demonstrate that CD68+ tumor-associated monocytes/macrophages (TAMs) represented the majority of CD1d-expressing cells in primary human neuroblastomas. TAMs stimulated neuroblastoma growth in human cell lines and their xenografts in NOD/SCID mice via IL-6 production. Indeed, TAMs produced IL-6 in primary tumors and in the BM of patients with metastatic neuroblastoma. Gene expression analysis using TaqMan low-density arrays of 129 primary human neuroblastomas without MYCN amplification revealed that high-level expression of TAM-specific genes (CD14, CD16, IL6, IL6R, and TGFB1) was associated with poor 5-year event-free survival. While NKTs were not cytotoxic against neuroblastoma cells, they effectively killed monocytes pulsed with tumor cell lysate. The killing of monocytes was CD1d restricted because it was inhibited by a CD1d-specific mAb. Cotransfer of human monocytes and NKTs to tumor-bearing NOD/SCID mice decreased monocyte number at the tumor site and suppressed tumor growth compared with mice transferred with monocytes alone. Thus, killing of TAMs reveals what we believe to be a novel mechanism of NKT antitumor activity that relates to the disease outcome.


European Journal of Cancer | 2000

Interactions between tumour cells and stromal cells and proteolytic modification of the extracellular matrix by metalloproteinases in cancer

Yves A. DeClerck

Over the last few years a growing number of matrix degrading metalloproteinases have been implicated in cancer. These include in particular the matrix metalloproteinases (MMPs) which have been shown to be associated with a large variety of human malignancies, and the metalloproteinases with a disintegrin domain (ADAMs) whose potential role in cancer has begun to be examined. The expression of MMPs in human cancer is the result of a complex interaction between tumour cells and non-malignant stromal cells including fibroblasts, endothelial cells and inflammatory cells which all actively participate in the production of MMPs in tumour tissue. The proteolytic modification of the extracellular matrix by these proteases does more than allow malignant cells to locally invade and form distant metastasis. It significantly alters the tumour micro-environment and modifies the contacts between tumour cells and extracellular matrix proteins. These changes can affect essential cellular functions such as growth, survival, migration and even drug resistance. As our understanding of the nature of the contacts between tumour cells and a proteolytically modified extracellular matrix continues to progress, it is likely that novel therapeutic approaches to modify tumour cell behaviour will be identified.


Cancer Research | 2005

The Contribution of Bone Marrow-Derived Cells to the Tumor Vasculature in Neuroblastoma Is Matrix Metalloproteinase-9 Dependent

Sonata Jodele; Christophe Chantrain; Laurence Blavier; Carolyn Lutzko; Hiroyuki Shimada; Lisa M. Coussens; Yves A. DeClerck

The contribution of the tumor stroma to cancer progression has been increasingly recognized. We had previously shown that in human neuroblastoma tumors orthotopically implanted in immunodeficient mice, stromal-derived matrix metalloproteinase-9 (MMP-9) contributes to the formation of a mature vasculature by promoting pericyte recruitment along endothelial cells. Here we show that MMP-9 is predominantly expressed by bone marrow-derived CD45-positive leukocytes. Using a series of bone marrow transplantation experiments in MMP-9(+/+) and MMP-9(-/-) mice xenotransplanted with human neuroblastoma tumors, we show that bone marrow-derived MMP-9 is critical for the recruitment of leukocytes from bone marrow into the tumor stroma and for the integration of bone marrow-derived endothelial cells into the tumor vasculature. Expression of MMP-9 by bone marrow-derived cells in the tumor stroma is also critical for the formation of a mature vasculature and coverage of endothelial cells with pericytes. Furthermore, in primary human neuroblastoma tumor specimens of unfavorable histology, we observed a higher level of tumor infiltration with MMP-9 expressing phagocytic cells and a higher degree of coverage of endothelial cells by pericytes when compared with tumor specimens with a favorable histology. Taken together, the data show that in neuroblastoma, MMP-9 plays a critical role in the recruitment of bone marrow-derived cells to the tumor microenvironment where they positively contribute to angiogenesis and tumor progression.

Collaboration


Dive into the Yves A. DeClerck's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laurence Blavier

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Robert C. Seeger

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Walter E. Laug

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Tasnim Ara

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Richard Sposto

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Lucia Borriello

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Susan Groshen

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

C. Patrick Reynolds

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge