1
|
Andreadi A, Lodeserto P, Todaro F, Meloni M, Romano M, Minasi A, Bellia A, Lauro D. Nanomedicine in the Treatment of Diabetes. Int J Mol Sci 2024; 25:7028. [PMID: 39000136 PMCID: PMC11241380 DOI: 10.3390/ijms25137028] [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] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Nanomedicine could improve the treatment of diabetes by exploiting various therapeutic mechanisms through the use of suitable nanoformulations. For example, glucose-sensitive nanoparticles can release insulin in response to high glucose levels, mimicking the physiological release of insulin. Oral nanoformulations for insulin uptake via the gut represent a long-sought alternative to subcutaneous injections, which cause pain, discomfort, and possible local infection. Nanoparticles containing oligonucleotides can be used in gene therapy and cell therapy to stimulate insulin production in β-cells or β-like cells and modulate the responses of T1DM-associated immune cells. In contrast, viral vectors do not induce immunogenicity. Finally, in diabetic wound healing, local delivery of nanoformulations containing regenerative molecules can stimulate tissue repair and thus provide a valuable tool to treat this diabetic complication. Here, we describe these different approaches to diabetes treatment with nanoformulations and their potential for clinical application.
Collapse
Affiliation(s)
- Aikaterini Andreadi
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Pietro Lodeserto
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Federica Todaro
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
| | - Marco Meloni
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Maria Romano
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Alessandro Minasi
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Alfonso Bellia
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Davide Lauro
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| |
Collapse
|
2
|
Duan K, Liu J, Zhang J, Chu T, Liu H, Lou F, Liu Z, Gao B, Wei S, Wei F. Advancements in innate immune regulation strategies in islet transplantation. Front Immunol 2024; 14:1341314. [PMID: 38288129 PMCID: PMC10823010 DOI: 10.3389/fimmu.2023.1341314] [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] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
As a newly emerging organ transplantation technique, islet transplantation has shown the advantages of minimal trauma and high safety since it was first carried out. The proposal of the Edmonton protocol, which has been widely applied, was a breakthrough in this method. However, direct contact between islets and portal vein blood will cause a robust innate immune response leading to massive apoptosis of the graft, and macrophages play an essential role in the innate immune response. Therefore, therapeutic strategies targeting macrophages in the innate immune response have become a popular research topic in recent years. This paper will summarize and analyze recent research on strategies for regulating innate immunity, primarily focusing on macrophages, in the field of islet transplantation, including drug therapy, optimization of islet preparation process, islet engineering and Mesenchymal stem cells cotransplantation. We also expounded the heterogeneity, plasticity and activation mechanism of macrophages in islet transplantation, providing a theoretical basis for further research.
Collapse
Affiliation(s)
- Kehang Duan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiao Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tongjia Chu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huan Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Fengxiang Lou
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ziyu Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Bing Gao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shixiong Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
Yan LL, Ye LP, Chen YH, He SQ, Zhang CY, Mao XL, Li SW. The Influence of Microenvironment on Survival of Intraportal Transplanted Islets. Front Immunol 2022; 13:849580. [PMID: 35418988 PMCID: PMC8995531 DOI: 10.3389/fimmu.2022.849580] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/03/2022] [Indexed: 12/21/2022] Open
Abstract
Clinical islet transplantation has the potential to cure type 1 diabetes. Despite recent therapeutic success, it is still uncommon because transplanted islets are damaged by multiple challenges, including instant blood mediated inflammatory reaction (IBMIR), inflammatory cytokines, hypoxia/reperfusion injury, and immune rejection. The transplantation microenvironment plays a vital role especially in intraportal islet transplantation. The identification and targeting of pathways that function as "master regulators" during deleterious inflammatory events after transplantation, and the induction of immune tolerance, are necessary to improve the survival of transplanted islets. In this article, we attempt to provide an overview of the influence of microenvironment on the survival of transplanted islets, as well as possible therapeutic targets.
Collapse
Affiliation(s)
- Ling-ling Yan
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Li-ping Ye
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Ya-hong Chen
- Health Management Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Sai-qin He
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Chen-yang Zhang
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Xin-li Mao
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Shao-wei Li
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
4
|
Yuan L, Wu XJ, Li WC, Zhuo C, Xu Z, Tan C, Ma R, Wang J, Pu J. SLC6A8 Knockdown Suppresses the Invasion and Migration of Human Hepatocellular Carcinoma Huh-7 and Hep3B Cells. Technol Cancer Res Treat 2020; 19:1533033820983029. [PMID: 33356959 PMCID: PMC7780307 DOI: 10.1177/1533033820983029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Liver cancer is considered the sixth most commonly diagnosed cancer and the fourth leading cause of cancer-related deaths worldwide. Currently, there is no specific and effective therapy for hepatocellular carcinoma. Therefore, developing novel diagnostic and therapeutic strategies against hepatocellular carcinoma is of paramount importance. Solute carrier family 6 member 8 (SLC6A8) encodes the solute carrier family 6-8 to transport creatine into cells in a Na+ and Cl-- dependent manner. SLC6A8 deficiency is characterized by intellectual disabilities, loss of speech, and behavioral abnormalities. Of concern, the association of SLC6A8 with hepatocellular carcinoma remains elusive. In this study, we revealed that SLC6A8 knockdown significantly induced apoptosis and suppressed the migration and invasion of Hep3B and Huh-7 cells. These findings depicted the vital role of SLC6A8 in the initiation and progression of human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Lu Yuan
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xian Jian Wu
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Wen Chuan Li
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chenyi Zhuo
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - ZuoMing Xu
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chuan Tan
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - RiHai Ma
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - JianChu Wang
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Jian Pu
- Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| |
Collapse
|
5
|
Figueiredo H, Figueroa ALC, Garcia A, Fernandez-Ruiz R, Broca C, Wojtusciszyn A, Malpique R, Gasa R, Gomis R. Targeting pancreatic islet PTP1B improves islet graft revascularization and transplant outcomes. Sci Transl Med 2020; 11:11/497/eaar6294. [PMID: 31217339 DOI: 10.1126/scitranslmed.aar6294] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/16/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Deficient vascularization is a major driver of early islet graft loss and one of the primary reasons for the failure of islet transplantation as a viable treatment for type 1 diabetes. This study identifies the protein tyrosine phosphatase 1B (PTP1B) as a potential modulator of islet graft revascularization. We demonstrate that grafts of pancreatic islets lacking PTP1B exhibit increased revascularization, which is accompanied by improved graft survival and function, and recovery of normoglycemia and glucose tolerance in diabetic mice transplanted with PTP1B-deficient islets. Mechanistically, we show that the absence of PTP1B leads to activation of hypoxia-inducible factor 1α-independent peroxisome proliferator-activated receptor γ coactivator 1α/estrogen-related receptor α signaling and enhanced expression and production of vascular endothelial growth factor A (VEGF-A) by β cells. These observations were reproduced in human islets. Together, these findings reveal that PTP1B regulates islet VEGF-A production and suggest that this phosphatase could be targeted to improve islet transplantation outcomes.
Collapse
Affiliation(s)
- Hugo Figueiredo
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,University of Barcelona, 08036 Barcelona, Spain.,Escuela de Medicina y Ciencias de la Salud, Dept. Medicina Cardiovascular y Metabolómica, Tecnológico de Monterrey, 66278 San Pedro Garza García, Nuevo León, Mexico
| | - Ana Lucia C Figueroa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,University of Barcelona, 08036 Barcelona, Spain
| | - Ainhoa Garcia
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Rebeca Fernandez-Ruiz
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Christophe Broca
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, 34295 Montpellier, France
| | - Anne Wojtusciszyn
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, 34295 Montpellier, France.,Department of Endocrinology, Diabetes and Nutrition, University Hospital of Montpellier, Lapeyronie Hospital, 34295 Montpellier, France.,Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Rita Malpique
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Rosa Gasa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Ramon Gomis
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain. .,University of Barcelona, 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.,Universitat Oberta de Catalunya (UOC), 08018 Barcelona, Spain.,Department of Endocrinology and Nutrition, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
6
|
Singh AP, Biswas A, Shukla A, Maiti P. Targeted therapy in chronic diseases using nanomaterial-based drug delivery vehicles. Signal Transduct Target Ther 2019; 4:33. [PMID: 31637012 PMCID: PMC6799838 DOI: 10.1038/s41392-019-0068-3] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/01/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
The application of nanomedicines is increasing rapidly with the promise of targeted and efficient drug delivery. Nanomedicines address the shortcomings of conventional therapy, as evidenced by several preclinical and clinical investigations indicating site-specific drug delivery, reduced side effects, and better treatment outcome. The development of suitable and biocompatible drug delivery vehicles is a prerequisite that has been successfully achieved by using simple and functionalized liposomes, nanoparticles, hydrogels, micelles, dendrimers, and mesoporous particles. A variety of drug delivery vehicles have been established for the targeted and controlled delivery of therapeutic agents in a wide range of chronic diseases, such as diabetes, cancer, atherosclerosis, myocardial ischemia, asthma, pulmonary tuberculosis, Parkinson's disease, and Alzheimer's disease. After successful outcomes in preclinical and clinical trials, many of these drugs have been marketed for human use, such as Abraxane®, Caelyx®, Mepact®, Myocet®, Emend®, and Rapamune®. Apart from drugs/compounds, novel therapeutic agents, such as peptides, nucleic acids (DNA and RNA), and genes have also shown potential to be used as nanomedicines for the treatment of several chronic ailments. However, a large number of extensive clinical trials are still needed to ensure the short-term and long-term effects of nanomedicines in humans. This review discusses the advantages of various drug delivery vehicles for better understanding of their utility in terms of current medical needs. Furthermore, the application of a wide range of nanomedicines is also described in the context of major chronic diseases.
Collapse
Affiliation(s)
- Akhand Pratap Singh
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi, 221005 India
| | - Arpan Biswas
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi, 221005 India
| | - Aparna Shukla
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi, 221005 India
| | - Pralay Maiti
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi, 221005 India
| |
Collapse
|
7
|
Fang Z, Liang W, Luo L. HSP27 promotes epithelial-mesenchymal transition through activation of the β-catenin/MMP3 pathway in pancreatic ductal adenocarcinoma cells. Transl Cancer Res 2019; 8:1268-1278. [PMID: 35116869 PMCID: PMC8799219 DOI: 10.21037/tcr.2019.07.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/24/2019] [Indexed: 11/06/2022]
Abstract
Background The precise role of heat shock protein 27 (HSP27), as a type of small molecular protein in HSPs, in pancreatic ductal adenocarcinoma (PDAC) remains to be elucidated. The aim of the present study was to investigate the expression and function of HSP27 in PDAC cells. Methods We first detected the expression of HSP27 in PDAC tissues. Combining with the clinical pathology characteristics of PDAC patients, the relationship between them was analyzed. Then, we knocked down HSP27 using short interfering RNA (siRNA) and observed its biological functions using scratch assay and matrigel invasion and migration assays in ASPC-1 and PANC-1 cells. In mechanism, we verified the β-catenin/MMP-3 pathway associated proteins in ASPC-1 and PANC-1 cells. Results We found that HSP27 was highly expression in PDAC tissues, and was positively correlated with tumor differentiation, TNM staging and poor prognosis of PDAC patients. In vitro, we down-regulated the expression of HSP27 in ASPC-1 and PANC-1 cells and found that the invasion and migration ability of PDAC cells were significantly depressed, meanwhile, the activation of the β-catenin/MMP-3 pathway was inhibited. Conclusions HSP27 may be used as a tumor biomarker for diagnosis of PDAC, and HSP27 can promote the invasion and migration of PDAC by activating the β-catenin/MMP3 Pathway. Therefore, inhibition of HSP27 has therapeutic potential for the treatment of PDAC.
Collapse
Affiliation(s)
- Zehong Fang
- Third Department of General Surgery, Department of Organ Transplant, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, China
| | - Wenjin Liang
- Hepatobiliary Disease Institution, Zhongnan Hospital of Wuhan University, Transplant Center of Wuhan University & Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan 430071, China
| | - Laibang Luo
- Third Department of General Surgery, Department of Organ Transplant, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, China
| |
Collapse
|
8
|
Tang B, Liang W, Liao Y, Li Z, Wang Y, Yan C. PEA15 promotes liver metastasis of colorectal cancer by upregulating the ERK/MAPK signaling pathway. Oncol Rep 2018; 41:43-56. [PMID: 30365128 PMCID: PMC6278416 DOI: 10.3892/or.2018.6825] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Liver metastasis is one of the major causes of death in patients with colorectal cancer, and although treatment has improved recently, the long‑term survival rate of patients has not improved significantly. In the present study, we used immunohistochemistry to determine that phosphoprotein enriched in astrocytes‑15 kDa (PEA15) was highly expressed in colorectal cancer tissues and liver metastatic cancer tissues. It was also highly expressed in metastatic colorectal cancer patients compared to non‑metastatic patients. Through clinicopathological data of patients with liver metastasis of colorectal cancer, we found that high expression of PEA15 was positively correlated with TNM staging, liver metastasis and poor prognosis of colorectal cancer patients. Using confocal immunofluorescence microscopy, western blotting and cell proliferation, migration and invasion assays, we also determined that PEA15 could promote cancer cell proliferation in vitro and in vivo, epithelial mesenchymal transition (EMT) and the characteristics of cancer stem cells in vitro, thus promoting the abilities of invasion and migration. In addition, we revealed that PEA15 promoted the liver metastasis of colorectal cancer cells in a xenograft tumor metastasis model. In addition, concerning the mechanism, we used gene chip analysis to determine that PEA15 upregulated the ERK/MAPK signaling pathway in colorectal cancer cells. Therefore, we concluded that PEA15 may be a potential biomarker for liver metastasis of colorectal cancer therapy. Collectively, PEA15 promoted the development of liver metastasis of colorectal cancer through the ERK/MAPK signaling pathway.
Collapse
Affiliation(s)
- Bo Tang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Wenjin Liang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi 541004, P.R. China
| | - Yong Liao
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi 541004, P.R. China
| | - Zeming Li
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi 541004, P.R. China
| | - Yan Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Chao Yan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| |
Collapse
|
9
|
Liang W, Liao Y, Li Z, Wang Y, Zheng S, Xu X, Ran F, Tang B, Wang Z. MicroRNA-644a promotes apoptosis of hepatocellular carcinoma cells by downregulating the expression of heat shock factor 1. Cell Commun Signal 2018; 16:30. [PMID: 29898735 PMCID: PMC6001142 DOI: 10.1186/s12964-018-0244-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/01/2018] [Indexed: 02/07/2023] Open
Abstract
In this study, we investigated the role of microRNA-644a (miR-644a) in the growth and survival of hepatocellular carcinoma (HCC) cells. MiR-644a levels were lower in HCC tissues than in adjacent peri-cancerous tissues (n = 135). MiR-644a expression was inversely correlated with heat shock factor 1 (HSF1) expression, tumour diameter and TNM stage. Moreover, HepG2 and SMMC-7721 cell lines showed lower miR-644a expression than normal L-O2 hepatocytes. MiR-644a overexpression in HepG2 and SMMC-7721 cells increased apoptosis by downregulating HSF1. Dual luciferase reporter assays confirmed the presence of a miR-644a binding site in the 3'-untranslated region (3'-UTR) of HSF1. Xenograft tumours derived from SMMC-7721 cells transfected with a miR-664a mimic showed less growth than tumours derived from untransfected controls. Protein chip analysis revealed that miR-644a-overexpressing SMMC-7721 and HepG2 cells strongly expressed pro-apoptotic BH3-only proteins, such as BID, BAD, BIM, SMAC, Apaf-1 and cleaved caspases-3 and -9. These findings suggest miR-644a promotes apoptosis in HCC cells by inhibiting HSF1.
Collapse
Affiliation(s)
- Wenjin Liang
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China
| | - Yong Liao
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China
| | - Zeming Li
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China
| | - Yan Wang
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China
| | - Siqi Zheng
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China
| | - Xiaochen Xu
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China
| | - Fulin Ran
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China
| | - Bo Tang
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China.
| | - Zhenran Wang
- Department of Gastrointestinal Surgery and Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi, 541001, People's Republic of China.
| |
Collapse
|
10
|
Liang W, Liao Y, Zhang J, Huang Q, Luo W, Yu J, Gong J, Zhou Y, Li X, Tang B, He S, Yang J. Heat shock factor 1 inhibits the mitochondrial apoptosis pathway by regulating second mitochondria-derived activator of caspase to promote pancreatic tumorigenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:64. [PMID: 28482903 PMCID: PMC5422968 DOI: 10.1186/s13046-017-0537-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/04/2017] [Indexed: 12/20/2022]
Abstract
Background As a relatively conservative transcriptional regulator in biological evolution, heat shock factor 1 (HSF1) is activated by, and regulates the expression of heat shock proteins (HSPs) in response to a variety of stress conditions. HSF1 also plays a key role in regulating the development of various tumors; however, its role in pancreatic cancer and the specific underlying mechanism are not clear. Methods We first examined HSF1 expression in pancreatic cancer tissues by immunohistochemistry, and then studied its clinical significance. We then constructed HSF1-siRNA to investigate the potential of HSF1 to regulate apoptosis, proliferation and the cell cycle of pancreatic cancer cells and the underlying mechanism both in vitro and in vivo. Protein chip analysis was used subsequently to explore the molecular regulation pathway. Finally, second mitochondria-derived activator of caspase (SMAC)-siRNA was used to validate the signaling pathway. Results HSF1 was highly expressed in pancreatic cancer tissues and the level of upregulation was found to be closely related to the degree of pancreatic cancer differentiation and poor prognosis. After HSF1-silencing, we found that pancreatic cancer cell proliferation decreased both in vitro and in vivo and the apoptotic cell ratio increased, while the mitochondrial membrane potential decreased, and the cells were arrested at the G0/G1 phase. In terms of the molecular mechanism, we confirmed that HSF1 regulated SMAC to inhibit mitochondrial apoptosis in pancreatic cancer cells, and to promote the occurrence of pancreatic tumors. SMAC silencing reversed the effects of HSF1 silencing. Conclusion Our study provides evidence that HSF1 functions as a novel oncogene in pancreatic tumors and is implicated as a target for the diagnosis and treatment of pancreatic cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0537-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenjin Liang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Yong Liao
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Jing Zhang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Qi Huang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Wei Luo
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Jidong Yu
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Jianhua Gong
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Yi Zhou
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Xuan Li
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China
| | - Bo Tang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China.
| | - Songqing He
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China.
| | - Jinghong Yang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, 541004, Guangxi, People's Republic of China.
| |
Collapse
|
11
|
Peng Y, Dong Y, Mahato RI. Synthesis and Characterization of a Novel Mycophenolic Acid-Quinic Acid Conjugate Serving as Immunosuppressant with Decreased Toxicity. Mol Pharm 2015; 12:4445-53. [PMID: 26529468 DOI: 10.1021/acs.molpharmaceut.5b00639] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mycophenolic acid (MPA) is one of the most commonly used immunosuppressive drugs for improving the outcome of cell and organ transplantations. However, an undesired adverse effect of MPA impedes its application in the clinics for post-transplant patients. By conjugating MPA to quinic acid (QA) via amide bonds, we synthesized a novel immunosuppressant, N-[2-[[(4E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-1-oxo-4-hexen-1-yl]amino]ethyl]-(1α,3R,4α,5R)-1,3,4,5-tetrakis(acetyloxy)cyclohexanecarboxamide (abbreviated as MQ4), which exhibits improved stability demonstrated by its incubation in vitro with human plasma, suggesting its better resistance to hydrolytic degradation induced by plasma enzyme. While the immunosuppressive effect of MQ4 on human lymphocyte proliferation was partially compromised as shown by flow cytometry, significant decrease in cytotoxicity of MQ4 to insulin producing β cells could compensate this drawback to some degree. There was a decreased level of apoptotic mediator caspase-3, which may contribute to the decreased toxicity of MQ4 to INS-1E cells. MQ4 could further improve insulin stimulation index and downregulate NFκB expression compared to physical mixing of QA to MPA. Taken together, MQ4 is a promising immunosuppressive agent for preventing and minimizing post-transplanted immune rejection.
Collapse
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198-6025, United States
| | - Yuxiang Dong
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198-6025, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198-6025, United States
| |
Collapse
|
12
|
Kim JY. Current Prospects of RNA Interference-based Therapy in Organ Transplantation. KOREAN JOURNAL OF TRANSPLANTATION 2015. [DOI: 10.4285/jkstn.2015.29.3.101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Jae Young Kim
- Department of Life Science, Gachon University, Seongnam, Korea
| |
Collapse
|
13
|
Stimac M, Dolinsek T, Lampreht U, Cemazar M, Sersa G. Gene Electrotransfer of Plasmid with Tissue Specific Promoter Encoding shRNA against Endoglin Exerts Antitumor Efficacy against Murine TS/A Tumors by Vascular Targeted Effects. PLoS One 2015; 10:e0124913. [PMID: 25909447 PMCID: PMC4409373 DOI: 10.1371/journal.pone.0124913] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/11/2015] [Indexed: 02/06/2023] Open
Abstract
Vascular targeted therapies, targeting specific endothelial cell markers, are promising approaches for the treatment of cancer. One of the targets is endoglin, transforming growth factor-β (TGF-β) co-receptor, which mediates proliferation, differentiation and migration of endothelial cells forming neovasculature. However, its specific, safe and long-lasting targeting remains the challenge. Therefore, in our study we evaluated the transfection efficacy, vascular targeted effects and therapeutic potential of the plasmid silencing endoglin with the tissue specific promoter, specific for endothelial cells marker endothelin-1 (ET) (TS plasmid), in comparison to the plasmid with constitutive promoter (CON plasmid), in vitro and in vivo. Tissue specificity of TS plasmid was demonstrated in vitro on several cell lines, and its antiangiogenic efficacy was demonstrated by reducing tube formation of 2H11 endothelial cells. In vivo, on a murine mammary TS/A tumor model, we demonstrated good antitumor effect of gene electrotransfer (GET) of either of both plasmids in treatment of smaller tumors still in avascular phase of growth, as well as on bigger tumors, already well vascularized. In support to the observations on predominantly vascular targeted effects of endoglin, histological analysis has demonstrated an increase in necrosis and a decrease in the number of blood vessels in therapeutic groups. A significant antitumor effect was observed in tumors in avascular and vascular phase of growth, possibly due to both, the antiangiogenic and the vascular disrupting effect. Furthermore, the study indicates on the potential use of TS plasmid in cancer gene therapy since the same efficacy as of CON plasmid was determined.
Collapse
Affiliation(s)
- Monika Stimac
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Tanja Dolinsek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Ursa Lampreht
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia; Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
14
|
Yang C, Zhang C, Zhao Z, Zhu T, Yang B. Fighting against kidney diseases with small interfering RNA: opportunities and challenges. J Transl Med 2015; 13:39. [PMID: 25637948 PMCID: PMC4354745 DOI: 10.1186/s12967-015-0387-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/12/2015] [Indexed: 11/21/2022] Open
Abstract
The significant improvements in siRNA therapy have been achieved, which have great potential applications in humans. The kidney is a comparatively easy target organ of siRNA therapy due to its unique structural and functional characteristics. Here, we reviewed recent achievements in siRNA design, delivery and application with focuses on kidney diseases, in particular kidney transplant-related injuries. In addition, the strategy for increasing serum stability and immune tolerance of siRNA was also discussed. At last, the future challenges of siRNA therapy including organ/tissue/cell-specific delivery and time-controlled silence, as well as selecting therapeutic targets, were addressed as well.
Collapse
Affiliation(s)
- Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.
| | - Chao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.
| | - Zitong Zhao
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.
| | - Bin Yang
- Transplant Group, Department of Infection, Immunity and Inflammation, University Hospitals of Leicester, University of Leicester, Leicester, UK. .,Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China. .,Basic Medical Research Centre, Medical School of Nantong University, Nantong, China.
| |
Collapse
|
15
|
Hwang HJ, Lee M, Park JH, Jung HS, Kang JG, Kim CS, Lee SJ, Ihm SH. Improved islet transplantation outcome by the co-delivery of siRNAs for iNOS and 17β-estradiol using an R3V6 peptide carrier. Biomaterials 2015; 38:36-42. [DOI: 10.1016/j.biomaterials.2014.10.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 10/19/2014] [Indexed: 01/06/2023]
|
16
|
Veiseh O, Tang BC, Whitehead KA, Anderson DG, Langer R. Managing diabetes with nanomedicine: challenges and opportunities. Nat Rev Drug Discov 2015; 14:45-57. [PMID: 25430866 PMCID: PMC4751590 DOI: 10.1038/nrd4477] [Citation(s) in RCA: 355] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanotechnology-based approaches hold substantial potential for improving the care of patients with diabetes. Nanoparticles are being developed as imaging contrast agents to assist in the early diagnosis of type 1 diabetes. Glucose nanosensors are being incorporated in implantable devices that enable more accurate and patient-friendly real-time tracking of blood glucose levels, and are also providing the basis for glucose-responsive nanoparticles that better mimic the body's physiological needs for insulin. Finally, nanotechnology is being used in non-invasive approaches to insulin delivery and to engineer more effective vaccine, cell and gene therapies for type 1 diabetes. Here, we analyse the current state of these approaches and discuss key issues for their translation to clinical practice.
Collapse
Affiliation(s)
- Omid Veiseh
- 1] Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [2] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [3] Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave., Boston, Massachusetts 02115, USA. [4]
| | - Benjamin C Tang
- 1] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [2] Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave., Boston, Massachusetts 02115, USA. [3]
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, 5000 Forbes Ave., Pittsburgh, Pennsylvania 15213, USA
| | - Daniel G Anderson
- 1] Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [2] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [3] Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave., Boston, Massachusetts 02115, USA. [4] Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. [5] Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Robert Langer
- 1] Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [2] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [3] Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave., Boston, Massachusetts 02115, USA. [4] Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. [5] Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
17
|
Mcam Silencing With RNA Interference Using Magnetofection has Antitumor Effect in Murine Melanoma. MOLECULAR THERAPY-NUCLEIC ACIDS 2014; 3:e205. [PMID: 25350580 PMCID: PMC4217080 DOI: 10.1038/mtna.2014.56] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/09/2014] [Indexed: 01/27/2023]
Abstract
The melanoma cell adhesion molecule (MCAM) is involved in melanoma development and its progression, including invasiveness, metastatic potential and angiogenesis. Therefore, MCAM represents a potential target for gene therapy of melanoma, whose expression could be hindered with posttranscriptional specific gene silencing with RNA interference technology. In this study, we constructed a plasmid DNA encoding short hairpin RNA against MCAM (pMCAM) to explore the antitumor and antiangiogenic effects. The experiments were performed in vitro on murine melanoma and endothelial cells, as well as in vivo on melanoma tumors in mice. The antiproliferative, antimigratory, antiangiogenic and antitumor effects were examined after gene therapy with pMCAM. Gene delivery was performed by magnetofection, and its efficacy compared to gene electrotransfer. Gene therapy with pMCAM has proved to be an effective approach in reducing the proliferation and migration of melanoma cells, as well as having antiangiogenic effect in endothelial cells and antitumor effect on melanoma tumors. Magnetofection as a developing nonviral gene delivery system was effective in the transfection of melanoma cells and tumors with pMCAM, but less efficient than gene electrotransfer in in vivo tumor gene therapy due to the lack of antiangiogenic effect after silencing Mcam by magnetofection.
Collapse
|
18
|
Tamaki S, Nye C, Slorach E, Scharp D, Blau HM, Whiteley PE, Pomerantz JH. Simultaneous silencing of multiple RB and p53 pathway members induces cell cycle reentry in intact human pancreatic islets. BMC Biotechnol 2014; 14:86. [PMID: 25305068 PMCID: PMC4287515 DOI: 10.1186/1472-6750-14-86] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 09/19/2014] [Indexed: 11/20/2022] Open
Abstract
Background Human pancreatic islet structure poses challenges to investigations that require specific modulation of gene expression. Yet dissociation of islets into individual cells destroys cellular interactions important to islet physiology. Approaches that improve transient targeting of gene expression in intact human islets are needed in order to effectively perturb intracellular pathways to achieve biological effects in the most relevant tissue contexts. Results Electroporation of intact human cadaveric islets resulted in robust and specific suppression of gene expression. Two genes were simultaneously suppressed by 80% from baseline levels. When multiple (up to 5) genes were simultaneously targeted, effective suppression of 3 of 5 genes occurred. Enzymatic pretreatment of islets was not required. Simultaneous targeting of RB and p53 pathway members resulted in cell cycle reentry as measured by EDU incorporation in 10% of islet nuclei. Conclusions At least three genes can be effectively suppressed simultaneously in cultured intact human pancreatic islets without disruption of islet architecture or overt alterations in function. This enabled the effective modulation of two central growth control pathways resulting in the phenotypic outcome of cell cycle reentry in postmitotic islet cells. Transient exposure to multiple siRNAs is an effective approach to modify islets for study with the potential to aid clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jason H Pomerantz
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Craniofacial and Mesenchymal Biology Program, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
19
|
Abstract
Prostate cancer is the second leading cause of cancer related death in American men. Androgen deprivation therapy (ADT) is used to treat patients with aggressive prostate cancers. After androgen deprivation therapy, prostate cancers slowly progress to an androgen-independent status. Taxanes (e.g., docetaxel) are used as standard treatments for androgen-independent prostate cancers. However, these chemotherapeutic agents will eventually become ineffective due to the development of drug resistance. A microRNA (miRNA) is a small noncoding RNA molecule, which can regulate gene expression at the post-transcription level. miRNAs elicit their effects by binding to the 3'-untranslated region (3'-UTR) of their target mRNAs, leading to the inhibition of translation or the degradation of the mRNAs. miRNAs have received increasing attention as targets for cancer therapy, as they can target multiple signaling pathways related to tumor progression, metastasis, invasion, and chemoresistance. Emerging evidence suggests that aberrant expression of miRNAs can lead to the development of resistant prostate cancers. Here, we discuss the roles of miRNAs in the development of resistant prostate cancers and their involvement in various drug resistant mechanisms including androgen signaling, apoptosis avoidance, multiple drug resistance (MDR) transporters, epithelialmesenchymal transition (EMT), and cancer stem cells (CSCs). In addition, we also discuss strategies for treating resistant prostate cancers by targeting specific miRNAs. Different delivery strategies are also discussed with focus on those that have been successfully used in human clinical trials.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University , Hampton, Virginia 23668, United States
| | | |
Collapse
|
20
|
Kandeel M, Kitade Y. In silico molecular docking analysis of the human Argonaute 2 PAZ domain reveals insights into RNA interference. J Comput Aided Mol Des 2013; 27:605-14. [PMID: 23877830 DOI: 10.1007/s10822-013-9665-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 07/05/2013] [Indexed: 12/01/2022]
Abstract
RNA interference (RNAi) is a critical cellular pathway activated by double stranded RNA and regulates the gene expression of target mRNA. During RNAi, the 3' end of siRNA binds with the PAZ domain, followed by release and rebinding in a cyclic manner, which deemed essential for proper gene silencing. Recently, we provided the forces underlying the recognition of small interfering RNA by PAZ in a computational study based on the structure of Drosophila Argonaute 2 (Ago2) PAZ domain. We have now reanalyzed these data within the view of the new available structures from human Argonauts. While the parameters of weak binding are correlated with higher (RNAi) in the Drosophila model, a different profile is predicted with the human Ago2 PAZ domain. On the basis of the human Ago2 PAZ models, the indicators of stronger binding as the total binding energy and the free energy were associated with better RNAi efficacy. This discrepancy might be attributable to differences in the binding site topology and the difference in the conformation of the bound nucleotides.
Collapse
Affiliation(s)
- Mahmoud Kandeel
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelshiekh University, Kafrelshiekh, Egypt.
| | | |
Collapse
|
21
|
Zhang RX, Yan XB, Gu YH, Huang D, Gan L, Han R, Huang LH. Gene silencing of NR2B-containing NMDA receptor by intrathecal injection of short hairpin RNA reduces formalin-induced nociception in C57BL/6 mouse. Int J Neurosci 2013; 123:650-6. [PMID: 23528046 DOI: 10.3109/00207454.2013.789873] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Spinal NR2B-containing N-methyl-D-aspartate receptors (NR2B) play a critical role in the formation of central sensitization and persistent pain. Previous studies show that gene silencing of the spinal NR2B subunit by small interfering RNA (siRNA) could alleviate nociception in animals. The siRNA is a 19- to 23-nt RNA duplex, which can be synthesized in vitro or derived from short hairpin RNAs (shRNAs). In the present study, we investigated whether intrathecal injection of shRNAs targeting NR2B (GRIN2B shRNA) could affect nociception on formalin-induced pain in mice. Our results showed that intrathecal injection of GRIN2B shRNA could decrease NR2B mRNA and protein expression levels and hence effectively relieve formalin-induced nociception in mice, suggesting that intrathecal delivery of GRIN2B shRNA can be an efficient way to silence the target gene and provide new insights into the treatment of chronic pain.
Collapse
Affiliation(s)
- Rao-Xiang Zhang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Saito M, Kakutani Y, Kaburagi M, Funabashi H, Matsuoka H. Development of a protocol for selection of genes fit for the in vivo knockdown method and its application to insulin receptor substrate genes in mice. Exp Anim 2013; 62:117-25. [PMID: 23615306 DOI: 10.1538/expanim.62.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Prediabetes model mice in which more than one gene associated with diabetes is knocked down simultaneously are potentially useful for pharmaceutical and medical studies of diabetes. However, the effective conditions for sufficient knockdown in vivo are dependent on the intrinsic properties of the target genes. It is necessary to investigate which genes are applicable or not to the in vivo knockdown method. In this study, insulin receptor substrate 1 and 2 (Irs-1, Irs-2) were selected as target genes. Effective siRNAs against the respective genes were designed, and their efficacy was confirmed by cell-based experiments. Based on the results of siRNAs, shRNA expression vectors against Irs-1 and Irs-2 were constructed, respectively. Their efficacy was also confirmed by cell-based experiments. A hydrodynamic method was applied to the delivery of the vectors to mice. This method was found to be effective for predominant delivery to the liver by demonstrative delivery of an EGFP expression vector and successive histochemical analysis. Fifty micrograms of the shRNA expression vector was injected into the tail vein. After 24 h, the liver, pancreas, and muscle were isolated, and the expression levels of Irs-1 and Irs-2 were analyzed by quantitative RT-PCR. In the liver, Irs-2 was effectively knocked down to 60% of the control level, but Irs-1 was not influenced even under the same conditions. The protocol developed here is feasible for the selection of genes fit for in vivo knockdown method.
Collapse
Affiliation(s)
- Mikako Saito
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | | | | | | | | |
Collapse
|
23
|
Development of novel cell lines of diabetic dysfunction model fit for cell-based screening tests of medicinal materials. Cytotechnology 2012; 65:105-18. [PMID: 22777586 DOI: 10.1007/s10616-012-9466-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/14/2012] [Indexed: 10/28/2022] Open
Abstract
Pdx-1 and Irs-1, genes highly associated with diabetes onset, were knocked down in mouse embryonic stem (ES) cells in order to develop cell line models for diabetes. ES cells with different gene knockdown levels were induced to differentiate to the stage of insulin production. Among the cell lines that differentiated, we identified two in which the levels of expression of both genes were 20-40 % of that of control cells. These cell lines showed appreciable deficiencies in three characteristic malfunctions associated with diabetes, namely, insulin production, insulin reception signaling, and glucose-stimulated insulin secretion. These dysfunctions were consistent with results reported elsewhere from in vivo and in vitro studies. Both cell lines did not show any abnormal morphology such as size, shape, color, and surface roughness. No abnormal expression profiles for 17 genes relevant to diabetes were observed. Therefore, these cell lines fulfilled the criteria for a validated cell model for diabetes. The model cell lines developed here are promising biomaterials for cell-based screening tests of new medicines that may be effective in treating diabetes.
Collapse
|