1
|
Li Q, Zhang L, Yang Q, Li M, Pan X, Xu J, Zhong C, Yao F, Zhang R, Zhou S, Dai X, Shi X, Dai Y, Xu J, Cheng X, Xiao W, She Z, Wang K, Qian X, Pu L, Zhang P, Wang X. Thymidine kinase 1 drives hepatocellular carcinoma in enzyme-dependent and -independent manners. Cell Metab 2023:S1550-4131(23)00095-5. [PMID: 37071992 DOI: 10.1016/j.cmet.2023.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/17/2022] [Accepted: 03/24/2023] [Indexed: 04/20/2023]
Abstract
Metabolic reprogramming plays a crucial role in the development of hepatocellular carcinoma (HCC). However, the key drivers of metabolic reprogramming underlying HCC progression remain unclear. Using a large-scale transcriptomic database and survival correlation screening, we identify thymidine kinase 1 (TK1) as a key driver. The progression of HCC is robustly mitigated by TK1 knockdown and significantly aggravated by its overexpression. Furthermore, TK1 promotes the oncogenic phenotypes of HCC not only through its enzymatic activity and production of deoxythymidine monophosphate (dTMP) but also by promoting glycolysis via binding with protein arginine methyltransferase 1 (PRMT1). Mechanistically, TK1 directly binds PRMT1 and stabilizes it by interrupting its interactions with tripartite-motif-containing 48 (TRIM48), which inhibits its ubiquitination-mediated degradation. Subsequently, we validate the therapeutic capacity of hepatic TK1 knockdown in a chemically induced HCC mouse model. Therefore, targeting both the enzyme-dependent and -independent activity of TK1 may be therapeutically promising for HCC treatment.
Collapse
Affiliation(s)
- Qing Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Liren Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Qin Yang
- Department of Cardiovascular Surgery, Huanggang Central Hospital, Huanggang Institute of Translational Medicine, Huanggang, China
| | - Mei Li
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Xiongxiong Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiali Xu
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Zhong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Feifan Yao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Ruizhi Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Suiqing Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Xinzheng Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Xiaoli Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Yongjiu Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Jing Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Cheng
- School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenchang Xiao
- Department of Cardiovascular Surgery, Huanggang Central Hospital, Huanggang Institute of Translational Medicine, Huanggang, China
| | - Zhigang She
- School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ke Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| | - Xiaofeng Qian
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| | - Peng Zhang
- School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| |
Collapse
|
2
|
Li M, Wang X, Wang Y, Bao S, Chang Q, Liu L, Zhang S, Sun L. Strategies for Remodeling the Tumor Microenvironment Using Active Ingredients of Ginseng-A Promising Approach for Cancer Therapy. Front Pharmacol 2022; 12:797634. [PMID: 35002732 PMCID: PMC8727883 DOI: 10.3389/fphar.2021.797634] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/06/2021] [Indexed: 12/21/2022] Open
Abstract
The tumor microenvironment (TME) plays a key role in promoting the initiation and progression of tumors, leading to chemoradiotherapy resistance and immunotherapy failure. Targeting of the TME is a novel anti-tumor therapeutic approach and is currently a focus of anti-tumor research. Panax ginseng C. A. Meyer (ginseng), an ingredient of well-known traditional Asia medicines, exerts beneficial anti-tumor effects and can regulate the TME. Here, we present a systematic review that describes the current status of research efforts to elucidate the functions and mechanisms of ginseng active components (including ginsenosides and ginseng polysaccharides) for achieving TME regulation. Ginsenosides have variety effects on TME, such as Rg3, Rd and Rk3 can inhibit tumor angiogenesis; Rg3, Rh2 and M4 can regulate the function of immune cells; Rg3, Rd and Rg5 can restrain the stemness of cancer stem cells. Ginseng polysaccharides (such as red ginseng acidic polysaccharides and polysaccharides extracted from ginseng berry and ginseng leaves) can regulate TME mainly by stimulating immune cells. In addition, we propose a potential mechanistic link between ginseng-associated restoration of gut microbiota and the tumor immune microenvironment. Finally, we describe recent advances for improving ginseng efficacy, including the development of a nano-drug delivery system. Taken together, this review provides novel perspectives on potential applications for ginseng active ingredients as anti-cancer adjuvants that achieve anti-cancer effects by reshaping the tumor microenvironment.
Collapse
Affiliation(s)
- Mo Li
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China.,Department of Thyroid Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xin Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Shunchao Bao
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Qing Chang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Linlin Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Shuai Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
3
|
Kim HK, Choi JS, Lee SW, Joo CK, Joe YA. A Novel Peptide Derived From Tissue-Type Plasminogen Activator Potently Inhibits Angiogenesis and Corneal Neovascularization. J Cell Biochem 2017; 118:1132-1143. [DOI: 10.1002/jcb.25732] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 09/09/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Hyun-Kyung Kim
- Cancer Research Institute; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Department of Medical Lifescience; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Cancer Evolution Research Center; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| | - Jun-Sub Choi
- Department of Ophthalmology and Visual Science; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| | - Seung Woo Lee
- Cancer Research Institute; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Department of Medical Lifescience; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Cancer Evolution Research Center; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| | - Choun-Ki Joo
- Department of Ophthalmology and Visual Science; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| | - Young Ae Joe
- Cancer Research Institute; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Department of Medical Lifescience; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Cancer Evolution Research Center; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| |
Collapse
|
4
|
The Kringle Domain of Tissue-Type Plasminogen Activator Inhibits Extracellular Matrix-Induced Adhesion and Migration of Endothelial Cells. Biosci Biotechnol Biochem 2014; 72:2303-8. [DOI: 10.1271/bbb.80152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
5
|
Endothelial progenitor cells: current development of their paracrine factors in cardiovascular therapy. J Cardiovasc Pharmacol 2012; 59:387-96. [PMID: 22157259 DOI: 10.1097/fjc.0b013e3182440338] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Endothelial progenitor cells were initially considered to radically alter the concepts of adult tissue angiogenesis for their contribution of incorporation into new blood vessels. Nevertheless, controversy arises over their mechanism of action due to rare cell population and decreased number and impaired activity under pathological changes. Recent studies show that endothelial progenitor cells also function in a paracrine manner by secreting multiple cytokines and growth factors, but the beneficial paracrine signals remain partially unidentified. In this review, we provide an overview of varieties and signal pathways of factors secreted by endothelial progenitor cells and further present the prospect of new ways to encourage cardiovascular protection such as neovascularization, reendothelialization of larger vessels, and myocardial remodeling based on the paracrine factors.
Collapse
|
6
|
Kim JW, Jung SY, Kwon YH, Lee JH, Lee YM, Lee BY, Kwon SM. Ginsenoside Rg3 attenuates tumor angiogenesis via inhibiting bioactivities of endothelial progenitor cells. Cancer Biol Ther 2012; 13:504-15. [PMID: 22406998 DOI: 10.4161/cbt.19599] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Accumulating evidence suggests that Ginsenoside Rg3 appears to inhibit tumor growth including Lewis lung carcinoma, intestinal adenocarcinomas or B16 melanoma by inhibiting cell proliferation, tumor cell invasion and metastasis. Endothelial progenitor cells (EPCs) appear to play a key role in the growth of early tumors by intervening with the angiogenic switch promoting tumor neovessel formation by producing angiogenic cytokines during tumor progression. This paper reports a novel mechanism of Ginsenoside Rg3, a candidate anticancer bio-molecule, on tumor angiogenesis by inhibiting the multiple bioactivities of EPCs. When Ginsenoside Rg3 was applied to the ex vivo cultured outgrowth ECs, a type of EPCs, it inhibited the cell proliferation, cell migration and tubular formation of EPCs. Importantly, Ginsenoside Rg3 attenuated the phosphorylation cascade of the VEGF dependent p38/ERK signaling in vitro. The xenograft tumor model clearly showed that Ginsenoside Rg3 suppresses tumor growth and tumor angiogenesis by inhibiting the mobilization of EPCs from the bone marrow microenvironment to the peripheral circulation and modulates VEGF-dependent tumor angiogenesis. In conclusion, this study provides a potential therapeutic molecule, Ginsenoside Rg3, as an anticancer drug by inhibiting the EPC bioactivities.
Collapse
Affiliation(s)
- Jae-Won Kim
- Department of Biomedical Science, Laboratory for Functional Foods & Nutrigenomics, CHA University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
7
|
Kwon YH, Jung SY, Kim JW, Lee SH, Lee JH, Lee BY, Kwon SM. Phloroglucinol inhibits the bioactivities of endothelial progenitor cells and suppresses tumor angiogenesis in LLC-tumor-bearing mice. PLoS One 2012; 7:e33618. [PMID: 22496756 PMCID: PMC3322124 DOI: 10.1371/journal.pone.0033618] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 02/14/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND There is increasing evidence that phloroglucinol, a compound from Ecklonia cava, induces the apoptosis of cancer cells, eventually suppressing tumor angiogenesis. METHODOLOGY/PRINCIPAL FINDINGS This is the first report on phloroglucinol's ability to potentially inhibit the functional bioactivities of endothelial progenitor cells (EPCs) and thereby attenuate tumor growth and angiogenesis in the Lewis lung carcinoma (LLC)-tumor-bearing mouse model. Although Phloroglucinol did not affect their cell toxicity, it specifically inhibited vascular endothelial growth factor (VEGF) dependent migration and capillary-like tube formation of EPCs. Our matrigel plug assay clearly indicated that orally injected phloroglucinol effectively disrupts VEGF-induced neovessel formation. Moreover, we demonstrated that when phloroglucinol is orally administered, it significantly inhibits tumor growth and angiogenesis as well as CD45(-)/CD34(+) progenitor mobilization into peripheral blood in vivo in the LLC-tumor-bearing mouse model. CONCLUSIONS/SIGNIFICANCE These results suggest a novel role for Phloroglucinol: Phloroglucinol might be a modulator of circulating EPC bioactivities, eventually suppressing tumorigenesis. Therefore, phloroglucinol might be a candidate compound for biosafe drugs that target tumor angiogenesis.
Collapse
MESH Headings
- Administration, Oral
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Apoptosis/drug effects
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/prevention & control
- Cell Movement/drug effects
- Cells, Cultured
- Collagen/metabolism
- Drug Combinations
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Indicators and Reagents/administration & dosage
- Laminin/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic/prevention & control
- Phloroglucinol/administration & dosage
- Proteoglycans/metabolism
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Yi-Hong Kwon
- Department of Biomedical Science, Laboratory for Functional Foods and Nutrigenomics, Department of Food Science and Biotechnology, CHA University, Kyunggi, Republic of Korea
| | - Seok-Yun Jung
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Medical Research Institute, Pusan National University, Gyeongsangnam-do, Republic of Korea
| | - Jae-Won Kim
- Department of Biomedical Science, Laboratory for Functional Foods and Nutrigenomics, Department of Food Science and Biotechnology, CHA University, Kyunggi, Republic of Korea
| | - Sang-Hun Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Medical Research Institute, Pusan National University, Gyeongsangnam-do, Republic of Korea
| | - Jun-Hee Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Medical Research Institute, Pusan National University, Gyeongsangnam-do, Republic of Korea
| | - Boo-Yong Lee
- Department of Biomedical Science, Laboratory for Functional Foods and Nutrigenomics, Department of Food Science and Biotechnology, CHA University, Kyunggi, Republic of Korea
- * E-mail: (B-YL); (S-MK)
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Medical Research Institute, Pusan National University, Gyeongsangnam-do, Republic of Korea
- * E-mail: (B-YL); (S-MK)
| |
Collapse
|
8
|
Enhancement of endothelial progenitor cell numbers and migration by H1152, a Rho kinase specific inhibitor. Biosci Biotechnol Biochem 2012; 76:172-5. [PMID: 22232255 DOI: 10.1271/bbb.110468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Endothelial progenitor cells (EPCs) are applied in the treatment of ischemic diseases. In ex vivo culture of human cord-blood derived EPCs, H1152, (S)-(+)-2-methyl-1-[(4-methyl-5-iso-quinolinyl) sulfonyl]-homopiperazine, markedly increased the number of EPCs. It also induced EPC migration, stimulated the phosphorylation of AKT, and reduced the expression of p27 in the EPCs. Thus H1152 can be used effectively in ex vivo expansion of EPCs.
Collapse
|
9
|
Chen L, Ackerman R, Guo AM. 20-HETE in neovascularization. Prostaglandins Other Lipid Mediat 2011; 98:63-8. [PMID: 22227460 DOI: 10.1016/j.prostaglandins.2011.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 12/09/2011] [Accepted: 12/19/2011] [Indexed: 12/18/2022]
Abstract
Cytochrome P450 4A/F (CYP4A/F) converts arachidonic acid (AA) to 20-HETE by ω-hydroxylation. The contribution of 20-HETE to the regulation of myogenic response, blood pressure, and mitogenic actions has been well summarized. This review focuses on the emerging role of 20-HETE in physiological and pathological vascularization. 20-HETE has been shown to regulate vascular smooth muscle cells (VSMC) and endothelial cells (EC) by affecting their proliferation, migration, survival, and tube formation. Furthermore, the proliferation, migration, secretion of proangiogenic molecules (such as HIF-1α, VEGF, SDF-1α), and tube formation of endothelial progenitor cells (EPC) are stimulated by 20-HETE. These effects are mediated through c-Src- and EGFR-mediated downstream signaling pathways, including MAPK and PI3K/Akt pathways, eNOS uncoupling, and NOX/ROS system activation. Therefore, the CYP4A/F-20-HETE system may be a therapeutic target for the treatment of abnormal angiogenic diseases.
Collapse
Affiliation(s)
- Li Chen
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | |
Collapse
|
10
|
Sueblinvong V, Weiss DJ. Stem cells and cell therapy approaches in lung biology and diseases. Transl Res 2010; 156:188-205. [PMID: 20801416 PMCID: PMC4201367 DOI: 10.1016/j.trsl.2010.06.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/14/2010] [Accepted: 06/16/2010] [Indexed: 12/19/2022]
Abstract
Cell-based therapies with embryonic or adult stem cells, including induced pluripotent stem cells, have emerged as potential novel approaches for several devastating and otherwise incurable lung diseases, including emphysema, pulmonary fibrosis, pulmonary hypertension, and the acute respiratory distress syndrome. Although initial studies suggested engraftment of exogenously administered stem cells in lung, this is now generally felt to be a rare occurrence of uncertain physiologic significance. However, more recent studies have demonstrated paracrine effects of administered cells, including stimulation of angiogenesis and modulation of local inflammatory and immune responses in mouse lung disease models. Based on these studies and on safety and initial efficacy data from trials of adult stem cells in other diseases, groundbreaking clinical trials of cell-based therapy have been initiated for pulmonary hypertension and for chronic obstructive pulmonary disease. In parallel, the identity and role of endogenous lung progenitor cells in development and in repair from injury and potential contribution as lung cancer stem cells continue to be elucidated. Most recently, novel bioengineering approaches have been applied to develop functional lung tissue ex vivo. Advances in each of these areas will be described in this review with particular reference to animal models.
Collapse
Key Words
- aec, alveolar epithelial cell
- ali, acute lung injury
- ards, acute respiratory distress syndrome
- basc, bronchioalveolar stem cell
- ccsp, clara cell secretory protein
- cf, cystic fibrosis
- cftr, cystic fibrosis transmembrane conductance regulator
- clp, cecal ligation and puncture
- copd, chronic obstructive pulmonary disease
- enos, endothelial nitric oxide synthetase
- epc, endothelial progenitor cell
- esc, embryonic stem cell
- fev1, forced expiratory volume in 1 second
- fvc, forced vital capacity
- gfp, green fluorescent protein
- hsc, hematopoietic stem cell
- ipf, idiopathic pulmonary fibrosis
- kgf, keratinocyte growth factor
- lps, lipopolysaccharide
- mct, monocrotaline
- mhc, major histocompatibility complex
- msc, mesenchymal stromal (stem) cell
- ph, pulmonary hypertension
- pro-spc, pro-surfactant protein c
- sca-1, stem cell antigen-1
Collapse
Affiliation(s)
- Viranuj Sueblinvong
- Division of Pulmonary, Critical Care and Allergy, Department of Medicine, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
11
|
Su L, Xu X, Zhao H, Gu Q, Zou H. In vitro and in vivo antiangiogenic activity of a novel deca-peptide derived from human tissue-type plasminogen activator kringle 2. Biochem Biophys Res Commun 2010; 396:1012-7. [PMID: 20471363 DOI: 10.1016/j.bbrc.2010.05.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 05/10/2010] [Indexed: 10/19/2022]
Abstract
A synthetic deca-peptide corresponding to the amino acid sequence Arg(54)-Trp(63) of human tissue-type plasminogen activator (t-PA) kringle 2 domain, named TKII-10, is produced and tested for its ability to inhibit endothelial cell proliferation, migration, tube formation in vitro, and angiogenesis in vivo. At the same time, another peptide TKII-10S composed of the same 10 amino acids as TKII-10, but in a different sequence, is also produced and tested. The results show that TKII-10 potently inhibits VEGF-stimulated endothelial cell migration and tube formation in a dose-dependent, as well as sequence-dependent, manner in vitro while it is inactive in inhibiting endothelial cell proliferation. Furthermore, TKII-10 potently inhibits angiogenesis in chick chorioallantoic membrane and mouse cornea. The middle four amino acids DGDA in their sequence play an important role in TKII-10 angiogenesis inhibition(.) These results suggest that TKII-10 is a novel angiogenesis inhibitor that may serve as a prototype for antiangiogenic drug development.
Collapse
Affiliation(s)
- Li Su
- Department of Ophthalmology, Shanghai First People's Hospital, Affiliate of Shanghai Jiaotong University, No. 100 Haining Road, Shanghai 200080, PR China
| | | | | | | | | |
Collapse
|
12
|
Kim HK, Joe YA. DGDA, a local sequence of the kringle 2 domain, is a functional motif of the tissue-type plasminogen activator’s antiangiogenic kringle domain. Biochem Biophys Res Commun 2010; 391:166-9. [DOI: 10.1016/j.bbrc.2009.11.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 11/05/2009] [Indexed: 10/20/2022]
|
13
|
Lucas T, Abraham D, Untergasser G, Zins K, Hofer E, Gunsilius E, Aharinejad S. Adenoviral-mediated endothelial precursor cell delivery of soluble CD115 suppresses human prostate cancer xenograft growth in mice. Stem Cells 2009; 27:2342-52. [PMID: 19522014 PMCID: PMC2879317 DOI: 10.1002/stem.145] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Prostate cancer tumor growth and neovascularization is promoted by an interplay between migratory tumor stromal cells such as specialized tumor-associated macrophages (TAMs) and circulating endothelial precursor cells (CEPs). As vehicles for tumor therapy, human CEPs are relatively easy to isolate from peripheral blood, are able to proliferate long-term in vitro, are amenable to viral manipulation, and preferentially home to regions of ischemia found in growing tumors. We show here that human peripheral blood CEPs expanded ex vivo migrate to prostate cancer cells in vitro and efficiently home to human prostate tumor xenografts in vivo. Infection of precursors ex vivo with an adenovirus constructed to secrete a soluble form of the colony-stimulating factor-1 receptor CD115 that inhibits macrophage viability and migration in vitro significantly decreases the number of TAMs in xenografts (p < .05), reduces proliferation (p < .01) and vascular density (p < .03), and suppresses the growth of xenografts (p < .03). These data show for the first time that targeting stromal cell processes with cellular therapy has the potential to retard prostate tumor growth.
Collapse
Affiliation(s)
- Trevor Lucas
- Laboratory for Cardiovascular Research, Department of Anatomy and Cell Biology, Vienna Medical University, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
14
|
Suuronen EJ, Zhang P, Kuraitis D, Cao X, Melhuish A, McKee D, Li F, Mesana TG, Veinot JP, Ruel M. An acellular matrix-bound ligand enhances the mobilization, recruitment and therapeutic effects of circulating progenitor cells in a hindlimb ischemia model. FASEB J 2009; 23:1447-58. [PMID: 19136616 DOI: 10.1096/fj.08-111054] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Circulating progenitor cells home to and engraft to sites of ischemia, mediated in part by the adhesion molecule L-selectin; however, accumulation in tissues such as the heart is low. In this study, an acellular collagen-based matrix containing sialyl Lewis(X) (sLe(X)), which binds L-selectin, was developed in order to enhance the endogenous progenitor cell therapeutic response. Its effect on progenitor cells and angiogenesis were assessed in vitro and using a hindlimb ischemia model with rats. In culture, the sLe(X)-collagen matrix recruited more CD133(+)CD34(+)L-selectin(+) cells than collagen-only matrix, with adhesion mediated by L-selectin binding. Increased angiogenic/chemotactic cytokine production and improved resistance to apoptosis appeared in cells cultured on sLe(X)-collagen matrix. In vivo, mobilization of endogenous circulating progenitor cells was increased, and greater recruitment of these and systemically injected human peripheral blood CXCR4(+)L-selectin(+) cells to sLe(X)-collagen treated limbs was observed compared to collagen-only. This condition was associated with differences in angiogenic/chemotactic cytokine levels, with greater arteriole density and increased perfusion in sLe(X)-collagen treated hindlimbs. With these factors taken together, we demonstrated that an acellular matrix-bound ligand approach can enhance the mobilization, recruitment, and therapeutic effects of endogenous and/or transplanted progenitor cells, possibly through paracrine and antiapoptotic mechanisms, and could be used to improve cell-based regenerative therapies.
Collapse
Affiliation(s)
- Erik J Suuronen
- Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y 4W7, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Bishop-Bailey D. Tumour vascularisation: a druggable target. Curr Opin Pharmacol 2008; 9:96-101. [PMID: 19056315 DOI: 10.1016/j.coph.2008.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 10/24/2008] [Accepted: 10/24/2008] [Indexed: 01/17/2023]
Abstract
Tumour growth, spreading and metastasis require the development of a local vasculature. There have been great advances in the understanding of how this new vasculature arises, particularly in our increased knowledge of the process of angiogenesis, Although, a vast number of pro-angiogenic and anti-angiogenic mediators have been identified, one of the key signalling processes in the development of the tumour vasculature is the hypoxia-induced stimulation of vascular endothelial cell growth factors (VEGFs) production. Anti-VEGF therapy therefore not only provides a new paradigm for limiting tumour growth via targeting angiogenesis, but also provides prototypic agents to test the hypothesis that by controlling the development of the tumour vasculature we are able to limit, and potentially stop, tumour growth and spreading.
Collapse
Affiliation(s)
- David Bishop-Bailey
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, United Kingdom.
| |
Collapse
|
16
|
Stem cells and cell therapies in lung biology and lung diseases. Ann Am Thorac Soc 2008; 5:637-67. [PMID: 18625757 DOI: 10.1513/pats.200804-037dw] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
17
|
Zwolak P, Jasinski P, Terai K, Gallus NJ, Ericson ME, Clohisy DR, Dudek AZ. Addition of receptor tyrosine kinase inhibitor to radiation increases tumour control in an orthotopic murine model of breast cancer metastasis in bone. Eur J Cancer 2008; 44:2506-17. [DOI: 10.1016/j.ejca.2008.07.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 06/10/2008] [Accepted: 07/01/2008] [Indexed: 01/10/2023]
|
18
|
Kim HK, Oh DS, Lee SB, Ha JM, Joe YA. Antimigratory effect of TK1-2 is mediated in part by interfering with integrin alpha2beta1. Mol Cancer Ther 2008; 7:2133-41. [PMID: 18645023 DOI: 10.1158/1535-7163.mct-07-2405] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The recombinant two kringle domain of human tissue-type plasminogen activator (TK1-2) has been shown to inhibit endothelial cell proliferation, angiogenesis, and tumor cell growth despite of sharing a low amino acid sequence homology with angiostatin. Here, we explored a possible inhibitory mechanism of action of TK1-2 by focusing on antimigratory effect. TK1-2 effectively inhibited endothelial cell migration induced by basic fibroblast growth factor or vascular endothelial growth factor in a dose-dependent manner and tube formation on Matrigel. It blocked basic fibroblast growth factor-induced or vascular endothelial growth factor-induced phosphorylation of extracellular signal-regulated kinase 1/2 and formation of actin stress fibers and focal adhesions. Interestingly, TK1-2 alone induced the weak phosphorylation of focal adhesion kinase, whereas it inhibited focal adhesion kinase phosphorylation induced by growth factors. When immobilized, TK1-2 promoted adhesion and spreading of endothelial cells compared with bovine serum albumin. However, treatment with anti-alpha(2)beta(1) blocking antibody markedly diminished endothelial cell adhesion to immobilized TK1-2 compared with anti-alpha(v)beta(3) or anti-alpha(5)beta(1) antibody. Pretreatment of soluble TK1-2 also altered the binding level of anti-alpha(2)beta(1) antibody to endothelial cells in fluorescence-activated cell sorting analysis. Indeed, a blocking antibody against integrin alpha(2)beta(1) or knocking down of integrin alpha(2) expression prevented the inhibitory effect of TK1-2 in cell migration. Therefore, these results suggest that TK1-2 inhibits endothelial cell migration through inhibition of signaling and cytoskeleton rearrangement in part by interfering with integrin alpha(2)beta(1).
Collapse
Affiliation(s)
- Hyun-Kyung Kim
- Cancer Research Institute and Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Banpo-dong 505, Seocho-ku, Seoul 137-701, Korea
| | | | | | | | | |
Collapse
|
19
|
Prieto J, Fernandez-Ruiz V, Kawa MP, Sarobe P, Qian C. Cells as vehicles for therapeutic genes to treat liver diseases. Gene Ther 2008; 15:765-71. [DOI: 10.1038/gt.2008.44] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
20
|
|