Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Cancer Sci ; 114(10): 3935-3945, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37482942

ABSTRACT

Tumors contain various stromal cells, such as immune cells, endothelial cells, and fibroblasts, which contribute to the development of a tumor-specific microenvironment characterized by hypoxia and inflammation, and are associated with malignant progression. In this study, we investigated the activity of intratumoral hypoxia-inducible factor (HIF), which functions as a master regulator of the cellular response to hypoxia and inflammation. We constructed the HIF activity-monitoring reporter gene hypoxia-response element-Venus-Akaluc (HVA) that expresses the green fluorescent protein Venus and modified firefly luciferase Akaluc in a HIF activity-dependent manner, and created transgenic mice harboring HVA transgene (HVA-Tg). In HVA-Tg, HIF-active cells can be visualized using AkaBLI, an ultra-sensitive in vivo bioluminescence imaging technology that produces an intense near-infrared light upon reaction of Akaluc with the D-luciferin analog AkaLumine-HCl. By orthotopic transplantation of E0771, a mouse triple negative breast cancer cell line without a reporter gene, into HVA-Tg, we succeeded in noninvasively monitoring bioluminescence signals from HIF-active stromal cells as early as 8 days after transplantation. The HIF-active stromal cells initially clustered locally and then spread throughout the tumors with growth. Immunohistochemistry and flow cytometry analyses revealed that CD11b+ F4/80+ macrophages were the predominant HIF-active stromal cells in E0771 tumors. These results indicate that HVA-Tg is a useful tool for spatiotemporal analysis of HIF-active tumor stromal cells, facilitating investigation of the roles of HIF-active tumor stromal cells in tumor growth and malignant progression.


Subject(s)
Endothelial Cells , Neoplasms , Mice , Animals , Stromal Cells , Hypoxia , Cell Hypoxia , Inflammation , Optical Imaging , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cell Line, Tumor , Tumor Microenvironment
2.
Ultrason Imaging ; 44(2-3): 96-104, 2022 05.
Article in English | MEDLINE | ID: mdl-35549598

ABSTRACT

Photoacoustic (PA) technology can be used for non-invasive imaging of blood vessels. In this paper, we report on our prototype PA imaging system with a newly designed ultrasound sensor and its visualization performance of microvascular in animal. We fabricated an experimental system for animals using a high-frequency sensor. The system has two modes: still image mode by wide scanning and moving image mode by small rotation of sensor array. Optical test target, euthanized mice and rats, and live mice were used as objects. The results of optical test target showed that the spatial resolution was about two times higher than that of our conventional prototype. The image performance in vivo was evaluated in euthanized healthy mice and rats, allowing visualization of detailed blood vessels in the liver and kidneys. In tumor-bearing mice, different results of vascular induction were shown depending on the type of tumor and the method of transplantation. By utilizing the video imaging function, we were able to observe the movement of blood vessels around the tumor. We have demonstrated the feasibility of the system as a less invasive animal experimental device, as it can acquire vascular images in animals in a non-contrast and non-invasive manner.


Subject(s)
Neoplasms , Photoacoustic Techniques , Animals , Imaging, Three-Dimensional/methods , Mice , Neoplasms/diagnostic imaging , Photoacoustic Techniques/methods , Rats , Ultrasonography
3.
Bioorg Med Chem ; 28(1): 115207, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31740202

ABSTRACT

HIF-1 is regarded as a promising target for the drugs used in cancer chemotherapy, and creating readily accessible templates for the development of synthetic drug candidates that could inhibit HIF-1 transcriptional activity is an important pursuit. In this study, indeno[2,1-c]pyrazolones were designed as readily available synthetic inhibitors of HIF-1 transcriptional activity. Nine compounds were synthesized in 4-5 steps from commercially available starting materials. In evaluations of the ability to inhibit the hypoxia-induced transcriptional activity of HIF-1, compound 3c showed a higher level compared with that of known inhibitor, YC-1. The compound 3c suppressed HIF-1α protein accumulation without affecting the levels of HIF-1α mRNA.


Subject(s)
Drug Design , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Pyrazolones/pharmacology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Molecular Structure , Pyrazolones/chemical synthesis , Pyrazolones/chemistry , Structure-Activity Relationship , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
4.
Bioconjug Chem ; 30(5): 1323-1330, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30848886

ABSTRACT

Single-walled carbon nanotubes (SWCNTs) show strong fluorescence in the 1000-1700 nm second near-infrared (NIR-II) wavelength range and are considered promising candidates for angiographic imaging probes. Oxygen-doped SWCNTs coated with phospholipid-polyethylene glycol (o-SWCNT-PEG) show exceptional potential, as they emit fluorescence at Ć¢ĀˆĀ¼1300 nm through excitation with 980 nm light. Here, with the aim of putting o-SWCNTs to practical use as an angiographic agent in animal experiments, the retention time after intravenous administration in the vasculature of mice and the biodistribution were studied. To provide bio affinity, the o-SWCNTs were coated with phospholipid polyethylene glycol. The intravenously injected o-SWCNT-PEG circulated within the vasculature for 3 h and cleared within 1 day. There was prominent fluorescence and Raman signals from the SWCNTs in the liver and spleen early in the experiment; the signals remained for 1 month. No apparent abnormalities in weight or appearance were observed after 2 months, suggesting low toxicity of o-SWCNT-PEG. These characteristics of o-SWCNT-PEG would make it useful as an angiographic imaging probe in the NIR-II wavelength range.


Subject(s)
Fluorescent Dyes/chemistry , Nanotubes, Carbon , Oxygen/chemistry , 3T3 Cells , Animals , Biological Assay , Cell Line, Tumor , Fluorescence , Humans , Mice , Polyethylene Glycols/chemistry , Tissue Distribution
5.
Cancer Sci ; 109(9): 2746-2756, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29927026

ABSTRACT

Lung metastasis is a major cause of mortality in patients with osteosarcoma (OS). A better understanding of the molecular mechanism of OS lung metastasis may facilitate development of new therapeutic strategies to prevent the metastasis. We have established high- and low-metastatic sublines (LM8-H and LM8-L, respectively) from Dunn OS cell line LM8 by using in vivo image-guided screening. Among the genes whose expression was significantly increased in LM8-H compared to LM8-L, the transcription factor lymphoid enhancer-binding factor 1 (LEF1) was identified as a factor that promotes LM8-H cell extravasation into the lungs. To identify downstream effectors of LEF1 that are involved in OS lung metastasis, 13 genes were selected based on LM8 microarray data and genomewide meta-analysis of a public database for OS patients. Among them, the cytoglobin (Cygb) gene was identified as a key effector in promoting OS extravasation into the lungs. CYGB overexpression increased the extravasation ability of LM8-L cells, whereas knocking out the Cygb gene in LM8-H cells reduced this ability. Our results showed a novel LEF1-CYGB axis in OS lung metastasis and may provide a new way of developing therapeutic strategies to prevent OS lung metastasis.


Subject(s)
Bone Neoplasms/pathology , Globins/physiology , Lung Neoplasms/secondary , Lymphoid Enhancer-Binding Factor 1/physiology , Osteosarcoma/pathology , Animals , Cell Line, Tumor , Cytoglobin , Globins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
6.
Cancer Sci ; 107(8): 1151-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27270607

ABSTRACT

Pancreatic cancer is one of the most lethal digestive system cancers with a 5-year survival rate of 4-7%. Despite extensive efforts, recent chemotherapeutic regimens have provided only limited benefits to pancreatic cancer patients. Gemcitabine and TS-1, the current standard-of-care chemotherapeutic drugs for treatment of this severe cancer, have a low response rate. Hypoxia is one of the factors contributing to treatment resistance. Specifically, overexpression of hypoxia-inducible factor, a master transcriptional regulator of cell adaption to hypoxia, is strongly correlated with poor prognosis in many human cancers. TAT-ODD-procaspase-3 (TOP3) is a protein prodrug that is specifically processed and activated in hypoxia-inducible factor-active cells in cancers, leading to cell death. Here, we report combination therapies in which TOP3 was combined with gemcitabine or TS-1. As monotherapy, gemcitabine and TS-1 showed a limited effect on hypoxic and starved pancreatic cancer cells, whereas co-treatment with TOP3 successfully overcame this limitation in vitro. Furthermore, combination therapies of TOP3 with these drugs resulted in a significant improvement in survival of orthotopic pancreatic cancer models involving the human pancreatic cancer cell line SUIT-2. Overall, our study indicates that the combination of TOP3 with current chemotherapeutic drugs can significantly improve treatment outcome, offering a promising new therapeutic option for patients with pancreatic cancer.


Subject(s)
Deoxycytidine/analogs & derivatives , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prodrugs/metabolism , Recombinant Fusion Proteins/metabolism , Silicates/pharmacology , Titanium/pharmacology , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Fluorouracil/pharmacology , Humans , Male , Mice , Mice, Inbred BALB C , Prodrugs/pharmacology , Recombinant Fusion Proteins/pharmacology , Silicates/administration & dosage , Survival Analysis , Titanium/administration & dosage , Tumor Hypoxia , Gemcitabine
7.
Inorg Chem ; 54(1): 215-20, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25494847

ABSTRACT

Protein crystals generally are stable solid protein assemblies. Certain protein crystals are suitable for use as nanovessels for immobilizing metal complexes. Here we report the preparation of ruthenium carbonyl-incorporated cross-linked hen egg white lysozyme crystals (RuĀ·CL-HEWL). RuĀ·CL-HEWL retains a Ru carbonyl moiety that can release CO, although a composite of Ru carbonyl-HEWL dissolved in buffer solution (RuĀ·HEWL) does not release CO. We found that treatment of cells with RuĀ·CL-HEWL significantly increased nuclear factor kappa B (NF-κB) activity as a cellular response to CO. These results demonstrate that RuĀ·CL-HEWL has potential for use as an artificial extracellular scaffold suitable for transport and release of a gas molecule.


Subject(s)
Carbon Monoxide/chemistry , Coordination Complexes/chemistry , Muramidase/chemistry , Ruthenium/chemistry , Animals , Biological Transport , Carbon Monoxide/pharmacology , Chickens , Coordination Complexes/pharmacology , Cross-Linking Reagents/chemistry , Crystallization , Crystallography, X-Ray , Egg White/chemistry , Gene Expression/drug effects , Genes, Reporter , Glutaral/chemistry , HEK293 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Myoglobin/chemistry , NF-kappa B/genetics , NF-kappa B/metabolism , Porosity
8.
J Am Chem Soc ; 136(48): 16902-8, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25352251

ABSTRACT

Protein cages have been utilized as templates in the development of biomaterials. Here we report protein engineering of the ferritin (Fr) cage for encapsulating carbon monoxide releasing molecules (CORMs) and release of CO gas which serves as a cell signaling molecule. The protein cages enable us to increase the half-life for CO release, providing a release rate that is 18-fold slower than the rate of a typical CORM, Ru(CO)3Cl(glycinate) (CORM-3). Moreover, the uptake ratio of the composite is about 4-fold greater than that of CORM-3. We found that these effects enhance the activation of nuclear factor κB 10-fold higher than CORM-3. The protein cage of Fr thus provides the basis for new CORMs that can be used for in vitro cell research.


Subject(s)
Carbon Monoxide/chemistry , Ferritins/chemistry , Organometallic Compounds/chemistry , Ruthenium/chemistry , Crystallography, X-Ray , Models, Molecular , Molecular Conformation , Organometallic Compounds/chemical synthesis
9.
Cancer Sci ; 105(5): 553-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24597654

ABSTRACT

Bone metastasis is a multistep process that includes cancer cell dissemination, colonization, and metastatic growth. Furthermore, this process involves complex, reciprocal interactions between cancer cells and the bone microenvironment. Bone resorption is known to be involved in both osteolytic and osteoblastic bone metastasis. However, the precise roles of the bone resorption in the multistep process of osteoblastic bone metastasis remain unidentified. In this study, we show that bone resorption plays important roles in cancer cell colonization during the initial stage of osteoblastic bone metastasis. We applied bioluminescence/X-ray computed tomography multimodal imaging that allows us to spatiotemporally analyze metastasized cancer cells and bone status in osteoblastic bone metastasis models. We found that treatment with receptor activator of factor-κB ligand (RANKL) increased osteoblastic bone metastasis when given at the same time as intracardiac injection of cancer cells, but failed to increase metastasis when given 4 days after cancer cell injection, suggesting that RANKL-induced bone resorption facilitates growth of cancer cells colonized in the bone. We show that insulin-like growth factor-1 released from the bone during bone resorption and hypoxia-inducible factor activity in cancer cells cooperatively promoted survival and proliferation of cancer cells in bone marrow. These results suggest a mechanism that bone resorption and hypoxic stress in the bone microenvironment cooperatively play an important role in establishing osteoblastic metastasis.


Subject(s)
Bone Neoplasms/secondary , Bone Resorption/metabolism , Bone Resorption/pathology , Hypoxia-Inducible Factor 1/metabolism , Osteosarcoma/secondary , RANK Ligand/pharmacology , Somatomedins/metabolism , Animals , Bone Marrow Cells/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Cell Survival , Enzyme Activation , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Transforming Growth Factor beta/metabolism , Transplantation, Heterologous , Tumor Microenvironment
10.
Blood ; 119(23): 5405-16, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22544701

ABSTRACT

HSC fate decisions are regulated by cell-intrinsic and cell-extrinsic cues. The latter cues are derived from the BM niche. Membrane-type 1 matrix metalloproteinase (MT1-MMP), which is best known for its proteolytic role in pericellular matrix remodeling, is highly expressed in HSCs and stromal/niche cells. We found that, in MT1-MMP(-/-) mice, in addition to a stem cell defect, the transcription and release of kit ligand (KitL), stromal cell-derived factor-1 (SDF-1/CXCL12), erythropoietin (Epo), and IL-7 was impaired, resulting in a trilineage hematopoietic differentiation block, while addition of exogenous KitL and SDF-1 restored hematopoiesis. Further mechanistic studies revealed that MT1-MMP activates the hypoxia-inducible factor-1 (HIF-1) pathway via factor inhibiting HIF-1 (FIH-1) within niche cells, thereby inducing the transcription of HIF-responsive genes, which induce terminal hematopoietic differentiation. Thus, MT1-MMP in niche cells regulates postnatal hematopoiesis, by modulating hematopoietic HIF-dependent niche factors that are critical for terminal differentiation and migration.


Subject(s)
Cytokines/genetics , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hypoxia-Inducible Factor 1/metabolism , Matrix Metalloproteinase 14/metabolism , Transcriptional Activation , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Line , Cells, Cultured , Chemokine CXCL12/metabolism , Chemokines/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Deletion , Hematopoietic Stem Cells/metabolism , Humans , Matrix Metalloproteinase 14/genetics , Mice , Mice, Inbred C57BL , Stem Cell Factor/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
11.
Int J Clin Oncol ; 19(3): 497-504, 2014.
Article in English | MEDLINE | ID: mdl-23739923

ABSTRACT

BACKGROUND: Despite the advent of targeted therapies, interferon-alpha (IFN-α) remains a therapeutic option for advanced renal cell carcinoma (RCC), especially in Japan, with a treatment response rate of 15-20Ā %. To improve the efficacy of IFN-α-based therapies, we evaluated a novel treatment strategy for RCC using an IFN-α2b gene construct with a repetitive hypoxia-inducible factor binding site. METHODS: We constructed an expression plasmid designated 5HREp-IFN-α2b containing the coding region of the IFN-α2b gene. Five copies of the hypoxia-response element (HRE) sequences were inserted upstream of the IFN-α2b gene, and the construct was transfected into human RCC cell lines ACHN, 786-O and KU19-20. The concentrations of IFN-α2b in the conditioned media were measured by enzyme-linked immunosorbent assay. Cell viabilities were determined by MTS assays. RESULTS: Construct-induced IFN-α secretion was confirmed in all three cell lines. IFN-α production was significantly enhanced by the hypoxia-mimicking agent deferoxamine mesylate in cell lines expressing the wild-type von Hippel-Lindau (VHL) gene (KU19-20 and ACHN) compared with cells expressing the mutant VHL gene (786-O). The construct exerted significant suppressive effects on the viabilities of all RCC cell lines. CONCLUSION: This is the first study to report on the construction of a cytokine gene with a repetitive hypoxia-inducible factor binding site and its application in the suppression of human cancer cells. Gene therapy using this IFN-α2b gene construct with HREs may represent a novel treatment modality for advanced RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Genetic Therapy/methods , Interferon-alpha/pharmacology , Kidney Neoplasms/pathology , Recombinant Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Survival/genetics , Culture Media, Conditioned/pharmacology , Humans , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interferon alpha-2 , Interferon-alpha/genetics , Interferon-alpha/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Recombinant Proteins/metabolism , Response Elements , Transfection , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
12.
Commun Biol ; 7(1): 652, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806676

ABSTRACT

Epitope binning, an approach for grouping antibodies based on epitope similarities, is a critical step in antibody drug discovery. However, conventional methods are complex, involving individual antibody production. Here, we established Epitope Binning-seq, an epitope binning platform for simultaneously analyzing multiple antibodies. In this system, epitope similarity between the query antibodies (qAbs) displayed on antigen-expressing cells and a fluorescentlyĀ labeled reference antibody (rAb) targeting a desired epitope is analyzed by flow cytometry. The qAbs with epitope similar to the rAb can be identified by next-generation sequencing analysis of fluorescence-negative cells. Sensitivity and reliability of this system are confirmed using rAbs, pertuzumab and trastuzumab, which target human epidermal growth factor receptor 2. Epitope Binning-seq enables simultaneous epitope evaluation of 14 qAbs at various abundances in libraries, grouping them into respective epitope bins. This versatile platform is applicable to diverse antibodies and antigens, potentially expediting the identification of clinically useful antibodies.


Subject(s)
Epitopes , Humans , Epitopes/immunology , Sequence Analysis, DNA/methods , High-Throughput Nucleotide Sequencing/methods , Animals , Receptor, ErbB-2/immunology , Receptor, ErbB-2/genetics , Flow Cytometry/methods , Trastuzumab/immunology , Epitope Mapping/methods , Antibodies/immunology , Antibodies/genetics , Antibodies, Monoclonal, Humanized/immunology
13.
Adv Biol (Weinh) ; 8(3): e2300159, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37986133

ABSTRACT

Myeloid-derived suppressor cell (MDSC)-like adherent cells (MLACs) are a recently identified CD11b+ F4/80- myeloid cell subset that can infiltrate tumors early in development and promote their growth. Because of these functions, MLACs play an important role in establishing an immunosuppressive tumor microenvironment (TME). However, the lack of MLAC-specific markers has hampered further characterization of this cell type. This study identifies the gene signature of MLACs by analyzing RNA-sequencing (RNA-seq) and public single-cell RNA-seq data, revealing that MLACs are an independent cell population that are distinct from other intratumoral myeloid cells. After combining proteome analysis of membrane proteins with RNA-seq data, H2-Ab1 and CD11c are indicated as marker proteins that can support the isolation of MLAC subsets from CD11b+ F4/80- myeloid cells by fluorescence-activated cell sorting. The CD11b+ F4/80- H2-Ab1+ and CD11b+ F4/80- CD11c+ MLAC subsets represent approximately half of the MLAC population that is isolated based on their adhesion properties and possess gene signatures and functional properties similar to those of the MLAC population. Additionally, membrane proteome analysis suggests that MLACs express highly heterogeneous surface proteins. This study facilitates an integrated understanding of heterogeneous intratumoral myeloid cells, as well as the molecular and cellular details of the development of an immunosuppressive TME.


Subject(s)
Myeloid-Derived Suppressor Cells , Myeloid-Derived Suppressor Cells/metabolism , Proteome/metabolism , Myeloid Cells , Flow Cytometry , Cell Line, Tumor
14.
Biochem Biophys Res Commun ; 433(1): 139-44, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23485455

ABSTRACT

Hypoxia inducible factor is a dominant regulator of adaptive cellular responses to hypoxia and controls the expression of a large number of genes regulating angiogenesis as well as metabolism, cell survival, apoptosis, and other cellular functions in an oxygen level-dependent manner. When a neoplasm is able to induce angiogenesis, tumor progression occurs more rapidly because of the nutrients provided by the neovasculature. Meningioma is one of the most hypervascular brain tumors, making anti-angiogenic therapy an attractive novel therapy for these tumors. HIF-3α has been conventionally regarded as a dominant-negative regulator of HIF-1α, and although alternative HIF-3α splicing variants are extensively reported, their specific functions have not yet been determined. In this study, we found that the transcription of HIF-3α4 was silenced by the promoter DNA methylation in meningiomas, and inducible HIF-3α4 impaired angiogenesis, proliferation, and metabolism/oxidation in hypervascular meningiomas. Thus, HIF-3α4 could be a potential molecular target in meningiomas.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Meningeal Neoplasms/blood supply , Meningeal Neoplasms/genetics , Meningioma/blood supply , Meningioma/genetics , Neovascularization, Pathologic/genetics , Alternative Splicing , Animals , Apoptosis Regulatory Proteins , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Glucose/metabolism , Humans , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningioma/metabolism , Meningioma/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins , Transplantation, Heterologous
15.
Proc Natl Acad Sci U S A ; 107(29): 13063-8, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20616008

ABSTRACT

Recent reports have suggested critical roles of myeloid cells in tumor invasion and metastasis, although these findings have not led to therapeutics. Using a mouse model for liver dissemination, we show that mouse and human colon cancer cells secrete CC-chemokine ligands CCL9 and CCL15, respectively, and recruit CD34(+) Gr-1(-) immature myeloid cells (iMCs). They express CCL9/15 receptor CCR1 and produce matrix metalloproteinases MMP2 and MMP9. Lack of the Ccr1, Mmp2, or Mmp9 gene in the host dramatically suppresses outgrowths of disseminated tumors in the liver. Importantly, CCR1 antagonist BL5923 blocks the iMC accumulation and metastatic colonization and significantly prolongs the survival of tumor-bearing mice. These results suggest that CCR1 antagonists can provide antimetastatic therapies for patients with disseminated colon cancer in the liver.


Subject(s)
Cell Differentiation , Colonic Neoplasms/pathology , Liver Neoplasms/secondary , Myeloid Cells/pathology , Receptors, CCR1/metabolism , Animals , Chemokines, CC/metabolism , Colonic Neoplasms/metabolism , Disease Models, Animal , Humans , Ligands , Liver/metabolism , Liver/pathology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Macrophage Inflammatory Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Myeloid Cells/metabolism , Receptors, CCR1/antagonists & inhibitors , Signal Transduction , Survival Analysis
16.
Commun Biol ; 6(1): 144, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36737474

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive and highly heterogenous disease with no well-defined therapeutic targets. Treatment options are thus limited and mortality is significantly higher compared with other breast cancer subtypes. Mammary gland tissue-resident macrophages (MGTRMs) are found to be the most abundant stromal cells in early TNBC before angiogenesis. We therefore aimed to explore novel therapeutic approaches for TNBC by focusing on MGTRMs. Local depletion of MGTRMs in mammary gland fat pads the day before TNBC cell transplantation significantly reduced tumor growth and tumor-associated macrophage (TAM) infiltration in mice. Furthermore, local depletion of MGTRMs at the site of TNBC resection markedly reduced recurrence and distant metastases, and improved chemotherapy outcomes. This study demonstrates that MGTRMs are a major TAM resource and play pivotal roles in the growth and malignant progression of TNBC. The results highlight a possible novel anti-cancer approach targeting tissue-resident macrophages.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Animals , Mice , Triple Negative Breast Neoplasms/drug therapy , Tumor-Associated Macrophages , Cell Line, Tumor
17.
Nat Commun ; 14(1): 8031, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38052804

ABSTRACT

Cancer cells inevitably interact with neighboring host tissue-resident cells during the process of metastatic colonization, establishing a metastatic niche to fuel their survival, growth, and invasion. However, the underlying mechanisms in the metastatic niche are yet to be fully elucidated owing to the lack of methodologies for comprehensively studying the mechanisms of cell-cell interactions in the niche. Here, we improve a split green fluorescent protein (GFP)-based genetically encoded system to develop secretory glycosylphosphatidylinositol-anchored reconstitution-activated proteins to highlight intercellular connections (sGRAPHIC) for efficient fluorescent labeling of tissue-resident cells that neighbor on and putatively interact with cancer cells in deep tissues. The sGRAPHIC system enables the isolation of metastatic niche-associated tissue-resident cells for their characterization using a single-cell RNA sequencing platform. We use this sGRAPHIC-leveraged transcriptomic platform to uncover gene expression patterns in metastatic niche-associated hepatocytes in a murine model of liver metastasis. Among the marker genes of metastatic niche-associated hepatocytes, we identify Lgals3, encoding galectin-3, as a potential pro-metastatic factor that accelerates metastatic growth and invasion.


Subject(s)
Liver Neoplasms , Humans , Mice , Animals , Liver Neoplasms/metabolism , Hepatocytes/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Extracellular Matrix/metabolism , Cell Communication
18.
J Cell Sci ; 123(Pt 5): 747-55, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20164304

ABSTRACT

Visualization and quantification of the dynamics of protein-protein interactions in living cells can be used to explore the macromolecular events involved in signal transduction processes. In this study, functional molecular imaging using a luciferase-based complementation method demonstrated how the integrin-linked kinase (ILK)-mediated protein complex controls downstream signals. The luciferase complementation assay showed that Akt1 preferentially binds to beta-parvin rather than to ILK within the complex. Moreover, photon flux from the interaction between beta-parvin and Akt1 increased following serum stimulation, and the beta-parvin-Akt1 interaction was dependent on phosphoinositide 3-kinase. Intriguingly, small interfering (si)RNA-mediated beta-parvin knockdown increased photon flux from the interaction between ILK and Akt1, leading to stabilization of hypoxia-inducible factor-1alpha and increased expression of vascular endothelial growth factor-A. These data from functional molecular imaging demonstrated that beta-parvin plays a regulatory role in the ILK-mediated Akt (also called protein kinase B) signaling cascades, suggesting that beta-parvin might be a crucial modulator of cell survival.


Subject(s)
Actinin/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Actinin/genetics , Animals , Blotting, Western , Cell Line , Cell Survival/genetics , Cell Survival/physiology , Humans , Hypoxia-Inducible Factor 1/metabolism , Immunoprecipitation , Mice , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism
19.
Bioconjug Chem ; 23(3): 324-9, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22335430

ABSTRACT

We developed a novel near-infrared (NIR) fluorescent probe, GPU-167, for in vivo imaging of tumor hypoxia. GPU-167 comprises a tricarbocyanine dye as an NIR fluorophore and two 2-nitroimidazole moieties as exogenous hypoxia markers that undergo bioreductive activation and then selective entrapment in hypoxic cells. After treatment with GPU-167, tumor cells contained significantly higher levels of fluorescence in hypoxia than in normoxia. In vivo fluorescence imaging specifically detected GPU-167 in tumors 24 h after administration. Ex vivo analysis revealed that fluorescence showed a strong correlation with hypoxia inducible factor (HIF)-1 active hypoxic regions. These data suggest that GPU-167 is a promising in vivo optical imaging probe for tumor hypoxia.


Subject(s)
Carbocyanines/chemistry , Cell Hypoxia , Fluorescent Dyes/chemistry , Neoplasms/pathology , Nitroimidazoles/chemistry , Spectroscopy, Near-Infrared/methods , Animals , Mice , Mice, Nude
20.
J Biomed Biotechnol ; 2012: 262741, 2012.
Article in English | MEDLINE | ID: mdl-22778544

ABSTRACT

PURPOSE: We aimed to clearly visualize heterogeneous distribution of hypoxia-inducible factor 1α (HIF) activity in tumor tissues in vivo. METHODS: We synthesized of (125)I-IPOS, a (125)I labeled chimeric protein probe, that would visualize HIF activity. The biodistribution of (125)I-IPOS in FM3A tumor-bearing mice was evaluated. Then, the intratumoral localization of this probe was observed by autoradiography, and it was compared with histopathological findings. The distribution of (125)I-IPOS in tumors was imaged by a small animal SPECT/CT scanner. The obtained in vivo SPECT-CT fusion images were compared with ex vivo images of excised tumors. Fusion imaging with MRI was also examined. RESULTS: (125)I-IPOS well accumulated in FM3A tumors. The intratumoral distribution of (125)I-IPOS by autoradiography was quite heterogeneous, and it partially overlapped with that of pimonidazole. High-resolution SPECT-CT fusion images successfully demonstrated the heterogeneity of (125)I-IPOS distribution inside tumors. SPECT-MRI fusion images could give more detailed information about the intratumoral distribution of (125)I-IPOS. CONCLUSION: High-resolution SPECT images successfully demonstrated heterogeneous intratumoral distribution of (125)I-IPOS. SPECT-CT fusion images, more favorably SPECT-MRI fusion images, would be useful to understand the features of heterogeneous intratumoral expression of HIF activity in vivo.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Imaging/methods , Molecular Probes/pharmacokinetics , Tomography, Emission-Computed, Single-Photon/methods , Animals , Female , Immunohistochemistry , Iodine Radioisotopes/pharmacokinetics , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred C3H , Neoplasms, Experimental/metabolism , Nitroimidazoles/pharmacokinetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL