Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Biol Toxicol ; 39(6): 2709-2724, 2023 12.
Article in English | MEDLINE | ID: mdl-36757501

ABSTRACT

Drug-induced liver injury (DILI) still poses a major clinical challenge and is a leading cause of acute liver failure. Inhibitor of nuclear factor kappa B kinase subunit epsilon (IKBKE) is essential for inflammation and metabolic disorders. However, it is unclear how IKBKE regulates cellular damage in acetaminophen (APAP)-induced acute liver injury. Here, we found that the deficiency of IKBKE markedly aggravated APAP-induced acute liver injury by targeting RIPK1. We showed that APAP-treated IKBKE-deficient mice exhibited severer liver injury, worse mitochondrial integrity, and enhanced glutathione depletion than wild-type mice. IKBKE deficiency may directly upregulate the expression of total RIPK1 and the cleaved RIPK1, resulting in sustained JNK activation and increased translocation of RIPK1/JNK to mitochondria. Moreover, deficiency of IKBKE enhanced the expression of pro-inflammatory factors and inflammatory cell infiltration in the liver, especially neutrophils and monocytes. Inhibition of RIPK1 activity by necrostatin-1 significantly reduced APAP-induced liver damage. Thus, we have revealed a negative regulatory function of IKBKE, which acts as an RIPK1/JNK regulator to mediate APAP-induced hepatotoxicity. Targeting IKBKE/RIPK1 may serve as a potential therapeutic strategy for acute or chronic liver injury.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Animals , Mice , Acetaminophen/toxicity , Liver , Glutathione/metabolism , Mitochondria/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Mice, Inbred C57BL , JNK Mitogen-Activated Protein Kinases/metabolism , Hepatocytes/metabolism , NF-kappaB-Inducing Kinase
2.
Cell Death Discov ; 7(1): 342, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34750342

ABSTRACT

CD5 molecule like (CD5L), a member of the scavenger receptor cysteine-rich domain superfamily, plays a critical role in immune homeostasis and inflammatory disease. Acetaminophen (APAP) is a safe and effective antipyretic analgesic. However, overdose may cause liver damage or even liver failure. APAP hepatotoxicity is characterized by extensive necrotic cell death and a sterile inflammatory response, in which the role of CD5L remains to be investigated. In this study, we found that the expression of CD5L was increased in the livers of mice after APAP overdose. Furthermore, CD5L deficiency reduced the increase of alanine transaminase (ALT) level, histopathologic lesion area, c-Jun N-terminal kinase (JNK)/extracellular signal-regulated kinase (ERK) phosphorylation level, Transferase-Mediated dUTP Nick End-Labeling positive (TUNEL+) cells proportion, vascular endothelial cell permeability and release of inflammatory cytokines induced by excess APAP. Therefore, our findings reveal that CD5L may be a potential therapeutic target for prevention and treatment of APAP-induced liver injury.

3.
Mol Med ; 27(1): 57, 2021 06 06.
Article in English | MEDLINE | ID: mdl-34092215

ABSTRACT

BACKGROUND: Acetaminophen (APAP) overdose causes hepatotoxicity and even acute liver failure. Recent studies indicate that sterile inflammation and innate immune cells may play important roles in damage-induced hepatocytes regeneration and liver repair. The scavenger receptor CD36 has its crucial functions in sterile inflammation. However, the roles of CD36 in APAP induced acute liver injury remain unclear and warrant further investigation. METHODS: WT C57BL/6 J and CD36-/- mice were intraperitoneally injected with APAP (300 mg/kg) after fasting for 16 h. Liver injury was evaluated by serum alanine aminotransferase (ALT) level and liver tissue hematoxylin and eosin (H&E) staining. Liver inflammatory factor expression was determined by real-time polymerase chain reaction (PCR). The protein adducts forming from the metabolite of APAP and the metabolism enzyme cytochrome P450 2E1 (CYP2E1) levels were measured by Western blot. Liver infiltrating macrophages and neutrophils were characterized by flow cytometry. RNA sequencing and Western blot were used to evaluate the effect of damage-associated molecular patterns (DAMP) molecule high mobility group B1 (HMGB1) on WT and CD36-/- macrophages. Moreover, PP2, a Src kinase inhibitor, blocking CD36 signaling, was applied in APAP model. RESULTS: The expression of CD36 was increased in the liver of mice after APAP treatment. Compared with WT mice, APAP treated CD36-/- mice show less liver injury. There was no significant difference in APAP protein adducts and CYP2E1 expression between these two strains. However, reduced pro-inflammatory factor mRNA expression and serum IL-1ß level were observed in APAP treated CD36-/- mice as well as infiltrating macrophages and neutrophils. Moreover, CD36 deficiency impaired the activation of c-Jun N-terminal kinase (JNK) caused by APAP. Interestingly, the lack of CD36 reduced the activation of extracellular regulated protein kinases (Erk) and v-akt murine thymoma viral oncogene homolog (Akt) induced by HMGB1. RNA transcription sequencing data indicated that HMGB1 has a different effect on WT and CD36-/- macrophages. Furthermore, treatment with PP2 attenuated APAP induced mouse liver injury. CONCLUSION: Our data demonstrated that CD36 deficiency ameliorated APAP-induced acute liver injury and inflammatory responses by decreasing JNK activation. CD36 might serve as a new target to reduce acute liver injury.


Subject(s)
CD36 Antigens/deficiency , Chemical and Drug Induced Liver Injury/etiology , Disease Susceptibility , Acetaminophen/adverse effects , Animals , Biomarkers , Biopsy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Gene Expression Regulation , Genetic Predisposition to Disease , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Knockout , src-Family Kinases/metabolism
4.
J Exp Clin Cancer Res ; 39(1): 172, 2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32859246

ABSTRACT

BACKGROUND: Lysine succinylation is an emerging posttranslational modification that has garnered increased attention recently, but its role in gastric cancer (GC) remains underexplored. METHODS: Proteomic quantification of lysine succinylation was performed in human GC tissues and adjacent normal tissues by mass spectrometry. The mRNA and protein levels of lactate dehydrogenase A (LDHA) in GC and adjacent normal tissues were analyzed by qRT-PCR and western blot, respectively. The expression of K222-succinylated LDHA was measured in GC tissue microarray by the K222 succinylation-specific antibody. The interaction between LDHA and sequestosome 1 (SQSTM1) was measured by co-immunoprecipitation (co-IP) and proximity ligation assay (PLA). The binding of carnitine palmitoyltransferase 1A (CPT1A) to LDHA was determined by co-IP. The effect of K222-succinylated LDHA on tumor growth and metastasis was evaluated by in vitro and in vivo experiments. RESULTS: Altogether, 503 lysine succinylation sites in 303 proteins were identified. Lactate dehydrogenase A (LDHA), the key enzyme in Warburg effect, was found highly succinylated at K222 in GC. Intriguingly, this modification did not affect LDHA ubiquitination, but reduced the binding of ubiquitinated LDHA to SQSTM1, thereby decreasing its lysosomal degradation. We demonstrated that CPT1A functions as a lysine succinyltransferase that interacts with and succinylates LDHA. Moreover, high K222-succinylation of LDHA was associated with poor prognosis in patients with GC. Finally, overexpression of a succinylation-mimic mutant of LDHA promoted cell proliferation, invasion, and migration. CONCLUSIONS: Our data revealed a novel lysosomal pathway of LDHA degradation, which is mediated by the binding of K63-ubiquitinated LDHA to SQSTM1. Strikingly, CPT1A succinylates LDHA on K222, which thereby reduces the binding and inhibits the degradation of LDHA, as well as promotes GC invasion and proliferation. This study thus uncovers a new role of lysine succinylation and the mechanism underlying LDHA upregulation in GC.


Subject(s)
Biomarkers, Tumor/metabolism , L-Lactate Dehydrogenase/metabolism , Lysine/chemistry , Lysosomes/metabolism , Protein Processing, Post-Translational , Stomach Neoplasms/pathology , Succinic Acid/chemistry , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Proliferation , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , L-Lactate Dehydrogenase/chemistry , L-Lactate Dehydrogenase/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Middle Aged , Prognosis , Proteolysis , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Cell Mol Med ; 23(1): 293-305, 2019 01.
Article in English | MEDLINE | ID: mdl-30394687

ABSTRACT

Gastric cancer (GC) is a malignancy of the lining of the stomach and is prone to distant metastasis, which involves a variety of complex molecules. The S100 proteins are a family of calcium-binding cytosolic proteins that possess a wide range of intracellular and extracellular functions and play pivotal roles in the invasion and migration of tumour cells. Among these, S100A10 is known to be overexpressed in GC. Lysine succinylation, a recently identified form of protein post-translational modification, is an important regulator of cellular processes. Here, we demonstrated that S100A10 was succinylated at lysine residue 47 (K47), and levels of succinylated S100A10 were increased in human GC. Moreover, K47 succinylation of S100A10 was stabilized by suppression of ubiquitylation and subsequent proteasomal degradation. Furthermore, carnitine palmitoyltransferase 1A (CPT1A) was found to function as a lysine succinyltransferase that interacts with S100A10. Succinylation of S100A10 is regulated by CPT1A, while desuccinylation is regulated by SIRT5. Overexpression of a succinylation mimetic mutant, K47E S100A10, increased cell invasion and migration. Taken together, this study reveals a novel mechanism of S100A10 accumulation mediated by succinylation in GC, which promotes GC progression and is regulated by the succinyltransferase CPT1A and SIRT5-mediated desuccinylation.


Subject(s)
Annexin A2/metabolism , Carnitine O-Palmitoyltransferase/metabolism , S100 Proteins/metabolism , Stomach Neoplasms/pathology , Animals , Annexin A2/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lysine/metabolism , Male , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Protein Processing, Post-Translational , S100 Proteins/genetics , Sirtuins/metabolism , Stomach Neoplasms/metabolism , Succinates/metabolism
6.
Drug Des Devel Ther ; 12: 4253-4260, 2018.
Article in English | MEDLINE | ID: mdl-30587921

ABSTRACT

PURPOSE: ICAM-1 plays a critical role in the development of acute respiratory distress syndrome (ARDS). MK2 regulates the expression of ICAM-1 in human pulmonary microvascular endothelial cells. To explore whether the inhibition of MK2 activation has the same effect in experimental animals, MMI-0100, a peptide-mediated inhibitor of MK2, was used to verify whether MMI-0100 can ameliorate lung inflammation in a mouse model of ARDS by reducing endothelial expression of ICAM-1. METHODS: In this study, C57BL/6 mice were randomly divided into three groups: a control group, an lipopolysaccharides (LPS) group, and an LPS plus MMI-0100 group. Mice were killed 24 hours after the administration of LPS and MMI-0100. The mouse lung tissue histopathology, wet/dry weight ratio (W/D), and the neutrophil count were used to measure the severity of lung inflammation in mice. The pulmonary microvascular endothelial cells (PMVECs) of the mice were isolated. The mRNA expression of ICAM-1 in mouse PMVECs was determined using RT-PCR, and the protein expression of MK2 and ICAM-1 in mouse PMVECs was analyzed using Western blotting and immunohistochemistry. RESULTS: We found that the level of phosphorylated MK2 in the LPS plus MMI-0100 group was reduced. Compared with the LPS group, the LPS plus MMI-0100 group of mice showed less severe inflammation, including a lower W/D and neutrophil count. The mRNA and protein expression of ICAM-1 in the LPS group was significantly higher than in the control group in mouse PMVECs, and the ICAM-1 level was reduced after the administration of MMI-0100. CONCLUSION: These data indicate that MMI-0100 ameliorates lung inflammation in a mouse model of ARDS by reducing endothelial expression of ICAM-1.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Endothelial Cells/drug effects , Intercellular Adhesion Molecule-1/metabolism , Lung/blood supply , Peptides/pharmacology , Pneumonia/prevention & control , Respiratory Distress Syndrome/prevention & control , Animals , Disease Models, Animal , Down-Regulation , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides , Male , Mice, Inbred C57BL , Phosphorylation , Pneumonia/chemically induced , Pneumonia/metabolism , Protein Serine-Threonine Kinases/metabolism , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism
7.
J Exp Clin Cancer Res ; 37(1): 208, 2018 Aug 30.
Article in English | MEDLINE | ID: mdl-30165890

ABSTRACT

BACKGROUND: Enzymatically inactive chitinase-like protein CHI3L1 drives inflammatory response and promotes tumor progression. However, its role in gastric cancer (GC) tumorigenesis and metastasis has not yet been fully elucidated. We determined the significance of CHI3L1 expression in patients with GC. We also explored an as-yet unknown receptor of CHI3L1 and investigated the involved signaling in GC metastasis. METHODS: CHI3L1 expression was evaluated by immunoblotting, tissue microarray-based immunohistochemistry analysis (n = 100), and enzyme linked immunosorbent assay (ELISA) (n = 150). The interactions between CD44 and CHI3L1 or Interleukin-13 receptor alpha 2 (IL-13Rα2) were analyzed by co-immunoprecipitation, immunofluorescence co-localization assay, ELISA, and bio-layer interferometry. The roles of CHI3L1/CD44 axis in GC metastasis were investigated in GC cell lines and experimental animal model by gain and loss of function. RESULTS: CHI3L1 upregulation occurred during GC development, and positively correlated with GC invasion depth, lymph node status, and tumor staging. Mechanically, CHI3L1 binding to CD44 activated Erk and Akt, along with ß-catenin signaling by phosphorylating ß-catenin at Ser552 and Ser675. CD44 also interacted with IL-13Rα2 to form a complex. Notably, CD44v3 peptide and protein, but not CD44v6 peptide or CD44s protein, bound to both CHI3L1 and IL-13Rα2. Our in vivo and in vitro data further demonstrated that CHI3L1 promoted GC cell proliferation, migration, and metastasis. CONCLUSIONS: CHI3L1 binding to CD44v3 activates Erk, Akt, and ß-catenin signaling, therefore enhances GC metastasis. CHI3L1 expression is a novel biomarker for the prognosis of GC, and these findings have thus identified CHI3L1/CD44 axis as a vital pathway and potential therapeutic target in GC.


Subject(s)
Biomarkers, Tumor/genetics , Chitinase-3-Like Protein 1/genetics , Hyaluronan Receptors/genetics , Stomach Neoplasms/genetics , Animals , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Interleukin-13 Receptor alpha1 Subunit/genetics , MAP Kinase Signaling System/genetics , Mice , Neoplasm Metastasis , Oncogene Protein v-akt/genetics , Prognosis , Signal Transduction/genetics , Stomach Neoplasms/pathology , Tissue Array Analysis , Xenograft Model Antitumor Assays , beta Catenin/genetics
8.
J Cell Mol Med ; 22(11): 5708-5719, 2018 11.
Article in English | MEDLINE | ID: mdl-30160350

ABSTRACT

Helicobacter pylori (H. pylori) infection triggers chronic inflammation that has been associated with gastric cancer (GC). Exosomes are small extracellular vesicles that have become the key mediators of intercellular communication. In this study, we investigated exosome-mediated communication between H. pylori-infected GC cells and macrophages, focusing on the transfer of activated mesenchymal-epithelial transition factor (MET). We observed a significant decrease in MET protein expression in GC cells after infection with H. pylori, whereas MET mRNA levels remained unchanged. Intriguingly, MET expression, specifically the phosphorylated active form, was increased in exosomes released from H. pylori-infected GC cells. Confocal microscopy and Western blotting analyses showed that these exosomes containing MET were delivered to and internalized by macrophages. Indeed, in human GC tissues positive for H. pylori, we also observed that activated MET was highly expressed in tumour-infiltrating macrophages. After internalization, exosomal MET then appeared to educate the macrophages towards a pro-tumorigenesis phenotype. This included exosomal MET-mediated stimulation of proinflammatory cytokine secretion IL-1ß, which subsequently promoted tumour growth and progression in vitro and in vivo. Taken together, these data were the first to demonstrate H. pylori infection-induced upregulation of activated MET in exosomes and the pro-tumorigenic effect on tumour-associated macrophages.


Subject(s)
Helicobacter Infections/genetics , Inflammation/genetics , Proto-Oncogene Proteins c-met/genetics , Stomach Neoplasms/genetics , Animals , Cell Line, Tumor , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Exosomes/genetics , Exosomes/microbiology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Heterografts , Humans , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/genetics , Macrophages/microbiology , Macrophages/pathology , Mice , Stomach Neoplasms/complications , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
9.
Cell Cycle ; 17(4): 428-438, 2018.
Article in English | MEDLINE | ID: mdl-29468929

ABSTRACT

Tumor-associated macrophages (TAM) are prominent components of tumor microenvironment (TME) and capable of promoting cancer progression. However, the mechanisms for the formation of M2-like TAMs remain enigmatic. Here, we show that lactate is a pivotal oncometabolite in the TME that drives macrophage M2-polarization to promote breast cancer proliferation, migration, and angiogenesis. In addition, we identified that the activation of ERK/STAT3, major signaling molecules in the lactate signaling pathway, deepens our molecular understanding of how lactate educates TAMs. Moreover, suppression of ERK/STAT3 signaling diminished tumor growth and angiogenesis by abolishing lactate-induced M2 macrophage polarization. Finally, research data of the natural compound withanolide D provide evidence for ERK/STAT3 signaling as a potential therapeutic strategy for the prevention and treatment of breast cancer. These findings suggest that the lactate-ERK/STAT3 signaling pathway is a driver of breast cancer progression by stimulating macrophage M2-like polarization and reveal potential new therapeutic targets for breast cancer treatment.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Lactic Acid/pharmacology , Macrophages/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Humans , Macrophages/cytology , Macrophages/drug effects , Membrane Glycoproteins , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , STAT3 Transcription Factor/genetics , Tumor Microenvironment , Withanolides/pharmacology , Withanolides/therapeutic use
10.
Hepatology ; 67(5): 1943-1955, 2018 05.
Article in English | MEDLINE | ID: mdl-29220536

ABSTRACT

The scavenger receptor CD36 recognizes a diverse set of ligands and has been implicated in a wide variety of normal and pathological processes, including lipid metabolism, angiogenesis, atherosclerosis, and phagocytosis. In particular, recent findings have demonstrated its crucial functions in sterile inflammation and tumor metastasis. However, the role of CD36 in immune-mediated hepatitis remains unclear. Concanavalin A (ConA)-induced liver injury is a well-established experimental T cell-mediated hepatitis. To understand the role of CD36 in hepatitis, we tested the susceptibility of CD36-deficient (CD36-/- ) mice to this model, evaluated by a liver enzyme test, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay, histological analysis, mononuclear cell (MNC) infiltration, and hepatic proinflammatory factor production. CD36-/- mice were less sensitive to ConA-induced hepatitis and had a significantly lower number of liver MNCs (LMNCs), including CD4+ cells, CD8+ T cells, natural killer cells, natural killer T cells, infiltrating macrophages, and neutrophils, as well as reduced expression of inflammatory mediators (tumor necrosis factor α, CXC chemokine ligand (CXCL) 10, interleukin (IL)-1α, monocyte chemotactic protein 1, and IL-6) compared with controls. Notably, we used bone marrow chimeric mice to demonstrate that CD36 expression on nonhematopoietic cells was required to drive ConA-induced liver injury. Furthermore, our data show that the CD36 receptor was essential for CXCL10-induced hepatocyte apoptosis and activation of IκB kinase, Akt, and Jun N-terminal kinase. Moreover, treatment of wild-type mice with genistein, a tyrosine kinase inhibitor that blocks CD36-Lyn signaling, attenuated ConA-induced liver injury and reduced the number of MNCs. CONCLUSIONS: Our findings suggest that CD36 plays an important proinflammatory role in ConA-induced liver injury by promoting hepatic inflammation and mediating the proapoptotic effect of chemokine CXCL10, and therefore, may be a potential therapeutic target for immune-mediated hepatitis. (Hepatology 2018;67:1943-1955).


Subject(s)
Blood Platelet Disorders/pathology , CD36 Antigens/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemokine CXCL10/metabolism , Genetic Diseases, Inborn/pathology , Hepatitis/metabolism , Animals , Apoptosis/drug effects , Blood Platelet Disorders/immunology , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/pharmacology , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry , Genetic Diseases, Inborn/immunology , Genistein/pharmacology , Hepatitis/immunology , Hepatitis/pathology , Hepatocytes/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Signal Transduction
11.
Oncotarget ; 8(43): 75298-75307, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-29088866

ABSTRACT

The tumor microenvironment is critical for tumor growth and metastasis, but the underlying molecular mechanisms are poorly understood. Recent studies have shown that IκB-kinase-ε (IKKε) is involved in the proliferation and migration of certain cancers. However, the functional role of IKKε in the progression of gastric cancer (GC) remains unknown. In this study, we found that high levels of IKKε expression in GC tumors were correlated with more advanced disease and poor overall survival of patients. Silencing of IKKε effectively suppressed the migratory and invasive capabilities of human GC cells in vitro and tumorigenicity and metastasis in vivo. Further analysis revealed that IKKε was also highly expressed in tumor-infiltrating lymphocytes. Moreover, it was involved in tumor-infiltrating T-cell-mediated invasion and metastasis. Knockdown of IKKε elevated T-cell antitumor immunity. These findings suggest that IKKε may be a novel prognostic marker and a potential therapeutic target in human GCs.

12.
Oncotarget ; 8(32): 53819-53828, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28881853

ABSTRACT

As an intermediate metabolite of the tricarboxylic acid cycle in mitochondria, succinate is widely investigated for its role in metabolism. In recent years, an increasing number of studies have concentrated on the unanticipated role of succinate outside metabolism, acting as, for instance, an inflammatory signal or a carcinogenic initiator. Actually, succinate dehydrogenase gene mutations and abnormal succinate accumulation have been observed in a battery of hereditary and sporadic malignancies. In this review, we discuss the unexpected role of succinate and possible mechanisms that may contribute to its accumulation. Additionally, we describe how the high concentration of succinate in the tumor microenvironment acts as an active participant in tumorigenesis, rather than a passive bystander or innocent victim. Focusing on mechanism-based research, we summarize some targeted therapies which have been applied to the clinic or are currently under development. Furthermore, we posit that investigational drugs with different molecular targets may expand our horizon in anticancer therapy.

13.
Oncotarget ; 8(8): 13174-13185, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28061458

ABSTRACT

Altered cellular metabolism is now generally acknowledged as a hallmark of cancer cells, the resultant abnormal oncometabolites cause both metabolic and nonmetabolic dysregulation and potential transformation to malignancy. A subset of cancers have been found to be associated with mutations in succinate dehydrogenase genes which result in the accumulation of succinate. However, the function of succinate in tumorigenesis remains unclear. In the present study, we aim to investigate the role of oncometabolite succinate in tumor angiogenesis. Our data demonstrated the accumulation of markedly elevated succinate in gastric cancer tissues compared with that in paracancerous tissues. Moreover, succinate was able to increase the chemotactic motility, tube-like structure formation and proliferation of primary human umbilical vascular endothelial cells (pHUVECs) in vitro, as well as promoting the blood vessel formation in transgenic zebrafish. Our mechanistic studies reveal that succinate upregulates vascular endothelial growth factor (VEGF) expression by activation of signal transducer and activator of transcription 3 (STAT3) and extracellular regulated kinase (ERK)1/2 via its receptor GPR91 in a HIF-1α independent mechanism. Taken together, these data indicate an important role of the succinate-GPR91 axis in tumor angiogenesis, which may enable development of a novel therapeutic strategy that targets cancer metabolism.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Physiologic/drug effects , Receptors, G-Protein-Coupled/metabolism , STAT3 Transcription Factor/metabolism , Succinates/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Animals, Genetically Modified , Cell Line , Cell Line, Tumor , Cells, Cultured , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Immunoblotting , Microscopy, Fluorescence , RNA Interference , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Succinates/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
14.
Appl Biochem Biotechnol ; 182(2): 482-494, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27933483

ABSTRACT

Human 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1), a potential target in breast cancer prevention and therapy, was extracted from human placenta and immobilized on nonporous silica (∼5 µm) with a covalent method for the first time. The optimum initial enzyme concentration and immobilization time during the immobilization process were 0.42 mg mL-1 and 12 h, repectively. The binding was confirmed by scanning electron microscope (SEM) and infrared spectroscopy (FT-IR). It could improve the pH, thermal and storage stability compared to free enzyme. Moreover, the immobilized enzyme could be reused at least four times. A screening method based on it coupled with liquid chromatography-time-of-flight mass spectrometer (LC-TOF/MS) was established, and the half-maximal inhibitory concentration (IC 50) of apigenin for the immobilized enzyme was 291 nM. Subsequently, 10 natural products were evaluated leading to inhibition of the activity of 17ß-HSD1 at the concentration of 25 µM, and six of them inhibit the activity over 50%.


Subject(s)
17-Hydroxysteroid Dehydrogenases/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Apigenin/chemistry , Enzymes, Immobilized/chemistry , Mass Spectrometry , Pregnancy Proteins/chemistry , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Chromatography, Liquid , Drug Screening Assays, Antitumor/methods , Female , Glutaral/chemistry , Humans , Silicon Dioxide/chemistry
15.
Asian Pac J Cancer Prev ; 13(4): 1097-104, 2012.
Article in English | MEDLINE | ID: mdl-22799288

ABSTRACT

BACKGROUND/AIM: Pristimerin isolated from Celastrus and Maytenus spp can inhibit proteasome activity. However, whether pristimerin can modulate cancer metastasis is unknown. METHODS: The impacts of pristimerin on the purified and intracellular chymotrypsin proteasomal activity, the levels of regulator of G protein signaling 4 (RGS 4) expression and breast cancer cell lamellipodia formation, and the migration and invasion were determined by enzymatic, Western blot, immunofluorescent, and transwell assays, respectively. RESULTS: We found that pristimerin inhibited human chymotrypsin proteasomal activity in MDA-MB-231 cells in a dose-dependent manner. Pristimerin also inhibited breast cancer cell lamellipodia formation, migration, and invasion in vitro by up-regulating RGS4 expression. Thus, knockdown of RGS4 attenuated pristimerin-mediated inhibition of breast cancer cell migration and invasion. Furthermore, pristimerin inhibited growth and invasion of implanted breast tumors in mice. CONCLUSION: Pristmerin inhibits proteasomal activity and increases the levels of RGS4, inhibiting the migration and invasion of breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/drug effects , Proteasome Inhibitors/pharmacology , RGS Proteins/metabolism , Triterpenes/pharmacology , Actins/metabolism , Analysis of Variance , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/enzymology , Cell Line, Tumor , Chymases/drug effects , Chymases/metabolism , Dose-Response Relationship, Drug , Female , Humans , Leupeptins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Pentacyclic Triterpenes , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , RGS Proteins/drug effects , RGS Proteins/genetics , RNA Interference , Random Allocation , Tumor Burden/drug effects , Up-Regulation
16.
Molecules ; 17(6): 6854-68, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22669041

ABSTRACT

Pristimerin is a triterpenoid isolated from Celastrus and Maytenus spp. that has been shown to possess a variety of biological activities, including anti-cancer activity. However, little is known about pristimerin's effects on tumor angiogenesis. In this study, we examined the function and the mechanism of this compound in tumor angiogenesis using multiple angiogenesis assays. We found that pristimerin significantly reduced both the volume and weight of solid tumors and decreased angiogenesis in a xenograft mouse tumor model in vivo. Pristimerin significantly inhibited the neovascularization of chicken chorioallantoic membrane (CAM) in vivo and abrogated vascular endothelial growth factor (VEGF)-induced microvessel sprouting in an ex vivo rat aortic ring assay. Furthermore, pristimerin inhibited the VEGF-induced proliferation, migration and capillary-like structure formation of human umbilical vascular endothelial cells (HUVECs) in a concentration-dependent manner. Mechanistic studies revealed that pristimerin suppressed the VEGF-induced phosphorylation of VEGF receptor 2 kinase (KDR/Flk-1) and the activity of AKT, ERK1/2, mTOR, and ribosomal protein S6 kinase. Taken together, our results provide evidence for the first time that pristimerin potently suppresses angiogenesis by targeting VEGFR2 activation. These results provide a novel mechanism of action for pristimerin which may be important in the treatment of cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Triterpenes/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Chick Embryo , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Pentacyclic Triterpenes , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Triterpenes/administration & dosage , Triterpenes/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
17.
Endocr J ; 59(6): 473-81, 2012.
Article in English | MEDLINE | ID: mdl-22447140

ABSTRACT

Triptolide, a major active component of Tripterygium wilfordii Hook F (TWHF), is known to have multiple pharmacological activities. However, studies have also shown that triptolide is highly toxic to the reproductive system by disrupting normal androgen and estrogen signaling. In the present study, we investigated the effect of triptolide (5, 10, or 20 nM for 24 h) on estradiol production by rat granulosa cells. Triptolide inhibited basal and human chorionic gonadotropin (HCG)- or 8-bromo-cAMP-stimulated estradiol production as revealed by RIA assay. Furthermore, the HCG-evoked increase in cellular cAMP content was also inhibited by triptolide, indicating that disruption of the cAMP/PKA signaling pathway may mediate the deleterious effects of triptolide on steroid hormone regulation. In addition, (3)H(2)O tests showed that aromatase activity was significantly inhibited by triptolide in granulosa cells. Western blot and quantitative real-time PCR (qRT-PCR) assays further revealed that triptolide decreased protein and mRNA expression of aromatase in granulosa cells. Moreover, mRNA expression of luteinizing hormone receptor (LHR) was induced by triptolide also using qRT-PCR method. In contrast, cell viability tests using Cell Counting Kit-8 (CCK-8) and 3-(4,5-dimethyl-thiazol-2-yl)-2,5- diphenyl-tetrazolium bromide (MTT) method indicated that triptolide did not cause measurable cell death at doses that suppressed steroidogenesis. The reproductive toxicity of triptolide may be mainly caused by disruption of cAMP/PKA-mediated expression of estrogen synthesis enzymes, leading to reduced estradiol synthesis and reproductive dysfunction.


Subject(s)
Diterpenes/pharmacology , Estradiol/biosynthesis , Granulosa Cells/drug effects , Immunosuppressive Agents/pharmacology , Phenanthrenes/pharmacology , RNA, Messenger/analysis , Animals , Aromatase/drug effects , Aromatase/genetics , Aromatase/metabolism , Cell Survival/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Diterpenes/toxicity , Epoxy Compounds/pharmacology , Epoxy Compounds/toxicity , Female , Gene Expression Profiling , Granulosa Cells/metabolism , Immunosuppressive Agents/toxicity , Phenanthrenes/toxicity , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, LH/drug effects , Receptors, LH/genetics , Receptors, LH/metabolism
18.
Arzneimittelforschung ; 61(10): 571-6, 2011.
Article in English | MEDLINE | ID: mdl-22164965

ABSTRACT

The protective effects of tripterygium glycoside-loaded solid lipid nanoparticles (TG-SLNs) on male reproductive toxicity were investigated in rats. Thirty-six male Sprague-Dawley rats were randomly divided into three groups of 12: control group, tripterygium glycoside (TG) group, and TG-SLN group. After the animals had been orally administered with the substances for 28 consecutive days, their sperm count and sperm motility, organ coefficients, serum testosterone levels, testicular ultrastructure, and reproductive ability were observed. The results showed that the sperm motility rate in the TG group was only 3%, whereas the rates in the TG-SLN and control groups were 33% and 71%, respectively. Compared with those in the control group, the motion counts of path velocity, track speed, progressive velocity, straightness, linearity, beat cross frequency, amplitude of lateral head displacement, and sperm concentrations in the TG-SLN group were not significantly different while those in the TG group significantly decreased (p < 0.01). TG-SLNs did not cause testicular atrophy and instead maintained normal serum testosterone levels. The effect of TG-SLNs on the testicular ultrastructure was very evident; the morphologies of Sertoli, spermatogonial, mitochondrial, and sperm cells were normal. In terms of reproductive ability, one rat (17%) from the TG-SLN group and five rats (83%) from the control group became pregnant, whereas none of the rats from the TG group became pregnant. These data indicate that TG-SLNs have potentially protective effects on male reproductive toxicity in rats.


Subject(s)
Infertility, Male/chemically induced , Tripterygium/toxicity , Animals , Drug Compounding , Drug Delivery Systems , Genitalia, Male/drug effects , Glycosides/chemistry , Glycosides/toxicity , Lipids , Male , Microscopy, Electron, Transmission , Nanoparticles , Rats , Rats, Sprague-Dawley , Reproduction/drug effects , Sperm Count , Sperm Motility/drug effects , Testis/drug effects , Testis/ultrastructure , Testosterone/blood , Tripterygium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...