1
|
Zarzycka M, Korzekwa AJ, Dulińska-Litewka J, Kaingu CK, Kotula-Balak M. Red deer (Cervus elaphus L.) antler stem cell culture medium inhibits prostate cancer cells. Histochem Cell Biol 2025; 163:41. [PMID: 40175729 DOI: 10.1007/s00418-025-02373-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2025] [Indexed: 04/04/2025]
Abstract
Prostate cancer is a frequently diagnosed cancer in men, but today the prognosis of local cancer and its metastasis has improved markedly owing to multiple treatment options. In therapy, various molecular entities that induce the reversion of malignancy with the restoration of mature cells of the same histological lineage are still under development. The antler of the red deer is a large and easily available source of stem cells, rich in products with promising applications in cancer treatments. Here, for the first time, antler stem cell culture medium was used for the treatment of prostate cancer cells in vitro to determine the cellular and molecular effects. Our data revealed that antler stem cell culture medium promotes pronounced changes in the expression of the vaspin receptor GRP78, intercellular adhesion molecule 1 (ICAM-1), androgen receptor (AR), leptin, vaspin, and SRC and AKT kinase signaling pathways (connected with autophagy and cell cycle inhibition and apoptosis induction). Moreover, the inhibition of cell migration was also revealed. These results indicate useful properties of red deer antler stem cell culture medium that can control molecular and cellular mechanisms that halt carcinogenesis. All these can be considered targets for further cancer stem cell treatment or cancer diagnostic improvements.
Collapse
Affiliation(s)
- Marta Zarzycka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kopernika 7, 31-034, Kraków, Poland.
| | - Anna Justyna Korzekwa
- Research Team of Biodiversity Protection, Institute of Animal, Reproduction and Food Research of Polish Academy of Sciences (IAR&FR PAS), Tuwima 10 Str., 10-748, Olsztyn, Poland.
| | - Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kopernika 7, 31-034, Kraków, Poland
| | - Catherine Kaluwa Kaingu
- Department of Veterinary Anatomy and Physiology, University of Nairobi, PR98+JC9, Nairobi, Kenya
| | - Małgorzata Kotula-Balak
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland
| |
Collapse
|
2
|
Luan Z, Zhang J, Wang Y. Identification of marker genes for spinal cord injury. Front Med (Lausanne) 2024; 11:1364380. [PMID: 38463490 PMCID: PMC10921937 DOI: 10.3389/fmed.2024.1364380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Spinal cord injury (SCI) is a profoundly disabling and devastating neurological condition, significantly impacting patients' quality of life. It imposes unbearable psychological and economic pressure on both patients and their families, as well as placing a heavy burden on society. Methods In this study, we integrated datasets GSE5296 and GSE47681 as training groups, analyzed gene variances between sham group and SCI group mice, and conducted Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis based on the differentially expressed genes. Subsequently, we performed Weighted Gene Correlation Network Analysis (WGCNA) and Lasso regression analyses. Results We identified four characteristic disease genes: Icam1, Ch25h, Plaur and Tm4sf1. We examined the relationship between SCI and immune cells, and validated the expression of the identified disease-related genes in SCI rats using PCR and immunohistochemistry experiments. Discussion In conclusion, we have identified and verified four genes related to SCI: Icam1, Ch25h, Plaur and Tm4sf1, which could offer insights for SCI treatment.
Collapse
Affiliation(s)
- Zhiwei Luan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Jiayu Zhang
- Department of Hygienic Toxicology, College of Public Health, Harbin Medical University, Harbin, China
| | - Yansong Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Xiao X, Han Y, Li Q, Zheng D, Cheng CY, Ni Y. Exploring the evolving function of soluble intercellular adhesion molecule-1 in junction dynamics during spermatogenesis. Front Endocrinol (Lausanne) 2024; 14:1281812. [PMID: 38260159 PMCID: PMC10801026 DOI: 10.3389/fendo.2023.1281812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is a transmembrane glycoprotein expressed on immune, endothelial, and epithelial cells. Its ectodomain can be proteolytically cleaved to release a circulating soluble form called sICAM-1. Clinical studies demonstrate sICAM-1 is upregulated in various diseases and associated with disease severity. Research has identified sICAM-1 as a regulator of the blood-testis barrier (BTB) and spermatogenesis. Overexpression of sICAM-1 weakened the BTB in vitro and in vivo, downregulated junction proteins including N-cadherin, γ-catenin, and connexin 43, and caused germ cell loss. This contrasts with barrier-strengthening effects of membrane-bound ICAM-1. sICAM-1 may act as a molecular switch enabling germ cells to open BTB and Sertoli-germ cell adhesion for transport across the seminiferous epithelium. While the mechanism remains unclear, reduced SRC family kinase (SFK) signaling was observed following sICAM-1 overexpression. SRC promotes BTB protein endocytosis and degradation, influences cytoskeletal dynamics, and affects cell polarity. As sICAM-1 overexpression phenocopies SRC inhibition, SRC may operate downstream of sICAM-1 in regulating BTB dynamics and spermatogenesis. Investigating sICAM-1's structure-function regions and downstream targets will elucidate the molecular mechanisms of junction disruption. This knowledge could enable strategies targeting sICAM-1/SRC to modulate BTB permeability and treat male infertility or diseases involving endothelial/epithelial barrier dysfunction.
Collapse
Affiliation(s)
- Xiang Xiao
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Yating Han
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Qin Li
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Dongwang Zheng
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - C. Yan Cheng
- Department of Urology and Andrology, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ya Ni
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| |
Collapse
|
4
|
Qiu Z, Wang Y, Zhang Z, Qin R, Peng Y, Tang W, Xi Y, Tian G, Zhang Y. Roles of intercellular cell adhesion molecule-1 (ICAM-1) in colorectal cancer: expression, functions, prognosis, tumorigenesis, polymorphisms and therapeutic implications. Front Oncol 2022; 12:1052672. [PMID: 36505809 PMCID: PMC9728583 DOI: 10.3389/fonc.2022.1052672] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Colorectal cancer (CRC) is a major global health problem and one of the major causes of cancer-related death worldwide. It is very important to understand the pathogenesis of CRC for early diagnosis, prevention strategies and identification of new therapeutic targets. Intercellular adhesion molecule-1 (ICAM-1, CD54) displays an important role in the the pathogenesis of CRC. It is a cell surface glycoprotein of the immunoglobulin (Ig) superfamily and plays an essential role in cell-cell, cell-extracellular matrix interaction, cell signaling and immune process. It is also expressed by tumor cells and modulates their functions, including apoptosis, cell motility, invasion and angiogenesis. The interaction between ICAM-1 and its ligand may facilitate adhesion of tumor cells to the vascular endothelium and subsequently in the promotion of metastasis. ICAM-1 expression determines malignant potential of cancer. In this review, we will discuss the expression, function, prognosis, tumorigenesis, polymorphisms and therapeutic implications of ICAM-1 in CRC.
Collapse
Affiliation(s)
- Zhiyuan Qiu
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yan Wang
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhao Zhang
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Rong Qin
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yong Peng
- Department of Oncology, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Weifeng Tang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Yan Xi
- Department of Geriatrics, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guangyu Tian
- Department of Oncology, Jiangdu People’s Hospital Affiliated to Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Yeqing Zhang
- Department of Vascular Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
5
|
Park KB, Kim EY, Chin H, Yoon DJ, Jun KH. Leptin stimulates migration and invasion and maintains cancer stem‑like properties in gastric cancer cells. Oncol Rep 2022; 48:162. [PMID: 35866593 PMCID: PMC9350977 DOI: 10.3892/or.2022.8377] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
Obesity is a risk factor for various types of cancer. Leptin, an adipocyte-derived hormone, may stimulate the proliferation of gastric cancer cells. However, the effect of leptin and underlying mechanism in gastric cancer remain unclear. In the present study, the role of leptin in gastric cancer was evaluated. The effect of leptin on the JAK-STAT and MEK signaling pathways was investigated in gastric cancer cells using wound-healing and cell invasion assays, immunoblotting and inhibition studies. Cancer-initiating cells derived from gastric cancer cells were used to investigate the effect of leptin on the maintenance of stemness and epithelial-mesenchymal transition (EMT) by immunoblotting. Clinicopathological characteristics including the serum leptin level and overall survival (OS) were analyzed in patients with (n=23) and without (n=23) obesity. Leptin induced the migration and invasion of gastric cancer cells by activating AKT and ERK and upregulating vascular endothelial growth factor (VEGF). Leptin increased the mRNA and protein levels of markers of stemness (CD44) and the EMT (Snail and N-cadherin). Pharmacological inhibitors of the JAK-STAT and MEK signaling pathways decreased leptin-induced migration and invasion, and the expression of VEGF. Obesity was associated with an elevated leptin level and body mass index was positively correlated with the leptin level (P=0.001 for both). The 5-year OS rate was not significantly different between the two groups (P=0.098). Leptin stimulates the migration and invasion of gastric cancer cells by activating the JAK-STAT and MEK pathways, and contributes to the maintenance of cancer stemness and metastatic potential. The present findings support an adverse effect of obesity in gastric cancer. Consequently, targeting of leptin-associated signaling pathways may have therapeutic potential for gastric cancer.
Collapse
Affiliation(s)
- Ki Bum Park
- Department of Surgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Paldal‑gu, Suwon‑si, Gyeonggi‑do 16247, Republic of Korea
| | - Eun Young Kim
- Department of Surgery, Uijeongbu St. Mary Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu‑si, Gyeonggi‑do 11765, Republic of Korea
| | - Hyungmin Chin
- Department of Surgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Paldal‑gu, Suwon‑si, Gyeonggi‑do 16247, Republic of Korea
| | - Dong Joon Yoon
- Clinical Medical Laboratory, St. Vincent's Hospital, Paldal‑gu, Suwon‑si, Gyeonggi‑do 16247, Republic of Korea
| | - Kyong-Hwa Jun
- Department of Surgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Paldal‑gu, Suwon‑si, Gyeonggi‑do 16247, Republic of Korea
| |
Collapse
|
6
|
Assumpção JAF, Pasquarelli-do-Nascimento G, Duarte MSV, Bonamino MH, Magalhães KG. The ambiguous role of obesity in oncology by promoting cancer but boosting antitumor immunotherapy. J Biomed Sci 2022; 29:12. [PMID: 35164764 PMCID: PMC8842976 DOI: 10.1186/s12929-022-00796-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
Abstract
Obesity is nowadays considered a pandemic which prevalence's has been steadily increasingly in western countries. It is a dynamic, complex, and multifactorial disease which propitiates the development of several metabolic and cardiovascular diseases, as well as cancer. Excessive adipose tissue has been causally related to cancer progression and is a preventable risk factor for overall and cancer-specific survival, associated with poor prognosis in cancer patients. The onset of obesity features a state of chronic low-grade inflammation and secretion of a diversity of adipocyte-derived molecules (adipokines, cytokines, hormones), responsible for altering the metabolic, inflammatory, and immune landscape. The crosstalk between adipocytes and tumor cells fuels the tumor microenvironment with pro-inflammatory factors, promoting tissue injury, mutagenesis, invasion, and metastasis. Although classically established as a risk factor for cancer and treatment toxicity, recent evidence suggests mild obesity is related to better outcomes, with obese cancer patients showing better responses to treatment when compared to lean cancer patients. This phenomenon is termed obesity paradox and has been reported in different types and stages of cancer. The mechanisms underlying this paradoxical relationship between obesity and cancer are still not fully described but point to systemic alterations in metabolic fitness and modulation of the tumor microenvironment by obesity-associated molecules. Obesity impacts the response to cancer treatments, such as chemotherapy and immunotherapy, and has been reported as having a positive association with immune checkpoint therapy. In this review, we discuss obesity's association to inflammation and cancer, also highlighting potential physiological and biological mechanisms underlying this association, hoping to clarify the existence and impact of obesity paradox in cancer development and treatment.
Collapse
Affiliation(s)
| | | | - Mariana Saldanha Viegas Duarte
- Immunology and Tumor Biology Program - Research Coordination, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Martín Hernan Bonamino
- Immunology and Tumor Biology Program - Research Coordination, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice - Presidency of Research and Biological Collections (VPPCB), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasília, DF, Brazil.
| |
Collapse
|
7
|
Zhang A, Wang S, Zhang F, Li W, Li Q, Liu X. The Prognosis of Leptin rs2167270 G > A (G19A) Polymorphism in the Risk of Cancer: A Meta-Analysis. Front Oncol 2021; 11:754162. [PMID: 34868961 PMCID: PMC8637904 DOI: 10.3389/fonc.2021.754162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/29/2021] [Indexed: 01/08/2023] Open
Abstract
Background Although the effect of the LEP G19A (rs2167270) polymorphism on cancers is assumed, the results of its influence have been contradictory. A meta-analysis was conducted to precisely verify the relationships between LEP G19A and the development of digestion-related cancers. Methods Investigators systematically searched the literature in PubMed, Embase, and Web of Science and used STATA software 14.0 for the meta-analysis. The odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to evaluate the associations. Subgroup analyses stratified by ethnicity, cancer type, and cancer system were further conducted to assess the relationship between the LEP G19A polymorphism and digestion-related cancers. Results In the overall population, we found a significant relationship with overall cancer (allele comparison: OR = 0.921, p = 0.000; dominant comparison: OR = 0.923, p = 0.004; recessive comparison: OR = 0.842, p = 0.000; homozygote model: OR = 0.0843, p = 0.001). In a subgroup analysis conducted by ethnicity, we obtained significant results in Asians (Asian allele comparison: OR = 0.885, p = 0.000; dominant comparison: OR = 0.862, p = 0.000; homozygote model: OR = 0.824, p = 0.039; and heterozygote comparison: OR = 0.868, p = 0.000) but not in Caucasians. In a subgroup analysis conducted by cancer type and cancer system, we obtained significant results that the LEP G19A polymorphism may decrease the risk of colorectal cancer, esophageal cancer, digestive system cancer, and urinary system cancer. Conclusions This meta-analysis revealed that the LEP G19A polymorphism may decrease the risk of cancer.
Collapse
Affiliation(s)
- Aiqiao Zhang
- Department of Neonatology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Department of Neonatology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shangren Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fujun Zhang
- Department of Neonatology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Department of Neonatology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wei Li
- Department of Neonatology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Department of Neonatology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qian Li
- Department of Neonatology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Department of Neonatology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
8
|
Umar MI, Hassan W, Murtaza G, Buabeid M, Arafa E, Irfan HM, Asmawi MZ, Huang X. The Adipokine Component in the Molecular Regulation of Cancer Cell Survival, Proliferation and Metastasis. Pathol Oncol Res 2021; 27:1609828. [PMID: 34588926 PMCID: PMC8473628 DOI: 10.3389/pore.2021.1609828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022]
Abstract
A hormonal imbalance may disrupt the rigorously monitored cellular microenvironment by hampering the natural homeostatic mechanisms. The most common example of such hormonal glitch could be seen in obesity where the uprise in adipokine levels is in virtue of the expanding bulk of adipose tissue. Such aberrant endocrine signaling disrupts the regulation of cellular fate, rendering the cells to live in a tumor supportive microenvironment. Previously, it was believed that the adipokines support cancer proliferation and metastasis with no direct involvement in neoplastic transformations and tumorigenesis. However, the recent studies have reported discrete mechanisms that establish the direct involvement of adipokine signaling in tumorigenesis. Moreover, the individual adipokine profile of the patients has never been considered in the prognosis and staging of the disease. Hence, the present manuscript has focused on the reported extensive mechanisms that culminate the basis of poor prognosis and diminished survival rate in obese cancer patients.
Collapse
Affiliation(s)
| | - Waseem Hassan
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Elshaimaa Arafa
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | | | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, University of Science Malaysia, Pulau Pinang, Malaysia
| | - Xianju Huang
- College of Pharmacy, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
9
|
Bacillus subtilis Spore-Trained Dendritic Cells Enhance the Generation of Memory T Cells via ICAM1. Cells 2021; 10:cells10092267. [PMID: 34571913 PMCID: PMC8469252 DOI: 10.3390/cells10092267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 11/22/2022] Open
Abstract
Immunological memory is a cardinal feature of the immune system. The intestinal mucosa is the primary exposure and entry site of infectious organisms. For an effective and long-lasting safeguard, a robust immune memory system is required, especially by the mucosal immunity. It is well known that tissue-resident memory T cells (Trms) provide a first response against infections reencountered at mucosal tissues surfaces, where they accelerate pathogen clearance. However, their function in intestinal immunization remains to be investigated. Here, we report enhanced local mucosal and systemic immune responses through oral administration of H9N2 influenza whole inactivated virus (H9N2 WIV) plus Bacillus subtilis spores. Subsequently, H9N2 WIV plus spores led to the generation of CD103+ CD69+ Trms, which were independent of circulating T cells during the immune period. Meanwhile, we also found that Bacillus subtilis spores could stimulate Acrp30 expression in 3T3-L1 adipocytes. Moreover, spore-stimulated adipocyte supernatant also upregulated the expression of intercellular adhesion molecule-1 (ICAM1) in dendritic cells (DCs). Furthermore, the proportion of HA-tetramer+ cells was severely curtailed upon suppressed ICAM1 expression, which also depended on HA-loaded DCs. Taken together, our data demonstrated that spore-promoted H9N2 WIV induced an immune response by enhancing Trms populations, which were associated with the activation of ICAM1 in DCs.
Collapse
|
10
|
Wang K, Jiang L, Hu A, Sun C, Zhou L, Huang Y, Chen Q, Dong J, Zhou X, Zhang F. Vertebral-specific activation of the CX3CL1/ICAM-1 signaling network mediates non-small-cell lung cancer spinal metastasis by engaging tumor cell-vertebral bone marrow endothelial cell interactions. Am J Cancer Res 2021; 11:4770-4789. [PMID: 33754027 PMCID: PMC7978319 DOI: 10.7150/thno.54235] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
Rationale: The spine is one of the most common metastatic sites of non-small cell lung cancer (NSCLC), and NSCLC spinal metastasis results in serious consequences. Metastatic extravasation of disseminated cancer cells including increased invasiveness, adhesion and transendothelial migration is crucial for tumor metastasis. This study aimed to investigate the mechanisms underlying NSCLC spinal metastasis based on the C-X3-C motif chemokine ligand 1- (CX3CL1) and intercellular adhesion molecule-1- (ICAM-1) mediated signaling network. Methods: Immunohistochemistry, western blotting, and reverse transcription-quantitative PCR were conducted to detect the distribution of CX3CL1/ICAM-1 in different organs. Transwell, adhesion, and transendothelial migration assays were performed to evaluate the regulatory effects of CX3CL1/ICAM-1 on NSCLC cell invasion, adhesion, and transendothelial migration in vitro. A spontaneous spinal metastasis mouse model was established via injection of NSCLC cells into the left cardiac ventricle of NOD/SCID mice. The effects of CX3CL1/ICAM-1 on NSCLC spinal metastasis in vivo were validated using bioluminescent, micro-computerized tomography, immunohistochemistry and histological analyses. Results: CX3CL1 expression was specifically higher in vertebral bone compared with limb bones and lung tissue, and was associated with NSCLC spinal metastasis. Mechanically, vertebral bone marrow endothelial cells (VBMECs) enhanced NSCLC cell invasion via CX3CL1 signaling-mediated activation of the PI3K/AKT pathway. Furthermore, we found that VBMECs effectively induced ICAM-1-dependent NSCLC cell adhesion in coordination with platelets through the CX3CL1/ICAM-1/LFA-1 pathway. Meanwhile, CX3CL1 enhanced NSCLC cell transendothelial migration by increasing permeability of VBMECs via ICAM-1-dependent activation of the Src/GEF-H1 pathway. Interestingly, NSCLC cells were indicated to promote CX3CL1 secretion of VBMECs through MAPK14/ADMA17-dependent CX3CL1 release and NF-κB-dependent CX3CL1 synthesis. Based on these findings, we revealed a novel feedback cycle between circulating NSCLC cells and VBMECs mediated by CX3CL1/ICAM-1 signaling. Further disengagement of the CX3CL1/ICAM-1-mediated feedback cycle in vivo significantly restricted metastasis and prolonged mouse survival. Conclusions: Our results indicated a unique feedback cycle between circulating NSCLC cells and VBMECs mediated by CX3CL1/ICAM-1 signaling, which is necessary for NSCLC spinal metastasis. This work provides a new perspective for underlying the mechanisms of NSCLC spinal metastasis and indicates potential novel targets for the prevention of NSCLC spinal metastasis.
Collapse
|
11
|
Perego S, Sansoni V, Ziemann E, Lombardi G. Another Weapon against Cancer and Metastasis: Physical-Activity-Dependent Effects on Adiposity and Adipokines. Int J Mol Sci 2021; 22:ijms22042005. [PMID: 33670492 PMCID: PMC7922129 DOI: 10.3390/ijms22042005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/15/2022] Open
Abstract
Physically active behavior has been associated with a reduced risk of developing certain types of cancer and improved psychological conditions for patients by reducing anxiety and depression, in turn improving the quality of life of cancer patients. On the other hand, the correlations between inactivity, sedentary behavior, and overweight and obesity with the risk of development and progression of various cancers are well studied, mainly in middle-aged and elderly subjects. In this article, we have revised the evidence on the effects of physical activity on the expression and release of the adipose-tissue-derived mediators of low-grade chronic inflammation, i.e., adipokines, as well as the adipokine-mediated impacts of physical activity on tumor development, growth, and metastasis. Importantly, exercise training may be effective in mitigating the side effects related to anti-cancer treatment, thereby underlining the importance of encouraging cancer patients to engage in moderate-intensity activities. However, the strong need to customize and adapt exercises to a patient’s abilities is apparent. Besides the preventive effects of physically active behavior against the adipokine-stimulated cancer risk, it remains poorly understood how physical activity, through its actions as an adipokine, can actually influence the onset and development of metastases.
Collapse
Affiliation(s)
- Silvia Perego
- Laboratory of Experimental Biochemistry and Molecular Biology, Milano, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); or
| | - Veronica Sansoni
- Laboratory of Experimental Biochemistry and Molecular Biology, Milano, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); or
- Correspondence: ; Tel.: +39-0266214068
| | - Ewa Ziemann
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, 61-871 Poznań, Poland; or
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, Milano, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); or
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, 61-871 Poznań, Poland; or
| |
Collapse
|
12
|
Ma R, He Q. A Variant of Leptin Gene Decreases the Risk of Gastric Cancer in Chinese Individuals: Evidence from a Case-Control Study. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:397-404. [PMID: 33061532 PMCID: PMC7519837 DOI: 10.2147/pgpm.s258672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022]
Abstract
Background A host of studies have explored the potential connection between leptin (LEP) G19A polymorphism and the risk of cancers, but the relationship between gastric cancer (GC) susceptibility and LEP G19A polymorphism was not revealed before. The aim of this study was to investigate this relationship in Chinese Han population. Methods Thus, this case–control study with 380 GC cases and 465 controls was designed to unearth the link between LEP G19A polymorphism and GC susceptibility. Genotyping was accomplished by a custom-made 48-Plex SNP scanTM kit. Relative LEP gene expression was detected by real-time reverse transcription-polymerase chain reaction. Results LEP G19A polymorphism was shown to relate with a decreased risk of GC. Subgroup analyses uncovered significant connections in the males, nondrinkers, and those at age <60 years. G19A polymorphism was also linked with tumor size and location and pathological type of GC. Last, LEP gene expression in gastric tissues was considerably less than in control tissues. Conclusion This study shows that G19A polymorphism of LEP gene is linked with a lower risk of GC in the tested Chinese Han individuals.
Collapse
Affiliation(s)
- Renjie Ma
- Department of Infectious Disease, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu Province, 212300, People's Republic of China
| | - Qi He
- Department of Infectious Disease, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu Province, 212300, People's Republic of China
| |
Collapse
|
13
|
Zhang Y, Ma S, Niu Q, Han Y, Liu X, Jiang J, Chen S, Lin H. Features of alternative splicing in stomach adenocarcinoma and their clinical implication: a research based on massive sequencing data. BMC Genomics 2020; 21:580. [PMID: 32831016 PMCID: PMC7443856 DOI: 10.1186/s12864-020-06997-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Alternative splicing (AS) offers a main mechanism to form protein polymorphism. A growing body of evidence indicates the correlation between splicing disorders and carcinoma. Nevertheless, an overall analysis of AS signatures in stomach adenocarcinoma (STAD) is absent and urgently needed. RESULTS 2042 splicing events were confirmed as prognostic molecular events. Furthermore, the final prognostic signature constructed by 10 AS events gave good result with an area under the curve (AUC) of receiver operating characteristic (ROC) curve up to 0.902 for 5 years, showing high potency in predicting patient outcome. We built the splicing regulatory network to show the internal regulation mechanism of splicing events in STAD. QKI may play a significant part in the prognosis induced by splicing events. CONCLUSIONS In our study, a high-efficiency prognostic prediction model was built for STAD patients, and the results showed that AS events could become potential prognostic biomarkers for STAD. Meanwhile, QKI may become an important target for drug design in the future.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Shengling Ma
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Niu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun Han
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xingyu Liu
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jie Jiang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Simiao Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haolong Lin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
14
|
Abbaszadegan MR, Mojarrad M, Moghbeli M. Role of extra cellular proteins in gastric cancer progression and metastasis: an update. Genes Environ 2020; 42:18. [PMID: 32467737 PMCID: PMC7227337 DOI: 10.1186/s41021-020-00157-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background Gastric cancer (GC) is one of the most common cancers in the world with a high ratio of mortality. Regarding the late diagnosis, there is a high ratio of distant metastasis among GC cases. Despite the recent progresses in therapeutic modalities, there is not still an efficient therapeutic method to increase survival rate of metastatic GC cases. Main body Apart from the various intracellular signaling pathways which are involved in tumor cell migration and metastasis, the local microenvironment is also a critical regulator of tumor cell migration. Indeed, the intracellular signaling pathways also exert their final metastatic roles through regulation of extra cellular matrix (ECM). Therefore, it is required to assess the role of extra cellular components in biology of GC. Conclusion In the present review, we summarize 48 of the significant ECM components including 17 ECM modifying enzymes, seven extracellular angiogenic factors, 13 cell adhesion and cytoskeletal organizers, seven matricellular proteins and growth factors, and four proteoglycans and extra cellular glycoproteins. This review paves the way of determination of a specific extra cellular diagnostic and prognostic panel marker for the GC patients.
Collapse
Affiliation(s)
| | - Majid Mojarrad
- 2Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- 2Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Idrizaj E, Garella R, Squecco R, Baccari MC. Adipocytes-released Peptides Involved in the Control of Gastrointestinal Motility. Curr Protein Pept Sci 2019; 20:614-629. [PMID: 30663565 DOI: 10.2174/1389203720666190121115356] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/18/2022]
Abstract
The present review focuses on adipocytes-released peptides known to be involved in the control of gastrointestinal motility, acting both centrally and peripherally. Thus, four peptides have been taken into account: leptin, adiponectin, nesfatin-1, and apelin. The discussion of the related physiological or pathophysiological roles, based on the most recent findings, is intended to underlie the close interactions among adipose tissue, central nervous system, and gastrointestinal tract. The better understanding of this complex network, as gastrointestinal motor responses represent peripheral signals involved in the regulation of food intake through the gut-brain axis, may also furnish a cue for the development of either novel therapeutic approaches in the treatment of obesity and eating disorders or potential diagnostic tools.
Collapse
Affiliation(s)
- Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - Maria Caterina Baccari
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| |
Collapse
|
16
|
Dong Z, Chen B, Pan H, Wang D, Liu M, Yang Y, Zou M, Yang J, Xiao K, Zhao R, Zheng X, Zhang L, Zhang Y. Detection of Microbial 16S rRNA Gene in the Serum of Patients With Gastric Cancer. Front Oncol 2019; 9:608. [PMID: 31338330 PMCID: PMC6629868 DOI: 10.3389/fonc.2019.00608] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/20/2019] [Indexed: 01/03/2023] Open
Abstract
Aberrance in the blood bacterial microbiome has been identified and validated in several non-infectious diseases, including cancer. The occurrence and progression of gastric cancer has been found to be associated with alterations in the microbiome composition. However, the composition of the blood microbiome in patients with gastric cancer is not well-characterized. To test this hypothesis, we conducted a case-control study to investigate the microbiota compositions in the serum of patients with gastric cancer. The serum microbiome was investigated in patients with gastric cancer, atypical hyperplasia, chronic gastritis, and in healthy controls using 16S rRNA gene sequencing targeting the V1-V2 region. Our results revealed that the structure of the serum microbiome in gastric cancer was significantly different from all other groups, and alpha diversity decreased from the healthy control to patients with gastric cancer. The serum microbiome correlated significantly with tumor-node-metastasis (TNM) stage, lymphatic metastasis, tumor diameter, and invasion depth in gastric cancer. Three genera or species, namely, Acinetobacter, Bacteroides, Haemophilus parainfluenzae, were enriched in patients with gastric cancer, whereas Sphingomonas, Comamonas, and Pseudomonas stutzeri were enriched in the healthy control. Furthermore, the structure of serum microbiota differed between gastric cancer lymphatic metastasis and non-lymphatic metastasis. As a pilot investigation to characterizing the serum microbiome in gastric cancer, our study provided a foundation for improving our understanding of the role of microbiota in the pathogenesis of gastric cancer.
Collapse
Affiliation(s)
- Zhaogang Dong
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Bin Chen
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
| | - Hongwei Pan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Ding Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Min Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Yongmei Yang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Mingjin Zou
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Junjie Yang
- College of Life Science, Qilu Normal University, Jinan, China
| | - Ke Xiao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Rui Zhao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Xin Zheng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Lei Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China.,Shandong Children's Microbiome Center, Qilu Children's Hospital of Shandong University, Jinan, China.,Qingdao Human Microbiome Center, The Affiliated Central Hospital of Qingdao University, Qingdao, China
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Overexpression of ICAM-1 Predicts Poor Survival in High-Grade Serous Ovarian Carcinoma: A Study Based on TCGA and GEO Databases and Tissue Microarray. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2867372. [PMID: 31312656 PMCID: PMC6595389 DOI: 10.1155/2019/2867372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/24/2019] [Accepted: 05/14/2019] [Indexed: 01/17/2023]
Abstract
Intercellular cell adhesion molecule-1 (ICAM-1), an important adhesion molecule in the immunoglobulin superfamily, is expressed on many cell types. Recent studies have identified ICAM-1 as a potential oncogene that promotes the development of epithelial ovarian cancer (EOC); it was also found to be associated with poor survival. However, the clinical significance of its expression in high-grade serous ovarian carcinoma (HGSOC) is unclear. Thus, this study aimed to investigate the significance of ICAM-1 expression in HGSOC. Data on ICAM1 expression and mutations in serous ovarian carcinoma (SOC) were obtained from the Cancer Genome Atlas (TCGA), and ICAM1 mRNA expression data in HGSOC were obtained from the Gene Expression Omnibus (GEO) database. ICAM-1 expression was evaluated by immunohistochemistry in HGSOC and normal fallopian tube tissues microarray. In TCGA data, amplification/mutation of ICAM1 was identified in 12% of serous ovarian carcinoma samples, and overexpression of ICAM1 mRNA predicted reduced overall survival in SOC. From TCGA and GEO data, SOC patients with ICAM1 mRNA overexpression treated with chemotherapeutic drugs that contained taxol or taxol and platin together had significantly reduced progression-free survival. According to GEO data, ICAM1 mRNA expression was found significantly higher in HGSOC than in control samples. In our study, ICAM-1 overexpression was observed in 63.1% (65/103) of HGSOCs. As a prognostic biomarker, overexpression of ICAM-1 predicted reduced recurrence-free and overall survival and is an independent risk factor for poor prognosis. These findings suggest that overexpression of ICAM-I is an independent indicator of poor prognosis for HGSOC and that it can serve as an effective clinical prognostic biomarker for this disease.
Collapse
|
18
|
Inagaki-Ohara K. Gastric Leptin and Tumorigenesis: Beyond Obesity. Int J Mol Sci 2019; 20:ijms20112622. [PMID: 31141984 PMCID: PMC6600422 DOI: 10.3390/ijms20112622] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022] Open
Abstract
Leptin, an adipocyte-derived hormone and its receptor (ObR) expressed in the hypothalamus are well known as an essential regulator of appetite and energy expenditure. Obesity induces abundant leptin production, however, reduced sensitivity to leptin leads to the development of metabolic disorders, so called leptin resistance. The stomach has been identified as an organ that simultaneously expresses leptin and ObR. Accumulating evidence has shown gastric leptin to perform diverse functions, such as those in nutrient absorption and carcinogenesis in the gastrointestinal system, independent of its well-known role in appetite regulation and obesity. Overexpression of leptin and phosphorylated ObR is implicated in gastric cancer in humans and in murine model, and diet-induced obesity causes precancerous lesions in the stomach in mice. While the underlying pathomechanisms remain unclear, leptin signaling can affect gastric mucosal milieu. In this review, we focus on the significant role of the gastric leptin signaling in neoplasia and tumorigenesis in stomach in the context of hereditary and diet-induced obesity.
Collapse
Affiliation(s)
- Kyoko Inagaki-Ohara
- Division of Host Defense, Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562 Nanatsuka, Shobara, Hiroshima 727-0023, Japan.
| |
Collapse
|
19
|
Tsai CF, Chen JH, Wu CT, Chang PC, Wang SL, Yeh WL. Induction of osteoclast-like cell formation by leptin-induced soluble intercellular adhesion molecule secreted from cancer cells. Ther Adv Med Oncol 2019; 11:1758835919846806. [PMID: 31205504 PMCID: PMC6535721 DOI: 10.1177/1758835919846806] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 03/13/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Leptin is considered a tumorigenic adipokine, suggested to promote tumorigenesis and progression in many cancers. On the other hand, intercellular adhesion molecule-1 (ICAM-1) shows altered expression in a variety of benign and malignant diseases. Histologically, ICAM-1 expression is reported as proportional to cancer stage and considered as a potential diagnosis biomarker. The altered expressions of ICAM-1 and its soluble form in malignant diseases have gained interests in recent years. Material and methods: The expression of ICAM-1 and its regulatory signaling were examined by Western blot or flow cytometry. The effect of soluble ICAM-1 on osteoclast formation was investigated by tartrate-resistance acid phosphatase staining of RAW cells and tumor-induced osteolysis in vivo. Results: In our study, we found that leptin enhanced soluble ICAM-1 production but not surface ICAM-1 expression in lung and breast cancer cells, and this effect was regulated through leptin receptor (ObR), while silencing ObR abrogated leptin-induced soluble ICAM-1 expression. In addition, we revealed that leptin administration provoked the JAK1/2, STAT3, FAK, ERK, and GSK3αβ signaling cascade, leading to the elevation of ICAM-1 expression. Moreover, soluble ICAM-1 secreted by leptin-stimulated cancer cells synergize with the receptor activator of nuclear factor kappa-B ligand (RANKL) in inducing osteoclast formation. Soluble ICAM also enhanced tumor-induced osteolysis in vivo. Conclusion: These findings suggest that soluble ICAM-1 produced under leptin treatment enhances osteoclast formation and is involved in tumor-induced osteolysis. Leptin plays an important role in physiology in health and diseases. Leptin affects immune responses that may induce inflammation and carcinogenesis. Leptin is also considered as a tumorigenic adipokine suggested to promote tumorigenesis and progression in many cancers. On the other hand, intercellular adhesion molecule-1 (ICAM-1) shows altered expression in a variety of benign and malignant diseases. Histologically, ICAM-1 expression is reported to be proportional to cancer stage and considered as a potential diagnosis biomarker. It has been reported that soluble ICAM-1 allows tumor cells to escape from immune recognition and stimulates angiogenesis and tumor growth. The altered expressions of ICAM-1 and its soluble form in malignant diseases have gained interests in recent years. In our study, we found that leptin enhanced soluble ICAM-1 production but not surface ICAM-1 expression in lung and breast cancer cells, and this effect was regulated through leptin receptor (ObR), while silencing ObR abrogated leptin-induced soluble ICAM-1 expression. In addition, we revealed that leptin administration provoked the JAK1/2, STAT3, FAK, ERK, and GSK3αβ signaling cascade, leading to the elevation of ICAM-1 expression. Moreover, soluble ICAM-1 secreted by leptin-stimulated cancer cells synergize with receptor activator of nuclear factor-kappa B ligand in inducing osteoclast formation. Soluble ICAM also enhanced tumor-induced osteolysis in vivo. These findings suggest that soluble ICAM-1 produced under leptin treatment is possibly involved in lung and breast cancer bone metastasis.
Collapse
Affiliation(s)
- Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, China
| | - Jia-Hong Chen
- Department of General Surgery, Buddhist Tzu Chi Medical Foundation, Taichung, China
| | - Chen-Teng Wu
- Department of Surgery, China Medical University Hospital, Taichung, China
| | - Pei-Chun Chang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, China
| | - Shu-Lin Wang
- Institute of New Drug Development, China Medical University, Taichung, China
| | - Wei-Lan Yeh
- Institute of New Drug Development, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402 China
| |
Collapse
|
20
|
Wang YX, Zhu N, Zhang CJ, Wang YK, Wu HT, Li Q, Du K, Liao DF, Qin L. Friend or foe: Multiple roles of adipose tissue in cancer formation and progression. J Cell Physiol 2019; 234:21436-21449. [PMID: 31054175 DOI: 10.1002/jcp.28776] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/19/2022]
Abstract
Obesity is well-known as the second factor for tumorigenesis after smoking and is bound up with the malignant progression of several kinds of cancers, including esophageal cancer, liver cancer, colorectal cancer, kidney cancer, and ovarian cancer. The increased morbidity and mortality of obesity-related cancer are mostly attributed to dysfunctional adipose tissue. The possible mechanisms connecting dysfunctional adipose tissue to high cancer risk mainly focus on chronic inflammation, obesity-related microenvironment, adipokine secretion disorder, and browning of adipose tissue, and so forth. The stromal vascular cells in adipose tissue trigger chronic inflammation through secreting inflammatory factors and promote cancer cell proliferation. Hypertrophic adipose tissues lead to metabolic disorders of adipocytes, such as abnormal levels of adipokines that mediate cancer progression and metastasis. Cancer patients often show adipose tissue browning and cancerous cachexia in an advanced stage, which lead to unsatisfied chemotherapy effect and poor prognosis. However, increasing evidence has shown that adipose tissue may display quite opposite effects in cancer development. Therefore, the interaction between cancers and adipose tissue exert a vital role in mediates adipose tissue dysfunction and further leads to cancer progression. In conclusion, targeting the dysfunction of adipose tissue provides a promising strategy for cancer prevention and therapy.
Collapse
Affiliation(s)
- Yu-Xiang Wang
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chan-Juan Zhang
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yi-Kai Wang
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia
| | - Hong-Tao Wu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qun Li
- Outpatient Department of Hanpu Campus, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ke Du
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, Key Lab for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Li Qin
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
21
|
Ghasemi A, Saeidi J, Azimi-Nejad M, Hashemy SI. Leptin-induced signaling pathways in cancer cell migration and invasion. Cell Oncol (Dordr) 2019; 42:243-260. [PMID: 30877623 DOI: 10.1007/s13402-019-00428-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Increasing evidence indicates that obesity is associated with tumor development and progression. Leptin is an adipocyte-related hormone with a key role in energy metabolism and whose circulating levels are elevated in obesity. The effect of leptin on cancer progression and metastasis and its underlying mechanisms are still unclear. Leptin can impact various steps in tumor metastasis, including epithelial-mesenchymal transition, cell adhesion to the extracellular matrix (ECM), and proteolysis of ECM components. To do so, leptin binds to its receptor (OB-Rb) to activate signaling pathways and downstream effectors that participate in tumor cell invasion as well as distant metastasis. CONCLUSIONS In this review, we describe metastasis steps in detail and characterize metastasis-related molecules activated by leptin, which may help to develop a roadmap that guides future work. In addition, we conclude that a profound understanding of the fundamental molecular processes that contribute to leptin-induced metastasis may pave the way for the development of new prognostic molecules and appropriate approaches to the treatment of obesity-related cancers.
Collapse
Affiliation(s)
- Ahmad Ghasemi
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jafar Saeidi
- Department of Physiology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Mohsen Azimi-Nejad
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Genetic, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
22
|
Hu Z, Yang D, Tang Y, Zhang X, Wei Z, Fu H, Xu J, Zhu Z, Cai Q. Five-long non-coding RNA risk score system for the effective prediction of gastric cancer patient survival. Oncol Lett 2019; 17:4474-4486. [PMID: 30988816 PMCID: PMC6447923 DOI: 10.3892/ol.2019.10124] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022] Open
Abstract
The prognosis for patients with gastric cancer (GC) is usually poor, as the majority of patients have reached the advanced stages of disease at the point of diagnosis. Therefore, revealing the mechanisms of GC is necessary for the identification of key biomarkers and the development of effective targeted therapies. The present study aimed to identify long non-coding RNAs (lncRNAs) prominently expressed in patients with GC. The GC dataset (including 384 GC samples) was downloaded from The Cancer Genome Atlas database as the training set. A number of other GC datasets were obtained from the Gene Expression Omnibus database as validation sets. Following data processing, lncRNAs were annotated, followed by co-expression module analysis to identify stable modules, using the weighted gene co-expression network analysis (WGCNA) package. Prognosis-associated lncRNAs were screened using the ‘survival’ package. Following the selection of the optimal lncRNA combinations using the ‘penalized’ package, risk score systems were constructed and assessed. Consensus differentially-expressed RNAs (DE-RNAs) were screened using the MetaDE package, and an lncRNA-mRNA network was constructed. Additionally, pathway enrichment analysis was conducted for the network nodes using gene set enrichment analysis (GSEA). A total of seven modules (blue, brown, green, grey, red, turquoise and yellow) were obtained following WGCNA analysis, among which the green and turquoise modules were stable and associated with the histological grade of GC. A total of 12 prognosis-associated lncRNAs were identified in the two modules. Combined with the optimal lncRNA combinations, risk score systems were constructed. The risk score system based on the green module [including ITPK1 antisense RNA 1 (ITPK1-AS1), KCNQ1 downstream neighbor (KCNQ1DN), long intergenic non-protein coding RNA 167 (LINC00167), LINC00173 and LINC00307] was the more efficient at predicting risk compared with those based on the turquoise, or the green + turquoise modules. A total of 1,105 consensus DE-RNAs were identified; GSEA revealed that LINC00167, LINC00173 and LINC00307 had the same association directions with 4 pathways and the 32 genes involved in those pathways. In conclusion, a risk score system based on the green module may be applied to predict the survival of patients with GC. Furthermore, ITPK1-AS1, KCNQ1DN, LINC00167, LINC00173 and LINC00307 may serve as biomarkers for GC pathogenesis.
Collapse
Affiliation(s)
- Zunqi Hu
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Dejun Yang
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yuan Tang
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Xin Zhang
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Ziran Wei
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Hongbing Fu
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Jiapeng Xu
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Zhenxin Zhu
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Qingping Cai
- Department of Gastrointestinal Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
23
|
Xu YXZ, Mishra S. Obesity-Linked Cancers: Current Knowledge, Challenges and Limitations in Mechanistic Studies and Rodent Models. Cancers (Basel) 2018; 10:E523. [PMID: 30567335 PMCID: PMC6316427 DOI: 10.3390/cancers10120523] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/09/2018] [Accepted: 12/15/2018] [Indexed: 02/07/2023] Open
Abstract
The worldwide prevalence of obesity has doubled during the last 50 years, and according to the World Obesity Federation, one third of the people on Earth will be obese by the year 2025. Obesity is described as a chronic, relapsing and multifactorial disease that causes metabolic, biomechanical, and psychosocial health consequences. Growing evidence suggests that obesity is a risk factor for multiple cancer types and rivals smoking as the leading preventable cause for cancer incidence and mortality. The epidemic of obesity will likely generate a new wave of obesity-related cancers with high aggressiveness and shortened latency. Observational studies have shown that from cancer risk to disease prognosis, an individual with obesity is consistently ranked worse compared to their lean counterpart. Mechanistic studies identified similar sets of abnormalities under obesity that may lead to cancer development, including ectopic fat storage, altered adipokine profiles, hormone fluctuations and meta-inflammation, but could not explain how these common mechanisms produce over 13 different cancer types. A major hurdle in the mechanistic underpinning of obesity-related cancer is the lack of suitable pre-clinical models that spontaneously develop obesity-linked cancers like humans. Current approaches and animal models fall short when discerning the confounders that often coexist in obesity. In this mini-review, we will briefly survey advances in the different obesity-linked cancers and discuss the challenges and limitations in the rodent models employed to study their relationship. We will also provide our perspectives on the future of obesity-linked cancer research.
Collapse
Affiliation(s)
- Yang Xin Zi Xu
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| | - Suresh Mishra
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
- Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| |
Collapse
|
24
|
Spyrou N, Avgerinos KI, Mantzoros CS, Dalamaga M. Classic and Novel Adipocytokines at the Intersection of Obesity and Cancer: Diagnostic and Therapeutic Strategies. Curr Obes Rep 2018; 7:260-275. [PMID: 30145771 DOI: 10.1007/s13679-018-0318-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW In this review, we investigate the role of classic and novel adipocytokines in cancer pathogenesis synopsizing the mechanisms underlying the association between adipocytokines and malignancy. Special emphasis is given on novel adipocytokines as new evidence is emerging regarding their entanglement in neoplastic development. RECENT FINDINGS Recent data have emphasized the role of the triad of overweight/obesity, insulin resistance and adipocytokines in cancer. In the setting of obesity, classic and novel adipocytokines present independent and joint effects on activation of major intracellular signaling pathways implicated in cell proliferation, expansion, survival, adhesion, invasion, and metastasis. Until now, more than 15 adipocytokines have been associated with cancer, and this list continues to expand. While the plethora of circulating pro-inflammatory adipocytokines, such as leptin, resistin, extracellular nicotinamide phosphoribosyl transferase, and chemerin are elevated in malignancies, some adipocytokines such as adiponectin and omentin-1 are generally decreased in cancers and are considered protective against carcinogenesis. Elucidating the intertwining of inflammation, cellular bioenergetics, and adiposopathy is significant for the development of preventive, diagnostic, and therapeutic strategies against cancer. Novel more effective and safe adipocytokine-centered therapeutic interventions may pave the way for targeted oncotherapy.
Collapse
Affiliation(s)
- Nikolaos Spyrou
- 251 Airforce General Hospital, Kanellopoulou 3, 11525, Athens, Greece
| | | | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
- Section of Endocrinology, VA Boston Healthcare System, Boston, MA, USA
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece.
| |
Collapse
|
25
|
Ray A, Cleary MP. The potential role of leptin in tumor invasion and metastasis. Cytokine Growth Factor Rev 2017; 38:80-97. [PMID: 29158066 DOI: 10.1016/j.cytogfr.2017.11.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
The adipocyte-released hormone-like cytokine/adipokine leptin behaves differently in obesity compared to its functions in the normal healthy state. In obese individuals, elevated leptin levels act as a pro-inflammatory adipokine and are associated with certain types of cancers. Further, a growing body of evidence suggests that higher circulating leptin concentrations and/or elevated expression of leptin receptors (Ob-R) in tumors may be poor prognostic factors. Although the underlying pathological mechanisms of leptin's association with poor prognosis are not clear, leptin can impact the tumor microenvironment in several ways. For example, leptin is associated with a number of biological components that could lead to tumor cell invasion and distant metastasis. This includes interactions with carcinoma-associated fibroblasts, tumor promoting effects of infiltrating macrophages, activation of matrix metalloproteinases, transforming growth factor-β signaling, etc. Recent studies also have shown that leptin plays a role in the epithelial-mesenchymal transition, an important phenomenon for cancer cell migration and/or metastasis. Furthermore, leptin's potentiating effects on insulin-like growth factor-I, epidermal growth factor receptor and HER2/neu have been reported. Regarding unfavorable prognosis, leptin has been shown to influence both adenocarcinomas and squamous cell carcinomas. Features of poor prognosis such as tumor invasion, lymph node involvement and distant metastasis have been recorded in several cancer types with higher levels of leptin and/or Ob-R. This review will describe the current scenario in a precise manner. In general, obesity indicates poor prognosis in cancer patients.
Collapse
Affiliation(s)
- Amitabha Ray
- Lake Erie College of Osteopathic Medicine, Seton Hill University, Greensburg, PA 15601, United States
| | - Margot P Cleary
- The Hormel Institute, University of Minnesota, Austin, MN 55912, United States.
| |
Collapse
|
26
|
Xiao S, Feng F, Sun L, Cai L, Liu Z, Liu S, Fan D, Zhang H. Blood type AB predicts promising prognosis in gastric cancer patients with positive preoperative serum CEA. Medicine (Baltimore) 2017; 96:e8496. [PMID: 29381925 PMCID: PMC5708924 DOI: 10.1097/md.0000000000008496] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Little is known about association between ABO blood groups and tumor markers in gastric cancer (GC) patients. The aim of the present study was to assess the prognostic value of ABO blood groups in GC patients with different levels of preoperative serum carcinoembryonic antigen (CEA).From September 2008 to April 2015, a total of 3234 GC patients who received radical gastrectomy were retrospectively analyzed. The clinicopathological characteristics including ABO blood groups and preoperative CEA were recorded. The prognostic value of ABO blood groups within different levels of serum CEA was analyzed.Overall, the ratio of male to female patients was 3.5:1; the median age was 57.4 years (range 20-87). The median overall survival (OS) for GC patients with blood type A, B, AB, and O were 52.6, 52.8, 53.8, and 53.6 months, respectively. There was no significant difference for the survival of patients among the 4 groups (P = .736). Also, no significant difference was found among the OS of the 4 blood types with negative (P = .875) and positive (P = .131) preoperative serum CEA. Further, we found that the OS of patients with positive preoperative serum CEA and blood type AB was significantly higher than that with blood type non-AB (P = .026). For patients with positive preoperative serum CEA, multivariate analysis showed that ABO blood groups were an independent prognostic factor.Blood type AB was a favorable prognostic factor for GC patients with positive preoperative serum CEA.
Collapse
|
27
|
Prospectively isolated mesenchymal stem/stromal cells are enriched in the CD73 + population and exhibit efficacy after transplantation. Sci Rep 2017; 7:4838. [PMID: 28684854 PMCID: PMC5500568 DOI: 10.1038/s41598-017-05099-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs), which reside in the bone marrow (BM) and various other tissues, can self-renew and differentiate into mesenchymal lineages. Many groups have harvested rat MSCs (rMSCs) from rat BM (rBM) by using a flush-out procedure and have evaluated surface marker expression after long-term culture. However, MSCs gradually differentiate during expansion and exhibit altered proliferation rates, morphological features and functions in vitro. Variations in MSC isolation methods may alter the effectiveness of therapeutic applications. Here, on the basis of CD29 (Itgb1) and CD54 (Icam1) expression, we prospectively isolated a population with a high colony-forming ability and multi-lineage potential from the rBM, and we demonstrated that most of these cells expressed CD73. Successful engraftment of rMSCs was achieved by using a fluorescence-conjugated anti-CD73 antibody. In humans and mice, MSCs were also purified by CD73, thus suggesting that CD73 may serve as a universal marker for prospective isolation of MSCs. Our results may facilitate investigations of MSC properties and function.
Collapse
|
28
|
miR 1296-5p Inhibits the Migration and Invasion of Gastric Cancer Cells by Repressing ERBB2 Expression. PLoS One 2017; 12:e0170298. [PMID: 28099468 PMCID: PMC5242522 DOI: 10.1371/journal.pone.0170298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/03/2017] [Indexed: 12/15/2022] Open
Abstract
The metastasis of gastric cancer, one of the most common tumors, has a molecular mechanism that is still largely unclear. Here we investigated the role of possible tumor-suppressor miR-1296-5p in the cell migration and invasion of ERBB2-positive gastric cancer. It found that miR-1296-5p was significantly down-regulated in gastric cancer tissues. Moreover, it was down-regulated in lymph node metastatic gastric cancer tissues compared with non-metastatic gastric cancer tissues. The luciferase activity of ERBB2 3'-untranslated region-based reporters constructed in SNU-216 and NUGC-4 gastric cancer cells suggested that ERBB2 was the target gene of miR-1296-5p. Overexpressed miR-1296-5p reduced its target protein level and Rac1 activation, and inhibited the migration and invasion of SNU-216 and NUGC-4 gastric cancer cells. MiR-1296-5p was down-regulated in ERBB2-positive gastric cancer tissues compared with ERBB2-negative gastric cancer tissues. In ERBB2-positive gastric cancers, the miR-1296-5p expression was suppressed in a majority of metastatic lymph node tissues compared to non-metastatic gastric cancer samples. The migration and invasion of gastric cancer cells was inhibited by miR-1296-5p overexpression or herceptin treatment, and rescued by the overexpression of constitutively active Rac1-Q61L or ERBB2. Taken together, our findings first suggest that miR-1296-5p might be involved in the regulation on the migration and invasion of human gastric cancer cells at least in part via targeting ERBB2/Rac1 signaling pathway.
Collapse
|
29
|
Ghasemi A, Hashemy SI, Aghaei M, Panjehpour M. RhoA/ROCK pathway mediates leptin-induced uPA expression to promote cell invasion in ovarian cancer cells. Cell Signal 2017; 32:104-114. [PMID: 28104444 DOI: 10.1016/j.cellsig.2017.01.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 01/09/2017] [Accepted: 01/14/2017] [Indexed: 12/11/2022]
Abstract
Previous studies have shown that leptin, an adipocyte-secreted hormone, stimulates ovarian cancer invasion. Here, we investigated the contribution of uPA in leptin-induced ovarian cancer cell invasion. The cell invasion and migration experiments were carried out using matrigel invasion and wound healing assays in ovarian cancer cell lines (OVCAR3, SKOV3and CaoV-3). The mechanism underlying the invasive effect of leptin was examined using cell transfection with Ob-Rb siRNA, pre-treatment with a specific inhibitor of RhoA and ROCK, RhoA activation assay, OB-Rb, Rock and upA protein expression. Our results show that leptin induced ovarian cancer cell invasion via up-regulating upA in a time and dose-dependent manner, which was attenuated using knockdown of OB-Rb by siRNA. Moreover, pre-incubation with C3 (inhibitor of RhoA) and Y-27632 (inhibitor of ROCK) effectively attenuated leptin-induced upA expression and inhibited invasive ability of ovarian cancer cells. We also found that pretreatment with inhibitors of PI3K/AKT (LY294002), JAK/STAT (AG490) and NF-kB (BAY 11-7082) significantly reduced leptin-induced upA expression. Collectively, our findings demonstrate that OB-Rb, RhoA/ROCK, PI3K/AKT, JAK/STAT pathways and NF-kB activation are involved in leptin-induced upA expression. These results may provide a new mechanism that facilitates leptin-induced ovarian cancer invasion.
Collapse
Affiliation(s)
- Ahmad Ghasemi
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Isaac Hashemy
- Surgical Oncology Research Center, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Aghaei
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mojtaba Panjehpour
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
30
|
|
31
|
Huang YK, Kang WM, Ma ZQ, Liu YQ, Zhou L, Yu JC. Body mass index, serum total cholesterol, and risk of gastric high-grade dysplasia: A case-control study among Chinese adults. Medicine (Baltimore) 2016; 95:e4730. [PMID: 27583914 PMCID: PMC5008598 DOI: 10.1097/md.0000000000004730] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Obesity is related to an increased risk of gastric cardia cancer. However, the influences of excess body weight and serum total cholesterol on the risk of gastric high-grade dysplasia have not been fully characterized.A case-control study was conducted to explore the relationships between body mass index (BMI), serum total cholesterol level, and the risk of gastric high-grade dysplasia in Chinese adults. A total of 893 consecutive patients with gastric high-grade dysplasia (537 men and 356 women) and 902 controls (543 men and 359 women) were enrolled from January 2000 to October 2015. Odds ratios (ORs) with 95% confidence intervals (CIs) were estimated, and a multivariate analysis was conducted.After adjusting for age, alcohol consumption, smoking status, family history of gastric cancer or esophageal cancer, and serum total cholesterol level, a BMI ranging from 27.5 to 29.9 was significantly related to an increased risk of gastric high-grade dysplasia in both men (adjusted OR = 1.87, 95% CI = 1.24-2.81) and women (adjusted OR = 2.72, 95% CI = 1.44-5.16). The 2 highest BMI categories (27.5-29.9 and ≥30.0) were identified as risk factors for gastric cardia high-grade dysplasia in both men (BMI = 27.5-29.9: adjusted OR = 1.78, 95% CI = 1.02-3.10; BMI ≥ 30.0: adjusted OR = 2.54, 95% CI = 1.27-5.08) and women (BMI = 27.5-29.9: adjusted OR = 2.88, 95% CI = 1.27-6.55; BMI ≥ 30.0: adjusted OR = 2.77, 95% CI = 1.36-5.64), whereas only a BMI ranging from 27.5 to 29.9 was a risk factor for gastric noncardia high-grade dysplasia in both men (adjusted OR = 1.98, 95% CI = 1.25-3.14) and women (adjusted OR = 2.88, 95% CI = 1.43-5.81). In addition, higher serum total cholesterol was associated with an increased risk of gastric noncardia high-grade dysplasia (adjusted OR = 1.83, 95% CI = 1.25-2.69) in women.Increased BMI was associated with an increased risk of gastric high-grade dysplasia in both men and women, and higher serum total cholesterol increased the risk of gastric noncardia high-grade dysplasia in women.
Collapse
Affiliation(s)
- Ya-Kai Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Wei-Ming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Zhi-Qiang Ma
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Yu-Qin Liu
- Cell Culture Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Jian-Chun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College
- Correspondence: Jian-Chun Yu, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing 100730, China (e-mail: )
| |
Collapse
|
32
|
Polvani S, Tarocchi M, Tempesti S, Bencini L, Galli A. Peroxisome proliferator activated receptors at the crossroad of obesity, diabetes, and pancreatic cancer. World J Gastroenterol 2016; 22:2441-2459. [PMID: 26937133 PMCID: PMC4768191 DOI: 10.3748/wjg.v22.i8.2441] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/17/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth cause of cancer death with an overall survival of 5% at five years. The development of PDAC is characteristically associated to the accumulation of distinctive genetic mutations and is preceded by the exposure to several risk factors. Epidemiology has demonstrated that PDAC risk factors may be non-modifiable risks (sex, age, presence of genetic mutations, ethnicity) and modifiable and co-morbidity factors related to the specific habits and lifestyle. Recently it has become evident that obesity and diabetes are two important modifiable risk factors for PDAC. Obesity and diabetes are complex systemic and intertwined diseases and, over the years, experimental evidence indicate that insulin-resistance, alteration of adipokines, especially leptin and adiponectin, oxidative stress and inflammation may play a role in PDAC. Peroxisome proliferator activated receptor-γ (PPARγ) is a nuclear receptor transcription factor that is implicated in the regulation of metabolism, differentiation and inflammation. PPARγ is a key regulator of adipocytes differentiation, regulates insulin and adipokines production and secretion, may modulate inflammation, and it is implicated in PDAC. PPARγ agonists are used in the treatment of diabetes and oxidative stress-associated diseases and have been evaluated for the treatment of PDAC. PPARγ is at the cross-road of diabetes, obesity, and PDAC and it is an interesting target to pharmacologically prevent PDAC in obese and diabetic patients.
Collapse
|
33
|
Downregulation of human intercellular adhesion molecule-1 attenuates the metastatic ability in human breast cancer cell lines. Oncol Rep 2016; 35:1541-8. [PMID: 26751847 DOI: 10.3892/or.2016.4543] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/09/2015] [Indexed: 11/05/2022] Open
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is a cell surface glycoprotein that belongs to immunoglobulin superfamily and plays an important role in tumor cell expansion or metastasis. However, the detailed mechanisms of ICAM-1 in breast cancer remain unclear. In this study, we evaluated the expression level of ICAM-1 in breast cancer using tissue microarray and clinical tissue specimens by immunohistochemical method, and the results revealed that ICAM-1 is highly expressed in the breast cancer tissues. To investigate whether ICAM-1 can affect the metastasis ability in breast cancer, we knocked down ICAM-1 expression in breast cancer cell line MCF-7 by using lentivirus-mediated RNA interference (RNAi). As a result, we stably silenced ICAM-1 expression in MCF-7 cells by infection with lentivirus expressing green fluorescent protein (GFP), the change of metastatic ability of MCF-7 cells was assessed by wound-healing assay, Transwell assay or clone formation assay. Our results showed that silencing of ICAM-1 can inhibit the metastatic ability of MCF-7 cell lines in vitro significantly, and the decreased migration and invasion was accompanied by a reduction of MMP-14. These results implying that ICAM-1 might be involved in the progression of breast cancer metastasis and lentivirus-mediated silencing of ICAM-1 might be a potential therapeutic approach for the treatment of breast cancer.
Collapse
|
34
|
Genetic variants within obesity-related genes are associated with tumor recurrence in patients with stages II/III colon cancer. Pharmacogenet Genomics 2015; 25:30-7. [PMID: 25379721 DOI: 10.1097/fpc.0000000000000101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Obesity is an established risk factor for colorectal cancer (CRC) incidence and it is also linked to CRC recurrence and survival. Polymorphisms located in obesity-related genes are associated with an increased risk of developing several cancer types including CRC. We evaluated whether single-nucleotide polymorphisms in obesity-related genes may predict tumor recurrence in colon cancer patients. MATERIALS AND METHODS Genotypes were obtained from germline DNA from 207 patients with stage II or III colon cancer at the Norris Comprehensive Cancer Center. Nine polymorphisms in eight obesity-related genes (PPAR, LEP, NFKB, CD36, DRG1, NGAL, REGIA, and DSCR1) were evaluated. The primary endpoint of the study was the 3-year recurrence rate. Positive associations were also tested in an independent Japanese cohort of 350 stage III CRC patients. RESULTS In univariate analysis, for PPARrs1801282, patients with a CC genotype had significantly lower recurrence probability (29 ± 4% SE) compared with patients with a CG genotype (48 ± 8% SE) [hazard ratio (HR): 1.77; 95% confidence interval (CI), 1.01-3.10; P = 0.040]. For DSCR1rs6517239, patients with an AA genotype had higher recurrence probability than patients carrying at least one allele G (37 ± 4% SE vs. 15 ± 6% SE) (HR: 0.51; 95% CI, 0.27-0.94; P = 0.027). This association was stronger in the patients bearing a left-sided tumor (HR: 0.34; 95% CI, 0.13-0.88; P = 0.018). In the Japanese cohort, no associations were found. CONCLUSION This hypothesis-generating study suggests a potential influence of polymorphisms within obesity-related genes in the recurrence probability of colon cancer. These interesting results should be evaluated further.
Collapse
|
35
|
Diedrich J, Gusky HC, Podgorski I. Adipose tissue dysfunction and its effects on tumor metabolism. Horm Mol Biol Clin Investig 2015; 21:17-41. [PMID: 25781550 DOI: 10.1515/hmbci-2014-0045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/14/2015] [Indexed: 12/12/2022]
Abstract
Growing by an alarming rate in the Western world, obesity has become a condition associated with a multitude of diseases such as diabetes, metabolic syndrome and various cancers. Generally viewed as an abnormal accumulation of hypertrophied adipocytes, obesity is also a poor prognostic factor for recurrence and chemoresistance in cancer patients. With more than two-thirds of the adult population in the United States considered clinically overweight or obese, it is critical that the relationship between obesity and cancer is further emphasized and elucidated. Adipocytes are highly metabolically active cells, which, through release of adipokines and cytokines and activation of endocrine and paracrine pathways, affect processes in neighboring and distant cells, altering their normal homeostasis. This work will examine specifically how adipocyte-derived factors regulate the cellular metabolism of malignant cells within the tumor niche. Briefly, tumor cells undergo metabolic pressure towards a more glycolytic and hypoxic state through a variety of metabolic regulators and signaling pathways, i.e., phosphoinositol-3 kinase (PI3K), hypoxia-inducible factor-1 alpha (HIF-1α), and c-MYC signaling. Enhanced glycolysis and high lactate production are hallmarks of tumor progression largely because of a process known as the Warburg effect. Herein, we review the latest literature pertaining to the body of work on the interactions between adipose and tumor cells, and underlining the changes in cancer cell metabolism that have been targeted by the currently available treatments.
Collapse
|
36
|
Suzukawa M, Koketsu R, Baba S, Igarashi S, Nagase H, Yamaguchi M, Matsutani N, Kawamura M, Shoji S, Hebisawa A, Ohta K. Leptin enhances ICAM-1 expression, induces migration and cytokine synthesis, and prolongs survival of human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2015; 309:L801-11. [PMID: 26276826 DOI: 10.1152/ajplung.00365.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 08/08/2015] [Indexed: 12/22/2022] Open
Abstract
There is rising interest in how obesity affects respiratory diseases, since epidemiological findings indicate a strong relationship between the two conditions. Leptin is a potent adipokine produced mainly by adipocytes. It regulates energy storage and expenditure and also induces inflammation. Previous studies have shown that leptin is able to activate inflammatory cells such as lymphocytes and granulocytes, but little is known about its effect on lung structural cells. The present study investigated the effects of leptin on human airway epithelial cells by using human primary airway epithelial cells and a human airway epithelial cell line, BEAS-2B. Flow cytometry showed enhanced ICAM-1 expression by both of those cells in response to leptin, and that effect was abrogated by dexamethasone or NF-κB inhibitor. Flow cytometry and quantitative PCR showed that airway epithelial cells expressed leptin receptor (Ob-R), whose expression level was downregulated by leptin itself. Multiplex cytokine analysis demonstrated enhanced production of CCL11, G-CSF, VEGF, and IL-6 by BEAS-2B cells stimulated with leptin. Furthermore, transfection of Ob-R small interference RNA decreased the effect of leptin on CCL11 production as assessed by quantitative PCR. Finally, leptin induced migration of primary airway epithelial cells toward leptin, suppressed BEAS-2B apoptosis induced with TNF-α and IFN-γ, and enhanced proliferation of primary airway epithelial cells. In summary, leptin was able to directly activate human airway epithelial cells by binding to Ob-R and by NF-κB activation, resulting in upregulation of ICAM-1 expression, induction of CCL11, VEGF, G-CSF, and IL-6 synthesis, induction of migration, inhibition of apoptosis, and enhancement of proliferation.
Collapse
Affiliation(s)
- Maho Suzukawa
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan;
| | | | - Shintaro Baba
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sayaka Igarashi
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Hiroyuki Nagase
- Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Masao Yamaguchi
- Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Noriyuki Matsutani
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Masafumi Kawamura
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Shunsuke Shoji
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Akira Hebisawa
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Ken Ohta
- National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| |
Collapse
|
37
|
Tian M, Li Y, Liu W, Jin L, Jiang X, Wang X, Ding Z, Peng Y, Zhou J, Fan J, Cao Y, Wang W, Shi Y. The nanomechanical signature of liver cancer tissues and its molecular origin. NANOSCALE 2015; 7:12998-3010. [PMID: 26168746 DOI: 10.1039/c5nr02192h] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Patients with cirrhosis are at higher risk of developing hepatocellular carcinoma (HCC), the second most frequent cause of cancer-related deaths. Although HCC diagnosis based on conventional morphological characteristics serves as the "gold standard" in the clinic, there is a high demand for more convenient and effective diagnostic methods that employ new biophysical perspectives. Here, we show that the nanomechanical signature of liver tissue is directly correlated with the development of HCC. Using indentation-type atomic force microscopy (IT-AFM), we demonstrate that the lowest elasticity peak (LEP) in the Young's modulus distribution of surgically removed liver cancer tissues can serve as a mechanical fingerprint to evaluate the malignancy of liver cancer. Cirrhotic tissues shared the same LEP as normal tissues. However, a noticeable downward shift in the LEP was detected when the cirrhotic tissues progressed to a malignant state, making the tumor tissues more prone to microvascular invasion. Cell-level mechanistic studies revealed that the expression level of a Rho-family effector (mDia1) was consistent with the mechanical trend exhibited by the tissue. Our findings indicate that the mechanical profiles of liver cancer tissues directly varied with tumor progression, providing an additional platform for the future diagnosis of HCC.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Elastic Modulus
- Formins
- Humans
- Liver/pathology
- Liver Cirrhosis/metabolism
- Liver Cirrhosis/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Microscopy, Atomic Force
- Microscopy, Electron, Scanning
- Nanotechnology
- RNA Interference
- RNA, Small Interfering/metabolism
- rho GTP-Binding Proteins/metabolism
- rhoC GTP-Binding Protein
Collapse
Affiliation(s)
- Mengxin Tian
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chang W, Kim R, Park SI, Jung YJ, Ham O, Lee J, Kim JH, Oh S, Lee MY, Kim J, Park MS, Chung YA, Hwang KC, Maeng LS. Enhanced Healing of Rat Calvarial Bone Defects with Hypoxic Conditioned Medium from Mesenchymal Stem Cells through Increased Endogenous Stem Cell Migration via Regulation of ICAM-1 Targeted-microRNA-221. Mol Cells 2015; 38:643-50. [PMID: 26062554 PMCID: PMC4507031 DOI: 10.14348/molcells.2015.0050] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/22/2015] [Accepted: 04/24/2015] [Indexed: 01/08/2023] Open
Abstract
The use of conditioned medium from mesenchymal stem cells may be a feasible approach for regeneration of bone defects through secretion of various components of mesenchymal stem cells such as cytokines, chemokines, and growth factors. Mesenchymal stem cells secrete and accumulate multiple factors in conditioned medium under specific physiological conditions. In this study, we investigated whether the conditioned medium collected under hypoxic condition could effectively influence bone regeneration through enhanced migration and adhesion of endogenous mesenchymal stem cells. Cell migration and adhesion abilities were increased through overexpression of intercellular adhesion molecule-1 in hypoxic conditioned medium treated group. Intercellular adhesion molecule-1 was upregulated by microRNA-221 in mesenchymal stem cells because microRNAs are key regulators of various biological functions via gene expression. To investigate the effects in vivo, evaluation of bone regeneration by computed tomography and histological assays revealed that osteogenesis was enhanced in the hypoxic conditioned medium group relative to the other groups. These results suggest that behavioral changes of endogenous mesenchymal stem cells through microRNA-221 targeted-intercellular adhesion molecule-1 expression under hypoxic conditions may be a potential treatment for patients with bone defects.
Collapse
Affiliation(s)
- Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Ran Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Sang In Park
- Institute of Catholic Integrative Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, College of Medicine, Incheon 403-720,
Korea
| | - Yu Jin Jung
- EIT/LOFUS Research Center, International St. Mary’s Hospital, Catholic Kwandong University, Incheon 404-834,
Korea
| | - Onju Ham
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752,
Korea
| | - Jihyun Lee
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Ji Hyeong Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Sekyung Oh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305,
USA
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu 702-701,
Korea
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women’s University, Seoul 140-742,
Korea
| | - Moon-Seo Park
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735,
Korea
| | - Yong-An Chung
- Institute of Catholic Integrative Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, College of Medicine, Incheon 403-720,
Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangwon 210-701,
Korea
- Catholic Kwandong University International, St. Mary’s Hospital, Incheon 404-834,
Korea
| | - Lee-So Maeng
- Institute of Catholic Integrative Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, College of Medicine, Incheon 403-720,
Korea
| |
Collapse
|