Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
World J Gastroenterol ; 30(23): 2959-2963, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38946873

ABSTRACT

In this editorial, we comment on the article entitled "Stage at diagnosis of colorectal cancer through diagnostic route: Who should be screened?" by Agatsuma et al. Colorectal cancer (CRC) is emerging as an important health issue as its incidence continues to rise globally, adversely affecting the quality of life. Although the public has become more aware of CRC prevention, most patients lack screening awareness. Some poor lifestyle practices can lead to CRC and symptoms can appear in the early stages of CRC. However, due to the lack of awareness of the disease, most of the CRC patients are diagnosed already at an advanced stage and have a poor prognosis.


Subject(s)
Colorectal Neoplasms , Early Detection of Cancer , Humans , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/prevention & control , Colorectal Neoplasms/epidemiology , Early Detection of Cancer/methods , Quality of Life , Neoplasm Staging , Mass Screening/methods , Mass Screening/standards , Prognosis , Colonoscopy , Incidence , Health Knowledge, Attitudes, Practice , Life Style
2.
World J Clin Oncol ; 15(5): 603-613, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38835843

ABSTRACT

Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a member of the type I receptor tyrosine kinase family. ROR1 is pivotal in embryonic development and cancer, and serves as a biomarker and therapeutic target. It has soluble and membrane-bound subtypes, with the latter highly expressed in tumors. ROR1 is conserved throughout evolution and may play a role in the development of gastrointestinal cancer through multiple signaling pathways and molecular mechanisms. Studies suggest that overexpression of ROR1 may increase tumor invasiveness and metastasis. Additionally, ROR1 may regulate the cell cycle, stem cell characteristics, and interact with other signaling pathways to affect cancer progression. This review explores the structure, expression and role of ROR1 in the development of gastrointestinal cancers. It discusses current antitumor strategies, outlining challenges and prospects for treatment.

3.
Cells ; 11(9)2022 05 07.
Article in English | MEDLINE | ID: mdl-35563879

ABSTRACT

We demonstrated previously that extracellular vesicles (EVs) released from mesenchymal stem cells (MSCs) play a critical role in angiogenesis. Here, we examine whether this pro-angiogenic efficacy is enhanced in EVs derived from MSCs overexpressing GATA-4 (MSCGATA-4). Methods and Results. EVs were isolated from MSCGATA-4 (EVGATA-4) and control MSCs transduced with an empty vector (EVnull). EVs from both cell types were of the same size and displayed similar molecular markers. Compared with EVnull, EVGATA-4 increased both a tube-like structure formation and spheroid-based sprouting of human umbilical vein endothelial cells (HUVECs). The EVGATA-4 increased the numbers of CD31-positive cells and hemoglobin content inside Matrigel plugs subcutaneously transplanted into mice for 2 weeks. Moreover, EVGATA-4 encapsulated higher levels of let-7 family miRs compared to EVnull. The transfer of exosomal let-7 miRs into HUVECs was recorded with an accompanied down-regulation of thrombospondin-1 (THBS1) expression, a major endogenous angiogenesis inhibitor. The loss-and-gain of function studies of let-7 miRs showed that let-7f knockdown significantly decreased EVGATA-4-mediated vascularization inside Matrigel plugs. In contrast, let-7f overexpression promoted HUVEC migration and tube formation. Conclusion. Our results indicate that EVs derived from genetically modified MSCs with GATA-4 overexpression had increased pro-angiogenic capacity due to the delivery of let-7 miRs that targeted THBS1 in endothelial cells.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Animals , Extracellular Vesicles/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Pathologic/metabolism
4.
World J Stem Cells ; 14(1): 41-53, 2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35126827

ABSTRACT

The transforming growth factor (TGF)-ß signaling pathway controls many cellular processes, including proliferation, differentiation, and apoptosis. Abnormalities in the TGF-ß signaling pathway and its components are closely related to the occurrence of many human diseases, including cancer. Mothers against decapentaplegic homolog 4 (Smad4), also known as deleted in pancreatic cancer locus 4, is a typical tumor suppressor candidate gene locating at q21.1 of human chromosome 18 and the common mediator of the TGF-ß/Smad and bone morphogenetic protein/Smad signaling pathways. It is believed that Smad4 inactivation correlates with the development of tumors and stem cell fate decisions. Smad4 also interacts with cytokines, miRNAs, and other signaling pathways, jointly regulating cell behavior. However, the regulatory function of Smad4 in tumorigenesis, stem cells, and drug resistance is currently controversial. In addition, Smad4 represents an attractive therapeutic target for cancer. Elucidating the specific role of Smad4 is important for understanding the mechanism of tumorigenesis and cancer treatment. Here, we review the identification and characterization of Smad4, the canonical TGF-ß/Smad pathway, as well as the multiple roles of Smad4 in tumorigenesis, stem cells, and drug resistance. Furthermore, we provide novel insights into the prospects of Smad4-targeted cancer therapy and the challenges that it will face in the future.

5.
Genes (Basel) ; 11(11)2020 10 29.
Article in English | MEDLINE | ID: mdl-33137935

ABSTRACT

BACKGROUND: We demonstrated that the transduction of Wnt11 into mesenchymal stem cells (MSCs) (MSCWnt11) promotes these cells differentiation into cardiac phenotypes. In the present study, we investigated the paracrine effects of MSCWnt11 on cardiac function and angiogenesis. METHODS AND RESULTS: Conditioned medium was collected from MSCWnt11 (CdMWnt11) and their control cells (CdMGFP). CdMWnt11, especially obtained from MSCWnt11 exposed to hypoxia, significantly promoted human umbilical vein endothelial cells (HUVECs) migration and increased capillary-like tube (CLT) formation, which was blocked by Wnt11 neutralizing antibody. Wnt11 protein was significantly higher in CdMWnt11 compared to that in CdMGFP. Directly treating HUVECs with recombinant Wnt11 protein significantly increased CLT formation, which was abrogated by treating cells with the JNK inhibitor SP600125, as well as the PKC inhibitor Calphostin-C. Moreover, the transfection of Wnt11 to HUVECs (HWnt11) significantly increased CLT formation and HUVEC migration, as well as upregulated p-pan-PKC and p-JNK expression. Injection of CdMWnt11 into the peri-infarct region in a rat acute myocardial infarction (AMI) model significantly improved cardiac function, reduced infarct size, and increased myocardial blood flow and blood vessel density in the ischemic area. CONCLUSION: Wnt11 released from MSCWnt11 increased angiogenesis and improved cardiac function via non-canonical Wnt-PKC-JNK dependent pathways.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Capillaries/cytology , Capillaries/growth & development , Capillaries/metabolism , Cells, Cultured , Culture Media, Conditioned , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells , Humans , MAP Kinase Signaling System , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/genetics , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics
6.
World J Stem Cells ; 12(6): 481-487, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32742565

ABSTRACT

Cancer cells possess metabolic properties that are different from those of benign cells. p21, encoded by CDKN1A gene, also named p21Cip1/WAF1, was first identified as a cyclin-dependent kinase regulator that suppresses cell cycle G1/S phase and retinoblastoma protein phosphorylation. CDKN1A (p21) acts as the downstream target gene of TP53 (p53), and its expression is induced by wild-type p53 and it is not associated with mutant p53. p21 has been characterized as a vital regulator that involves multiple cell functions, including G1/S cell cycle progression, cell growth, DNA damage, and cell stemness. In 1994, p21 was found as a tumor suppressor in brain, lung and colon cancer by targeting p53 and was associated with tumorigenesis and metastasis. Notably, p21 plays a significant role in tumor development through p53-dependent and p53-independent pathways. In addition, expression of p21 is closely related to the resting state or terminal differentiation of cells. p21 is also associated with cancer stem cells and acts as a biomarker for such cells. In cancer therapy, given the importance of p21 in regulating the G1/S and G2 check points, it is not surprising that p21 is implicated in response to many cancer treatments and p21 promotes the effect of oncolytic virotherapy.

7.
J Am Heart Assoc ; 8(15): e012089, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31315484

ABSTRACT

Background Myocardial infarction results in a large-scale cardiomyocyte loss and heart failure due to subsequent pathological remodeling. Whereas zebrafish and neonatal mice have evident cardiomyocyte expansion following injury, adult mammalian cardiomyocytes are principally nonproliferative. Despite historical presumptions of stem cell-mediated cardiac regeneration, numerous recent studies using advanced lineage-tracing methods demonstrated that the only source of cardiomyocyte renewal originates from the extant myocardium; thus, the augmented proliferation of preexisting adult cardiomyocytes remains a leading therapeutic approach toward cardiac regeneration. In the present study we investigate the significance of suppressing cell cycle inhibitors Rb1 and Meis2 to promote adult cardiomyocyte reentry to the cell cycle. Methods and Results In vitro experiments with small interfering RNA-mediated simultaneous knockdown of Rb1 and Meis2 in both adult rat cardiomyocytes, isolated from 12-week-old Fischer rats, and human induced pluripotent stem cell-derived cardiomyocytes showed a significant increase in cell number, a decrease in cell size, and an increase in mononucleated cardiomyocytes. In vivo, a hydrogel-based delivery method for small interfering RNA-mediated silencing of Rb1 and Meis2 is utilized following myocardial infarction. Immunofluorescent imaging analysis revealed a significant increase in proliferation markers 5-ethynyl-2'-deoxyuridine, PH3, KI67, and Aurora B in adult cardiomyocytes as well as improved cell survivability with the additional benefit of enhanced peri-infarct angiogenesis. Together, this intervention resulted in a reduced infarct size and improved cardiac function post-myocardial infarction. Conclusions Silencing of senescence-inducing pathways in adult cardiomyocytes via inhibition of Rb1 and Meis2 results in marked cardiomyocyte proliferation and increased protection of cardiac function in the setting of ischemic injury.


Subject(s)
Cell Cycle/physiology , Homeodomain Proteins/genetics , Myocardial Infarction , Myocytes, Cardiac/cytology , Retinoblastoma Binding Proteins/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Age Factors , Animals , Homeodomain Proteins/physiology , Humans , Male , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats , Rats, Inbred F344 , Retinoblastoma Binding Proteins/physiology , Transcription Factors/physiology , Ubiquitin-Protein Ligases/physiology
8.
Acta Pharmacol Sin ; 40(9): 1168-1183, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30858476

ABSTRACT

Understanding the dynamics of the immune response following late myocardial reperfusion is critical for the development of immunomodulatory therapy for myocardial infarction (MI). Cyclosporine A (CSA) possesses multiple therapeutic applications for MI, but its effects on the inflammation caused by acute MI are not clear. This study aimed to determine the dynamics of the immune response following myocardial ischemia/reperfusion (I/R) and the effects of CSA in a mouse model of prolonged myocardial ischemia designated to represent the human condition of late reperfusion. Adult C57BL/6 mice were subjected to 90 min of closed-chest myocardial I/R, which induced severe myocardial injury and excessive inflammation in the heart. Multicomponent analysis of the immune response caused by prolonged I/R revealed that the peak of cytokines/chemokines in the systemic circulation was synchronized with the maximal influx of neutrophils and T-cells in the heart 1 day after MI. The peak of cytokine/chemokine secretion in the infarcted heart coincided with the maximal macrophage and natural killer cell infiltration on day 3 after MI. The cellular composition of the mediastinal lymph nodes changed similarly to that of the infarcted hearts. CSA (10 mg/kg/day) given after prolonged I/R impaired heart function, enlarged the resulting scar, and reduced heart vascularization. It did not change the content of immune cells in hearts exposed to prolonged I/R, but the levels of MCP-1 and MIP-1α (hearts) and IL-12 (hearts and serum) were significantly reduced in the CSA-treated group in comparison to the untreated group, indicating alterations in immune cell function. Our findings provide new knowledge necessary for the development of immunomodulatory therapy targeting the immune response after prolonged myocardial ischemia/reperfusion.


Subject(s)
Cyclosporine/pharmacology , Immunity, Cellular/drug effects , Immunity, Cellular/physiology , Myocardial Reperfusion Injury/physiopathology , Animals , Chemokines/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Killer Cells, Natural/metabolism , Lymph Nodes/drug effects , Lymph Nodes/physiology , Male , Mice, Inbred C57BL , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Neutrophils/metabolism , T-Lymphocytes/metabolism , Time Factors , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
9.
World J Gastroenterol ; 24(41): 4643-4651, 2018 Nov 07.
Article in English | MEDLINE | ID: mdl-30416312

ABSTRACT

Autophagy is a "self-degradative" process and is involved in the maintenance of cellular homeostasis and the control of cellular components by facilitating the clearance or turnover of long-lived or misfolded proteins, protein aggregates, and damaged organelles. Autophagy plays a dual role in cancer, including in tumor progression and tumor promotion, suggesting that autophagy acts as a double-edged sword in cancer cells. Liver cancer is one of the greatest leading causes of cancer death worldwide due to its high recurrence rate and poor prognosis. Especially in China, liver cancer has become one of the most common cancers due to the high infection rate of hepatitis virus. In primary liver cancer, hepatocellular carcinoma (HCC) is the most common type. Considering the perniciousness and complexity of HCC, it is essential to elucidate the function of autophagy in HCC. In this review, we summarize the physiological function of autophagy in cancer, analyze the role of autophagy in tumorigenesis and metastasis, discuss the therapeutic strategies targeting autophagy and the mechanisms of drug-resistance in HCC, and provide potential methods to circumvent resistance and combined anticancer strategies for HCC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Carcinoma, Hepatocellular/pathology , Hepatocytes/pathology , Liver Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Autophagy/genetics , Beclin-1/genetics , Beclin-1/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Disease Progression , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genetic Therapy/methods , Humans , Liver/cytology , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Oncolytic Virotherapy/methods
10.
Acta Pharmacol Sin ; 39(3): 405-414, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28880012

ABSTRACT

GOLPH2 (also called GP73) is a Golgi glycoprotein, which has been identified as a novel tumor marker upregulated in various cancers, including prostate cancer (PCa). GD55 is a novel GOLPH2-regulated oncolytic adenovirus that exhibits a strong killing effect on hepatoma cells. Here, we investigate the antitumor effect of GD55 on prostate cancer stem cell (CSC)-like cells in vitro and in vivo. Prostate CSC-like sphere cells were acquired and enriched by culturing DU145, LNCap or P3 prostate cancer cells in suspension. The prostate CSC-like sphere cells were capable of self-renewal, differentiation and quiescence, displaying tumorigenic feature and chemo-resistance to 5-FU, doxorubicin and DDP. Treatment with GD55 (1, 5, 10 MOI) dose-dependently suppressed the viability of DU145 sphere cells, which was a more pronounced compared to its cytotoxic action on the parental DU145 cells. In a mouse xenograft prostate CSC-like model, intratumoral injection of GD55 markedly suppressed the growth rate of xenograft tumors and induced higher levels of cell death and necrosis within the tumor tissues. Our results demonstrate that GD55 infection exerts strong anticancer effects on prostate CSC-like cells in vitro and in vivo, and has a potential to be used in the clinical therapy of PCa.


Subject(s)
Adenoviridae , Membrane Proteins/biosynthesis , Neoplastic Stem Cells/virology , Oncolytic Viruses , Animals , Apoptosis , Cell Line, Tumor , Female , Humans , Male , Mice , Oncolytic Virotherapy/methods , Prostate/pathology , Xenograft Model Antitumor Assays
11.
Cardiovasc Res ; 113(13): 1585-1602, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29016740

ABSTRACT

AIMS: Fibroblast growth factor 1 (FGF1), a heparin/heparan sulfate-binding growth factor, is a potent cardioprotective agent against myocardial infarction (MI). The impact of heparin, the standard of care for MI patients entering the emergency room, on cardioprotective effects of FGF1 is unknown, however. METHODS AND RESULTS: To address this, a rat model of MI was employed to compare cardioprotective potentials (lower infarct size and improve post-ischemic function) of native FGF1 and an engineered FGF1 (FGF1ΔHBS) with reduced heparin-binding affinity when given at the onset of reperfusion in the absence or presence of heparin. FGF1 and FGF1ΔHBS did not alter heparin's anticoagulant properties. Treatment with heparin alone or native FGF1 significantly reduced infarct size compared to saline (P < 0.05). Surprisingly, treatment with FGF1ΔHBS markedly lowered infarct size compared to FGF1 (P < 0.05). Both native and modified FGF1 restored contractile and relaxation function (P < 0.05 versus saline or heparin). Furthermore, FGF1ΔHBS had greater improvement in cardiac function compared to FGF1 (P < 0.05). Heparin negatively impacted the cardioprotective effects (infarct size, post-ischemic recovery of function) of FGF1 (P < 0.05) but not of FGF1ΔHBS. Heparin also reduced the biodistribution of FGF1, but not FGF1ΔHBS, to the left ventricle. FGF1 and FGF1ΔHBS bound and triggered FGFR1-induced downstream activation of ERK1/2 (P < 0.05); yet, heparin co-treatment decreased FGF1-produced ERK1/2 activation, but not that activated by FGF1ΔHBS. CONCLUSION: These findings demonstrate that modification of the heparin-binding region of FGF1 significantly improves the cardioprotective efficacy, even in the presence of heparin, identifying a novel FGF ligand available for therapeutic use in ischemic heart disease.


Subject(s)
Cardiovascular Agents/pharmacology , Fibroblast Growth Factor 1/pharmacology , Heparin/pharmacology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Animals , Cardiovascular Agents/metabolism , Cardiovascular Agents/pharmacokinetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 1/pharmacokinetics , Heparin/metabolism , Humans , Ligands , Male , Mutation , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Protein Binding , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recovery of Function , Tissue Distribution , Ventricular Function, Left/drug effects
12.
World J Gastroenterol ; 22(35): 7999-8009, 2016 Sep 21.
Article in English | MEDLINE | ID: mdl-27672294

ABSTRACT

Gastrointestinal cancer has been one of the five most commonly diagnosed and leading causes of cancer mortality over the past few decades. Great progress in traditional therapies has been made, which prolonged survival in patients with early cancer, yet tumor relapse and drug resistance still occurred, which is explained by the cancer stem cell (CSC) theory. Oncolytic virotherapy has attracted increasing interest in cancer because of its ability to infect and lyse CSCs. This paper reviews the basic knowledge, CSC markers and therapeutics of gastrointestinal cancer (liver, gastric, colon and pancreatic cancer), as well as research advances and possible molecular mechanisms of various oncolytic viruses against gastrointestinal CSCs. This paper also summarizes the existing obstacles to oncolytic virotherapy and proposes several alternative suggestions to overcome the therapeutic limitations.


Subject(s)
Gastrointestinal Neoplasms/therapy , Neoplasm Recurrence, Local , Neoplastic Stem Cells/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses , Pancreatic Neoplasms/therapy , Adenoviridae , Alphavirus , Animals , Gastrointestinal Neoplasms/virology , Humans , Measles virus , Newcastle disease virus , Pancreatic Neoplasms/virology , Reoviridae , Simplexvirus , Vaccinia virus , Vesiculovirus
13.
PLoS One ; 11(3): e0151542, 2016.
Article in English | MEDLINE | ID: mdl-26962868

ABSTRACT

BACKGROUND: Clusterin (Clu) is a stress-responding protein with multiple biological functions. Our preliminary microarray studies show that clusterin was prominently upregulated in mesenchymal stem cells (MSCs) overexpressing GATA-4 (MSCGATA-4). We hypothesized that the upregulation of clusterin is involved in overexpression of GATA-4 mediated cytoprotection. METHODS: MSCs harvested from bone marrow of rats were transduced with GATA-4. The expression of clusterin in MSCs was further confirmed by real-time PCR and western blotting. Simulation of ischemia was achieved by exposure of MSCs to a hypoxic environment. Lactate dehydrogenase (LDH) released from MSCs was served as a biomarker of cell injury and MTs uptake was used to estimate cell viability. Mitochondrial function was evaluated by measuring mitochondrial membrane potential (ΔΨm) and caspase 3/7 activity. RESULTS: (1) Clusterin expression was up-regulated in MSCGATA-4 compared to control MSCs transfected with empty-vector (MSCNull). MSCGATA-4 were tolerant to 72 h hypoxia exposure as shown by reduced LDH release and higher MTs uptake. This protection was abrogated by transfecting Clu-siRNA into MSCGATA-4. (2) Exogenous clusterin significantly decreased LDH release and increased MSC survival in hypoxic environment. Moreover, ΔΨm was maintained and caspase 3/7 activity was reduced by clusterin in a concentration-dependent manner. (3) p-Akt expression in MSCs was upregulated following pre-treatment with clusterin, with no change in total Akt. Moreover, cytoprotection mediated by clusterin was partially abrogated by Akt inhibitor LY294002. CONCLUSIONS: Clusterin/Akt signaling pathway is involved in GATA-4 mediated cytoprotection against hypoxia stress. It is suggested that clusterin may be therapeutically exploited in MSC based therapy for cardiovascular diseases.


Subject(s)
Clusterin/biosynthesis , GATA4 Transcription Factor/metabolism , Mesenchymal Stem Cells/metabolism , Oncogene Protein v-akt/biosynthesis , Signal Transduction , Up-Regulation , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Cell Hypoxia , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley
14.
World J Gastroenterol ; 22(1): 326-37, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26755879

ABSTRACT

Human hepatocellular carcinoma (HCC) heavily endangers human heath worldwide. HCC is one of most frequent cancers in China because patients with liver disease, such as chronic hepatitis, have the highest cancer susceptibility. Traditional therapeutic approaches have limited efficacy in advanced liver cancer, and novel strategies are urgently needed to improve the limited treatment options for HCC. This review summarizes the basic knowledge, current advances, and future challenges and prospects of adeno-associated virus (AAV) and adenoviruses as vectors for gene therapy of HCC. This paper also reviews the clinical trials of gene therapy using adenovirus vectors, immunotherapy, toxicity and immunological barriers for AAV and adenoviruses, and proposes several alternative strategies to overcome the therapeutic barriers to using AAV and adenoviruses as vectors.


Subject(s)
Carcinoma, Hepatocellular/therapy , Genetic Therapy/methods , Liver Neoplasms/therapy , Adenoviridae/genetics , Dependovirus/genetics , Genetic Therapy/adverse effects , Genetic Therapy/trends , Genetic Vectors , Humans , Immunotherapy , Oncolytic Virotherapy
15.
Stem Cells ; 34(1): 148-59, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26390028

ABSTRACT

Previously, we reported that a novel subpopulation of young mesenchymal stem cells (YMSCs) existed in old bone marrow, which possessed high antiaging properties as well as excellent efficacy for cardiac repair. MicroRNAs (miRNAs) have emerged as key regulators in post-transcriptional gene expression programs, and however, it is unknown whether miRNAs directly control stem cell senescence. Here we present the first evidence that miR-195 overexpressed in old MSCs (OMSCs) induces stem cell senescence deteriorating their regenerative ability by directly deactivating telomerase reverse transcriptase (Tert), and abrogation of miR-195 can reverse stem cell aging. MiRNAs profiling analysis in YMSCs and OMSCs by microarray showed that miR-140, miR-146a/b, and miR-195 were significantly upregulated in OMSCs, which led us to hypothesize that these are age-induced miRNAs involved in stem cell senescence. Of these miRNAs, we found miR-195 directly targeted 3'-untranslated region of Tert gene by computational target prediction analysis and luciferase assay, and knockdown of miR-195 significantly increased Tert expression in OMSCs. Strikingly, miR-195 inhibition significantly induced telomere relengthening in OMSCs along with reduced expression of senescence-associated ß-galactosidase. Moreover, silencing miR-195 in OMSCs by transfection of miR-195 inhibitor significantly restored antiaging factors expression including Tert and Sirt1 as well as phosphorylation of Akt and FOXO1. Notably, abrogation of miR-195 markedly restored proliferative abilities in OMSCs. Transplantation of OMSCs with knocked out miR-195 reduced infarction size and improved LV function. In conclusion, rejuvenation of aged stem cells by miR-195 inhibition would be a promising autologous therapeutic strategy for cardiac repair in the elderly patients.


Subject(s)
Aging/genetics , Cellular Senescence , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Telomerase/metabolism , Animals , Base Sequence , Biomarkers/metabolism , Cellular Senescence/genetics , Gene Expression Regulation, Developmental , Mesenchymal Stem Cell Transplantation , Mice, Inbred C57BL , MicroRNAs/genetics , Molecular Sequence Data , Treatment Outcome , Up-Regulation/genetics , Wound Healing
16.
J Mol Cell Cardiol ; 77: 102-12, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25451385

ABSTRACT

Although ischemia/reperfusion (I/R)-induced myocardial contractile dysfunction is associated with a prominent decrease in myofilament Ca(2+) sensitivity, the underlying mechanisms have not yet been fully clarified. Phosphorylation of ventricular myosin light chain 2 (MLC-2v) facilitates actin-myosin interactions and enhances contractility, however, its level and regulation by cardiac MLC kinase (cMLCK) and cMLC phosphatase (cMLCP) in I/R hearts are debatable. In this study, the levels and/or effects of MLC-2v phosphorylation, cMLCK, cMLCP, and proteases during I/R were determined. Global myocardial I/R-suppressed cardiac performance in isolated rat hearts was concomitant with decreases of MLC-2v phosphorylation, myofibrillar Ca(2+)-stimulated ATPase activity, and cMLCK content, but not cMLCP proteins. Consistently, simulated I/R in isolated cardiomyocytes inhibited cell shortening, Ca(2+) transients, MLC-2v phosphorylation, and myofilament sensitivity to Ca(2+). These observations were reversed by cMLCK overexpression, while the specific cMLCK knockdown by short hairpin RNA (shRNA) had the opposite effect. Moreover, the inhibition of matrix metalloproteinase-2 (MMP-2, a zinc-dependent endopeptidase) reversed IR-decreased cMLCK, MLC-2v phosphorylation, myofibrillar Ca(2+)-stimulated ATPase activity, myocardial contractile function, and myofilament sensitivity to Ca(2+), while the inhibition or knockdown of cMLCK by ML-9 or specific shRNA abolished MMP-2 inhibition-induced cardioprotection. Finally, the co-localization in cardiomyocytes and interaction in vivo of MMP-2 and cMLCK were observed. Purified recombinant rat cMLCK was concentration- and time-dependently degraded by rat MMP-2 in vitro, and this was prevented by the inhibition of MMP-2. These findings reveal that the I/R-activated MMP-2 leads to the degradation of cMLCK, resulting in a reduction of MLC-2v phosphorylation, and myofibrillar Ca(2+)-stimulated ATPase activity, which subsequently suppresses myocardial contractile function through a decrease of myofilament Ca(2+) sensitivity.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Myocardial Reperfusion Injury/enzymology , Myosin-Light-Chain Kinase/metabolism , Animals , Cells, Cultured , Male , Myocardial Contraction , Myocardial Ischemia/enzymology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/enzymology , Protein Processing, Post-Translational , Proteolysis , Rats, Sprague-Dawley
17.
Int J Clin Exp Pathol ; 7(7): 4067-75, 2014.
Article in English | MEDLINE | ID: mdl-25120785

ABSTRACT

Current metabolomic studies of ischemic brain mainly attach importance on a certain ischemic period, are lack of data about dynamic metabolites in ischemic stroke process, especially early period. Thus, in this study, (1)H NMR spectroscopy was used to investigate biochemical changes in the early stages of rats' focal cerebral ischemia reperfusion (I/R) injury. Serum samples of 0, 0.5, 1, 3, 6, 12, 24 h of reperfusion, based on multivariate data analyses, were tested to analyze the changing of metabolites during the early disease process. Partial least squares-discriminant analysis scores plots of the (1)H NMR data revealed clear differences among the experiment groups. Combination the results of loading plot and t-test, we found that 13 metabolites were changed significantly. Among that, malonic acid and glycine are the most noticeable variable metabolites. Dramatic changed malonic acid and glycine most probably served as biomarkers in this study. These findings help us understand the biochemical metabolite changes in early ischemic stroke stages, especially different periods. That may be conducive to distinguish at-risk individuals, benefit early diagnosis and understand the dynamic pathogenesis of early cerebral ischemia.


Subject(s)
Biomarkers/blood , Brain Ischemia/metabolism , Metabolomics/methods , Reperfusion Injury/metabolism , Stroke/metabolism , Animals , Disease Models, Animal , Glycine/blood , Magnetic Resonance Spectroscopy , Male , Malonates/blood , Rats , Rats, Sprague-Dawley
18.
Orthop Surg ; 6(1): 38-41, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24590992

ABSTRACT

OBJECTIVE: To investigate the effects of arthroscopic partial meniscectomy of torn discoid lateral meniscus on patellar tracking. METHODS: In all, 112 patients (112 knees) who underwent arthroscopic partial meniscectomy participated in the study. All subjects were examined with standing weight-bearing magnetic resonance imaging (MRI) preoperatively and one month postoperation. Axial-plane images through the maximum width of the patella and the maximum dorsal area of the femoral condyles were superimposed and the bisect offset index used to quantify patellar translation. Differences between pre- and postoperation in the bisect offset indexes were calculated and compared by Student's paired t-test. P < 0.05 was considered statistically significant. RESULTS: Before surgery, the mean bisect offset index was 0.574 (0.437-0.692). One month postoperation, the mean bisect offset index was 0.622 (0.510-0.801). The postoperation bisect offset index increased by an average of 0.048 (-0.018 to 0.129) compared with the preoperation value; this difference is statistically significant result (t = 18.33, P < 0.01). CONCLUSION: After arthroscopic partial meniscectomy, the patella translates more lateral to the femoral trochlear groove. These results suggest that arthroscopic partial meniscectomy may result in patellar maltracking.


Subject(s)
Arthroscopy/methods , Tibial Meniscus Injuries , Adolescent , Female , Humans , Magnetic Resonance Imaging , Male , Menisci, Tibial/pathology , Menisci, Tibial/surgery , Patella/pathology , Postoperative Care , Rupture/pathology , Rupture/surgery , Weight-Bearing , Young Adult
20.
Int J Biochem Cell Biol ; 45(12): 2724-35, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24070634

ABSTRACT

UNLABELLED: We reported previously that pre-programming mesenchymal stem cells with the GATA-4 gene increases significantly cell survival in an ischemic environment. In this study, we tested whether regulation of microRNAs and their target proteins was associated with the cytoprotective effects of GATA-4. METHODS AND RESULTS: Mesenchymal stem cells were harvested from adult rat bone marrow and transduced with GATA-4 (MSC(GATA-4)) using the murine stem cell virus retroviral expression system. Cells transfected with empty vector (MSC(Null)) were used as controls. Quantitative real-time PCR data showed that the expression levels of miR-15 family members (miR-15b, miR-16, and miR-195) were significantly down-regulated in MSC(GATA-4). The protein expression of Bcl-w (Bcl-2-like-2), an anti-apoptotic Bcl-2 family protein, was increased in MSC(GATA-4). Hypoxic culture (low glucose and low oxygen) induced the release of lactate dehydrogenase from mesenchymal stem cells and reduced cell survival. Compared to MSC(Null), MSC(GATA-4) showed less lactate dehydrogenase release and greater cell survival following 72 h hypoxia exposure. The mitochondrial membrane potential, detected with the dye JC-1, was well maintained, and mitochondrial membrane permeability, expressed as caspase 3 and 7 activities in response to the ischemic environment was lower in MSC(GATA-4). Moreover, transfection with miR-195 significantly down-regulated Bcl-w expression in mesenchymal stem cells through a binding site in the 3'-UTR of the Bcl-w mRNA and reduced mesenchymal stem cell resistance to ischemic injury. CONCLUSIONS: The overexpression of GATA-4 in mesenchymal stem cells down-regulates miR-15 family members, causing increased resistance to ischemia through the up-regulation of anti-apoptotic proteins in the Bcl-2 family.


Subject(s)
GATA4 Transcription Factor/genetics , Mesenchymal Stem Cells/physiology , MicroRNAs/metabolism , Animals , Cell Survival/physiology , Cells, Cultured , GATA4 Transcription Factor/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/pharmacology , Rats , Rats, Sprague-Dawley , Transduction, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...