1
|
Chick Embryo Experimental Platform for Micrometastases Research in a 3D Tissue Engineering Model: Cancer Biology, Drug Development, and Nanotechnology Applications. Biomedicines 2021; 9:biomedicines9111578. [PMID: 34829808 PMCID: PMC8615510 DOI: 10.3390/biomedicines9111578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/06/2021] [Accepted: 10/16/2021] [Indexed: 12/31/2022] Open
Abstract
Colonization of distant organs by tumor cells is a critical step of cancer progression. The initial avascular stage of this process (micrometastasis) remains almost inaccessible to study due to the lack of relevant experimental approaches. Herein, we introduce an in vitro/in vivo model of organ-specific micrometastases of triple-negative breast cancer (TNBC) that is fully implemented in a cost-efficient chick embryo (CE) experimental platform. The model was built as three-dimensional (3D) tissue engineering constructs (TECs) combining human MDA-MB-231 cells and decellularized CE organ-specific scaffolds. TNBC cells colonized CE organ-specific scaffolds in 2–3 weeks, forming tissue-like structures. The feasibility of this methodology for basic cancer research, drug development, and nanomedicine was demonstrated on a model of hepatic micrometastasis of TNBC. We revealed that MDA-MB-231 differentially colonize parenchymal and stromal compartments of the liver-specific extracellular matrix (LS-ECM) and become more resistant to the treatment with molecular doxorubicin (Dox) and Dox-loaded mesoporous silica nanoparticles than in monolayer cultures. When grafted on CE chorioallantoic membrane, LS-ECM-based TECs induced angiogenic switch. These findings may have important implications for the diagnosis and treatment of TNBC. The methodology established here is scalable and adaptable for pharmacological testing and cancer biology research of various metastatic and primary tumors.
Collapse
|
2
|
Coughlin MF, Kamm RD. The Use of Microfluidic Platforms to Probe the Mechanism of Cancer Cell Extravasation. Adv Healthc Mater 2020; 9:e1901410. [PMID: 31994845 PMCID: PMC7274859 DOI: 10.1002/adhm.201901410] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/23/2019] [Indexed: 01/15/2023]
Abstract
Powerful experimental tools have contributed a wealth of novel insight into cancer etiology from the organ to the subcellular levels. However, these advances in understanding have outpaced improvements in clinical outcomes. One possible reason for this shortcoming is the reliance on animal models that do not fully replicate human physiology. An alternative in vitro approach that has recently emerged features engineered microfluidic platforms to investigate cancer progression. These devices allow precise control over cellular components, extracellular constituents, and physical forces, while facilitating detailed microscopic analysis of the metastatic process. This review focuses on the recent use of microfluidic platforms to investigate the mechanism of cancer cell extravasation.
Collapse
Affiliation(s)
- Mark F Coughlin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Roger D Kamm
- Cecil and Ida Green Distinguished Professor of Biological and Mechanical Engineering, Department of Mechanical Engineering, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
3
|
Ding W, Liu S, Li S, Ge D, Li F, Gao D. Simulation of blood and oxygen distributions in a hepatic lobule with sinusoids obstructed by cancer cells. J Theor Biol 2018; 446:229-237. [PMID: 29548738 DOI: 10.1016/j.jtbi.2018.03.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 02/01/2018] [Accepted: 03/12/2018] [Indexed: 01/26/2023]
Abstract
The liver is one of the common metastatic sites for many cancers. The obstruction of sinusoids by circulating tumor cells changes liver microenvironments and is thus considered a source of hepatic metastases. To date, few studies provide detailed information, either experimentally or theoretically, concerning the changes in blood and oxygen distributions induced by the obstruction of sinusoids. In this study, we utilized a 3D porous medium-vascular tree geometric structure to mimic the hepatic lobule and studied theoretical blood flow and oxygen transport in the lobule. The simulation was validated with data from the literature. Then, the distributions of blood and oxygen in the presence of the obstruction by cancer cells were simulated. The area and degree of the liver damage induced by the obstruction were analyzed by comparing the difference of liver microenvironments between physiological (non-blocked sinusoid) and pathological (fully or partially blocked sinusoid) conditions and the minimum cancer cell sizes causing liver damage for various obstruction positions were obtained. The work presented in this study can be used to predict the degree of liver damage induced by the local ischemia caused by the obstruction of sinusoids and to characterize the relationship between hepatic metastases and liver microenvironments.
Collapse
Affiliation(s)
- Weiping Ding
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China; Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Sen Liu
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China; Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shibo Li
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China; Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Duobiao Ge
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China; Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Fenfen Li
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China; Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, WA 98195, USA
| |
Collapse
|
4
|
Alcala MA, Park K, Yoo J, Lee DH, Park BH, Lee BC, Bartlett DL, Lee YJ. Effect of hyperthermia in combination with TRAIL on the JNK-Bim signal transduction pathway and growth of xenograft tumors. J Cell Biochem 2010; 110:1073-81. [PMID: 20544795 DOI: 10.1002/jcb.22619] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Approximately 25% of patients with colorectal cancer develop metastases to the liver, and surgery is currently the best treatment available. But there are several patients who are unresectable, and isolated hepatic perfusion (IHP) offers a different approach in helping to treat these patients. IHP is a method used for isolating the liver and delivering high doses of chemotherapeutic agents. The efficacy of IHP has been improved by combining hyperthermia not only with chemotherapeutics but with other deliverable agents such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). In this study, we used human colorectal cancer CX-1 cells and treated them with hyperthermia and TRAIL, causing cytotoxicity. We were able to demonstrate that the numbers of live cells were significantly reduced with hyperthermia and 10 ng/ml of TRAIL combined. We also showed that the effect of hyperthermia on TRAIL in our studies was enhancement of the apoptotic pathway by the promotion of JNK and Bim(EL) activity as well as PARP cleavage. We have also used our CX-1 cells to generate tumors in Balb/c nude mice. With intratumoral injections of TRAIL combined with hyperthermia at 42 degrees C, we were able to show a delayed onset of tumor growth in our xenograft model.
Collapse
Affiliation(s)
- Marco A Alcala
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
5
|
The prometastatic microenvironment of the liver. CANCER MICROENVIRONMENT 2008; 1:113-29. [PMID: 19308690 PMCID: PMC2654354 DOI: 10.1007/s12307-008-0011-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Accepted: 03/13/2008] [Indexed: 02/07/2023]
Abstract
The liver is a major metastasis-susceptible site and majority of patients with hepatic metastasis die from the disease in the absence of efficient treatments. The intrahepatic circulation and microvascular arrest of cancer cells trigger a local inflammatory reaction leading to cancer cell apoptosis and cytotoxicity via oxidative stress mediators (mainly nitric oxide and hydrogen peroxide) and hepatic natural killer cells. However, certain cancer cells that resist or even deactivate these anti-tumoral defense mechanisms still can adhere to endothelial cells of the hepatic microvasculature through proinflammatory cytokine-mediated mechanisms. During their temporary residence, some of these cancer cells ignore growth-inhibitory factors while respond to proliferation-stimulating factors released from tumor-activated hepatocytes and sinusoidal cells. This leads to avascular micrometastasis generation in periportal areas of hepatic lobules. Hepatocytes and myofibroblasts derived from portal tracts and activated hepatic stellate cells are next recruited into some of these avascular micrometastases. These create a private microenvironment that supports their development through the specific release of both proangiogenic factors and cancer cell invasion- and proliferation-stimulating factors. Moreover, both soluble factors from tumor-activated hepatocytes and myofibroblasts also contribute to the regulation of metastatic cancer cell genes. Therefore, the liver offers a prometastatic microenvironment to circulating cancer cells that supports metastasis development. The ability to resist anti-tumor hepatic defense and to take advantage of hepatic cell-derived factors are key phenotypic properties of liver-metastasizing cancer cells. Knowledge on hepatic metastasis regulation by microenvironment opens multiple opportunities for metastasis inhibition at both subclinical and advanced stages. In addition, together with metastasis-related gene profiles revealing the existence of liver metastasis potential in primary tumors, new biomarkers on the prometastatic microenvironment of the liver may be helpful for the individual assessment of hepatic metastasis risk in cancer patients.
Collapse
|
6
|
Kruskal JB, Azouz A, Korideck H, El-Hallak M, Robson SC, Thomas P, Goldberg SN. Hepatic colorectal cancer metastases: imaging initial steps of formation in mice. Radiology 2007; 243:703-11. [PMID: 17431127 DOI: 10.1148/radiol.2432060604] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE To prospectively use optical imaging to study the cell-specific mechanisms of entrapment and subsequent growth of two human colon cancer cell lines differing in their propensity to form hepatic metastases. MATERIALS AND METHODS In this Animal Care Committee-approved study, intravital optical imaging was performed in exteriorized livers of three groups of mice after intrasplenic inoculation of human colon cancer cells. Group 1 mice (control group; n=12) received a cell-maintaining solution only. Groups 2 and 3 (n=12 in each) were administered poorly (MIP-101 colon cancer cells) or highly (CX-1 colon cancer cells) metastatic cells. Imaging was performed on postinoculation days 0, 1, 3, and 6 to document sites and mechanisms of tumor cell entrapment and presence and sites of endothelial cell activation and of tumor cell interactions with systemic macrophages and Kupffer cells. Fluorescence intensity of Kupffer cells was compared by using the Mann-Whitney test. Immunohistochemistry served as the reference standard for all in vivo observations. RESULTS Whereas both MIP-101 and CX-1 colon cancer cells adhered to periportal Kupffer cells, the CX-1 cells resulted in Kupffer cell activation, evidenced in vivo by increased visible peroxidase activity (P<.05). Only CX-1 cells were associated with subsequent downstream endothelial cell activation, evidenced by in vivo expression of E-selectin. By day 6, regression of periportal MIP-101 tumor growth correlated with ingrowth of systemic macrophages, while CX-1 tumor growth, originating in the outflow venous regions, correlated with translobular migration and ingrowth of activated Kupffer cells. CONCLUSION Formation of hepatic colon cancer metastases is cancer cell-type specific, with cell lines differing in their mechanisms and intrahepatic locations of initial entrapment and Kupffer cell activation and subsequent growth.
Collapse
Affiliation(s)
- Jonathan B Kruskal
- Department of Radiology, Beth Israel Deaconess Medical Center and Harvard Medical School, West Clinical Center-CC302B, 1 Deaconess Rd, Boston, MA 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
7
|
Jessup JM, Samara R, Battle P, Laguinge LM. Carcinoembryonic antigen promotes tumor cell survival in liver through an IL-10-dependent pathway. Clin Exp Metastasis 2005; 21:709-17. [PMID: 16035616 DOI: 10.1007/s10585-004-7705-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Most circulating tumor cells die within 24 h of entering the hepatic microvasculature because their arrest initiates an ischemia-reperfusion (I/R) injury that is cytotoxic. Human colorectal carcinomas (CRC) produce the glycoprotein Carcinoembryonic Antigen (CEA) that increases experimental liver metastasis in nude mice. Since CEA induces release of IL-6 and IL-10, we hypothesized that CEA inhibits the I/R injury through a Kupffer cell-mediated cytokine-dependent pathway. We assessed cytokine effects in CRC co-cultured with liver and in vivo. Human CRC prelabeled with fluorescent dyes were incubated with a reoxygenated suspension of ischemic nude mouse liver fragments in a bioreactor. CEA, rhIL-6 or rhIL-10 were either administered to the donor mice prior to hepatic ischemia or during co-culture. Liver donors were athymic nude or iNOS, IL-6 or IL-10 knock out mice. Ischemic-reoxygenated liver kills Clone A CRC through production of nitric oxide (NO) and superoxide anion. Treatment of liver donors with CEA prior to hepatic ischemia inhibited this in vitro cytotoxicity through an IL-10 and Kupffer cell dependent pathway that inhibited NF-kappaB activation, NO production and iNOS upregulation. IL-10 but not IL-6 enhanced CRC survival in nude mouse liver in vivo. Thus, CEA enhanced metastasis by inducing IL-10 to inhibit iNOS upregulation in host liver.
Collapse
Affiliation(s)
- J Milburn Jessup
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA.
| | | | | | | |
Collapse
|
8
|
Timmers M, Vekemans K, Vermijlen D, Asosingh K, Kuppen P, Bouwens L, Wisse E, Braet F. Interactions between rat colon carcinoma cells and Kupffer cells during the onset of hepatic metastasis. Int J Cancer 2004; 112:793-802. [PMID: 15386374 DOI: 10.1002/ijc.20481] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Liver sinusoids harbor populations of 2 important types of immunocompetent cells, Kupffer cells (KCs) and natural killer (NK) cells, which are thought to play an important role in controlling hepatic metastasis in the first 24 hr upon arrival of the tumor cells in the liver. We studied the early interaction of KCs, NK and CC531s colon carcinoma cells in a syngeneic rat model by confocal laser scanning microscopy. Results showed a minority of KCs (19% periportal and 7% pericentral) involved in the interaction with 94% of tumor cells and effecting the phagocytosis of 92% of them. NK cell depletion decreased the phagocytosis of tumor cells by KCs by 33% over a period of 24 hr, leaving 35% of the cancer cells free, as compared to 6% in NK-positive rats. Surviving cancer cells were primarily located close to the Glisson capsule, suggesting that metastasis would initiate from this region.
Collapse
Affiliation(s)
- Maarten Timmers
- Laboratory for Cell Biology and Histology, Free University of Brussels, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Jessup JM, Laguinge L, Lin S, Samara R, Aufman K, Battle P, Frantz M, Edmiston KH, Thomas P. Carcinoembryonic antigen induction of IL-10 and IL-6 inhibits hepatic ischemic/reperfusion injury to colorectal carcinoma cells. Int J Cancer 2004; 111:332-7. [PMID: 15221959 DOI: 10.1002/ijc.20264] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Tumor cells cause ischemia/reperfusion (I/R) injury as they arrest within the hepatic microvasculature with the production of nitric oxide (NO) and reactive oxygen species (ROS) that kill both host liver and implanting tumor cells. Carcinoembryonic antigen (CEA) both facilitates the survival of experimental metastasis to nude mouse liver by weakly metastatic human colorectal carcinomas (CRCs) and induces the release of the proinflammatory cytokine IL-6. We hypothesized that CEA also stimulates the release of the antiinflammatory cytokine IL-10 causing inhibition of the toxicity of hepatic I/R injury and indirect stimulation of tumor cell colonization of the liver. Intravenous injection of CEA produced more than 1 ng/ml of IL-10 in the systemic circulation within 1 hr which subsided by 8 hr. The IL-10 response is specific to CEA since the pentapeptide sequence in CEA that binds to the CEA receptor stimulated isolated Kupffer cells to produce IL-10. IL-10, but not IL-6, increased the survival of weakly metastatic CRC cocultured with ischemic-reoxygenated liver fragments but did not affect the survival of CRC exposed to oxidative stress in the absence of any host cells. CEA, IL-6 and IL-10 pretreatment reduced expression of iNOS but only CEA and IL-10 strongly inhibited NO and total reactive species production by ischemic-rexoygenated liver. IL-6 was toxic to CRC exposed to oxidative stress while IL-10 did not have a direct effect on CRC. Thus, CEA stimulates production of IL-10 that may enhance metastasis by promoting the ability of circulating CRC cells to survive the I/R injury of implantation.
Collapse
Affiliation(s)
- John Milburn Jessup
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Hwang TS, Han HS, Choi HK, Lee YJ, Kim YJ, Han MY, Park YM. Differential, stage-dependent expression of Hsp70, Hsp110 and Bcl-2 in colorectal cancer. J Gastroenterol Hepatol 2003; 18:690-700. [PMID: 12753152 DOI: 10.1046/j.1440-1746.2003.03011.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The presence of hypoxic cells in solid tumors has been suggested to contribute to the malignant progression of various tumors. Recently, we reported an activation of heat shock transcription factor (HSF) and expression of heat shock proteins (Hsp) in murine tumor cells by hypoxia. METHODS To search for a possible role of Hsp in the malignant progression of human tumors, we analyzed the expression profiles of Hsp family proteins in weakly and highly metastatic human colorectal cancer (CRC) cell lines. We also analyzed the expression profiles of Bcl-2 family proteins because the altered expression of these proteins has been demonstrated in various solid tumors. RESULTS In the present paper we showed among various Hsp and Bcl-2 family proteins that the expression of Hsp70 and Hsp110 was elevated in highly metastatic CX-1 and HT-29 cells, while the expression of Bcl-2 was elevated in weakly metastatic MIP-101 and Clone A cells. Subsequent immunohistochemical analysis of 81 primary human CRC tissues demonstrated that the expression of Hsp70 and Hsp110 was highly correlated with the advanced clinical stages and positive lymph node involvement. The expression of Bcl-2, in contrast, was correlated to the early clinical stage and negative lymphovascular invasion. CONCLUSION Taken together, our study demonstrated for the first time a differential, stage-dependent expression of Hsp70, Hsp110 and Bcl-2 in CRC. We suggest that the molecular mechanisms underlying the differential expression of Hsp and Bcl-2 family members deserves a more rigorous future study, the results of which might offer novel modes of rationale and strategy to predict and manipulate the malignant progression of colorectal cancers.
Collapse
Affiliation(s)
- Tae Sook Hwang
- Department of Pathology, Inha University College of Medicine, Incheon, Korea
| | | | | | | | | | | | | |
Collapse
|
11
|
Song YK, Billiar TR, Lee YJ. Role of galectin-3 in breast cancer metastasis: involvement of nitric oxide. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:1069-75. [PMID: 11891203 PMCID: PMC1867157 DOI: 10.1016/s0002-9440(10)64927-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We investigated the role of galectin-3 in metastasis of human breast carcinoma BT549 cells using the experimental liver metastasis model. Underlying mechanisms were then elucidated using the liver/tumor co-culture and cell culture systems. After intrasplenic injection, galectin-3 cDNA transfected BT549 cells (BT549(gal-3 wt)) formed metastatic colonies in the liver, while galectin-3 null BT549 cells (BT549(par)) did not, demonstrating that galectin-3 enhances metastatic potential. More than 90% of BT549(gal-3 wt) cells survived after 24 hours-co-culture with the liver fragments isolated following ischemia treatment. In contrast, more than half of BT549(par) cells showed metabolic death following co-culture with the liver fragments. When the liver from inducible nitric oxide synthase (iNOS) knockout mice was used, no cytotoxicity to BT549(par) cells was observed. Thus, iNOS exerts cytotoxicity on BT549(par) cells and galectin-3 can protect against iNOS-induced cytotoxicity. BT549(gal-3 wt) also exhibited enhanced survival against peroxynitrite (up to 400 micromol/L) in vitro. A single mutation in the NWGR motif of galectin-3 obliterated both metastatic capability and cell survival, indicating that the antiapoptotic function of galectin-3 is involved in enhanced metastasis. In conclusion, galectin-3 enhances the metastatic potential of BT549 cells through resistance to the products of iNOS, possibly through its bcl-2-like antiapoptotic function.
Collapse
Affiliation(s)
- Young K Song
- Department of Pharmacology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
12
|
Moon BK, Lee YJ, Battle P, Jessup JM, Raz A, Kim HR. Galectin-3 protects human breast carcinoma cells against nitric oxide-induced apoptosis: implication of galectin-3 function during metastasis. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:1055-60. [PMID: 11549597 PMCID: PMC1850442 DOI: 10.1016/s0002-9440(10)61780-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Galectin-3 is a beta-galactoside-binding protein which regulates many biological processes including cell adhesion, migration, cell growth, tumor progression, metastasis, and apoptosis. Although the exact function of galectin-3 in cancer development is unclear, galectin-3 expression is associated with neoplastic progression and metastatic potential. Since studies have suggested that tumor cell survival in microcirculation determines the metastatic outcome, we examined the effect of galectin-3 overexpression in human breast carcinoma cell survival using the liver ischemia/reperfusion metastasis model. While the majority of control cells died by hepatic ischemia/reoxygenation, nearly all of galectin-3 overexpressing cells survived. We showed that galectin-3 inhibits nitrogen free radical-mediated apoptosis, one of the major death pathways induced during hepatic ischemia/reperfusion. Galectin-3 inhibition of apoptosis involved protection of mitochondrial integrity, inhibition of cytochrome c release and caspase activation. Taking these results together with the previous observation that galectin-3 inhibits apoptosis induced by loss of cell adhesion, we propose that galectin-3 is a critical determinant for anchorage-independent and free radical-resistant cell survival during metastasis.
Collapse
Affiliation(s)
- B K Moon
- Department of Anesthesiology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | |
Collapse
|
13
|
Ito S, Nakanishi H, Ikehara Y, Kato T, Kasai Y, Ito K, Akiyama S, Nakao A, Tatematsu M. Real-time observation of micrometastasis formation in the living mouse liver using a green fluorescent protein gene-tagged rat tongue carcinoma cell line. Int J Cancer 2001; 93:212-7. [PMID: 11410868 DOI: 10.1002/ijc.1318] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Initial arrest, attachment, extravasation and subsequent extravascular growth of tumor cells in the secondary organs are believed to be crucial events for hematogenous metastasis, but the actual processes in living animals remain unclear. For the present study, we established green fluorescent protein (GFP)-expressing rat tongue carcinoma cell lines (RSC3) that permit real-time analysis of micrometastasis formation in combination with intravital video microscopy (IVVM). With this system, GFP-expressing metastatic (LM-EGFP) and non-metastatic (E2-EGFP) cell lines could be visualized at the cellular level in live mice for more than 1 month. Real-time IVVM analysis of liver metastases after intraportal injection of cells via a mesenteric vein revealed that both LM-EGFP and E2-EGFP tumor cells arrest similarly in sinusoidal vessels near terminal portal venules within 0.4 sec, during which time no evidence of a "rolling"-like movement along endothelial cell surfaces was observed. Quantitative analysis of GFP-positive foci showed that E2-EGFP cells were completely sheared from the liver sinusoid within 3 days, with no solitary dormant cells, whereas a substantial number of LM-EGFP cells remained in the liver, probably due to stable attachment to the sinusoidal wall. Confocal laser scanning microscopic study in combination with laminin immunohistochemistry revealed that only LM-EGFP cells started growth at 3 to 4 days after inoculation and that most of the growing foci were surrounded by subsinusoidal basement membrane. Our results suggest that micrometastasis formation by LM-EGFP cells consists of initial tumor cell arrest due to size constraints of the vessel, stable attachment to subsinusoidal basement membrane and subsequent intravascular growth before extravasation. The difference in metastatic potential between the 2 lines may reside in their capacity to attach stably to the vessel wall rather than their potential for initial cell arrest or subsequent growth. The system used in the present study may be a powerful tool for analyzing targets for various anti-metastatic agents in the sequential process of metastasis.
Collapse
Affiliation(s)
- S Ito
- Laboratory of Pathology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kruskal JB, Thomas P, Nasser I, Cay O, Kane RA. Hepatic colon cancer metastases in mice: dynamic in vivo correlation with hypoechoic rims visible at US. Radiology 2000; 215:852-7. [PMID: 10831710 DOI: 10.1148/radiology.215.3.r00jn16852] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE To use videomicroscopy of tumor-bearing livers of live mice to depict tumors directly to determine the exact nature of rims seen on corresponding ultrasonographic (US) scans. MATERIALS AND METHODS Seventy-six hepatic colorectal cancer metastases were studied in exteriorized livers of 18 mice by using intravital microscopy, US, and histologic examination of the same tumors. RESULTS Hypoechoic rims correlated with distended sinusoidal spaces in vivo. These spaces surrounded only locally invasive tumors (mean diameter, 0.85 mm) that had obstructed the supplying terminal portal venules. These spaces, containing adherent leukocytes and tumor cells, gave rise to new tumor vasculature. Results of histologic examination of rims (portal inflammation, congested or compressed sinusoids, cell atrophy) correlated with leukocyte endothelial adherence, occluded sinusoids, and new vessel formation in vivo. CONCLUSION Unlike results from previous studies, dynamic in vivo observations of peritumoral rims demonstrated distended sinusoidal spaces giving rise to new tumor-penetrating vessels. These sinusoids arose around locally invasive tumors and were associated with more advanced intrahepatic disease. These dynamic observations provide a pathophysiologic explanation for previous histologic correlates of peritumoral rims.
Collapse
Affiliation(s)
- J B Kruskal
- Department of Radiology, Beth Israel Deaconess Medical Center-West, Boston, MA 02215, USA.
| | | | | | | | | |
Collapse
|
15
|
Jessup JM, Ishii S, Mitzoi T, Edmiston KH, Shoji Y. Carcinoembryonic antigen facilitates experimental metastasis through a mechanism that does not involve adhesion to liver cells. Clin Exp Metastasis 2000; 17:481-8. [PMID: 10763913 DOI: 10.1023/a:1006685817395] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Carcinoembryonic antigen (CEA) injected intravenously into athymic nude mice increases the ability of weakly metastatic human colorectal carcinoma (CRC) cells to colonize liver in an experimental metastasis assay. Since CEA acts as an intercellular adhesion molecule in vitro, several investigators have postulated that this facilitation of experimental metastasis may be mediated through adhesion between CEA on CRC and CEA-binding proteins on Kupffer or other cells lining the hepatic sinusoid. The present work tested this postulate both by intravital fluorescence videomicroscopy in vivo and in adhesion assays in vitro to enriched populations of Kupffer cells and hepatic sinusoidal endothelial cells (SEC). The data indicate that CEA expression does not effect adhesion to enriched Kupffer cells or SEC in vitro. These data suggest that CEA enhances liver colonization through another mechanism, possibly one that involves modulation of the hepatic response to tumor cell implantation.
Collapse
Affiliation(s)
- J M Jessup
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA. jjessup+@pitt.edu
| | | | | | | | | |
Collapse
|
16
|
Schultze RL, Gangopadhyay A, Cay O, Lazure D, Thomas P. Tyrosine kinase activation in LPS stimulated rat Kupffer cells. Cell Biochem Biophys 1999; 30:287-301. [PMID: 10356646 DOI: 10.1007/bf02738071] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Kupffer cells, a majority of the body's fixed macrophages, are a major site of bacterial lipopolysaccharide (LPS) metabolism and are mediators in the body's response to sepsis. Uptake of LPS is different in Kupffer cells than other macrophages. Signal transduction in other macrophages in response to LPS involves phosphorylation of proteins in the 50-60 kDa range. We hypothesized that Kupffer cells may have unique signal transduction pathways in response to LPS. Rat Kupffer cells were exposed to LPS (1 microgram/mL) for varying times ranging from 15 to 90 min. Cell lysates were Western blotted using an anti-phosphotyrosine antibody. The blots showed an increase in the amount of tyrosine phosphorylation on two proteins of 119 kDa and 83 kDa. The effects of varying LPS concentration (1 ng/mL-1 microgram/mL) showed an increasing amount of phosphorylation with increasing LPS concentration. To associate the importance of tyrosine phosphorylation in the response of Kupffer cells to LPS, the tyrosine kinase inhibitors, tyrphostin, lavendustin, and genisten were used to study the effects of inhibiting phosphorylation on TNF-alpha production. Kupffer cells were preincubated in the presence of the inhibitor and exposed to LPS (1 microgram/mL). TNF-alpha was measured in the conditioned media by ELISA. A 70% or greater decrease in TNF-alpha production was observed. When phagocytosis of latex beads by rat Kupffer cells was measured in vivo using intravital video microscopy, LPS treatment significantly increased uptake. This increase in phagocytosis was inhibited by tyrphostin. These results show what may be unique phosphorylation events in Kupffer cells that are related to LPS induced production of TNF-alpha.
Collapse
Affiliation(s)
- R L Schultze
- Department of Surgery, Beth Israel-Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
17
|
Ogoshi T, Ishii S, Mizoi T, Harada N, Sato W, Saito K, Ogawa H, Inabe K, Shiiba K, Matsuno S. CD44H participates in the intrahepatic growth of murine colon 26 adenocarcinoma cells. Jpn J Cancer Res 1998; 89:1160-8. [PMID: 9914785 PMCID: PMC5921726 DOI: 10.1111/j.1349-7006.1998.tb00511.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study was to determine if CD44, a metastasis-associated cell adhesion molecule, is involved in the hepatic colonization by murine colon 26 adenocarcinoma cells. Indirect membrane immunofluorescence and FACS analysis showed strong expressions of CD44 and integrin beta 1 on colon 26 cells. Injection of 1 x 10(5) colon 26 cells into the superior mesenteric vein of syngeneic BALB/c mice produced macroscopic hepatic nodules in 92% (22/24) of the mice 14 days after inoculation. When colon 26 cells were pretreated with an anti-CD44 monoclonal antibody (mAb), IM7, only 30% (3/10) of the mice produced minute nodules in the liver on day 14 (P < 0.001), though IM7 did not inhibit growth of the cells in vitro. Pretreatment of colon 26 cells with an anti-integrin beta 1 mAb did not significantly block the hepatic metastasis. Histologically, microcolonies of tumor cells were detected in all of the livers on day 14 including the IM7-pretreatment mice that were free of gross nodules. However, percentages of tumor-occupied areas in the liver were consistently lower in IM7-pretreatment mice than in control mice (0.82% vs. 5.0% on day 14; P < 0.005). Reverse transcription-polymerase chain reaction (RT-PCR) amplification of mRNA revealed that colon 26 cells and splenocytes only expressed the hematopoietic isoform of CD44 (CD44H), which had no insertion of variant exons, while normal colonocytes expressed possible variant isoforms. These data suggest that malignant transformation of murine colonic epithelium altered the expression pattern of CD44 isoforms and that CD44H participates in the intrahepatic growth of colon 26 cells.
Collapse
Affiliation(s)
- T Ogoshi
- First Department of Surgery, Tohoku University School of Medicine, Sendai
| | | | | | | | | | | | | | | | | | | |
Collapse
|