Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(757): eadg0338, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39047116

ABSTRACT

Donor organ shortages for transplantation remain a serious global concern, and alternative treatment is in high demand. Fetal cells and tissues have considerable therapeutic potential as, for example, organoid technology that uses human induced pluripotent stem cells (hiPSCs) to generate unlimited human fetal-like cells and tissues. We previously reported the in vivo vascularization of early fetal liver-like hiPSC-derived liver buds (LBs) and subsquent improved survival of recipient mice with subacute liver failure. Here, we show hiPSC-liver organoids (LOs) that recapitulate midgestational fetal liver promote de novo liver generation when grafted onto the surface of host livers in chemical fibrosis models, thereby recovering liver function. We found that fetal liver, a hematopoietic tissue, highly expressed macrophage-recruiting factors and antifibrotic M2 macrophage polarization factors compared with the adult liver, resulting in fibrosis reduction because of CD163+ M2-macrophage polarization. Next, we created midgestational fetal liver-like hiPSC-LOs by fusion of hiPSC-LBs to induce static cell-cell interactions and found that these contained complex structures such as hepatocytes, vasculature, and bile ducts after transplantation. This fusion allowed the generation of a large human tissue suitable for transplantation into immunodeficient rodent models of liver fibrosis. hiPSC-LOs showed superior liver function compared with hiPSC-LBs and improved survival and liver function upon transplantation. In addition, hiPSC-LO transplantation ameliorated chemically induced liver fibrosis, a symptom of liver cirrhosis that leads to organ dysfunction, through immunomodulatory effects, particularly on CD163+ phagocytic M2-macrophage polarization. Together, our results suggest hiPSC-LO transplantation as a promising therapeutic option for liver fibrosis.


Subject(s)
Immunomodulation , Induced Pluripotent Stem Cells , Liver Cirrhosis , Liver , Organoids , Humans , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Animals , Liver/pathology , Macrophages , Liver Transplantation , Mice
2.
Cell Rep ; 43(3): 113918, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38451817

ABSTRACT

Maximizing the potential of human liver organoids (LOs) for modeling human septic liver requires the integration of innate immune cells, particularly resident macrophage Kupffer cells. In this study, we present a strategy to generate LOs containing Kupffer cells (KuLOs) by recapitulating fetal liver hematopoiesis using human induced pluripotent stem cell (hiPSC)-derived erythro-myeloid progenitors (EMPs), the origin of tissue-resident macrophages, and hiPSC-derived LOs. Remarkably, LOs actively promote EMP hematopoiesis toward myeloid and erythroid lineages. Moreover, supplementing with macrophage colony-stimulating factor (M-CSF) proves crucial in sustaining the hematopoietic population during the establishment of KuLOs. Exposing KuLOs to sepsis-like endotoxins leads to significant organoid dysfunction that closely resembles the pathological characteristics of the human septic liver. Furthermore, we observe a notable functional recovery in KuLOs upon endotoxin elimination, which is accelerated by using Toll-like receptor-4-directed endotoxin antagonist. Our study represents a comprehensive framework for integrating hematopoietic cells into organoids, facilitating in-depth investigations into inflammation-mediated liver pathologies.


Subject(s)
Induced Pluripotent Stem Cells , Liver Diseases , Sepsis , Humans , Kupffer Cells , Liver/pathology , Liver Diseases/pathology , Organoids , Sepsis/pathology , Endotoxins , Cell Differentiation
3.
Front Biosci (Landmark Ed) ; 29(1): 34, 2024 01 19.
Article in English | MEDLINE | ID: mdl-38287837

ABSTRACT

Establishing reliable and reproducible animal models for disease modelling, drug screening and the understanding of disease susceptibility and pathogenesis is critical. However, traditional animal models differ significantly from humans in terms of physiology, immune response, and pathogenesis. As a result, it is difficult to translate laboratory findings into biomedical applications. Although several animal models with human chimeric genes, organs or systems have been developed in the past, their limited engraftment rate and physiological functions are a major obstacle to realize convincing models of humans. The lack of human transplantation resources and insufficient immune tolerance of recipient animals are the main challenges that need to be overcome to generate fully humanized animals. Recent advances in gene editing and pluripotent stem cell-based xenotransplantation technologies offer opportunities to create more accessible human-like models for biomedical research. In this article, we have combined our laboratory expertise to summarize humanized animal models, with a focus on hematopoietic/immune system and liver. We discuss their generation strategies and the potential donor cell sources, with particular attention given to human pluripotent stem cells. In particular, we discuss the advantages, limitations and emerging trends in their clinical and pharmaceutical applications. By providing insights into the current state of humanized animal models and their potential for biomedical applications, this article aims to advance the development of more accurate and reliable animal models for disease modeling and drug screening.


Subject(s)
Induced Pluripotent Stem Cells , Animals , Humans , Models, Animal , Transplantation, Heterologous , Disease Models, Animal
4.
Hepatology ; 76(4): 1030-1045, 2022 10.
Article in English | MEDLINE | ID: mdl-35243665

ABSTRACT

BACKGROUND AND AIMS: The loss of liver regenerative capacity is the most dramatic age-associated alteration. Because of an incomplete mechanistic understanding of the liver aging process, a successful therapeutic strategy to improve liver regeneration in the elderly has not been developed so far. Hepatocyte plasticity is a principal mechanism for producing new hepatocytes and cholangiocytes during regeneration. This study aims to promote the repopulation capacity of elderly hepatocytes by decoding the underlying mechanism about the regulation of aging on human hepatocyte plasticity. APPROACH AND RESULTS: To understand the age-related mechanisms, we established a hepatocyte aging model from human-induced pluripotent stem cells and developed a method for ex vivo characterization of hepatocyte plasticity. We found that hepatocyte plasticity was gradually diminished with aging, and the impaired plasticity was caused by age-induced histone hypoacetylation. Notably, selective inhibition of histone deacetylases could markedly restore aging-impaired plasticity. Based on these findings, we successfully improved the plasticity of elderly primary human hepatocytes that enhanced their repopulation capacity in the liver injury model. CONCLUSIONS: This study suggests that age-induced histone hypoacetylation impairs hepatocyte plasticity, and hepatocyte plasticity might be a therapeutic target for promoting the regenerative capacity of the elderly liver.


Subject(s)
Hepatocytes , Histones , Aged , Aging , Histone Deacetylases , Humans , Liver , Liver Regeneration/physiology
5.
Int J Mol Sci ; 22(19)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34638810

ABSTRACT

Liver disease is a global health issue that has caused an economic burden worldwide. Organ transplantation is the only effective therapy for end-stage liver disease; however, it has been hampered by a shortage of donors. Human pluripotent stem cells (hPSCs) have been widely used for studying liver biology and pathology as well as facilitating the development of alternative therapies. hPSCs can differentiate into multiple types of cells, which enables the generation of various models that can be applied to investigate and recapitulate a range of biological activities in vitro. Here, we summarize the recent development of hPSC-derived hepatocytes and their applications in disease modeling, cell therapy, and drug discovery. We also discuss the advantages and limitations of these applications and critical challenges for further development.


Subject(s)
Drug Discovery , Hepatocytes/metabolism , Liver Diseases , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Humans , Liver Diseases/metabolism , Liver Diseases/therapy
6.
Cancers (Basel) ; 13(16)2021 Aug 08.
Article in English | MEDLINE | ID: mdl-34439154

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common form of liver cancer. This study aims to develop a new method to generate an HCC mouse model with a human tumor, and imitates the tumor microenvironment (TME) of clinical patients. Here, we have generated functional, three-dimensional sheet-like human HCC organoids in vitro, using luciferase-expressing Huh7 cells, human iPSC-derived endothelial cells (iPSC-EC), and human iPSC-derived mesenchymal cells (iPSC-MC). The HCC organoid, capped by ultra-purified alginate gel, was implanted into the disrupted liver using an ultrasonic homogenizer in the immune-deficient mouse, which improved the survival and engraftment rate. We successfully introduced different types of controllable TME into the model and studied the roles of TME in HCC tumor growth. The results showed the role of the iPSC-EC and iPSC-MC combination, especially the iPSC-MC, in promoting HCC growth. We also demonstrated that liver fibrosis could promote HCC tumor growth. However, it is not affected by non-alcoholic fatty liver disease. Furthermore, the implantation of HCC organoids to humanized mice demonstrated that the immune response is important in slowing down tumor growth at an early stage. In conclusion, we have created an HCC model that is useful for studying HCC development and developing new treatment options in the future.

7.
EBioMedicine ; 35: 114-123, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30120080

ABSTRACT

Therapies against hepatitis B virus (HBV) have improved in recent decades; however, the development of individualized treatments has been limited by the lack of individualized infection models. In this study, we used human induced pluripotent stem cell (hiPSC) to generate a functional liver organoid (LO) that inherited the genetic background of the donor, and evaluated its application in modeling HBV infection and exploring virus-host interactions. To establish a functional hiPSC-LO, we cultured hiPSC-derived endodermal, mesenchymal, and endothelial cells with a chemically defined medium in a three-dimensional microwell culture system. Based on cell-cell interactions, these cells could organize themselves and gradually differentiate into a functional organoid, which exhibited stronger hepatic functions than hiPSC derived hepatic like cell (HLC). Moreover, the functional LO demonstrated more susceptibility to HBV infection than hiPSC-HLC, and could maintain HBV propagation and produce infectious virus for a prolonged duration. Furthermore, we found that virus infection could cause hepatic dysfunction of hiPSC-LOs, with down-regulation of hepatic gene expression, induced release of early acute liver failure markers, and altered hepatic ultrastructure. Therefore, our study demonstrated that HBV infection in hiPSC-LOs could recapitulate virus life cycle and virus induced hepatic dysfunction, suggesting that hiPSC-LOs may provide a promising individualized infection model for the development of individualized treatment for hepatitis.


Subject(s)
Hepatitis B virus/physiology , Host-Pathogen Interactions , Induced Pluripotent Stem Cells/virology , Liver/virology , Organoids/virology , Cell Line , Hepatitis B/pathology , Hepatitis B/virology , Humans , Induced Pluripotent Stem Cells/ultrastructure , Liver/pathology , Liver/physiopathology , Organoids/ultrastructure
8.
Stem Cell Res Ther ; 9(1): 29, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29402311

ABSTRACT

BACKGROUND: Mature human hepatocytes are critical in preclinical research and therapy for liver disease, but are difficult to manipulate and expand in vitro. Hepatic stem cells (HpSCs) may be an alternative source of functional hepatocytes for cell therapy and disease modeling. Since these cells play an import role in regenerative medicine, the precise characterization that determines specific markers used to isolate these cells as well as whether they contribute to liver regeneration still remain to be shown. METHOD: In this study, human HpSCs were isolated from human primary fetal liver cells (FLCs) by flow cytometry using CDCP1, CD90, and CD66 antibodies. The isolated CDCP1+CD90+CD66- HpSCs were cultured on dishes coated with type IV collagen in DMEM nutrient mixture F-12 Ham supplemented with FBS, human γ-insulin, nicotinamide, dexamethasone, and L-glutamine for at least 2 weeks, and were characterized by transcriptomic profiling, quantitative real-time PCR, immunocytochemistry, and in-vivo transplantation. RESULTS: The purified CDCP1+CD90+CD66- subpopulation exhibited clonal expansion and self-renewal capability, and bipotential capacity was further identified in single cell-derived colonies containing distinct hepatocytes and cholangiocytes. Moreover, in-vivo liver repopulation assays demonstrated that human CDCP1+CD90+CD66- HpSCs repopulated over 90% of the mouse liver and differentiated into functional hepatocytes with drug metabolism activity. CONCLUSIONS: We identified a human hepatic stem/progenitor population in the CDCP1+CD90+CD66- subpopulation in human FLCs, indicating CDCP1 marker could potentially be utilized to identify and isolate HpSCs for further cytotherapy of liver disease.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Fetus/metabolism , Liver/metabolism , Neoplasm Proteins/metabolism , Stem Cells/metabolism , Antigens, Neoplasm , Cell Culture Techniques , Cells, Cultured , Fetus/cytology , Humans , Liver/cytology , Stem Cells/cytology
9.
Stem Cell Res Ther ; 9(1): 5, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321049

ABSTRACT

BACKGROUND: Acute liver failure (ALF) is a life-threatening disease with a high mortality rate. However, there are limited treatments or devices available for ALF therapy. Here, we aimed to develop a new strategy for ALF treatment by transplanting functional liver organoids (LOs) generated from single donor-derived human induced pluripotent stem cell (hiPSC) endoderm, endothelial cells (ECs), and mesenchymal cells (MCs). METHODS: First, we isolated ECs and MCs from a single donor umbilical cord (UC) through enzyme digestion and characterized the UC-ECs and UC-MCs by flow cytometry. Second, using a nonviral reprogramming method, we generated same donor-derived hiPSCs from the UC-ECs and investigated their hepatic differentiation abilities. Finally, we simultaneously plated EC-hiPSC endoderm, UC-ECs, and UC-MCs in a three-dimensional (3D) microwell culture system, and generated single donor cell-derived differentiated LOs for ALF mouse treatment. RESULTS: We obtained ECs and MCs from a single donor UC with high purity, and these cells provided a multicellular microenvironment that promoted LO differentiation. hiPSCs from the same donor were generated from UC-ECs, and the resultant EC-hiPSCs could be differentiated efficiently into pure definitive endoderm and further into hepatic lineages. Simultaneous plating of EC-hiPSC endoderm, UC-ECs, and UC-MCs in the 3D microwell system generated single donor cell-derived LOs (SDC-LOs) that could be differentiated into functional LOs with enhanced hepatic capacity as compared to that of EC-hiPSC-derived hepatic-like cells. When these functional SDC-LOs were transplanted into the renal subcapsules of ALF mice, they rapidly assumed hepatic functions and improved the survival rate of ALF mice. CONCLUSION: These results demonstrate that functional LOs generated from single donor cells can improve the condition of ALF mice. Functional SDC-LO transplantation provides a promising novel approach for ALF therapy.


Subject(s)
Endothelial Cells/transplantation , Liver Failure, Acute/therapy , Liver Regeneration/physiology , Liver/pathology , Mesenchymal Stem Cell Transplantation , Organoids/cytology , Pluripotent Stem Cells/transplantation , Animals , Cell Differentiation , Cells, Cultured , Endothelial Cells/cytology , Humans , Liver/cytology , Mesenchymal Stem Cells/cytology , Mice , Pluripotent Stem Cells/cytology , Umbilical Cord/cytology
10.
Stem Cell Res Ther ; 6: 71, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25889203

ABSTRACT

INTRODUCTION: Previous studies have produced controversial results regarding whether mesenchymal stem cells (MSCs) promote or inhibit tumor development. Given the dual role of MSCs in inflammation and cancer, in this study the colitis-associated colorectal cancer (CAC) model was used to examine whether umbilical cord tissue-derived MSCs could prevent neoplasm by inhibiting chronic inflammation. METHODS: MSCs were obtained and identified using flow cytometry. Colitis-associated colorectal cancer model was induced using azoxymethane (AOM) and dextran sulfate sodium (DSS) and MSCs were injected intravenously twice. Levels of immune cells in mesenteric lymph node including regulatory T (Treg) cells were detected using flow cytometry. Naïve T cells and Jurkat cells were co-cultured with MSCs and the effect of MSCs on Treg cells differentiation was evaluated. RESULTS: After injection through tail vein, MSCs could migrate to colon and suppress colitis-related neoplasm. This tumor suppressive effect was characterized by longer colon length, decreased tumor numbers and decreased expression of Ki-67. Moreover, MSCs alleviated the pathology of inflammation in the colitis stage of CAC model and inhibited inflammation cytokines both in colon and serum. Furthermore, Treg cells were accumulated in mesenteric lymph node of MSCs-treated mice while the percentage of T helper cells 2 (Th2) and Th17 were not changed. Of note, MSCs secreted transforming growth factor-ß (TGF-ß) enhanced the induction of Treg cells from naïve T cells. The conditioned medium of MSCs also activated Smad2 signaling, which has been reported to regulate Treg cells. CONCLUSIONS: These results proved that MSCs could migrate to colon tissues and induce the differentiation of Treg cells via Smad2 as so to inhibit the colitis and suppress the development of CAC.


Subject(s)
Colitis/pathology , Colorectal Neoplasms/prevention & control , Mesenchymal Stem Cells/metabolism , Smad2 Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Azoxymethane , CD4 Lymphocyte Count , Cell Differentiation , Cell Line , Cell Movement , Cell- and Tissue-Based Therapy/methods , Colitis/chemically induced , Colitis/immunology , Colon/cytology , Colon/immunology , Colon/pathology , Colorectal Neoplasms/pathology , Cytokines/blood , Dextran Sulfate , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Jurkat Cells , Lymphocyte Activation/immunology , Lymphocyte Count , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes, Regulatory/cytology , Th17 Cells/immunology , Th2 Cells/immunology , Umbilical Cord/cytology
11.
Stem Cell Res Ther ; 6: 49, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25889844

ABSTRACT

INTRODUCTION: Chimeric mice with humanized livers were recently established by transplanting human hepatocytes. This mouse model that is repopulated with functional human hepatocytes could be a useful tool for investigating human hepatic cell biology and drug metabolism and for other preclinical applications. Successfully transplanting human hepatocytes into mice requires that recipient mice with liver failure do not reject these human cells and provide a suitable microenvironment (supportive niche) to promote human donor cell expansion and differentiation. To overcome the limitations of current mouse models, we used Alb-TRECK/SCID mice for in vivo human immature hepatocyte differentiation and humanized liver generation. METHODS: 1.5 µg/kg diphtheria toxin was administrated into 8-week-old Alb-TRECK/SCID mice, and the degree of liver damage was assessed by serum aspartate aminotransferase activity levels. Forty-eight hours later, mice livers were sampled for histological analyses, and the human donor cells were then transplanted into mice livers on the same day. Chimeric rate and survival rate after cell transplantation was evaluated. Expressions of human hepatic-related genes were detected. A human albumin enzyme-linked immunosorbent assay was performed after 50 days of transplantation. On day 60 after transplantation, drug metabolism was examined in mice. RESULTS: Both human primary fetal liver cells and hepatic stem cells were successfully repopulated in the livers of Alb-TRECK/SCID mice that developed lethal fulminant hepatic failure after administering diphtheria toxin; the repopulation rate in some mice was nearly 100%. Compared with human primary fetal liver cells, human hepatic stem cell transplantation rescued Alb-TRECK/SCID mice with lethal fulminant hepatic failure, and human hepatic stem cell-derived humanized livers secreted more human albumin into mouse sera and also functioned as a "human liver" that could metabolize the drugs ketoprofen and debrisoquine. CONCLUSION: Our model of a humanized liver in Alb-TRECK/SCID mice may provide for functional applications such as drug metabolism, drug to drug interactions, and promote other in vivo and in vitro studies.


Subject(s)
Hepatocytes/transplantation , Inactivation, Metabolic/physiology , Liver Failure, Acute/therapy , Liver Regeneration/physiology , Stem Cell Transplantation/methods , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chimera , Debrisoquin/metabolism , Diphtheria Toxin/administration & dosage , Disease Models, Animal , Hepatocytes/cytology , Humans , Ketoprofen/metabolism , Liver/cytology , Liver Failure, Acute/chemically induced , Mice , Mice, Knockout , Mice, SCID , Stem Cells/metabolism , Transplantation, Heterologous
12.
Stem Cell Res Ther ; 6: 51, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25881300

ABSTRACT

INTRODUCTION: The therapeutic potential of acyclic retinoid (ACR), a synthetic retinoid, has been confirmed in experimental and clinical studies. Therapeutic targets include precancerous and cancer stem cells. As ACR is also involved in developmental processes, its effect on normal hepatic stem cells (HpSCs) should be investigated for understanding the underlying mechanisms. Here, we examined effects of the acyclic retinoid peretinoin on fresh isolated murine HpSCs. METHODS: We isolated c-kit-CD29+CD49f+/lowCD45-Ter119- cells from murine fetal livers using flow cytometry. To evaluate the effect of ACR, we traced clonal expansion and analyzed cell differentiation as well as apoptosis during the induction process by immunofluorescent staining and marker gene expression. RESULTS: ACR dose-dependently inhibited HpSCs expansion. Stem cell clonal expansion was markedly inhibited during the culture period. Moreover, ACR showed a significant promotion of HpSC differentiation and induction of cellular apoptosis. The expression of stem cell marker genes, Afp, Cd44, and Dlk, was downregulated, while that of mature hepatocyte genes, Alb and Tat, and apoptosis-related genes, Annexin V and Caspase-3, were upregulated. Flow cytometry showed that the proportion of Annexin V-positive cells increased after ACR incubation compared with the control. Data obtained by immunofluorescent staining for albumin and Caspase-3 corroborated the data on gene expression. Finally, we found that ACR directly regulates the expression of retinoic acid receptors and retinoid X receptors. CONCLUSIONS: These findings indicate that ACR inhibits the clonal expansion of normal HpSCs in vitro and promotes the differentiation of immature cells by regulating receptors of retinoic acid.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Liver Regeneration/physiology , Liver/cytology , Tretinoin/analogs & derivatives , Animals , Annexin A5/biosynthesis , Calcium-Binding Proteins , Caspase 3/biosynthesis , Cells, Cultured , Down-Regulation , Flow Cytometry , Gene Products, tat/biosynthesis , Hyaluronan Receptors/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Receptors, Retinoic Acid/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tretinoin/pharmacology , Up-Regulation , alpha-Fetoproteins/biosynthesis
13.
Mol Cancer Ther ; 14(2): 491-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25670331

ABSTRACT

Lung cancer is the deadliest type of cancer for both men and women. In this study, we evaluate the in vitro and in vivo efficacy of a biotherapeutic agent composed of a lysosomal protein (Saposin C, SapC) and a phospholipid (dioleoylphosphatidylserine, DOPS), which can be assembled into nanovesicles (SapC-DOPS) with selective antitumor activity. SapC-DOPS targets phosphatidylserine, an anionic phospholipid preferentially exposed in the surface of cancer cells and tumor-associated vasculature. Because binding of SapC to phosphatidylserine is favored at acidic pHs, and the latter characterizes the milieu of many solid tumors, we tested the effect of pH on the binding capacity of SapC-DOPS to lung tumor cells. Results showed that SapC-DOPS binding to cancer cells was more pronounced at low pH. Viability assays on a panel of human lung tumor cells showed that SapC-DOPS cytotoxicity was positively correlated with cell surface phosphatidylserine levels, whereas mitochondrial membrane potential measurements were consistent with apoptosis-related cell death. Using a fluorescence tracking method in live mice, we show that SapC-DOPS specifically targets human lung cancer xenografts, and that systemic therapy with SapC-DOPS induces tumor apoptosis and significantly inhibits tumor growth. These results suggest that SapC-DOPS nanovesicles are a promising treatment option for lung cancer.


Subject(s)
Lung Neoplasms/drug therapy , Molecular Targeted Therapy , Nanostructures/chemistry , Phosphatidylserines/chemistry , Saposins/therapeutic use , Unilamellar Liposomes/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Mice, Nude , Saposins/pharmacology , Xenograft Model Antitumor Assays
14.
Cell Mol Immunol ; 12(6): 768-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25363527

ABSTRACT

Estrogen is involved in promoting lung cancer cell division and metastasis. MICA and MICB function as ligands for NKG2D, an important immunoreceptor expressed on natural killer (NK) cells. However, whether estrogen regulates MICA/B expression and affects tumor immune escape remains unknown. In this study, we measured the mRNA levels of MICA, MICB and ADAM17in non-small cell lung cancer (NSCLC) cell lines treated with estrogen. Surface expression of MICA/B on LTEP-a2 and A549 was detected using flow cytometry. We demonstrate that both mRNA and secretory protein levels of MICA/B in lung adenocarcinoma cell lines were upregulated by estradiol. Estradiol enhanced the expression of ADAM17, which was associated with the secretion of MICA/B. This secretion of MICA/B downregulated the NKG2D receptor on the surface of NK92 cells and impaired the cytotoxic activity of NK cells. Estradiol enhanced the expression of ADAM17, which was associated with the secretion of MICA/B. Furthermore, a significant correlation between the concentration of estradiol and the expression of MICA was found in tumor tissues of NSCLC patients. Therefore, we conclude that estrogen can regulate the expression and secretion of MICA/B through ADAM17, which helps lung cancer cells escape NKG2D-mediated immune surveillance.


Subject(s)
ADAM Proteins/genetics , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Lung Neoplasms/genetics , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/immunology , ADAM17 Protein , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Female , Histocompatibility Antigens Class I/immunology , Humans , Immunologic Surveillance , Killer Cells, Natural , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasm Staging , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tumor Escape
15.
Cell Mol Immunol ; 11(5): 495-502, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24813230

ABSTRACT

MicroRNAs (miRNAs) are a class of small non-coding regulatory RNAs, and changes in miRNAs are involved in tumor origin and progression. Studies have shown that miR-20a is overexpressed in human ovarian cancer tissues and that this miRNA enhances long-term cellular proliferation and invasion capabilities. In this study, a positive correlation between serum miR-20a expression and ovarian cancer stage was observed. We found that miR-20a binds directly to the 3'-untranslated region of MICA/B mRNA, resulting in its degradation and reducing its protein levels on the plasma membrane. Reduction of membrane-bound MICA/B proteins, which are ligands of the natural killer group 2 member D (NKG2D) receptor found on natural killer (NK) cells, γδ(+) T cells and CD8(+) T cells, allows tumor cells to evade immune-mediated killing. Notably, antagonizing miR-20a action enhanced the NKG2D-mediated killing of tumor cells in both in vitro and in vivo models of tumors. Taken together, our data indicate that increased levels of miR-20a in tumor cells may indirectly suppress NK cell cytotoxicity by downregulating MICA/B expression. These data provide a potential link between metastasis capability and immune escape of tumor cells from NK cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma/diagnosis , Carcinoma/immunology , Killer Cells, Natural/immunology , MicroRNAs/biosynthesis , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/immunology , 3' Untranslated Regions/genetics , Carcinoma/mortality , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Down-Regulation , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , MicroRNAs/blood , MicroRNAs/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasm Staging , Ovarian Neoplasms/mortality , Prognosis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Survival Analysis , Tumor Escape/genetics
16.
BMC Cancer ; 13: 582, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24314291

ABSTRACT

BACKGROUND: We previously found that the low frequency magnetic fields (LF-MF) inhibited gastric and lung cancer cell growth. We suppose that exposure to LF-MF may modulate immune function so as to inhibit tumor. We here investigated whether LF-MF can inhibit the proliferation and metastasis of melanoma and influence immune function. METHODS: The effect of MF on the proliferation, cell cycle and ultrastracture of B16-F10 in vitro was detected by cell counting Kit-8 assay, flow cytometry, and transmission electron microscopy. Lung metastasis mice were prepared by injection of 2 × 105 B16-F10 melanoma cells into the tail vein in C57BL/6 mice. The mice were then exposed to an LF-MF (0.4 T, 7.5 Hz) for 43 days. Survival rate, tumor markers and the innate and adaptive immune parameters were measured. RESULTS: The growth of B16-F10 cells was inhibited after exposure to the LF-MF. The inhibition was related to induction of cell cycle arrest and decomposition of chromatins. Moreover, the LF-MF prolonged the mouse survival rate and inhibited the proliferation of B16-F10 in melanoma metastasis mice model. Furthermore, the LF-MF modulated the immune response via regulation of immune cells and cytokine production. In addition, the number of Treg cells was decreased in mice with the LF-MF exposure, while the numbers of T cells as well as dendritic cells were significantly increased. CONCLUSION: LF-MF inhibited the growth and metastasis of melanoma cancer cells and improved immune function of tumor-bearing mice. This suggests that the inhibition may be attributed to modulation of LF-MF on immune function and LF-MF may be a potential therapy for treatment of melanoma.


Subject(s)
Magnetic Fields , Melanoma/therapy , Animals , Apoptosis/radiation effects , CD40 Antigens/metabolism , Cell Cycle Checkpoints/radiation effects , Cell Differentiation/immunology , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cytokines/blood , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/radiation effects , Female , Inflammation Mediators/blood , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Melanoma/immunology , Melanoma/mortality , Melanoma/pathology , Melanoma, Experimental , Mice , Spleen/immunology , Spleen/metabolism , Spleen/radiation effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/radiation effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects , Tumor Burden/radiation effects
17.
PLoS One ; 8(11): e72411, 2013.
Article in English | MEDLINE | ID: mdl-24278103

ABSTRACT

OBJECTIVE: Many studies have shown that magnetic fields (MF) inhibit tumor growth and influence the function of immune system. However, the effect of MF on mechanism of immunological function in tumor-bearing mice is still unclear. METHODS: In this study, tumor-bearing mice were prepared by subcutaneously inoculating Balb/c mice with hepatocarcinoma cell line H22. The mice were then exposed to a low frequency MF (0.4 T, 7.5 Hz) for 30 days. Survival rate, tumor growth and the innate and adaptive immune parameters were measured. RESULTS: MF treatment could prolong survival time (n = 28, p<0.05) and inhibit tumor growth (n = 9, p<0.01) in tumor-bearing mice. Moreover, this MF suppressed tumor-induced production of cytokines including interleukin-6 (IL-6), granulocyte colony- stimulating factor (G-CSF) and keratinocyte-derived chemokine (KC) (n = 9-10, p<0.05 or 0.01). Furthermore, MF exposure was associated with activation of macrophages and dendritic cells, enhanced profiles of CD4(+) T and CD8(+) T lymphocytes, the balance of Th17/Treg and reduced inhibitory function of Treg cells (n = 9-10, p<0.05 or 0.01) in the mice model. CONCLUSION: The inhibitory effect of MF on tumor growth was related to the improvement of immune function in the tumor-bearing mice.


Subject(s)
Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Magnetic Fields , Adaptive Immunity/physiology , Animals , Carcinoma, Hepatocellular/blood , Cell Line, Tumor , Cell Survival/physiology , Cytokines/blood , Female , Flow Cytometry , Immunity, Innate/physiology , Immunohistochemistry , Liver Neoplasms/blood , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , T-Lymphocytes/immunology
18.
World J Gastroenterol ; 19(47): 8850-60, 2013 Dec 21.
Article in English | MEDLINE | ID: mdl-24379607

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common cancers, and is also the leading cause of death worldwide. Studies have shown that cellular reprogramming contributes to chemotherapy and/or radiotherapy resistance and the recurrence of cancers. In this article, we summarize and discuss the latest findings in the area of cellular reprogramming in HCC. The aberrant expression of transcription factors OCT4, KLF4, SOX2, c-MYC, NANOG, and LIN28 have been also observed, and the expression of these transcription factors is associated with unfavorable clinical outcomes in HCC. Studies indicate that cellular reprogramming may play a critical role in the occurrence and recurrence of HCC. Recent reports have shown that DNA methylation, miRNAs, tumor microenvironment, and signaling pathways can induce the expression of stemness transcription factors, which leads to cellular reprogramming in HCC. Furthermore, studies indicate that therapies based on cellular reprogramming could revolutionize HCC treatment. Finally, a novel therapeutic concept is discussed: reprogramming control therapy. A potential reprogramming control therapy method could be developed based on the reprogramming demonstrated in HCC studies and applied at two opposing levels: differentiation and reprogramming. Our increasing understanding and control of cellular programming should facilitate the exploitation of this novel therapeutic concept and its application in clinical HCC treatment, which may represent a promising strategy in the future that is not restricted to liver cancer.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , Cellular Reprogramming , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Genetic Therapy/methods , Humans , Kruppel-Like Factor 4 , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Mol Endocrinol ; 26(8): 1304-15, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22669742

ABSTRACT

Epithelial-mesenchymal transition (EMT) plays a specific role in the migration of tumor cells. Both estrogen and midkine (MK) have been thought to be important factors in promoting the progression of non-small-cell lung cancer (NSCLC) and can enhance EMT. Some evidence indicated the correlation between estradiol (E2) and MK, but the precise mechanism on their interreaction is unknown. Here, we try to clarify whether and how E2 regulates MK expression to promote EMT. We found that E2 increased MK mRNA expression in lung adenocarcinoma cells LTEP-a2 and A549 in a time-dependent manner. E2-induced MK expression was inhibited by the estrogen receptor (ER) antagonist ICI 182,780 and tamoxifen but not by phosphoinositide-3 kinase and MAPK inhibitors, suggesting a genomic mechanism of E2 on the regulation of MK transcription. Moreover, luciferase reporter and chromatin immunoprecipitation assays exhibited that E2 induced ERß recruitment to the estrogen response element in the MK promoter. Small interfering RNA to ERα and ERß revealed that ERß mainly mediated E2-induced MK transcription. Interestingly, E2 enhanced MK expression in accordance with increase of EMT, whereas knockdown of MK could block EMT under E2 stimulation. Importantly, through analyzing lung adenocarcinoma tissues, there was indeed a correlation among levels of E2, MK, and EMT-related protein expression. Taken together, we reported a previously unrecognized mechanism on E2 in the regulation of MK expression and proved that MK plays a pivotal role in progression of E2-regulated EMT.


Subject(s)
Adenocarcinoma/pathology , Epithelial-Mesenchymal Transition , Estradiol/physiology , Estrogen Receptor beta/metabolism , Lung Neoplasms/pathology , Nerve Growth Factors/genetics , Transcription, Genetic , Adenocarcinoma/blood , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Adult , Aged , Base Sequence , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Estradiol/blood , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Male , Middle Aged , Midkine , Nerve Growth Factors/blood , Nerve Growth Factors/metabolism , Nitriles/pharmacology , Phenols , Propionates/pharmacology , Protein Binding , Pyrazoles/pharmacology , RNA Interference , Response Elements , Signal Transduction , Statistics, Nonparametric
20.
Cancer Immunol Immunother ; 61(10): 1745-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22415659

ABSTRACT

Midkine (MK) is a heparin-binding growth factor overexpressed in various human cancers. In the current study, a positive correlation was observed between MK expression and MICA/B serum levels of gastric cancer patients. In addition, MK transfection significantly increased MICA/B expression in gastric cancer cells. The soluble MICA/B expression was also elevated. Furthermore, MK transfection inhibited CD107a and Granzyme B expression, thereby suppressing the natural killer (NK) cell cytotoxicity in vitro. The phosphorylation of p38 MAPK and its promotion of CHOP expression were also observed after MK treatment and transfection. CHOP was indirectly bound to the MICA/B promoter region by interacting with AP-1, leading to MICA/B transcription. Overall, the current study shows that MK expression in tumor cells indirectly suppresses NK cytotoxicity by inducing MICA/B expression and suppressing NKG2D expression.


Subject(s)
Adenocarcinoma/metabolism , Histocompatibility Antigens Class I/biosynthesis , Killer Cells, Natural/immunology , Nerve Growth Factors/metabolism , Stomach Neoplasms/metabolism , Adenocarcinoma/blood , Adenocarcinoma/genetics , Cell Line, Tumor , Female , Granzymes/biosynthesis , Histocompatibility Antigens Class I/blood , Histocompatibility Antigens Class I/genetics , Humans , Killer Cells, Natural/metabolism , Lysosomal-Associated Membrane Protein 1/biosynthesis , Male , Middle Aged , Midkine , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Nerve Growth Factors/blood , Nerve Growth Factors/genetics , Phosphorylation , Promoter Regions, Genetic , Stomach Neoplasms/blood , Stomach Neoplasms/genetics , Transcription Factor AP-1/chemistry , Transcription Factor AP-1/metabolism , Transcription Factor CHOP/biosynthesis , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL