Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Exp Mol Med ; 56(1): 19-31, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38172594

ABSTRACT

Cancer immunotherapy has revolutionized the approach to cancer treatment of malignant tumors by harnessing the body's immune system to selectively target cancer cells. Despite remarkable advances, there are still challenges in achieving successful clinical responses. Recent evidence suggests that immune cell-derived exosomes modulate the immune system to generate effective antitumor immune responses, making them a cutting-edge therapeutic strategy. However, natural exosomes are limited in clinical application due to their low drug delivery efficiency and insufficient antitumor capacity. Technological advancements have allowed exosome modifications to magnify their intrinsic functions, load different therapeutic cargoes, and preferentially target tumor sites. These engineered exosomes exert potent antitumor effects and have great potential for cancer immunotherapy. In this review, we describe ingenious modification strategies to attain the desired performance. Moreover, we systematically summarize the tumor-controlling properties of engineered immune cell-derived exosomes in innate and adaptive immunity. Collectively, this review provides a comprehensive and intuitive guide for harnessing the potential of modified immune cell-derived exosome-based approaches, offering valuable strategies to enhance and optimize cancer immunotherapy.


Subject(s)
Exosomes , Neoplasms , Humans , Exosomes/pathology , Immunotherapy , Neoplasms/pathology , Adaptive Immunity , Immune System
2.
Mol Med Rep ; 28(5)2023 11.
Article in English | MEDLINE | ID: mdl-37732549

ABSTRACT

Wear particle­induced osteolysis is a serious complication that occurs in individuals with titanium (Ti)­based implants following long­term usage due to loosening of the implants. The control of excessive osteoclast differentiation and inflammation is essential for protecting against wear particle­induced osteolysis. The present study evaluated the effect of britanin, a pseudoguaianolide sesquiterpene isolated from Inula japonica, on osteoclastogenesis in vitro and Ti particle­induced osteolysis in vivo. The effect of britanin was examined in the osteoclastogenesis of mouse bone marrow­derived macrophages (BMMs) using TRAP staining, RT­PCR, western blotting and immunocytochemistry. The protective effect of britanin was examined in a mouse calvarial osteolysis model and evaluated using micro­CT and histomorphometry. Britanin inhibited osteoclast differentiation and F­actin ring formation in the presence of macrophage colony­stimulating factor and receptor activator of nuclear factor kB ligand in BMMs. The expression of osteoclast­specific marker genes, including tartrate­resistant acid phosphatase, cathepsin K, dendritic cell­specific transmembrane protein, matrix metallopeptidase 9 and nuclear factor of activated T­cells cytoplasmic 1, in the BMMs was significantly reduced by britanin. In addition, britanin reduced the expression of B lymphocyte­induced maturation protein­1, which is a transcriptional repressor of negative osteoclastogenesis regulators, including interferon regulatory factor­8 and B­cell lymphoma 6. Conversely, britanin increased the expression levels of anti­oxidative stress genes, namely nuclear factor erythroid­2­related factor 2, NAD(P)H quinone oxidoreductase 1 and heme oxygenase 1 in the BMMs. Furthermore, the administration of britanin significantly reduced osteolysis in a Ti particle­induced calvarial osteolysis mouse model. Based on these findings, it is suggested that britanin may be a potential therapeutic agent for wear particle­induced osteolysis and osteoclast­associated disease.


Subject(s)
Osteogenesis , Osteolysis , Humans , Animals , Mice , Osteolysis/drug therapy , Osteolysis/etiology , Titanium/adverse effects , Osteoclasts , Actin Cytoskeleton , Disease Models, Animal
3.
Proc Natl Acad Sci U S A ; 120(33): e2300036120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37549292

ABSTRACT

While the world is rapidly transforming into a superaging society, pharmaceutical approaches to treat sarcopenia have hitherto not been successful due to their insufficient efficacy and failure to specifically target skeletal muscle cells (skMCs). Although electrical stimulation (ES) is emerging as an alternative intervention, its efficacy toward treating sarcopenia remains unexplored. In this study, we demonstrate a silver electroceutical technology with the potential to treat sarcopenia. First, we developed a high-throughput ES screening platform that can simultaneously stimulate 15 independent conditions, while utilizing only a small number of human-derived primary aged/young skMCs (hAskMC/hYskMC). The in vitro screening showed that specific ES conditions induced hypertrophy and rejuvenation in hAskMCs, and the optimal ES frequency in hAskMCs was different from that in hYskMCs. When applied to aged mice in vivo, specific ES conditions improved the prevalence and thickness of Type IIA fibers, along with biomechanical attributes, toward a younger skMC phenotype. This study is expected to pave the way toward an electroceutical treatment for sarcopenia with minimal side effects and help realize personalized bioelectronic medicine.


Subject(s)
Sarcopenia , Animals , Humans , Mice , Muscle Fibers, Skeletal , Muscle, Skeletal/physiology , Phenotype , Sarcopenia/therapy , Silver
4.
Cytokine ; 169: 156249, 2023 09.
Article in English | MEDLINE | ID: mdl-37290276

ABSTRACT

Lung cancer is a common and highly malignant tumor. Although lung cancer treatments continue to advance, conventional therapies are limited and the response rate of patients to immuno-oncology drugs is low. This phenomenon raises an urgent need to develop effective therapeutic strategies for lung cancer. In this study, we genetically modified human primary CD8+ T cells and obtained antitumor extracellular vesicles (EVs) from them. The engineered EVs, containing interlekin-2 and the anti-epidermal growth factor receptor (EGFR) antibody cetuximab on their surfaces, exhibited direct cytotoxicity against A549 human lung cancer cells and increased cancer cell susceptibility to human peripheral blood mononuclear cell-mediated cytotoxicity. In addition, the engineered EVs specifically targeted the lung cancer cells in an EGFR-dependent manner. Taken together, these findings show that surface engineering of cytokines and antibodies on CD8+ T cell-derived EVs not only enhances their antitumor effects but also confers target specificity, suggesting a potential of modifying the immune cell-derived EVs in cancer treatment.


Subject(s)
Extracellular Vesicles , Lung Neoplasms , Humans , CD8-Positive T-Lymphocytes , Leukocytes, Mononuclear/metabolism , Cell Line, Tumor , Lung Neoplasms/therapy , Lung Neoplasms/metabolism , ErbB Receptors/metabolism , Extracellular Vesicles/metabolism
5.
BMC Cancer ; 23(1): 490, 2023 May 31.
Article in English | MEDLINE | ID: mdl-37259024

ABSTRACT

BACKGROUND: Thrombocytopenia is a common complication in cancer patients undergoing chemotherapy. Chemotherapy-induced thrombocytopenia (CIT) leads to dose reduction and treatment delays, lowering chemotherapy efficacy and survival rate. Thus, rapid recovery and continuous maintenance of platelet count during chemotherapy cycles are crucial in patients with CIT. Thrombopoietin (TPO) and its receptor, myeloid proliferative leukemia (MPL) protein, play a major role in platelet production. Although several MPL agonists have been developed to regulate thrombopoiesis, none have been approved for the management of CIT due to concerns regarding efficacy or safety. Therefore, the development of effective MPL agonists for treating CIT needs to be further expanded. METHODS: Anti-MPL antibodies were selected from the human combinatorial antibody phage libraries using phage display. We identified 2R13 as the most active clone among the binding antibodies via cell proliferation assay using BaF3/MPL cells. The effect of 2R13 on megakaryocyte differentiation was evaluated in peripheral blood CD34+ cells by analyzing megakaryocyte-specific differentiation markers (CD41a+ and CD42b+) and DNA ploidy using flow cytometry. The 2R13-induced platelet production was examined in 8- to 10-week-old wild-type BALB/c female mice and a thrombocytopenia mouse model established by intraperitoneal injection of 5-fluorouracil (150 mg/kg). The platelet counts were monitored twice a week over 14 days post-initiation of treatment with a single injection of 2R13, or recombinant human TPO (rhTPO) for seven consecutive days. RESULTS: We found that 2R13 specifically interacted with MPL and activated its signaling pathways. 2R13 stimulated megakaryocyte differentiation, evidenced by increasing the proportion of high-ploidy (≥ 8N) megakaryocytes in peripheral blood-CD34+ cells. The platelet count was increased by a single injection of 2R13 for up to 14 days. Injection of 5-fluorouracil considerably reduced the platelet count by day 4, which was recovered by 2R13. The platelets produced by 2R13 sustained a higher count than that achieved using seven consecutive injections of rhTPO. CONCLUSIONS: Our findings suggest that 2R13 is a promising therapeutic agent for CIT treatment.


Subject(s)
Antineoplastic Agents , Thrombocytopenia , Mice , Animals , Humans , Female , Receptors, Thrombopoietin , Blood Platelets/metabolism , Thrombopoiesis , Antibodies , Recombinant Proteins/adverse effects , Antigens, CD34 , Fluorouracil/therapeutic use , Thrombocytopenia/chemically induced , Thrombocytopenia/drug therapy , Antineoplastic Agents/adverse effects
6.
J Cachexia Sarcopenia Muscle ; 14(3): 1441-1453, 2023 06.
Article in English | MEDLINE | ID: mdl-37017344

ABSTRACT

BACKGROUND: Patients with cancer undergoing chemotherapy experience cachexia with anorexia, body weight loss, and the depletion of skeletal muscles and adipose tissues. Effective treatment strategies for chemotherapy-induced cachexia are scarce. The growth differentiation factor 15 (GDF15)/GDNF family receptor alpha-like (GFRAL)/rearranged during transfection (RET) axis is a critical signalling pathway in chemotherapy-induced cachexia. In this study, we developed a fully human GFRAL antagonist antibody and investigated whether it inhibits the GDF15/GFRAL/RET axis, thereby alleviating chemotherapy-induced cachexia in tumour-bearing mice. METHODS: Anti-GFRAL antibodies were selected via biopanning, using a human combinatorial antibody phage library. The potent GFRAL antagonist antibody A11 was selected via a reporter cell assay and its inhibitory activity of GDF15-induced signalling was evaluated using western blotting. To investigate the in vivo function of A11, a tumour-bearing mouse model was established by inoculating 8-week-old male C57BL/6 mice with B16F10 cells (n = 10-16 mice per group). A11 was administered subcutaneously (10 mg/kg) 1 day before intraperitoneal treatment with cisplatin (10 mg/kg). Animals were assessed for changes in food intake, body weight, and tumour volume. Plasma and key metabolic tissues such as skeletal muscles and adipose tissues were collected for protein and mRNA expression analysis. RESULTS: A11 reduced serum response element-luciferase reporter activity up to 74% (P < 0.005) in a dose-dependent manner and blocked RET phosphorylation up to 87% (P = 0.0593), AKT phosphorylation up to 28% (P = 0.0593) and extracellular signal regulatory kinase phosphorylation up to 75% (P = 0.0636). A11 inhibited the action of cisplatin-induced GDF15 on the brainstem and decreased GFRAL-positive neuron population expressing c-Fos in the area postrema and nucleus of the solitary tract by 62% in vivo (P < 0.05). In a melanoma mouse model treated with cisplatin, A11 recovered anorexia by 21% (P < 0.05) and tumour-free body weight loss by 13% (P < 0.05). A11 significantly improved the cisplatin-induced loss of skeletal muscles (quadriceps: 21%, gastrocnemius: 9%, soleus: 13%, P < 0.05) and adipose tissues (epididymal white adipose tissue: 37%, inguinal white adipose tissue: 51%, P < 0.05). CONCLUSIONS: Our study suggests that GFRAL antagonist antibody may alleviate chemotherapy-induced cachexia, providing a novel therapeutic approach for patients with cancer experiencing chemotherapy-induced cachexia.


Subject(s)
Antineoplastic Agents , Melanoma , Mice , Humans , Male , Animals , Cachexia/chemically induced , Cachexia/drug therapy , Glial Cell Line-Derived Neurotrophic Factor , Anorexia/metabolism , Cisplatin , Mice, Inbred C57BL , Antineoplastic Agents/adverse effects
7.
J Extracell Vesicles ; 11(12): e12287, 2022 12.
Article in English | MEDLINE | ID: mdl-36447429

ABSTRACT

T cell-derived small extracellular vesicles (sEVs) exhibit anti-cancer effects. However, their anti-cancer potential should be reinforced to enhance clinical applicability. Herein, we generated interleukin-2-tethered sEVs (IL2-sEVs) from engineered Jurkat T cells expressing IL2 at the plasma membrane via a flexible linker to induce an autocrine effect. IL2-sEVs increased the anti-cancer ability of CD8+ T cells without affecting regulatory T (Treg ) cells and down-regulated cellular and exosomal PD-L1 expression in melanoma cells, causing their increased sensitivity to CD8+ T cell-mediated cytotoxicity. Its effect on CD8+ T and melanoma cells was mediated by several IL2-sEV-resident microRNAs (miRNAs), whose expressions were upregulated by the autocrine effects of IL2. Among the miRNAs, miR-181a-3p and miR-223-3p notably reduced the PD-L1 protein levels in melanoma cells. Interestingly, miR-181a-3p increased the activity of CD8+ T cells while suppressing Treg cell activity. IL2-sEVs inhibited tumour progression in melanoma-bearing immunocompetent mice, but not in immunodeficient mice. The combination of IL2-sEVs and existing anti-cancer drugs significantly improved anti-cancer efficacy by decreasing PD-L1 expression in vivo. Thus, IL2-sEVs are potential cancer immunotherapeutic agents that regulate both immune and cancer cells by reprogramming miRNA levels.


Subject(s)
Extracellular Vesicles , Melanoma , MicroRNAs , Mice , Animals , Interleukin-2 , MicroRNAs/genetics , B7-H1 Antigen , CD8-Positive T-Lymphocytes , Melanoma/therapy
8.
Biochem Biophys Res Commun ; 629: 34-39, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36099782

ABSTRACT

Tongue epithelium is one of the most proliferative and regenerative epithelia in our body. However, tongue stem cell research is hampered partly by the lack of optimal animal models to study tongue injury, repair, and regeneration. Here, we establish a novel chemically induced tongue injury-recovery mouse model. Focal application of sodium hydroxide for a limited time led to the denudation of suprabasal layers, leaving the basal layer. Time course study revealed that tongue epithelial cells robustly proliferate over one week after the tongue injury. Importantly, we demonstrated that our novel mouse model could be employed in the lineage tracing of the tongue stem cells under the injury and repair process and further showed that tongue stem cells proliferate faster and generate larger clones in the injury condition than in the steady state condition. Our data indicate the development of a novel chemically induced tongue injury-recovery mouse model for tongue stem cell research, which will significantly facilitate the preclinical study for the pathogenesis and treatment of caustic ingestion.


Subject(s)
Caustics , Animals , Epithelial Cells , Epithelium , Mice , Sodium Hydroxide , Tongue
9.
Biomaterials ; 289: 121765, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36067566

ABSTRACT

Extracellular vesicles (EVs) mediate cell-cell crosstalk by carrying bioactive molecules derived from cells. Recently, immune cell-derived EVs have been reported to regulate key biological functions such as tumor progression. CD4+ T cells orchestrate overall immunity; however, the biological role of their EVs is unclear. This study reveals that EVs derived from CD4+ T cells increase the antitumor response of CD8+ T cells by enhancing their proliferation and activity without affecting regulatory T cells (Tregs). Moreover, EVs derived from interleukin-2 (IL2)-stimulated CD4+ T cells induce a more enhanced antitumor response of CD8+ T cells compared with that of IL2-unstimulated CD4+ T cell-derived EVs. Mechanistically, miR-25-3p, miR-155-5p, miR-215-5p, and miR-375 within CD4+ T cell-derived EVs are responsible for the induction of CD8+ T cell-mediated antitumor responses. In a melanoma mouse model, the EVs potently suppress tumor growth through CD8+ T cell activation. This study demonstrates that the EVs, in addition to IL2, are important mediators between CD4+ and CD8+ T cells. Furthermore, unlike IL2, clinically used as an antitumor agent, CD4+ T cell-derived EVs stimulate CD8+ T cells without activating Tregs. Therefore, CD4+ T cell-derived EVs may provide a novel direction for cancer immunotherapy by inducing a CD8+ T cell-mediated antitumor response.


Subject(s)
Extracellular Vesicles , MicroRNAs , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Interleukin-2 , Mice , T-Lymphocytes, Regulatory
10.
BMB Rep ; 55(1): 48-56, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34353429

ABSTRACT

Small extracellular vesicles (sEVs) secreted by most cells carry bioactive macromolecules including proteins, lipids, and nucleic acids for intercellular communication. Given that some immune cell-derived sEVs exhibit anti-cancer properties, these sEVs have received scientific attention for the development of novel anticancer immunotherapeutic agents. In this paper, we reviewed the latest advances concerning the biological roles of immune cell-derived sEVs for cancer therapy. sEVs derived from immune cells including dendritic cells (DCs), T cells, natural-killer (NK) cells, and macrophages are good candidates for sEV-based cancer therapy. Besides their role of cancer vaccines, DC-shed sEVs activated cytotoxic lymphocytes and killed tumor cells. sEVs isolated from NK cells and chimeric antigen receptor (CAR) T cells exhibited cytotoxicity against cancer cells. sEVs derived from CD8+ T and CD4+ T cells inhibited cancer-associated cells in tumor microenvironment (TME) and activated B cells, respectively. M1-macrophage-derived sEVs induced M2 to M1 repolarization and also created a pro-inflammatory environment. Hence, these sEVs, via mono or combination therapy, could be considered in the treatment of cancer patients in the future. In addition, sEVs derived from cytokine-stimulated immune cells or sEV engineering could improve their anti-tumor potency. [BMB Reports 2022; 55(1): 48-56].


Subject(s)
Extracellular Vesicles , Neoplasms , Cell Communication , Cytokines/metabolism , Extracellular Vesicles/metabolism , Humans , Macrophages/metabolism , Neoplasms/metabolism , Neoplasms/therapy
11.
Cytokine ; 143: 155542, 2021 07.
Article in English | MEDLINE | ID: mdl-33926775

ABSTRACT

Interferon-γ (IFNG) is one of the key cytokines that regulates both innate and adaptive immune responses in the body. However, the role of IFNG in the regulation of vascularization, especially in the context of Vascular endothelial growth factor A (VEGFa)-induced angiogenesis is not clarified. Here, we report that IFNG shows potent anti-angiogenic potential against VEGFa-induced angiogenesis. IFNG significantly inhibited proliferation, migration, and tube formation of Human umbilical vein endothelial cells (HUVECs) both under basal and VEGFa-treated conditions. Intriguingly, Knockdown (KD) of STAT1 abolished the inhibitory effect of IFNG on VEGFa-induced angiogenic processes in HUVECs. Furthermore, IFNG exhibited potent anti-angiogenic efficacy in the mouse model of oxygen-induced retinopathy (OIR), an in vivo model for hypoxia-induced retinal neovascularization, without induction of functional side effects. Taken together, these results show that IFNG plays a crucial role in the regulation of VEGFa-dependent angiogenesis, suggesting its potential therapeutic applicability in neovascular diseases.


Subject(s)
Interferon-gamma/therapeutic use , Ischemia/complications , Retinal Neovascularization/complications , Retinal Neovascularization/drug therapy , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia/complications , Interferon-gamma/administration & dosage , Interferon-gamma/pharmacology , Intravitreal Injections , Mice , Neovascularization, Physiologic/drug effects , Retina/drug effects , Retina/pathology , Retina/physiopathology , Retinal Neovascularization/physiopathology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects
12.
Biochem Biophys Res Commun ; 558: 29-35, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33895548

ABSTRACT

Estrogen therapy is used to treat patients with post-menopausal symptoms, such as hot flashes and dyspareunia. Estrogen therapy also decreases the risk of fractures from osteoporosis in post-menopausal women. However, estrogen increases the risk of venous thromboembolic events, such as pulmonary embolism, but the pathways through which estrogen increase the risk of thromboembolism is unknown. Here, we show that estrogen elicits endothelial exocytosis, the key step in vascular thrombosis and inflammation. Exogenous 17ß-estradiol (E2) stimulated endothelial exocytosis of Weibel-Palade bodies (WPBs), releasing von Willebrand factor (vWF) and interleukin-8 (IL-8). Conversely, the estrogen antagonist ICI-182,780 interfered with E2-induced endothelial exocytosis. The ERα agonist propyl pyrazole triol (PPT) but not the ERß agonist diarylpropionitrile (DPN) induced vWF release, while ERα silencing counteracted vWF release by E2, suggesting that ERα mediates this effect. Exocytosis triggered by E2 occurred rapidly within 15 min and was not inhibited by either actinomycin D or cycloheximide. On the contrary, it was inhibited by the pre-treatment of U0126 or SB203580, an ERK or a p38 inhibitor, respectively, suggesting that E2-induced endothelial exocytosis is non-genomically mediated by the MAP kinase pathway. Finally, E2 treatment enhanced platelet adhesion to endothelial cells ex vivo, which was interfered with the pre-treatment of ICI-182,780 or U0126. Taken together, our data show that estrogen activates endothelial exocytosis non-genomically through the ERα-MAP kinase pathway. Our data suggest that adverse cardiovascular effects such as vascular inflammation and thrombosis should be considered in patients before menopausal hormone treatment.


Subject(s)
Endothelial Cells/drug effects , Estradiol/adverse effects , Exocytosis/drug effects , Endothelial Cells/pathology , Endothelial Cells/physiology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Replacement Therapy/adverse effects , Exocytosis/physiology , Female , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/physiology , Postmenopause/drug effects , Postmenopause/physiology , Risk Factors , Thromboembolism/etiology , Weibel-Palade Bodies/drug effects , Weibel-Palade Bodies/pathology , Weibel-Palade Bodies/physiology
13.
Biochem Biophys Res Commun ; 529(2): 169-174, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703406

ABSTRACT

Supernumerary tooth (ST) may arise from uncertain developmental abnormalities or underlying genetic causes, and the extraction at the early age is recommended. Dental pulp stem cells (DPSCs) are the valuable resource for the regeneration of tooth and related craniofacial structures. DPSCs isolated from ST (sDPSCs) have not been fully characterized despite the potential in the applications. The objectives of this study are the efficient isolation of sDPSCs and the analysis of the properties as stem cells. sDPSCs were established by hammer-cracking and separation of the intact pulp from ST. sDPSCs in the culture were examined by light microscope and flow cytometer for the morphology and the surface marker expression. sDPSCs exhibited the cellular morphology of typical mesenchymal stem cells and expressed CD44, CD73, CD90, CD105 and CD166, but not CD14, CD34 or CD45. sDPSCs showed the differentiation potential toward osteogenic, chondrogenic and adipogenic lineages. During osteogenic differentiation, the stimulation by Oncostatin M enhanced the differentiation and significantly increased the expression of genes involved in the hard tissue repair, such as BMP2, BMP4, BMP6 and RUNX2. sDPSCs can be effectively derived from ST and displays the characteristics of mesenchymal stem cells in the maintenance and the differentiation. sDPSCs satisfies the quality as DPSCs thus provide the valuable resource to the regenerative therapy.


Subject(s)
Dental Pulp/cytology , Oncostatin M/metabolism , Osteogenesis , Stem Cells/cytology , Tooth, Supernumerary/metabolism , Cell Differentiation , Cells, Cultured , Dental Pulp/metabolism , Humans , Stem Cells/metabolism
14.
FASEB J ; 34(1): 1270-1287, 2020 01.
Article in English | MEDLINE | ID: mdl-31914593

ABSTRACT

Dysregulation of the adipo-osteogenic differentiation balance of mesenchymal stem cells (MSCs), which are common progenitor cells of adipocytes and osteoblasts, has been associated with many pathophysiologic diseases, such as obesity, osteopenia, and osteoporosis. Growing evidence suggests that lipid metabolism is crucial for maintaining stem cell homeostasis and cell differentiation; however, the detailed underlying mechanisms are largely unknown. Here, we demonstrate that glucosylceramide (GlcCer) and its synthase, glucosylceramide synthase (GCS), are key determinants of MSC differentiation into adipocytes or osteoblasts. GCS expression was increased during adipogenesis and decreased during osteogenesis. Targeting GCS using RNA interference or a chemical inhibitor enhanced osteogenesis and inhibited adipogenesis by controlling the transcriptional activity of peroxisome proliferator-activated receptor γ (PPARγ). Treatment with GlcCer sufficiently rescued adipogenesis and inhibited osteogenesis in GCS knockdown MSCs. Mechanistically, GlcCer interacted directly with PPARγ through A/B domain and synergistically enhanced rosiglitazone-induced PPARγ activation without changing PPARγ expression, thereby treatment with exogenous GlcCer increased adipogenesis and inhibited osteogenesis. Animal studies demonstrated that inhibiting GCS reduced adipocyte formation in white adipose tissues under normal chow diet and high-fat diet feeding and accelerated bone repair in a calvarial defect model. Taken together, our findings identify a novel lipid metabolic regulator for the control of MSC differentiation and may have important therapeutic implications.


Subject(s)
Adipocytes/metabolism , Cell Differentiation , Glucosylceramides/metabolism , Glucosyltransferases/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis , PPAR gamma/metabolism , Animals , Glucosylceramides/genetics , Glucosyltransferases/genetics , Humans , Mice , PPAR gamma/genetics
15.
Cytokine ; 126: 154863, 2020 02.
Article in English | MEDLINE | ID: mdl-31629112

ABSTRACT

Adiponectin (Ad) is a representative adipocytokine that regulates energy homeostasis including glucose transport and lipid oxidation through activation of AMP-activated protein kinase (AMPK) pathways. Plasma levels of Ad are reduced in obesity, which contributes to type 2 diabetes. Therefore, agents that activate the Ad signaling pathway could ameliorate metabolic diseases such as type 2 diabetes. Here, we report the identification of a high-affinitive agonist antibody against Ad receptors. The antibody was selected by using phage display of human combinatorial antibody libraries. The selected antibody induced phosphorylation of the acetyl-CoA carboxylase (ACC) and AMPK in skeletal muscle cells and stimulated glucose uptake and fatty-acid oxidation (FAO) in myotubes. In addition, the antibody significantly lowered blood glucose levels during a glucose challenge in normal mice as well as basal blood glucose levels in a type 2 diabetic mouse model. Taken together, these results suggest that the agonist antibody could be a promising therapeutic agent for the treatment of metabolic syndrome such as type 2 diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Receptors, Adiponectin/agonists , Receptors, Adiponectin/immunology , Acetyl-CoA Carboxylase/metabolism , Adiponectin/metabolism , Animals , Blood Glucose/metabolism , Cell Line , Diabetes Mellitus, Type 2/metabolism , Gene Knockdown Techniques , Glucose/pharmacology , Humans , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , NF-kappa B/metabolism , Oxidation-Reduction , Phosphorylation , RNA, Small Interfering , Receptors, Adiponectin/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Proc Natl Acad Sci U S A ; 117(1): 426-431, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31871166

ABSTRACT

Herein we present a concept in cancer where an immune response is detrimental rather than helpful. In the cancer setting, the immune system is generally considered to be helpful in curtailing the initiation and progression of tumors. In this work we show that a patient's immune response to their tumor can, in fact, either enhance or inhibit tumor cell growth. Two closely related autoantibodies to the growth factor receptor TrkB were isolated from cancer patients' B cells. Although highly similar in sequence, one antibody was an agonist while the other was an antagonist. The agonist antibody was shown to increase breast cancer cell growth both in vitro and in vivo, whereas the antagonist antibody inhibited growth. From a mechanistic point of view, we showed that binding of the agonist antibody to the TrkB receptor was functional in that it initiated downstream signaling identical to its natural growth factor ligand, brain-derived neurotrophic factor (BDNF). Our study shows that individual autoantibodies may play a role in cancer patients.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Breast Neoplasms/pathology , Membrane Glycoproteins/immunology , Neoplasm Metastasis/immunology , Receptor, trkB/immunology , Animals , Autoantibodies/blood , Autoantibodies/isolation & purification , Autoantibodies/metabolism , Autoantigens/blood , Autoantigens/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Brain-Derived Neurotrophic Factor/immunology , Brain-Derived Neurotrophic Factor/metabolism , Breast Neoplasms/blood , Breast Neoplasms/immunology , Cell Line, Tumor , Cell Movement/immunology , Cell Proliferation , Female , Humans , Membrane Glycoproteins/agonists , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/blood , Mice , Receptor, trkB/agonists , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/blood , Signal Transduction/immunology
17.
Anal Chem ; 91(22): 14214-14219, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31631648

ABSTRACT

Pipetting techniques play a crucial role in obtaining reproducible and reliable results, especially when seeding cells on small target areas, such as on microarrays, biochips or microfabricated cell culture systems. For very rare cells, such as human primary skeletal muscle cells (skMCs), manual (freehand) cell seeding techniques invariably result in nonuniform cell spreading and heterogeneous cell densities, giving rise to undesirable variations in myogenesis and differentiation. To prevent such technique-dependent variation, we have designed and fabricated a simple, low-cost pipet guidance device (PGD), and holder that works with hand-held pipettes. This work validates the accuracy and reproducibility of the PGD platform and compares its effectiveness with manual and robotic seeding techniques. The PGD system ensures reproducibility of cell seeding, comparable to that of more expensive robotic dispensing systems, resulting in a high degree of cell uniformity and homogeneous cell densities, while also enabling cell community studies. As compared to freehand pipetting, PGD-assisted seeding of C2C12 mouse myoblasts showed 5.3 times more myotube formation and likewise myotubes derived from PGD-seeded human primary skMCs were 3.6 times thicker and 2.2 times longer. These results show that this novel, yet simple PGD-assisted pipetting technique provides precise cell seeding on small targets, ensuring reproducible and reliable high-throughput cell assays.


Subject(s)
Cell Culture Techniques/instrumentation , Muscle, Skeletal/cytology , Tissue Array Analysis/instrumentation , Cell Count , Cell Differentiation , Cell Proliferation , Cells, Cultured , Equipment Design , Humans , Microarray Analysis
18.
PLoS One ; 14(10): e0223193, 2019.
Article in English | MEDLINE | ID: mdl-31647823

ABSTRACT

Microchips are widely used to separate circulating tumor cells (CTCs) from whole blood by virtues of sophisticated manipulation for microparticles. Here, we present a chip with an 8 µm high and 27.9 mm wide slit to capture cancer cells bound to 3 µm beads. Apart from a higher purity and recovery rate, the slit design allows for simplified fabrication, easy cell imaging, less clogging, lower chamber pressure and, therefore, higher throughput. The beads were conjugated with anti-epithelial cell adhesion molecules (anti-EpCAM) to selectively bind to breast cancer cells (MCF-7) used to spike the whole blood. The diameter of the cell-bead construct was in average 23.1 µm, making them separable from other cells in the blood. As a result, the cancer cells were separated from 5 mL of whole blood with a purity of 52.0% and a recovery rate of 91.1%, and also we confirmed that the device can be applicable to clinical samples of human breast cancer patients. The simple design with microslit, by eliminating any high-aspect ratio features, is expected to reduce possible defects on the chip and, therefore, more suitable for mass production without false separation outputs.


Subject(s)
Antigens, Neoplasm/blood , Breast Neoplasms/blood , Microfluidic Analytical Techniques , Neoplastic Cells, Circulating , Breast Neoplasms/genetics , Female , Humans , MCF-7 Cells , Microspheres , Precancerous Conditions/blood , Precancerous Conditions/genetics
19.
Sci Rep ; 9(1): 3494, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30837591

ABSTRACT

Previously, we reported an agonist antibody to a cytokine receptor, Thrombopoietin receptor (TPOR) that effectively induces cytotoxic killer cells from precursor tumor cells isolated from newly diagnosed AML patients. Here, we show that the TPOR agonist antibody can induce even relapsed AML cells into killer cells more potently than newly diagnosed AML cells. After stimulation by the agonist antibody, these relapsed leukemic cells enter into a differentiation process of killer cells. The antibody-induced killer cells express, Granzyme B and Perforin that assault and kill other members of the AML cell population. Particularly, the agonist antibody showed potent efficacy on the AML xenograft model in mice using the NOD/LtSz-scid IL2Rγc null (NSG) mice. These results show that the TPOR agonist antibody that induces AML cells to kill each other is effective on both relapsed AML cells and in vivo. Therefore, this study suggests a new strategy for the treatment of cancer relapse after chemotherapy.


Subject(s)
Antibodies/immunology , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/pathology , Animals , Antibodies/therapeutic use , Cell Line, Tumor , Humans , Killer Cells, Natural/pathology , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Thrombopoietin/agonists , Receptors, Thrombopoietin/immunology , Receptors, Thrombopoietin/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Recurrence , Signal Transduction/drug effects , Thrombopoietin/genetics , Thrombopoietin/metabolism , Thrombopoietin/pharmacology , Transplantation, Heterologous
20.
Oncotarget ; 9(69): 33244, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30237865

ABSTRACT

[This corrects the article DOI: 10.18632/oncotarget.23401.].

SELECTION OF CITATIONS
SEARCH DETAIL
...