851
|
Abstract
The increasing incidence of obesity and its co-morbid conditions poses a great challenge to global health. In addition to cardiovascular disease and diabetes, epidemiological data demonstrate a link between obesity and multiple types of cancer. The molecular mechanisms underlying how obesity causes an increased risk of cancer are poorly understood. Obesity disrupts the dynamic role of the adipocyte in energy homeostasis, resulting in inflammation and alteration of adipokine (for example, leptin and adiponectin) signalling. Additionally, obesity causes secondary changes that are related to insulin signalling and lipid deregulation that may also foster cancer development. Understanding these molecular links may provide an avenue for preventive and therapeutic strategies to reduce cancer risk and mortality in an increasingly obese population.
Collapse
Affiliation(s)
- Melin J Khandekar
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
852
|
Rosenfeld S. Biomolecular self-defense and futility of high-specificity therapeutic targeting. GENE REGULATION AND SYSTEMS BIOLOGY 2011; 5:89-104. [PMID: 22272063 PMCID: PMC3236005 DOI: 10.4137/grsb.s8542] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Robustness has been long recognized to be a distinctive property of living entities. While a reasonably wide consensus has been achieved regarding the conceptual meaning of robustness, the biomolecular mechanisms underlying this systemic property are still open to many unresolved questions. The goal of this paper is to provide an overview of existing approaches to characterization of robustness in mathematically sound terms. The concept of robustness is discussed in various contexts including network vulnerability, nonlinear dynamic stability, and self-organization. The second goal is to discuss the implications of biological robustness for individual-target therapeutics and possible strategies for outsmarting drug resistance arising from it. Special attention is paid to the concept of swarm intelligence, a well studied mechanism of self-organization in natural, societal and artificial systems. It is hypothesized that swarm intelligence is the key to understanding the emergent property of chemoresistance.
Collapse
Affiliation(s)
- Simon Rosenfeld
- National Cancer Institute, EPN 3108, 6130 Executive Blvd., Rockville, Maryland 20892, USA
| |
Collapse
|
853
|
Yu D, Zhan XH, Zhao XF, Williams MS, Carey GB, Smith E, Scott D, Zhu J, Guo Y, Cherukuri S, Civin CI, Zhan X. Mice deficient in MIM expression are predisposed to lymphomagenesis. Oncogene 2011; 31:3561-8. [PMID: 22081072 DOI: 10.1038/onc.2011.509] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Missing in metastasis (MIM) is a member of newly emerged inverse Bin-Amphiphysin-Rvs (BAR) domain protein family and a putative metastasis suppressor. Although reduced MIM expression has been associated with bladder, breast and gastric cancers, evidence for the role of MIM in tumor progression remains scarce and controversial. Herein we characterized a MIM knockout mouse strain and observed that MIM-deficient mice often developed enlarged spleens. Autopsy and histological analysis revealed that nearly 78% of MIM(-/-) mice developed tumors with features similar to diffuse large B lymphoma during a period from 1 to 2 years. MIM(-/-) mice also exhibited abnormal distribution of B cells in lymphoid organs with decrease in the spleen but increase in the bone marrow and the peripheral blood. Furthermore, the bone marrow of MIM(-/-) mice contained a higher percentage of pre-B2 cells but fewer immature B-cells than wild-type mice. In response to CXCL13, a B-cell chemokine released from splenic stromal cells, MIM-deficient B-cells did not undergo chemotaxis or morphological changes in response to the chemokine and also did not internalize CXCR5, the receptor of CXCL13. Microarray analyses demonstrated that MIM is the only member of the I-BAR domain family that was highly expressed in human B cells. However, low or absent MIM expression was common in either primary B-cell malignancies or established B-cell acute lymphocytic leukemia or lymphomas. Thus, our data demonstrate for the first time an important role for MIM in B-cell development and suggest that predisposition of MIM-null mice to lymphomagenesis may involve aberrant interactions between B lineage cells and the lymphoid microenvironment.
Collapse
Affiliation(s)
- D Yu
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
854
|
Goulet B, Chan G, Chambers AF, Lewis JD. An emerging role for the nuclear localization of maspin in the suppression of tumor progression and metastasis. Biochem Cell Biol 2011; 90:22-38. [PMID: 22047058 DOI: 10.1139/o11-053] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Maspin, a member of the serpin family of serine protease inhibitors, was originally identified as a tumor suppressor that is expressed in normal mammary epithelial cells but is reduced or absent in breast carcinomas. Early enthusiasm for maspin as a biomarker for disease progression has been tempered by clinical data that associates maspin with favourable outcomes in some studies and poor prognosis in others. Here, we review all of the published clinical studies for maspin in breast and ovarian cancers and propose that the apparent discordance between clinical reports is a consequence of differential cellular distribution of maspin. Indeed, it was thought that an extracellular pool of maspin possessed tumor suppressor activity, acting by inhibiting migration and increasing cell adhesion. Recent evidence from our group and others indicates, however, that the nuclear localization of maspin in cancer cells is necessary for its tumor suppressor activity. We provide additional data here to demonstrate that nuclear-localized maspin binds to chromatin and is required to effectively prevent cells from metastasizing. Our knowledge of other serpins that localize to the nucleus should help to inform future studies of nuclear maspin. Elucidation of the molecular mechanisms regulating the localization and activities of maspin should pave the way for the development of improved diagnostics and therapies for cancer.
Collapse
Affiliation(s)
- Brigitte Goulet
- London Regional Cancer Program, Translational Prostate Cancer Research Group, London, ON N6A 4L6, Canada
| | | | | | | |
Collapse
|
855
|
Wolf K, Friedl P. Extracellular matrix determinants of proteolytic and non-proteolytic cell migration. Trends Cell Biol 2011; 21:736-44. [PMID: 22036198 DOI: 10.1016/j.tcb.2011.09.006] [Citation(s) in RCA: 235] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 09/20/2011] [Accepted: 09/22/2011] [Indexed: 11/25/2022]
Abstract
Cell invasion into the 3D extracellular matrix (ECM) is a multistep biophysical process involved in inflammation, tissue repair, and metastatic cancer invasion. Migrating cells navigate through tissue structures of complex and often varying physicochemical properties, including molecular composition, porosity, alignment and stiffness, by adopting strategies that involve deformation of the cell and engagement of matrix-degrading proteases. We review how the ECM determines whether or not pericellular proteolysis is required for cell migration, ranging from protease-driven invasion and secondary tissue destruction, to non-proteolytic, non-destructive movement that solely depends on cell deformability and available tissue space. These concepts call for therapeutic targeting of proteases to prevent invasion-associated tissue destruction rather than the migration process per se.
Collapse
Affiliation(s)
- Katarina Wolf
- Department of Cell Biology, Nijmegen Center for Molecular Life Science, Radboud University Nijmegen, 6500 HB Nijmegen, The Netherlands.
| | | |
Collapse
|
856
|
Fujita Y. Interface between normal and transformed epithelial cells: a road to a novel type of cancer prevention and treatment. Cancer Sci 2011; 102:1749-55. [PMID: 21692919 PMCID: PMC11159947 DOI: 10.1111/j.1349-7006.2011.02011.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell transformation arises from activation of oncoproteins and ⁄ or inactivation of tumor suppressor proteins. During the initial stage of carcinogenesis, transformation occurs in a single cell within an epithelial monolayer. However, it is not known what happens at the interface between normal and transformed cells once the initial transformation has occurred. Using elaborate cell culture systems, recent reports have shown that interactions between normal and transformed epithelial cells can induce various phenomena. For example, when Ras- or Src-transformed cells are surrounded by normal epithelial cells, multiple signaling pathways are activated and the transformed cells are apically extruded from the epithelium. In addition, normal and certain types of transformed cells compete with each other for cell survival, and the transformed cells undergo apoptosis. Importantly, when transformed cells alone are present, neither apoptosis nor elimination from epithelia occurs, indicating that the presence of surrounding normal cells influences the signaling pathways and fate of transformed cells. Comparable phenomena are also observed in zebrafish and mice in vivo model systems. In this review, I will introduce this newly emerging research field and discuss how these studies can potentially lead to establishment of novel types of cancer prevention and treatment.
Collapse
Affiliation(s)
- Yasuyuki Fujita
- MRC Laboratory for Molecular Cell Biology and Cell Biology Unit, Department of Cell and Developmental Biology, University College London, London, UK.
| |
Collapse
|
857
|
Arcangeli A. Ion channels and transporters in cancer. 3. Ion channels in the tumor cell-microenvironment cross talk. Am J Physiol Cell Physiol 2011; 301:C762-71. [DOI: 10.1152/ajpcell.00113.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The traditional view of cancer as a collection of proliferating cells must be reconsidered, and cancer must be viewed as a “tissue” constituted by both transformed cells and a heterogeneous microenvironment, that tumor cells construct and remodel during multistep tumorigenesis. The “tumor microenvironment” (TM) is formed by mesenchymal, endothelial, and immune cells immersed in a network of extracellular matrix (ECM) proteins and soluble factors. The TM strongly contributes to tumor progression, through long distance, cell-to-cell or cell-to-matrix signals, which influence different aspects of tumor cell behavior. Understanding the relationships among the different components of the cancer tissue is crucial to design and develop new therapeutic strategies. Ion channels are emerging as relevant players in the cross talk between tumor cells and their TM. Ion channels are expressed on tumor cells, as well as in the different cellular components of the TM. In all these cells, ion channels are in a strategic position to sense and transmit extracellular signals into the intracellular machinery. Often, this transmission is mediated by integrin adhesion receptors, which can be functional partners of ion channels since they form molecular complexes with the channel protein in the context of the plasma membrane. The same relevant role is exerted by ion transporters, which also contribute to determine two facets of the cancer tissue: hypoxia and the acidic extracellular pH. On the whole, it is conceivable to prospect the targeting of ion channels for new therapeutic strategies aimed at better controlling the malignant progression of the cancer tissue.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental Pathology and Oncology, University of Firenze, and Istituto Toscano Tumori, Firenze, Italy
| |
Collapse
|
858
|
Liguori M, Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages as incessant builders and destroyers of the cancer stroma. Cancers (Basel) 2011; 3:3740-61. [PMID: 24213109 PMCID: PMC3763394 DOI: 10.3390/cancers3043740] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 09/15/2011] [Accepted: 09/19/2011] [Indexed: 12/15/2022] Open
Abstract
Tumor-Associated Macrophages (TAM) are key components of the reactive stroma of tumors. In most, although not all cancers, their presence is associated with poor patient prognosis. In addition to releasing cytokines and growth factors for tumor and endothelial cells, a distinguished feature of TAM is their high-rate degradation of the extra-cellular matrix. This incessant stroma remodelling favours the release of matrix-bound growth factors and promotes tumor cell motility and invasion. In addition, TAM produce matrix proteins, some of which are typical of the neoplastic tissues. The gene expression profile of TAM isolated from human tumors reveals a matrix-related signature with the up-regulation of genes coding for different matrix proteins, as well as several proteolytic enzymes. Among ECM components are: osteopontin, osteoactivin, collagens and fibronectin, including also a truncated isoform of fibronectin termed migration stimulation factor. In addition to serve as structural proteins, these matrix components have key functions in the regulation of the vessel network, in the inductionof tumor cell motility and degradation of cellular debris. Among proteolytic enzymes are: matrix metalloproteases, cathepsins, lysosomal and ADAM proteases, and the urokinase-type plasminogen activator. The degrading activity of TAM, coupled to the production of bio-active ECM proteins, co-operate to the build-up and maintenance of an inflammatory micro-environment which eventually promotes tumor progression.
Collapse
Affiliation(s)
- Manuela Liguori
- Department of Immunology and Inflammation Istituto Clinico Humanitas, Via Manzoni 113, Rozzano-Milano 20089, Italy; E-Mails: (M.L.); (S.G.); (G.G.); (M.A.)
| | - Graziella Solinas
- Department of Immunology and Inflammation Istituto Clinico Humanitas, Via Manzoni 113, Rozzano-Milano 20089, Italy; E-Mails: (M.L.); (S.G.); (G.G.); (M.A.)
| | - Giovanni Germano
- Department of Immunology and Inflammation Istituto Clinico Humanitas, Via Manzoni 113, Rozzano-Milano 20089, Italy; E-Mails: (M.L.); (S.G.); (G.G.); (M.A.)
| | - Alberto Mantovani
- Department of Immunology and Inflammation Istituto Clinico Humanitas, Via Manzoni 113, Rozzano-Milano 20089, Italy; E-Mails: (M.L.); (S.G.); (G.G.); (M.A.)
- Department of Translational Medicine, University of Milano, Milano 20089, Italy
| | - Paola Allavena
- Department of Immunology and Inflammation Istituto Clinico Humanitas, Via Manzoni 113, Rozzano-Milano 20089, Italy; E-Mails: (M.L.); (S.G.); (G.G.); (M.A.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +39-02-8224-5112; Fax: +39-02-8224-5101
| |
Collapse
|
859
|
Chlenski A, Guerrero LJ, Salwen HR, Yang Q, Tian Y, Morales La Madrid A, Mirzoeva S, Bouyer PG, Xu D, Walker M, Cohn SL. Secreted protein acidic and rich in cysteine is a matrix scavenger chaperone. PLoS One 2011; 6:e23880. [PMID: 21949685 PMCID: PMC3174944 DOI: 10.1371/journal.pone.0023880] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 07/26/2011] [Indexed: 11/23/2022] Open
Abstract
Secreted Protein Acidic and Rich in Cysteine (SPARC) is one of the major non-structural proteins of the extracellular matrix (ECM) in remodeling tissues. The functional significance of SPARC is emphasized by its origin in the first multicellular organisms and its high degree of evolutionary conservation. Although SPARC has been shown to act as a critical modulator of ECM remodeling with profound effects on tissue physiology and architecture, no plausible molecular mechanism of its action has been proposed. In the present study, we demonstrate that SPARC mediates the disassembly and degradation of ECM networks by functioning as a matricellular chaperone. While it has low affinity to its targets inside the cells where the Ca(2+) concentrations are low, high extracellular concentrations of Ca(2+) activate binding to multiple ECM proteins, including collagens. We demonstrated that in vitro, this leads to the inhibition of collagen I fibrillogenesis and disassembly of pre-formed collagen I fibrils by SPARC at high Ca(2+) concentrations. In cell culture, exogenous SPARC was internalized by the fibroblast cells in a time- and concentration-dependent manner. Pulse-chase assay further revealed that internalized SPARC is quickly released outside the cell, demonstrating that SPARC shuttles between the cell and ECM. Fluorescently labeled collagen I, fibronectin, vitronectin, and laminin were co-internalized with SPARC by fibroblasts, and semi-quantitative Western blot showed that SPARC mediates internalization of collagen I. Using a novel 3-dimensional model of fluorescent ECM networks pre-deposited by live fibroblasts, we demonstrated that degradation of ECM depends on the chaperone activity of SPARC. These results indicate that SPARC may represent a new class of scavenger chaperones, which mediate ECM degradation, remodeling and repair by disassembling ECM networks and shuttling ECM proteins into the cell. Further understanding of this mechanism may provide insight into the pathogenesis of matrix-associated disorders and lead to the novel treatment strategies.
Collapse
Affiliation(s)
- Alexandre Chlenski
- Department of Pediatrics, University of Chicago, Chicago, Illinois, United States of America.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
860
|
Abstract
PURPOSE OF REVIEW This review intends to describe recent studies on the interaction between pancreatic cancer cells and tumor stroma, and potential opportunities and limitations to therapeutically targeting the stroma. RECENT FINDINGS Pancreatic cancer is characterized by densely desmoplastic stroma. It is becoming increasingly clear that there are complex and mutually supportive interactions between cancer cells and the stroma. Specific signaling pathways exist between cancer cells and cancer-associated fibroblasts that contribute to hypoxic desmoplasia. Recent developments in therapeutic approaches to targeting the stroma have demonstrated potential for enhancing efficacy of cytotoxic therapies. However, the heterogeneity and genomic complexity between tumors has also become more evident based on recent findings. There is increasing evidence for hierarchy of cancer cells with identification of a subpopulation of cancer stem cells that are inherently resistant to traditional therapies. SUMMARY Targeting pancreatic cancer stroma is a novel therapeutic strategy that appears justified based on recent studies; however, continued focus is needed to develop more effective therapies against cells resistant to standard chemotherapy.
Collapse
|
861
|
Fröhlich C, Nehammer C, Albrechtsen R, Kronqvist P, Kveiborg M, Sehara-Fujisawa A, Mercurio AM, Wewer UM. ADAM12 produced by tumor cells rather than stromal cells accelerates breast tumor progression. Mol Cancer Res 2011; 9:1449-61. [PMID: 21875931 DOI: 10.1158/1541-7786.mcr-11-0100] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Expression of ADAM12 is low in most normal tissues but is markedly increased in numerous human cancers, including breast carcinomas. We have previously shown that overexpression of ADAM12 accelerates tumor progression in a mouse model of breast cancer (PyMT). In this study, we found that ADAM12 deficiency reduces breast tumor progression in the PyMT model. However, the catalytic activity of ADAM12 seems to be dispensable for its tumor-promoting effect. Interestingly, we show that ADAM12 endogenously expressed in tumor-associated stroma in the PyMT model does not influence tumor progression, but that ADAM12 expression by tumor cells is necessary for tumor progression in these mice. This finding is consistent with our observation that in human breast carcinoma, ADAM12 is almost exclusively located in tumor cells and, only rarely, seen in the tumor-associated stroma. We hypothesized, however, that the tumor-associated stroma may stimulate ADAM12 expression in tumor cells, on the basis of the fact that TGF-β1 stimulates ADAM12 expression and is a well-known growth factor released from tumor-associated stroma. TGF-β1 stimulation of ADAM12-negative Lewis lung tumor cells induced ADAM12 synthesis, and growth of these cells in vivo induced more than 200-fold increase in ADAM12 expression. Our observation that ADAM12 expression is significantly higher in the terminal duct lobular units (TDLU) adjacent to human breast carcinoma compared with TDLUs found in normal breast tissue supports our hypothesis that tumor-associated stroma triggers ADAM12 expression.
Collapse
Affiliation(s)
- Camilla Fröhlich
- Department of Biomedical Sciences and Biotech Research & Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
862
|
The liver prometastatic reaction of cancer patients: implications for microenvironment-dependent colon cancer gene regulation. CANCER MICROENVIRONMENT 2011; 4:163-80. [PMID: 21870094 DOI: 10.1007/s12307-011-0084-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 08/08/2011] [Indexed: 02/07/2023]
Abstract
Colon cancer frequently metastasizes to the liver but the genetic and phenotypic properties of specific cancer cells able to implant and grow in this organ have not yet been established. The contribution of the patient's genetic, physiologic and pathologic backgrounds to the incidence and development of hepatic colon cancer metastases is also presently misunderstood. At a transcriptional level, hepatic metastasis development is in part associated with marked changes in gene expression of colon cancer cells that may originate in the primary tumor. Other changes occur in the liver and are regulated by hepatic cells, which represent the new microenvironment for metastatic colon cancer cells. However, hepatic parenchymal and non-parenchymal cell functions are also affected by both tumor-derived factors and systemic host factors, which suggests that the hepatic metastasis microenvironment is a functional linkage between the hepatic pathophysiology of the colon cancer patient and the biology of its cancer cells. Therefore, together with metastasis-related gene profiles suggesting the existence of liver metastasis potential in primary tumors, new biomarkers of the prometastatic microenvironment supported by the liver reaction to colon cancer factors may be helpful for the individual assessment of hepatic metastasis risk in colon cancer patients. In addition, knowledge on hepatic metastasis gene regulation by the hepatic microenvironment may open multiple opportunities for therapeutic intervention during colon cancer metastasis at both subclinical and advanced stages.
Collapse
|
863
|
Golubkov VS, Chernov AV, Strongin AY. Intradomain cleavage of inhibitory prodomain is essential to protumorigenic function of membrane type-1 matrix metalloproteinase (MT1-MMP) in vivo. J Biol Chem 2011; 286:34215-23. [PMID: 21832072 DOI: 10.1074/jbc.m111.264036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Invasive cancers use pericellular proteolysis to breach the extracellular matrix and basement membrane barriers and invade the surrounding tissue. Proinvasive membrane type-1 matrix metalloproteinase (MT1-MMP) is the primary mediator of proteolytic events on the cancer cell surface. MT1-MMP is synthesized as a zymogen. The latency of MT1-MMP is maintained by its N-terminal inhibitory prodomain. In the course of MT1-MMP activation, the R(108)RKR(111) ↓ Y(112) prodomain sequence is processed by furin. The intact prodomain released by furin alone, however, is a potent inhibitor of the emerging MT1-MMP enzyme. Evidence suggests that the prodomain undergoes intradomain cleavage at the PGD ↓ L(50) site followed by the release of the degraded prodomain by furin cleavage that finalizes the two-step activation event. These cleavages, only if combined, cause the activation of MT1-MMP. The significance of the intradomain cleavage in the protumorigenic program of MT1-MMP, however, remained unidentified. To identify this important parameter, in our current study, we used the cells that expressed the wild-type prodomain-based fluorescent biosensor and the mutant biosensor with the inactivated PGD↓L(50) cleavage site (L50D mutant) and also the cells with the enforced expression of the wild-type and L50D mutant MT1-MMP. Using cell-based tests, orthotopic breast cancer xenografts in mice, and genome-wide transcriptional profiling of cultured cells and tumor xenografts, we demonstrated that the intradomain cleavage of the PGD ↓ L(50) sequence of the prodomain is essential for the protumorigenic function of MT1-MMP. Our results emphasize the importance of the intradomain cleavages resulting in the inactivation of the respective inhibitory prodomains not only for MT1-MMP but also for other MMP family members.
Collapse
Affiliation(s)
- Vladislav S Golubkov
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | | | | |
Collapse
|
864
|
Mandl SJ, Rountree RB, Dalpozzo K, Do L, Lombardo JR, Schoonmaker PL, Dirmeier U, Steigerwald R, Giffon T, Laus R, Delcayre A. Immunotherapy with MVA-BN®-HER2 induces HER-2-specific Th1 immunity and alters the intratumoral balance of effector and regulatory T cells. Cancer Immunol Immunother 2011; 61:19-29. [PMID: 21822917 PMCID: PMC3249207 DOI: 10.1007/s00262-011-1077-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 07/12/2011] [Indexed: 11/30/2022]
Abstract
MVA-BN®-HER2 is a new candidate immunotherapy designed for the treatment of HER-2-positive breast cancer. Here, we demonstrate that a single treatment with MVA-BN®-HER2 exerts potent anti-tumor efficacy in a murine model of experimental pulmonary metastasis. This anti-tumor efficacy occurred despite a strong tumor-mediated immunosuppressive environment characterized by a high frequency of regulatory T cells (Treg) in the lungs of tumor-bearing mice. Immunogenicity studies showed that treatment with MVA-BN®-HER2 induced strongly Th1-dominated HER-2-specific antibody and T-cell responses. MVA-BN®-HER2-induced anti-tumor activity was characterized by an increased infiltration of lungs with highly activated, HER-2-specific, CD8+CD11c+ T cells accompanied by a decrease in the frequency of Treg cells in the lung, resulting in a significantly increased ratio of effector T cells to Treg cells. In contrast, administration of HER2 protein formulated in Complete Freund’s Adjuvant (CFA) induced a strongly Th2-biased immune response to HER-2. However, this did not lead to significant infiltration of the tumor-bearing lungs by CD8+ T cells or the decrease in the frequency of Treg cells nor did it result in anti-tumor efficacy. In vivo depletion of CD8+ cells confirmed that CD8 T cells were required for the anti-tumor activity of MVA-BN®-HER2. Furthermore, depletion of CD4+ or CD25+ cells demonstrated that tumor-induced Treg cells promoted tumor growth and that CD4 effector cells also contribute to MVA-BN®-HER2-mediated anti-tumor efficacy. Taken together, our data demonstrate that treatment with MVA-BN®-HER2 controls tumor growth through mechanisms including the induction of Th1-biased HER-2-specific immune responses and the control of tumor-mediated immunosuppression.
Collapse
Affiliation(s)
- Stefanie J Mandl
- Department of Research, BN ImmunoTherapeutics, 2425 Garcia Ave, Mountain View, CA 94043, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
865
|
Abstract
Chemotherapeutic regimens involve the systemic administration of genotoxic compounds that induce cancer cell death via well-established DNA damage response signaling networks. Less understood is how the treatment of other cell types within the tumor microenvironment affects the therapeutic response. Here we discuss recent work that shows that tumor-adjacent cells can respond to genotoxic stress by activating a paracrine secretory program. Although this secretory response serves to protect progenitor cells and promote tissue regeneration in conditions of cellular stress, it can also be coopted by tumor cells to survive frontline chemotherapy. Thus, local prosurvival signaling may present a fundamental barrier to tumor clearance by genotoxic agents, suggesting that effective treatments need to target both cancer cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Luke A Gilbert
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | | |
Collapse
|
866
|
Arwert EN, Mentink RA, Driskell RR, Hoste E, Goldie SJ, Quist S, Watt FM. Upregulation of CD26 expression in epithelial cells and stromal cells during wound-induced skin tumour formation. Oncogene 2011; 31:992-1000. [PMID: 21765471 DOI: 10.1038/onc.2011.298] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have previously described InvEE transgenic mice in which non-dividing, differentiating epidermal cells express oncogenically activated MAPK kinase 1 (MEK1). Skin wounding triggers tumour formation in InvEE mice via a mechanism that involves epidermal release of IL-1α and attraction of a pro-tumorigenic inflammatory infiltrate. To look for potential effects on the underlying connective tissue, we screened InvEE and wild-type epidermis for differential expression of cytokines and immune modulators. We identified a single protein, CD26 (dipeptidyl peptidase-4). CD26 serum levels were not increased in InvEE mice. In contrast, CD26 was upregulated in keratinocytes expressing mutant MEK1 and in the epithelial compartment of InvEE tumours, where it accumulated at cell-cell borders. CD26 expression was increased in dermal fibroblasts following skin wounding but was downregulated in tumour stroma. CD26 activity was stimulated by calcium-induced intercellular adhesion in keratinocytes, suggesting that the upregulation of CD26 in InvEE epidermis is due to expansion of the differentiated cell layers. IL-1α treatment of dermal fibroblasts stimulated CD26 activity, and therefore epidermal IL-1α release may contribute to the upregulation of CD26 expression in wounded dermis. Pharmacological blockade of CD26, via Sitagliptin, reduced growth of InvEE tumours, while combined inhibition of IL-1α and CD26 delayed tumour onset and reduced tumour incidence. Our results demonstrate that inappropriate activation of MEK1 in the epidermis leads to changes in dermal fibroblasts that, like the skin inflammatory infiltrate, contribute to tumour formation.
Collapse
Affiliation(s)
- E N Arwert
- Cancer Research UK Cambridge Research Institute, Cambridge, UK
| | | | | | | | | | | | | |
Collapse
|
867
|
Nieto MA. The ins and outs of the epithelial to mesenchymal transition in health and disease. Annu Rev Cell Dev Biol 2011; 27:347-76. [PMID: 21740232 DOI: 10.1146/annurev-cellbio-092910-154036] [Citation(s) in RCA: 567] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The epithelial to mesenchymal transition (EMT) converts epithelial cells into migratory and invasive cells and is a fundamental event in morphogenesis. Although its relevance in the progression of cancer and organ fibrosis had been debated until recently, the EMT is now established as an important step in the metastatic cascade of epithelial tumors. The similarities between pathological and developmental EMTs validate the embryo as the best model to understand the molecular and cellular mechanisms involved in this process, identifying those that are hijacked during the progression of cancer and organ degeneration. Our ever-increasing understanding of how transcription factors regulate the EMT has revealed complex regulatory loops coupled to posttranscriptional and epigenetic regulatory programs. The EMT is now integrated into the systemic activities of whole organisms, establishing links with cell survival, stemness, inflammation, and immunity. In addition, the EMT now constitutes a promising target for the treatment of cancer and organ-degenerative diseases.
Collapse
Affiliation(s)
- M Angela Nieto
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, San Juan de Alicante 03550, Spain.
| |
Collapse
|
868
|
Guo J, Bourre L, Soden DM, O'Sullivan GC, O'Driscoll C. Can non-viral technologies knockdown the barriers to siRNA delivery and achieve the next generation of cancer therapeutics? Biotechnol Adv 2011; 29:402-17. [DOI: 10.1016/j.biotechadv.2011.03.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/08/2011] [Accepted: 03/13/2011] [Indexed: 12/22/2022]
|
869
|
Tumor-initiating stem cells of squamous cell carcinomas and their control by TGF-β and integrin/focal adhesion kinase (FAK) signaling. Proc Natl Acad Sci U S A 2011; 108:10544-9. [PMID: 21670270 DOI: 10.1073/pnas.1107807108] [Citation(s) in RCA: 206] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cancer stem cells (CSCs) sustain tumor growth through their ability to self-renew and to generate differentiated progeny. These functions endow CSCs with the potential to initiate secondary tumors bearing characteristics similar to those of the parent. Recently the hair follicle stem cell marker CD34 was used to purify a CSC-like cell population from early skin tumors arising from treatment with 7,12-dimethylbenz[α]anthracene/12-o-tetradecanoylphorbol-13-acetate, which typically generates benign papillomas that occasionally progress to squamous cell carcinomas (SCCs). In the present study, we identify and characterize CSCs purified from malignant SCCs. We show that SCCs contain two highly tumorigenic CSC populations that differ in CD34 levels but are enriched for integrins and coexist at the SCC-stroma interface. Intriguingly, whether CD34(lo) or CD34(hi), α6(hi)β1(hi) populations can initiate secondary tumors by serial limit-dilution transplantation assays, but α6(lo)β1(lo) populations cannot. Moreover, secondary tumors generated from a single CSC of either subtype contain both CD34(lo) and CD34(hi) α6(hi)β1(hi)CSCs, indicating their nonhierarchical organization. Genomic profiling and hierarchical cluster analysis show that these two CSC subtypes share a molecular signature distinct from either the CD34(-) epidermal or the CD34(hi) hair follicle stem cell signature. Although closely related, α6(hi)β1(hi)CD34(lo) and α6(hi)β1(hi)CD34(hi) CSCs differ in cell-cycle gene expression and proliferation characteristics. Indeed, proliferation and expansion of α6(hi)β1(hi)CD34(hi) CSCs is sensitive to whether they can initiate a TGF-β receptor II-mediated response to counterbalance elevated focal adhesion kinase-mediated integrin signaling within the tumor. Overall, the coexistence and interconvertibility of CSCs with differing sensitivities to their microenvironment pose challenges and opportunities for SCC cancer therapies.
Collapse
|
870
|
Parvani JG, Taylor MA, Schiemann WP. Noncanonical TGF-β signaling during mammary tumorigenesis. J Mammary Gland Biol Neoplasia 2011; 16:127-46. [PMID: 21448580 PMCID: PMC3723114 DOI: 10.1007/s10911-011-9207-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Accepted: 03/08/2011] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is a heterogeneous disease comprised of at least five major tumor subtypes that coalesce as the second leading cause of cancer death in women in the United States. Although metastasis clearly represents the most lethal characteristic of breast cancer, our understanding of the molecular mechanisms that govern this event remains inadequate. Clinically, ~30% of breast cancer patients diagnosed with early-stage disease undergo metastatic progression, an event that (a) severely limits treatment options, (b) typically results in chemoresistance and low response rates, and (c) greatly contributes to aggressive relapses and dismal survival rates. Transforming growth factor-β (TGF-β) is a pleiotropic cytokine that regulates all phases of postnatal mammary gland development, including branching morphogenesis, lactation, and involution. TGF-β also plays a prominent role in suppressing mammary tumorigenesis by preventing mammary epithelial cell (MEC) proliferation, or by inducing MEC apoptosis. Genetic and epigenetic events that transpire during mammary tumorigenesis conspire to circumvent the tumor suppressing activities of TGF-β, thereby permitting late-stage breast cancer cells to acquire invasive and metastatic phenotypes in response to TGF-β. Metastatic progression stimulated by TGF-β also relies on its ability to induce epithelial-mesenchymal transition (EMT) and the expansion of chemoresistant breast cancer stem cells. Precisely how this metamorphosis in TGF-β function comes about remains incompletely understood; however, recent findings indicate that the initiation of oncogenic TGF-β activity is contingent upon imbalances between its canonical and noncanonical signaling systems. Here we review the molecular and cellular contributions of noncanonical TGF-β effectors to mammary tumorigenesis and metastatic progression.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Disease Progression
- Epithelial-Mesenchymal Transition
- Female
- Humans
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Glands, Human/growth & development
- Mammary Glands, Human/metabolism
- Mammary Glands, Human/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Signal Transduction
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Jenny G Parvani
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
871
|
Grunewald TGP, Herbst SM, Heinze J, Burdach S. Understanding tumor heterogeneity as functional compartments--superorganisms revisited. J Transl Med 2011; 9:79. [PMID: 21619636 PMCID: PMC3118334 DOI: 10.1186/1479-5876-9-79] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 05/27/2011] [Indexed: 02/07/2023] Open
Abstract
Compelling evidence broadens our understanding of tumors as highly heterogeneous populations derived from one common progenitor. In this review we portray various stages of tumorigenesis, tumor progression, self-seeding and metastasis in analogy to the superorganisms of insect societies to exemplify the highly complex architecture of a neoplasm as a system of functional "castes." Accordingly, we propose a model in which clonal expansion and cumulative acquisition of genetic alterations produce tumor compartments each equipped with distinct traits and thus distinct functions that cooperate to establish clinically apparent tumors. This functional compartment model also suggests mechanisms for the self-construction of tumor stem cell niches. Thus, thinking of a tumor as a superorganism will provide systemic insight into its functional compartmentalization and may even have clinical implications.
Collapse
Affiliation(s)
- Thomas G P Grunewald
- Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, Munich, Germany.
| | | | | | | |
Collapse
|
872
|
Rodriguez-Canales J, Eberle FC, Jaffe ES, Emmert-Buck MR. Why is it crucial to reintegrate pathology into cancer research? Bioessays 2011; 33:490-8. [PMID: 21590787 DOI: 10.1002/bies.201100017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The integration of pathology with molecular biology is vital if we are to enhance the translational value of cancer research. Pathology represents a bridge between medicine and basic biology, it remains the gold standard for cancer diagnosis, and it plays an important role in discovery studies. In the past, pathology and cancer research were closely associated; however, the molecular biology revolution has shifted the focus of investigators toward the molecular alterations of tumors. The reductionist approach taken in molecular studies is producing great insight into the inner workings of neoplasia, but it can also minimize the importance of histopathology and of understanding the disease as a whole. In turn, pathologists can underestimate the role of molecular studies in developing new ancillary techniques for clinical diagnosis. A multidisciplinary approach that integrates pathology and molecular biology within a translational research system is needed. This process will require overcoming cultural barriers and can be achieved through education, a more effective incorporation of pathology into biological research, and conversely an integration of biological research into the pathology laboratory.
Collapse
Affiliation(s)
- Jaime Rodriguez-Canales
- Laser Capture Microdissection Core and Pathogenetics Unit, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | |
Collapse
|
873
|
Calvo F, Sahai E. Cell communication networks in cancer invasion. Curr Opin Cell Biol 2011; 23:621-9. [PMID: 21570276 DOI: 10.1016/j.ceb.2011.04.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 04/14/2011] [Accepted: 04/18/2011] [Indexed: 01/07/2023]
Abstract
The invasion of cancer is a major clinical problem. It is now apparent that invasion is not a simply a cancer cell autonomous process but relies on a complex network of paracrine interactions. Furthermore, this network can change as cancer cells disseminate. Here we summarise the key components of the network and their mechanisms of communication. Finally, we discuss the difficulties and opportunities that this complex network of interactions presents during cancer therapy.
Collapse
Affiliation(s)
- Fernando Calvo
- Tumour Cell Biology Laboratory, London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, UK
| | | |
Collapse
|
874
|
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144:646-74. [PMID: 21376230 DOI: 10.1016/j.cell.2011.02.013] [Citation(s) in RCA: 46535] [Impact Index Per Article: 3323.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 02/06/2011] [Accepted: 02/07/2011] [Indexed: 11/26/2022]
Abstract
The hallmarks of cancer comprise six biological capabilities acquired during the multistep development of human tumors. The hallmarks constitute an organizing principle for rationalizing the complexities of neoplastic disease. They include sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. Underlying these hallmarks are genome instability, which generates the genetic diversity that expedites their acquisition, and inflammation, which fosters multiple hallmark functions. Conceptual progress in the last decade has added two emerging hallmarks of potential generality to this list-reprogramming of energy metabolism and evading immune destruction. In addition to cancer cells, tumors exhibit another dimension of complexity: they contain a repertoire of recruited, ostensibly normal cells that contribute to the acquisition of hallmark traits by creating the "tumor microenvironment." Recognition of the widespread applicability of these concepts will increasingly affect the development of new means to treat human cancer.
Collapse
Affiliation(s)
- Douglas Hanahan
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne CH-1015, Switzerland.
| | | |
Collapse
|
875
|
Hurst DR, Welch DR. Metastasis suppressor genes at the interface between the environment and tumor cell growth. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 286:107-80. [PMID: 21199781 DOI: 10.1016/b978-0-12-385859-7.00003-3] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms and genetic programs required for cancer metastasis are sometimes overlapping, but components are clearly distinct from those promoting growth of a primary tumor. Every sequential, rate-limiting step in the sequence of events leading to metastasis requires coordinated expression of multiple genes, necessary signaling events, and favorable environmental conditions or the ability to escape negative selection pressures. Metastasis suppressors are molecules that inhibit the process of metastasis without preventing growth of the primary tumor. The cellular processes regulated by metastasis suppressors are diverse and function at every step in the metastatic cascade. As we gain knowledge into the molecular mechanisms of metastasis suppressors and cofactors with which they interact, we learn more about the process, including appreciation that some are potential targets for therapy of metastasis, the most lethal aspect of cancer. Until now, metastasis suppressors have been described largely by their function. With greater appreciation of their biochemical mechanisms of action, the importance of context is increasingly recognized especially since tumor cells exist in myriad microenvironments. In this chapter, we assemble the evidence that selected molecules are indeed suppressors of metastasis, collate the data defining the biochemical mechanisms of action, and glean insights regarding how metastasis suppressors regulate tumor cell communication to-from microenvironments.
Collapse
Affiliation(s)
- Douglas R Hurst
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
876
|
Squadrito ML, De Palma M. Macrophage regulation of tumor angiogenesis: Implications for cancer therapy. Mol Aspects Med 2011; 32:123-45. [DOI: 10.1016/j.mam.2011.04.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 04/27/2011] [Indexed: 12/24/2022]
|
877
|
Watt FM, Fujiwara H. Cell-extracellular matrix interactions in normal and diseased skin. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a005124. [PMID: 21441589 DOI: 10.1101/cshperspect.a005124] [Citation(s) in RCA: 258] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mammalian skin comprises a multi-layered epithelium, the epidermis, and an underlying connective tissue, the dermis. The epidermal extracellular matrix is a basement membrane, whereas the dermal ECM comprises fibrillar collagens and associated proteins. There is considerable heterogeneity in ECM composition within both epidermis and dermis. The functional significance of this extends beyond cell adhesion to a range of cell autonomous and nonautonomous processes, including control of epidermal stem cell fate. In skin, cell-ECM interactions influence normal homeostasis, aging, wound healing, and disease. Disturbed integrin and ECM signaling contributes to both tumor formation and fibrosis. Strategies for manipulating cell-ECM interactions to repair skin defects and intervene in a variety of skin diseases hold promise for the future.
Collapse
Affiliation(s)
- Fiona M Watt
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, United Kingdom.
| | | |
Collapse
|
878
|
Suetsugu A, Osawa Y, Nagaki M, Saji S, Moriwaki H, Bouvet M, Hoffman RM. Imaging the recruitment of cancer-associated fibroblasts by liver-metastatic colon cancer. J Cell Biochem 2011; 112:949-53. [DOI: 10.1002/jcb.23011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
879
|
Ilina O, Bakker GJ, Vasaturo A, Hofmann RM, Friedl P. Two-photon laser-generated microtracks in 3D collagen lattices: principles of MMP-dependent and -independent collective cancer cell invasion. Phys Biol 2011; 8:015010. [DOI: 10.1088/1478-3975/8/1/015010] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
880
|
Ewald AJ, Werb Z, Egeblad M. Dynamic, long-term in vivo imaging of tumor-stroma interactions in mouse models of breast cancer using spinning-disk confocal microscopy. Cold Spring Harb Protoc 2011; 2011:pdb.top97. [PMID: 21285277 PMCID: PMC3533368 DOI: 10.1101/pdb.top97] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
INTRODUCTIONTumors contain many components in addition to the cancer cells, including blood vessels, fibroblasts, and immune cells. Genetic studies and tumor biopsies have generated insights into the importance of these stromal components for cancer progression. However, it remains a challenge to reveal the dynamic interactions among the distinct tumor components within live animals. Studies involving multiphoton microscopy allow direct imaging of cellular movement in live mice, but multiphoton microscopy is expensive, complex, and usually relies on a single excitation wavelength for all fluorophores. This article describes a method for intravital imaging using a microlensed spinning-disk confocal microscope. Although tissue penetration with spinning-disk confocal microscopy is lower than with multiphoton microscopy, image acquisition with this method is very rapid, so artifacts from respiratory motion are avoided. Photobleaching and phototoxicity are low, and multicolor acquisition is cheaper and easier than with multiphoton microscopy. This article discusses various aspects of experimental setup, as well as methods for addressing technical barriers, such as generating and working with multiple tumor microenvironments within individual live mice, image collection, and long-term anesthesia.
Collapse
|
881
|
Veiseh M, Turley EA. Hyaluronan metabolism in remodeling extracellular matrix: probes for imaging and therapy of breast cancer. Integr Biol (Camb) 2011; 3:304-15. [PMID: 21264398 DOI: 10.1039/c0ib00096e] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Clinical and experimental evidence increasingly support the concept of cancer as a disease that emulates a component of wound healing, in particular abnormal stromal extracellular matrix remodeling. Here we review the biology and function of one remodeling process, hyaluronan (HA) metabolism, which is essential for wound resolution but closely linked to breast cancer (BCA) progression. Components of the HA metabolic cycle (HAS2, SPAM1 and HA receptors CD44, RHAMM/HMMR and TLR2) are discussed in terms of their known functions in wound healing and in breast cancer progression. Finally, we discuss recent advances in the use of HA-based platforms for developing nanoprobes to image areas of active HA metabolism and for therapeutics in breast cancer.
Collapse
Affiliation(s)
- M Veiseh
- Life Sciences Division, Lawrence Berkeley National Laboratories, Berkeley, CA, USA.
| | | |
Collapse
|
882
|
Abstract
The non-white blood cell mesenchymal elements of the intestinal lamina propria are the myofibroblasts, fibroblasts, pericytes, stromal stem cells, muscularis mucosae, and the smooth muscle of the villus core associated with the lymphatic lacteal. We review the functional anatomy of these mesenchymal cells, what is known about their origin in the embryo and their replacement in adults, their putative role in intestinal mucosal morphogenesis, and the intestinal stem cell niche, and we consider new information about myofibroblasts as nonprofessional immune cells. Although our knowledge of the function of mesenchymal cells in intestinal disease is rudimentary, we briefly consider here their roles in cancer and intestinal inflammation.
Collapse
Affiliation(s)
- I V Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555-0764, USA.
| | | | | | | |
Collapse
|
883
|
Abstract
The mesenchymal elements of the intestinal lamina propria reviewed here are the myofibroblasts, fibroblasts, mural cells (pericytes) of the vasculature, bone marrow-derived stromal stem cells, smooth muscle of the muscularis mucosae, and smooth muscle surrounding the lymphatic lacteals. These cells share similar marker molecules, origins, and coordinated biological functions previously ascribed solely to subepithelial myofibroblasts. We review the functional anatomy of intestinal mesenchymal cells and describe what is known about their origin in the embryo and their replacement in adults. As part of their putative role in intestinal mucosal morphogenesis, we consider the intestinal stem cell niche. Lastly, we review emerging information about myofibroblasts as nonprofessional immune cells that may be important as an alarm system for the gut and as a participant in peripheral immune tolerance.
Collapse
Affiliation(s)
- D.W. Powell
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0764
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-0764
| | - I.V. Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0764
| | - J.I. Saada
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0764
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94143
| | - R.C. Mifflin
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0764
| |
Collapse
|
884
|
de Sousa EMF, Vermeulen L, Richel D, Medema JP. Targeting Wnt Signaling in Colon Cancer Stem Cells. Clin Cancer Res 2010; 17:647-53. [DOI: 10.1158/1078-0432.ccr-10-1204] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
885
|
Tumor formation initiated by nondividing epidermal cells via an inflammatory infiltrate. Proc Natl Acad Sci U S A 2010; 107:19903-8. [PMID: 21041641 DOI: 10.1073/pnas.1007404107] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In mammalian epidermis, integrin expression is normally confined to the basal proliferative layer that contains stem cells. However, in epidermal hyperproliferative disorders and tumors, integrins are also expressed by suprabasal cells, with concomitant up-regulation of Erk mitogen-activated protein kinase (MAPK) signaling. In transgenic mice, expression of activated MAPK kinase 1 (MEK1) in the suprabasal, nondividing, differentiated cell layers (InvEE transgenics) results in epidermal hyperproliferation and skin inflammation. We now demonstrate that wounding induces benign tumors (papillomas and keratoacanthomas) in InvEE mice. By generating chimeras between InvEE mice and mice that lack the MEK1 transgene, we demonstrate that differentiating, nondividing cells that express MEK1 stimulate adjacent transgene-negative cells to divide and become incorporated into the tumor mass. Dexamethasone treatment inhibits tumor formation, suggesting that inflammation is involved. InvEE skin and tumors express high levels of IL1α; treatment with an IL1 receptor antagonist delays tumor onset and reduces incidence. Depletion of γδ T cells and macrophages also reduces tumor incidence. Because a hallmark of cancer is uncontrolled proliferation, it is widely assumed that tumors arise only from dividing cells. In contrast, our studies show that differentiated epidermal cells can initiate tumor formation without reacquiring the ability to divide and that they do so by triggering an inflammatory infiltrate.
Collapse
|
886
|
Gray RS, Cheung KJ, Ewald AJ. Cellular mechanisms regulating epithelial morphogenesis and cancer invasion. Curr Opin Cell Biol 2010; 22:640-50. [PMID: 20832275 DOI: 10.1016/j.ceb.2010.08.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 08/12/2010] [Accepted: 08/12/2010] [Indexed: 12/21/2022]
Abstract
The cellular mechanisms driving mammalian epithelial morphogenesis are of significant fundamental and practical interest. Historically, these processes have been difficult to study directly, owing to the opacity and relative inaccessibility of mammalian tissues. Recent experimental advances in timelapse imaging and in 3D organotypic culture have enabled direct observation of epithelial morphogenesis. In the mammary gland, branching morphogenesis is observed to proceed through a novel form of collective epithelial migration. The active unit of morphogenesis is a multilayered epithelium with reduced apico-basal polarity, within which cells rearranged vigorously. From within this multilayered state, new ducts initiate and elongate into the matrix without leading cellular extensions or dedicated leaders. We discuss the implications of these findings on our understanding of epithelial morphogenesis in other organs and in cancer progression.
Collapse
Affiliation(s)
- Ryan S Gray
- Department of Cell Biology, Johns Hopkins University, 855 N. Wolfe St, Rangos 452, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
887
|
Dynamic interplay between the collagen scaffold and tumor evolution. Curr Opin Cell Biol 2010; 22:697-706. [PMID: 20822891 DOI: 10.1016/j.ceb.2010.08.015] [Citation(s) in RCA: 675] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 08/10/2010] [Accepted: 08/11/2010] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) is a key regulator of cell and tissue function. Traditionally, the ECM has been thought of primarily as a physical scaffold that binds cells and tissues together. However, the ECM also elicits biochemical and biophysical signaling. Controlled proteolysis and remodeling of the ECM network regulate tissue tension, generate pathways for migration, and release ECM protein fragments to direct normal developmental processes such as branching morphogenesis. Collagens are major components of the ECM of which basement membrane type IV and interstitial matrix type I are the most prevalent. Here we discuss how abnormal expression, proteolysis and structure of these collagens influence cellular functions to elicit multiple effects on tumors, including proliferation, initiation, invasion, metastasis, and therapy response.
Collapse
|