Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Oncol (Dordr) ; 2023 Nov 04.
Article in English | MEDLINE | ID: mdl-37924456

ABSTRACT

BACKGROUND: Cancer/testis antigen-45A1 (CT45A1) is overexpressed in various types of cancer but is not expressed in healthy women. The role of CT45A1 in cervical cancer has not yet been described in the literature. PURPOSE: The aim of this research was to study the role of CT45A1 in cervical cancer progression and drug resistance, elucidate the mechanisms underlying CT45A1-mediated tumorigenesis and investigate CT45A1 as a biomarker for cervical cancer diagnosis, prognostic prediction, and targeted therapy. METHODS: The CT45A1 levels in the tumors from cervical cancer patients were measured using immunohistochemical staining. The role and mechanisms underlying CT45A1-mediated cervical cancer cell tumor growth, invasion, and drug resistance were studied using xenograft mice, cervical cancer cells, immunohistochemistry, RNA-seq, real-time qPCR, Chromatin immunoprecipitation and Western blotting. RESULTS: CT45A1 levels were notably high in the tumor tissues of human cervical cancer patients compared to the paracancerous tissues (p < 0.001). Overexpression of CT45A1 was closely associated with poor prognosis in cervical cancer patients. CT45A1 promoted cervical cancer cell tumor growth, invasion, neovascularization, and drug resistance. Mechanistically, CT45A1 promoted the expression of 128 pro-tumorigenic genes and concurrently activated key signaling pathways, including the oncogenic SRC, ERK, CREB, and YAP/TAZ signaling pathways. Furthermore, CT45A1-mediated tumorigenesis and drug resistance were markedly inhibited by the small molecule lycorine. CONCLUSION: CT45A1 promotes cervical cancer cell tumorigenesis, neovascularization, and drug resistance by activating oncogenic SRC and downstream tumorigenic signaling pathways. These findings provide new insight into the pathogenesis of cervical cancer and offer a new platform for the development of novel therapeutics against cervical cancer.

2.
Cell Death Dis ; 14(4): 293, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185462

ABSTRACT

Expression of the long non-coding RNA (lncRNA) keratin-7 antisense (KRT7-AS) is downregulated in various types of cancer; however, the impact of KRT7-AS deficiency on tumorigenesis and apoptosis is enigmatic. We aim to explore the influence of KRT7-AS in carcinogenesis and apoptosis. We found that KRT7-AS was deficient in breast and lung cancers, and low levels of KRT7-AS were a poor prognostic factor in breast cancer. Cellular studies showed that silencing of KRT7-AS in lung cancer cells increased oncogenic Keratin-7 levels and enhanced tumorigenesis, but diminished cancer apoptosis of the cancer cells; by contrast, overexpression of KRT7-AS inhibited lung cancer cell tumorigenesis. Additionally, KRT7-AS sensitized cancer cells to the anti-cancer drug cisplatin, consequently enhancing cancer cell apoptosis. In vivo, KRT7-AS overexpression significantly suppressed tumor growth in xenograft mice, while silencing of KRT7-AS promoted tumor growth. Mechanistically, KRT7-AS reduced the levels of oncogenic Keratin-7 and significantly elevated amounts of the key tumor suppressor PTEN in cancer cells through directly binding to PTEN protein via its core nucleic acid motif GGCAAUGGCGG. This inhibited the ubiquitination-proteasomal degradation of PTEN protein, therefore elevating PTEN levels in cancer cells. We also found that KRT7-AS gene transcription was driven by the transcription factor RXRα; intriguingly, the small molecule berberine enhanced KRT7-AS expression, reduced tumorigenesis, and promoted apoptosis of cancer cells. Collectively, KRT7-AS functions as a new tumor suppressor and an apoptosis enhancer in lung and breast cancers, and we unraveled that the RXRα-KRT7-AS-PTEN signaling axis controls carcinogenesis and apoptosis. Our findings highlight a tumor suppressive role of endogenous KRT7-AS in cancers and an important effect the RXRα-KRT7-AS-PTEN axis on control of cancer cell tumorigenesis and apoptosis, and offer a new platform for developing novel therapeutics against cancers.


Subject(s)
Breast Neoplasms , Lung Neoplasms , RNA, Long Noncoding , Humans , Animals , Mice , Female , Breast Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Keratin-7/genetics , Keratin-7/metabolism , Cell Line, Tumor , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Apoptosis/genetics , Lung Neoplasms/genetics , Lung/metabolism , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic
3.
Theranostics ; 12(5): 2063-2079, 2022.
Article in English | MEDLINE | ID: mdl-35265199

ABSTRACT

Background: Dietary tyrosine regulating melanoma progression has been well-recognized. However, whether tyrosine-based melanin anabolism contributes to pulmonary and cerebral organotropic colonization of melanoma remains elusive. Furthermore, approaches based on targeting tyrosinase activity to inhibiting multi-organ metastasis of melanoma cells need to be designed and validated. Methods: Patients derived melanoma cells and mouse B16 melanoma cells with different pigmentation were employed in this investigation. Tyrosine content dynamics in tumors and multiple organs during the melanoma progression was monitored, and tyrosine-based melanin synthesis of melanoma cells derived from multi-organ was determined. Additionally, we also adopted RNA-seq, flow cytometry, real-time PCR and composite metastasis mouse model to analyze organotropic colonization and to validate designed therapeutic strategies. Results: B16 melanoma cells with high activity of tyrosinase and sensitivity of tyrosine utilization for melanin synthesis (Tyr-H cells) easily colonized in the lung, while B16 melanoma cells lacking above characteristics (Tyr-L cells) exhibited potent proliferation in the brain. Mechanistically, Tyr-H cells recruited and trained neutrophils and macrophages to establish pulmonary metastatic niche dependent on highly secreted CXCL1 and CXCL2 and an excessive melanosome accumulation-induced cell death. Tyr-L cells enhanced PD-L1 expression in tumor-infiltrated macrophages when they are progressing in the brain. Accordingly, intervention of tyrosinase activity (2-Ethoxybenzamide or hydroquinone) in combination with inhibitors of phagocytosis (GSK343) or chemotaxis (SB225002) suppressed organotropic colonization and significantly improved the survival of melanoma- bearing mice treated with immune checkpoint blockade (PD1 antibody). Conclusions: The heterogeneity of melanoma cells in utilization of tyrosine is associated with organotropic colonization, providing the basis for developing new strategies to combat melanoma.


Subject(s)
Melanins , Melanoma, Experimental , Animals , Cell Line, Tumor , Humans , Lung/pathology , Mice , Monophenol Monooxygenase/metabolism , Tumor Microenvironment , Tyrosine
4.
Cell Death Dis ; 10(6): 440, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31165736

ABSTRACT

The reconstitution of the T-cell repertoire and quantity is a major challenge in the clinical management of HIV infection/AIDS, cancer, and aging-associated diseases. We previously showed that autologous bone marrow transfusion (BMT) via the hepatic portal vein could effectively restore CD4+ T-cell count in AIDS patients also suffering from decompensated liver cirrhosis. In the current study, we characterized T-cell reconstitution in a mouse model of liver fibrosis induced by CCl4 and found that T-cell reconstitution after BMT via hepatic portal vein was also greatly enhanced. The expression of Dll4 (Delta-like 4), which plays an important role in T-cell progenitor expansion, was elevated in hepatocytes of fibrotic livers when compared to normal livers. This upregulation of Dll4 expression was found to be induced by TNFα in an NFκB-dependent manner. Liver fibroblasts transfected with Dll4 (LF-Dll4) also gained the capacity to promote T-cell lineage development from hematopoietic stem cells (HSCs), resulting in the generation of DN2 (CD4 and CD8 DN 2) and DN3 T-cell progenitors in vitro, which underwent a normal maturation program when adoptively transferred into Rag-2 deficient hosts. We also demonstrated a pivotal role of SDF-1 produced by primary liver fibroblasts (primary LF) in T-lineage differentiation from HSCs. These results suggest that Dll4 and SDF-1 in fibrotic liver microenvironment could promote extrathymic T-cell lineage development. These results expand our knowledge of T-cell development and reconstitution under pathological conditions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Cell Differentiation , Chemokine CXCL12/metabolism , Hematopoietic Stem Cells/metabolism , Liver Cirrhosis/metabolism , T-Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Transplantation , Calcium-Binding Proteins/genetics , Carbon Tetrachloride , Cell Differentiation/genetics , Cells, Cultured , Chemokine CXCL12/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , HIV Infections/metabolism , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , NIH 3T3 Cells , Tumor Necrosis Factor-alpha/metabolism
5.
Stem Cell Res Ther ; 10(1): 18, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635041

ABSTRACT

BACKGROUND: Mesenchymal stem/stromal cells (MSCs) have been widely used to treat various inflammatory diseases. The immunomodulatory capabilities of MSCs are usually licensed by inflammatory cytokines and may vary depending on the levels and the types of inflammatory cytokines. However, how the inflammatory microenvironment affects the fate of MSCs remains elusive. Here we characterized the molecular mechanism underlying the apoptosis of mouse MSCs triggered by the synergistic action of IFNγ and TNFα. METHODS: We isolated and expanded MSCs by flushing the femoral and tibial bone marrow of wild-type, iNOS-/-, and Fas-/- mice. BM-MSCs were treated with IFNγ and TNFα in vitro, and cell viability was evaluated by a CCK-8 kit. Apoptosis was assessed by Annexin V/propidium iodide-stained flow cytometry. Expression of genes related to apoptosis and endoplasmic reticulum (ER) stress was measured by reverse transcription-polymerase chain reaction (RT-PCR). Apoptosis and autophagy-related proteins were examined by Western blot analysis. RESULTS: IFNγ and TNFα synergistically trigger apoptosis of mouse BM-MSCs. The two cytokines were shown to stimulate the expression of inducible nitric oxide synthase (iNOS) and consequently the generation of nitric oxide (NO), which is required for the apoptosis of mouse BM-MSCs. The two cytokines similarly induced apoptosis in Fas-/- BM-MSCs. iNOS and NO were shown to upregulate Fas in mouse MSCs and sensitize them to Fas agonist-induced apoptosis. Moreover, NO stimulated by IFNγ/TNFα impairs autophagy, which aggravates ER stress and promotes apoptosis. CONCLUSIONS: IFNγ/TNFα-induced apoptosis in mouse MSCs is mediated by NO. Our findings shed new light on cytokine-induced apoptosis of MSCs and have implications in MSC-based therapy of inflammatory diseases.


Subject(s)
Inflammation/therapy , Interferon-gamma/genetics , Mesenchymal Stem Cells/metabolism , Tumor Necrosis Factor-alpha/genetics , Animals , Apoptosis/genetics , Autophagy/genetics , Cell Proliferation/genetics , Cellular Microenvironment/genetics , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Inflammation/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide Synthase Type II/genetics , fas Receptor/genetics
6.
Curr Cancer Drug Targets ; 14(6): 549-56, 2014.
Article in English | MEDLINE | ID: mdl-25088038

ABSTRACT

The Neuronally expressed developmentally downregulated 4 (NEDD4), functioning largely as an E3 ubiquitin ligase, has been demonstrated to play a critical role in the development and progression of human cancers. In this review, to understand the regulatory mechanism(s) of NEDD4 as well as the signaling pathways controlled by NEDD4, we briefly describe the NEDD4 upstream regulators and its downstream ubiquitin substrates. Moreover, we further discuss its oncogenic roles in human malignancies. Therefore, targeting NEDD4 could be a potential therapeutic strategy for treatment of human cancers.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Design , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/therapeutic use , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Antineoplastic Agents/adverse effects , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Nedd4 Ubiquitin Protein Ligases , Neoplasms/enzymology , Neoplasms/pathology , Proteasome Inhibitors/adverse effects , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
7.
Biochim Biophys Acta ; 1836(2): 273-86, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23933263

ABSTRACT

Robust neovascularization and lymphangiogenesis have been found in a variety of aggressive and metastatic tumors. Endothelial sprouting angiogenesis is generally considered to be the major mechanism by which new vasculature forms in tumors. However, increasing evidence shows that tumor vasculature is not solely composed of endothelial cells (ECs). Some tumor cells acquire processes similar to embryonic vasculogenesis and produce new vasculature through vasculogenic mimicry, trans-differentiation of tumor cells into tumor ECs, and tumor cell-EC vascular co-option. In addition, tumor cells secrete various vasculogenic factors that induce sprouting angiogenesis and lymphangiogenesis. Vasculogenic tumor cells actively participate in the formation of vascular cancer stem cell niche and a premetastatic niche. Therefore, tumor cell-mediated neovascularization and lymphangiogenesis are closely associated with tumor progression, cancer metastasis, and poor prognosis. Vasculogenic tumor cells have emerged as key players in tumor neovascularization and lymphangiogenesis and play pivotal roles in tumor progression and cancer metastasis. However, the mechanisms underlying tumor cell-mediated vascularity as they relate to tumor progression and cancer metastasis remain unclear. Increasing data have shown that various intrinsic and extrinsic factors activate oncogenes and vasculogenic genes, enhance vasculogenic signaling pathways, and trigger tumor neovascularization and lymphangiogenesis. Collectively, tumor cells are the instigators of neovascularization. Therefore, targeting vasculogenic tumor cells, genes, and signaling pathways will open new avenues for anti-tumor vasculogenic and metastatic drug discovery. Dual targeting of endothelial sprouting angiogenesis and tumor cell-mediated neovascularization and lymphangiogenesis may overcome current clinical problems with anti-angiogenic therapy, resulting in significantly improved anti-angiogenesis and anti-cancer therapies.


Subject(s)
Cell Transformation, Neoplastic/pathology , Lymphangiogenesis , Neoplasms/pathology , Neovascularization, Pathologic , Tumor Microenvironment , Animals , Disease Progression , Humans , Neoplasm Metastasis , Neoplasms/blood supply , Signal Transduction
8.
Toxicol Lett ; 218(2): 174-85, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23376478

ABSTRACT

Uncontrolled tumor cell proliferation and robust neovascularization are prominent features of aggressive ovarian cancers. Although great efforts in anti-ovarian cancer therapy have been made in the past 4 decades, the 5-year survival rates for ovarian cancer patients are still poor, and effective drugs to cure ovarian cancer patients are absent. In this study, we evaluated the anti-cancer effects of lycorine hydrochloride (LH), a novel anti-ovarian cancer agent, using the highly-invasive ovarian cancer cell line, Hey1B, as a model. Our data showed that LH effectively inhibited mitotic proliferation of Hey1B cells (half maximal inhibitory concentration=1.2µM) with very low toxicity, resulting in cell cycle arrest at the G2/M transition through enhanced expression of the cell cycle inhibitor p21 and marked down-regulation of cyclin D3 expression. Moreover, LH suppressed both the formation of capillary-like tubes by Hey1B cells cultured in vitro and the ovarian cancer cell-dominant neovascularization in vivo when administered to Hey1B-xenotransplanted mice. LH also suppressed the expression of several key angiogenic genes, including VE-cadherin, vascular endothelial growth factor, and Sema4D, and reduced Akt phosphorylation in Hey1B cells. These results suggest that LH selectively inhibits ovarian cancer cell proliferation and neovascularization and is a potential drug candidate for anti-ovarian cancer therapy.


Subject(s)
Amaryllidaceae Alkaloids/pharmacology , Cell Proliferation/drug effects , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/pathology , Phenanthridines/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cadherins/genetics , Cadherins/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin D3/genetics , Cyclin D3/metabolism , Down-Regulation , Female , Humans , Mice , Mice, Inbred BALB C , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Semaphorins/genetics , Semaphorins/metabolism
9.
Pigment Cell Melanoma Res ; 25(5): 630-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22781316

ABSTRACT

Melanoma cells actively participate in tumor angiogenesis and vasculogenic mimicry. However, anti-angiogenic therapy in patients with melanoma has not shown a significant survival gain. Thus, new anti-melanoma angiogenic and vasculogenic drugs are highly desired. Using the metastatic melanoma cell line C8161 as a model, we explored melanoma vasculogenic inhibitors and found that lycorine hydrochloride (LH) effectively suppressed C8161 cell-dominant formation of capillary-like tubes in vitro and generation of tumor blood vessels in vivo with low toxicity. Mechanistic studies revealed that LH markedly hindered expression of VE-cadherin in C8161 cells, but did not affect expression of six other important angiogenic and vasculogenic genes. Luciferase assays showed that LH significantly impeded promoter activity of the VE-cadherin gene in a dose-dependent manner. Together, these data suggest that LH inhibits melanoma C8161 cell-dominant vasculogenic mimicry by reducing VE-cadherin gene expression and diminishing cell surface exposure of the protein.


Subject(s)
Amaryllidaceae Alkaloids/therapeutic use , Melanoma/blood supply , Melanoma/drug therapy , Neovascularization, Pathologic/drug therapy , Phenanthridines/therapeutic use , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy , Amaryllidaceae Alkaloids/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Models, Biological , Neoplasm Metastasis , Neovascularization, Pathologic/pathology , Phenanthridines/pharmacology , Promoter Regions, Genetic/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology
10.
Hybridoma (Larchmt) ; 31(3): 214-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22741587

ABSTRACT

SZ117 is a monoclonal antibody against matrix metalloproteinase-2 (MMP-2) and exhibits anti-tumor angiogenic effect. In this study, we observed that SZ117 bound to a 280 kDa protein, which was detected in tumor cell-derived Matrigel and various tumor cells. Using immunoprecipitation, mass spectrometry analysis, and Western blot analysis, we identified the 280 kDa protein as filamin A and found that filamin A and its degraded products, notably a 53 kDa fragment, were released from a variety of tumor cells. This suggests that SZ117 is useful in the study of the pathogenesis of filamin A and that blockage of filamin A by SZ117 might contribute to the anti-tumor angiogenic effect of the monoclonal antibody.


Subject(s)
Antibodies, Monoclonal/immunology , Contractile Proteins/immunology , Microfilament Proteins/immunology , Peptide Fragments/immunology , Amino Acid Sequence , Contractile Proteins/chemistry , Contractile Proteins/metabolism , Filamins , Humans , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Molecular Sequence Data , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Tumor Cells, Cultured
11.
Leuk Lymphoma ; 53(12): 2456-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22582896

ABSTRACT

Anti-lymphoma therapy continues to present a major challenge. Even though cytotoxic therapy, immunotherapy and molecularly targeted therapy have been used in the clinic to treat the disease, effective anti-lymphoma drugs are still needed. In this study, we explored novel anti-lymphoma agents and found that scutellarin, an active component of a traditional Chinese medicinal herb Erigeron breviscapus, executed an anti-lymphoma effect. Scutellarin diminished the proliferation of B-lymphoma Namalwa cells in vitro and inhibited lymphoma growth in Namalwa cell-xenotransplanted mice without obvious toxicity. A mechanism study showed that scutellarin at doses of less than 10 µM induced cell cycle arrest at G0/G1 transition without the induction of cell apoptosis, which was accompanied by down-regulation of cyclin D1 and CDK4 expression. In contrast, scutellarin at concentrations of 15 µM or above promoted Namalwa cell apoptosis, which was partially associated with the activation of caspases. These results suggest that scutellarin is a new potential anti-lymphoma candidate.


Subject(s)
Apigenin/pharmacology , Apoptosis/drug effects , Burkitt Lymphoma/prevention & control , Cell Proliferation/drug effects , Glucuronates/pharmacology , Animals , Apigenin/chemistry , Blotting, Western , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Dose-Response Relationship, Drug , Erigeron/chemistry , Female , Gene Expression , Glucuronates/chemistry , HL-60 Cells , HeLa Cells , Humans , Jurkat Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Phytotherapy , Reverse Transcriptase Polymerase Chain Reaction , U937 Cells , Xenograft Model Antitumor Assays
12.
Cancer Microenviron ; 5(3): 211-23, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22592483

ABSTRACT

Tumor microenvironment (TME) is important in tumor development and may be a target for anti-cancer therapy. The genesis of TME is a dynamic process that is regulated by intrinsic and extrinsic factors and coordinated by multiple genes, cells, and signal pathways. Cancer anaerobic metabolism and various oncogenes may stimulate the genesis of TME. Tumor cells and cancer stem cells actively participate in the genesis of the cancer stem cell niche and tumor neovascularization, important in the initiation of the TME. Various cancer-associated stromal cells, derived niche factors, and tumor-associated macrophages may function as promoters in the genesis of the TME. Dicer1 gene-deleted stromal cells can induce generation of cancer stem cells and initiate tumorigenesis, suggesting that stromal cells also may promote the genesis of the TME. Therefore, the key features of TME include niche-driving oncogenes, cancer anaerobic metabolism, niche-driving cancer stem cells, neovascularization, tumor-associated inflammatory cells, and cancer-associated stromal cells. These features are potential targets for normalization of the malignant TME and effective anti-cancer therapy.

13.
Front Med ; 6(1): 67-78, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22460450

ABSTRACT

Antitumor angiogenic therapy has been shown promising in the treatment of several advanced cancers since the approval of the first antiangiogenic drug Avastin in 2004. Although the current antiangiogenic drugs reduce the density of tumor blood vessels and result in tumor shrinkage at the early stage of treatment, recent studies have shown that antiangiogenic therapy has transient and insufficient efficacy, resulting in tumor recurrence in patients after several months of treatment. Blockage of blood and oxygen supplies creates a hypoxic and acidic microenvironment in the tumor tissues, which fosters tumor cells to become more aggressive and metastatic. In 2001, Jain proposed tumor vascular normalization as an alternative approach to treating cancers based on the pioneering work on tumor blood vessels by several other researchers. At present, normalizing the disorganized tumor vasculature, rather than disrupting or blocking them, has emerged as a new option for anticancer therapy. Preclinical and clinical data have shown that tumor vascular normalization using monoclonal antibodies, proteins, peptides, small molecules, and pericytes resulted in decreased tumor size and reduced metastasis. However, current tumor vascular normalizing drugs display moderate anticancer efficacy. Accumulated data have shown that a variety of vasculogenic/angiogenic tumor cells and genes play important roles in tumor neovascularization, growth, and metastasis. Therefore, multiple-targeting of vasculogenic tumor cells and genes may improve the efficacy of tumor vascular normalization. To this end, the combination of antiangiogenic drugs with tumor vascular normalizing therapeutics, as well as the integration of Western medicine with traditional Chinese medicine, may provide a good opportunity for discovering novel tumor vascular normalizing drugs for an effective anticancer therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Angiogenesis Inhibitors/pharmacology , Cadherins/metabolism , Drugs, Chinese Herbal , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Macrophages/metabolism , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Procollagen-Proline Dioxygenase/metabolism , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
14.
Hybridoma (Larchmt) ; 31(1): 63-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22142190

ABSTRACT

In this study, using an in vitro tube formation model, we observed that SZ117, a monoclonal antibody against matrix metalloproteinase-2 (MMP2), attenuated a capillary-like tube structure formed by tumor endothelial cell 3B11 and human sarcoma cell MG63. In addition, gelatin zymography showed that SZ117 markedly inhibited MMP2 activity, but did not affect the capability of MMP9-mediated gelatin degradation. These data suggest that SZ117 might have an anti-tumor angiogenic effect and that angiogenic tumor cells and MMP2 may be targeted by monoclonal antibodies for novel anti-tumor angiogenic and anti-cancerous drug discovery.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Matrix Metalloproteinase 2/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Hybridomas , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/immunology
15.
Stem Cells Dev ; 21(6): 955-64, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22185567

ABSTRACT

Induced pluripotent stem cells (iPSCs) have recently boomed enthusiasm in stem cell therapy, whereas high potential tumorigenesis of iPSCs has become the biggest obstacle for clinic application and the tumorigenic genes in iPSCs have not been well documented. In this investigation, using tools of bioinformatics, we analyzed the all available datasets regarded to iPSCs from 11 differentiated cell lines and revealed 593 iPSC consensus genes. Notably, of the 593 genes, 209 were expressed in human tumor cell lines and cancer tissues, and some of them were expressed in the iPSC-differentiated hepatocytes; remarkably, 5 oncogenes were overexpressed in the iPSCs and an oncogene RAB25 in the iPSC-differentiated cells, suggesting that these iPSC consensus genes are implicated with the risk of tumorigenesis and cancers. This investigation provides useful information for designing new strategies and methods to curtail the expression of oncogenic genes in iPSCs and produce safe iPSC derivatives for stem cell therapy.


Subject(s)
Cell Transformation, Neoplastic/pathology , Consensus Sequence/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Neoplasms/genetics , Cell Line , Cell Transformation, Neoplastic/genetics , Computational Biology , Genes , Hepatocytes , Humans , Induced Pluripotent Stem Cells/transplantation , Neoplasms/pathology , Oncogenes , Risk , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...