Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Immunity ; 54(9): 2042-2056.e8, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34407391

ABSTRACT

Recruitment of immune cells to the site of inflammation by the chemokine CCL1 is important in the pathology of inflammatory diseases. Here, we examined the role of CCL1 in pulmonary fibrosis (PF). Bronchoalveolar lavage fluid from PF mouse models contained high amounts of CCL1, as did lung biopsies from PF patients. Immunofluorescence analyses revealed that alveolar macrophages and CD4+ T cells were major producers of CCL1 and targeted deletion of Ccl1 in these cells blunted pathology. Deletion of the CCL1 receptor Ccr8 in fibroblasts limited migration, but not activation, in response to CCL1. Mass spectrometry analyses of CCL1 complexes identified AMFR as a CCL1 receptor, and deletion of Amfr impaired fibroblast activation. Mechanistically, CCL1 binding triggered ubiquitination of the ERK inhibitor Spry1 by AMFR, thus activating Ras-mediated profibrotic protein synthesis. Antibody blockade of CCL1 ameliorated PF pathology, supporting the therapeutic potential of targeting this pathway for treating fibroproliferative lung diseases.


Subject(s)
Chemokine CCL1/metabolism , Fibroblasts/metabolism , Membrane Proteins/metabolism , Myofibroblasts/metabolism , Phosphoproteins/metabolism , Pulmonary Fibrosis/metabolism , Receptors, Autocrine Motility Factor/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Differentiation/physiology , Fibroblasts/pathology , Humans , Mice , Myofibroblasts/pathology , Pulmonary Fibrosis/pathology , Signal Transduction/physiology
2.
Immunity ; 51(3): 522-534.e7, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471107

ABSTRACT

Although recent progress provides mechanistic insights into the pathogenesis of pulmonary fibrosis (PF), rare anti-PF therapeutics show definitive promise for treating this disease. Repeated lung epithelial injury results in injury-repairing response and inflammation, which drive the development of PF. Here, we report that chronic lung injury inactivated the ubiquitin-editing enzyme A20, causing progressive accumulation of the transcription factor C/EBPß in alveolar macrophages (AMs) from PF patients and mice, which upregulated a number of immunosuppressive and profibrotic factors promoting PF development. In response to chronic lung injury, elevated glycogen synthase kinase-3ß (GSK-3ß) interacted with and phosphorylated A20 to suppress C/EBPß degradation. Ectopic expression of A20 or pharmacological restoration of A20 activity by disturbing the A20-GSK-3ß interaction accelerated C/EBPß degradation and showed potent therapeutic efficacy against experimental PF. Our study indicates that a regulatory mechanism of the GSK-3ß-A20-C/EBPß axis in AMs may be a potential target for treating PF and fibroproliferative lung diseases.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Macrophages/metabolism , Pulmonary Fibrosis/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Animals , Cell Line , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Up-Regulation/physiology
4.
Adv Exp Med Biol ; 1208: 131-173, 2021.
Article in English | MEDLINE | ID: mdl-34260026

ABSTRACT

Macroautophagy is an important biological process in eukaryotic cells by which longevity proteins, misfolded proteins, and damaged organelles are degraded. The autophagy process consists of three key steps: (1) the formation of autophagosomes; (2) the fusion of the autophagosomes with lysosomes; and (3) the degradation of the contents of autolysosomes. If any of the three steps is impaired, autophagy will not be able to complete its biological function. Dysfunctional or blocked autophagy is closely involved in the pathogenesis of a variety of diseases. The accurate determination of the autophagy activity in vivo and in vitro has become a challenge in the field of autophagy research. At present, the most widely used detection method to determine autophagy activity in mammalian cells is to quantify LC3B in the cells by Western blot, or to observe the formation and changes of autophagosomes and autolysosomes by immunofluorescence and electron microscopy. However, ignoring the dynamic characteristics of autophagy and only evaluating the number of autophagosomes or the presence of LC3B cannot completely reflect the activation or a blockage of the autophagy system, and objectively analyze its real role in the occurrence and development of a disease. For example, the accumulation of autophagosomes and autolysosomes can occur through an increase in substrate to be degraded after the activation of autophagy, or it may be caused by the partial obstruction or blockage of autophagy. In this chapter, new and familiar ways to detect the autophagic flux are methodically summarized to provide researchers with a multi-angled viewpoint.


Subject(s)
Autophagosomes , Autophagy , Animals , Eukaryotic Cells , Lysosomes
5.
Gastroenterology ; 156(3): 708-721.e15, 2019 02.
Article in English | MEDLINE | ID: mdl-30365932

ABSTRACT

BACKGROUND & AIMS: Activation of Wnt signaling to ß-catenin contributes to the development of colorectal cancer (CRC). Expression of tribbles pseudo-kinase 3 (TRIB3) is increased in some colorectal tumors and associated with poor outcome. We investigated whether increased TRIB3 expression promotes stem cell features of CRC cells and tumor progression by interacting with the Wnt signaling pathway. METHODS: We performed studies with C57BL/6J-ApcMin/J mice injected with an adeno-associated virus vector that expresses a small hairpin RNA against Trib3 mRNA (ApcMin/J-Trib3KD) or a control vector (ApcMin/J-Ctrl). We created BALB/c mice that overexpress TRIB3 from an adeno-associated virus vector and mice with small hairpin RNA-mediated knockdown of ß-catenin. The mice were given azoxymethane followed by dextran sodium sulfate to induce colitis-associated cancer. Intestinal tissues were collected and analyzed by histology, gene expression profiling, immunohistochemistry, and immunofluorescence. Leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5)-positive (LGR5Pos) and LGR5-negative (LGR5Neg) HCT-8 CRC cells, with or without knockdown or transgenic expression of TRIB3, were sorted and analyzed in sphere-formation assays. We derived organoids from human and mouse colorectal tumors to analyze the function of TRIB3 and test the effect of a peptide inhibitor. Wnt signaling to ß-catenin was analyzed in dual luciferase reporter, chromatin precipitation, immunofluorescence, and immunoblot assays. Proteins that interact with TRIB3 were identified by immunoprecipitation. CRC cell lines were grown in nude mice as xenograft tumors. RESULTS: At 10 weeks of age, more than half the ApcMin/J-Ctrl mice developed intestinal high-grade epithelial neoplasia, whereas ApcMin/J-Trib3KD mice had no intestinal polyps and normal histology. Colon tissues from ApcMin/J-Trib3KD mice expressed lower levels of genes regulated by ß-catenin and genes associated with cancer stem cells. Mice with overexpression of Trib3 developed more tumors after administration of azoxymethane and dextran sodium sulfate than BALB/c mice. Mice with knockdown of ß-catenin had a lower tumor burden after administration of azoxymethane and dextran sodium sulfate, regardless of Trib3 overexpression. Intestinal tissues from mice with overexpression of Trib3 and knockdown of ß-catenin did not have activation of Wnt signaling or expression of genes regulated by ß-catenin. LGR5Pos cells sorted from HCT-8 cells expressed higher levels of TRIB3 than LGR5Neg cells. CRC cells that overexpressed TRIB3 had higher levels of transcription by ß-catenin and formed larger spheroids than control CRC cells; knockdown of ß-catenin prevented the larger organoid size caused by TRIB3 overexpression. TRIB3 interacted physically with ß-catenin and transcription factor 4 (TCF4). TRIB3 overexpression increased, and TRIB3 knockdown decreased, recruitment of TCF4 and ß-catenin to the promoter region of genes regulated by Wnt. Activated ß-catenin increased expression of TRIB3, indicating a positive-feedback loop. A peptide (P2-T3A6) that bound ß-catenin disrupted its interaction with TRIB3 and TCF4. In primary CRC cells and HCT-8 cells, P2-T3A6 decreased expression of genes regulated by ß-catenin and genes associated with cancer stem cells and decreased cell viability and migration. Injection of C57BL/6J-ApcMin/J mice with P2-T3A6 decreased the number and size of tumor nodules and colon expression of genes regulated by ß-catenin. P2-T3A6 increased 5-fluorouracil-induced death of CRC cells and survival times of mice with xenograft tumors. CONCLUSION: TRIB3 interacts with ß-catenin and TCF4 in intestine cells to increase expression of genes associated with cancer stem cells. Knockdown of TRIB3 decreases colon neoplasia in mice, migration of CRC cells, and their growth as xenograft tumors in mice. Strategies to block TRIB3 activity might be developed for treatment of CRC.


Subject(s)
Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/genetics , beta Catenin/metabolism , Animals , Cell Communication/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplastic Stem Cells/metabolism , Random Allocation , Sensitivity and Specificity , Up-Regulation , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays
6.
J Asian Nat Prod Res ; 19(4): 305-313, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28347180

ABSTRACT

Natural products are an important original source of many widely used drugs, including anti-cancer drugs. Early research efforts for seeking anti-cancer therapy from the natural products are mainly focused on the compounds with cytotoxicity capability. The good examples include vinblastine, vincristine, the camptothecin derivatives; topotecan, irinotecan, epipodophyllotoxin derivatives and paclitaxel. In a recent decade, the fundamental progression has been made in the understanding of molecular and cellular mechanisms regarding tumor initiation, metastasis, therapeutic resistance, immune escape, and relapse, which provide a great opportunity for the development of new mechanism-based anticancer drugs, especially drugs against new molecular and cellular targets. Autophagy, a critical cell homeostasis mechanism and promising drug target involved in a verity of human diseases including cancer, can be modulated by many compounds derived from natural products. In this review, we'll give a short introduction of autophagy and discuss the roles of autophagy in the tumorigenesis and progression. And then, we summarize the accumulated evidences to show the anti-tumor effects of several compounds derived from natural products through modulation of autophagy activity.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Biological Products/pharmacology , Stilbenes/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Humans , Irinotecan , Molecular Structure , Paclitaxel/pharmacology , Resveratrol
7.
J Asian Nat Prod Res ; 19(2): 101-108, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28252344

ABSTRACT

Tissue fibrosis is a common pathologic change of many chronic diseases, which is characterized by extracellular matrix accumulation in tissues and dysfunction of the injured organs. Despite there recently gain mechanistic insight into the pathogenesis of tissue fibrosis, therapeutics for tissue fibrosis and thus many chronic diseases remain a significant clinical unmet need. Recent progressions indicate that autophagy, a conserved lysosomal degradation process in eukaryotic cells, not only plays an important regulatory role in maintaining cellular and tissue homeostasis, but also contributes to the development and progression of tissue fibrosis in a diversity of organs. Interestingly, a number of natural compounds derived from plant or Chinese Herb Medicines (CHM), have been identified as modulators of autophagy, and may function as potential therapeutic agents for the treatment of different fibrotic diseases. In this review, we focus on several plant natural compounds that have well-known anti-fibrotic effects through regulating autophagic signal pathways or autophagy activity. These findings should provide important therapeutic clues and strategy for the development of new anti-fibrosis drugs.


Subject(s)
Autophagy/drug effects , Fibrosis/drug therapy , Humans , Molecular Structure , Signal Transduction/drug effects
8.
J Asian Nat Prod Res ; 19(4): 314-319, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28347174

ABSTRACT

Patients with diabetes have a high level of blood glucose because their body cannot produce enough insulin or properly respond to this hormone. In both situations, it has become evident that persistent high concentrations of glucose, insulin, insulin-like growth factor, and insulin resistance lead to dysfunction and destruction of autophagic activity in the cells of islet and other organs involved in complications of diabetes, including the liver, cardiovascular, and nervous systems. Accumulating evidences have revealed that autophagy is a novel therapeutic target with a wide range of beneficial effects on diabetes and that plenty of drugs and natural products are involved in autophagy modulation, either inducing or inhibiting autophagy, through multiple signaling pathways. In this review, we summarize the roles of several clinical drugs and compounds derived from natural products in diabetes and its complications through regulation of autophagy, expecting to inspire further investigation of the underlying mechanisms of these compounds and to facilitate their better clinical application.


Subject(s)
Autophagy/drug effects , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/pharmacology , Animals , Blood Glucose/analysis , Humans , Insulin Resistance/physiology , Molecular Structure , Signal Transduction/drug effects
9.
Yao Xue Xue Bao ; 51(1): 39-44, 2016 Jan.
Article in Zh | MEDLINE | ID: mdl-27405160

ABSTRACT

Autophagy is an important homeostatic cellular recycling mechanism responsible for degrading injured or dysfunctional subcellular organelles and proteins in all living cells. The process of autophagy can be divided into three relatively independent steps: the initiation of phagophore, the formation of autophagosome and the maturation/degradation stage. Different morphological characteristics and molecular marker changes can be observed at these stages. Morphological approaches are useful to produce novel knowledge that would not be achieved through other experimental methods. Here we summarize the morphological methods in monitoring autophagy, the principles in data interpretation and the cautions that should be considered in the study of autophagy.


Subject(s)
Autophagy , Phagosomes , Homeostasis , Humans , Organelles
10.
Yao Xue Xue Bao ; 51(1): 45-51, 2016 Jan.
Article in Zh | MEDLINE | ID: mdl-27405161

ABSTRACT

Autophagy is a crucial biological process of eukaryotes, which is involved in cell growth, survival and energy metabolism, while the premise of the autophagy function is activated autophagic flux. It has been confirmed that impaired autophagic flux promotes pathogenesis of many chronic inflammatory diseases, especially cancer, neurodegenerative disease and tissue fibrosis, therefore the analysis of autophagic flux state is important for revealing autophagy function and the mechanism of autophagy related diseases. Given that autophagy is a dynamic process with multiple steps, it is very hard to observe the real state of autophagic flux. Summarized here is the novel concept and current approach to detect autophagic flux. This knowledge is crucial for the researching of the biological function of autophagy, and may provide some strategies for developing autophagy-related drug.


Subject(s)
Autophagy , Fibrosis , Humans , Inflammation/pathology , Neoplasms/pathology , Neurodegenerative Diseases/pathology
11.
Yao Xue Xue Bao ; 51(1): 52-8, 2016 Jan.
Article in Zh | MEDLINE | ID: mdl-27405162

ABSTRACT

Autophagy is an active research area in the biomedical field as its role has been identified in many physiological and pathological processes. Accordingly, there is a growing demand to identify, quantify and manipulate the process accurately. Meanwhile, there is great interest in identifying compounds that modulate autophagy because they may have applications in the treatment of a variety of autophagy-related diseases. In this review, we summarize the current status of autophagy screening systems to facilitate identification of autophagy modulators.


Subject(s)
Autophagy , Humans
12.
Yao Xue Xue Bao ; 51(7): 1017-24, 2016 07.
Article in Zh | MEDLINE | ID: mdl-29896947

ABSTRACT

Diabetes and cancer are two major chronic diseases with tremendous impact on human health worldwide. Clinical and basic studies demonstrate that diabetes can promote carcinogenesis and tumor progression. High insulin and high insulin-like growth factor are considered to be the major risk factors for cancer. Chronic inflammation and aberrant metabolism also participate in cancer development. It is noteworthy that therapies used for treatment of diabetes may increase or decrease the risk of cancer. Revealing the mechanisms that connect diabetes to cancer will be crucial for prevention and treatment of diabetes-related cancers.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Insulin/blood , Neoplasms/physiopathology , Somatomedins/metabolism , Humans , Hyperinsulinism , Inflammation
13.
Int J Cancer ; 134(3): 692-702, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23852533

ABSTRACT

Cell-penetrating peptides provide a unique platform to create a new generation of cancer therapeutics with enhanced efficacy and diminished toxicity. In our study, enhanced expression of toll-like receptor 2 (TLR2) was observed in acute myeloid leukemia (AML) cells. Screening of a phage display peptide library using Biopanning and Rapid Analysis of Selective Interactive Ligands (BRASIL) identified a TLR2-binding peptide motif, Pep2. We show that the TLR2-binding peptide motif targeted and penetrated into leukemia cells in a TLR2-dependent manner. Moreover, a synthetic, chimeric peptide composed of the TLR2-binding motif linked to a programmed cell death-inducing sequence, D(KLAKLAK)2, induced apoptosis in AML cells with high TLR2 expression (TLR2(high)) but not in chronic myeloid leukemia (CML) cells with low TLR2 expression (TLR2(low)). The antileukemia activity of this chimeric peptide was confirmed in leukemia patient samples and an animal model of myeloid leukemia, as the development of leukemia was significantly delayed in mice with TLR2(high) AML compared to TLR2(low) CML NOD/SCID mice. TUNEL assays on bone marrow tissue slices revealed that the chimerical peptide induced leukemia cell apoptosis in a TLR2-dependent manner. Together, our findings indicate that TLR2 is a potential therapeutic target for the prevention and treatment of AML, and the prototype, Pep2-D(KLAKLAK)2, is a promising drug candidate in this setting.


Subject(s)
Apoptosis , Leukemia, Myeloid, Acute/drug therapy , Peptides/therapeutic use , Toll-Like Receptor 2/chemistry , Cell Line, Tumor , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/pathology , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacokinetics , Surface Plasmon Resonance , Toll-Like Receptor 2/metabolism
14.
Hepatology ; 57(1): 171-82, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22859216

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is a complication at the endstage of chronic inflammatory liver diseases with dismal prognosis. Targeting of Toll-like receptor (TLR) 2 attenuates tumor metastases; we hypothesized that blocking TLR2 might also play a crucial role in reducing hepatocarcinogenesis. Surprisingly, we found that the genetic deletion of TLR2 increased susceptibility to diethylnitrosamine (DEN), a genotoxic carcinogen that can induce HCC. Indeed, TLR2-deficient mice showed a significant increase in carcinogenesis and progression of HCC as indicated by increases in tumor nodule size, tumor volume, and animal death. The enhanced susceptibility to DEN-induced HCC was associated with a broad-spectrum reduction in the immune response to DEN-induced liver injury. We found that TLR2 deficiency caused a decrease in the infiltration of macrophages and an attenuation of apoptosis signal regulating kinase 1 (ASK1) / p38 mitogen-activated protein kinase (p38 MAPK) / nuclear factor kappa B (NF-κB) signaling, which led to a decrease in the expression of interferon-gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), interleukin (IL)-1α/ß, IL-6, and Cxcl-2 as well as suppression of autophagy flux and increases in oxidative stress and p62 aggregation in liver tissue. The defects in immune networks resulted in suppressed p21- and p16/pRb-dependent senescence, which caused an increase in proliferation and a decrease in apoptotic and autophagy-associated cell death in mouse livers. Restoring cellular senescence and autophagy flux by treating TLR2-deficient mice with IFN-γ, a T helper 1 (Th1) cytokine and positive modulator of senescence and autophagy, could attenuate the carcinogenesis and progression of HCC associated with TLR2-deficient animals. CONCLUSION: The loss of immune networks supporting cellular senescence and autophagy flux is attributed to enhanced susceptibility to DEN-induced hepatocellular carcinogenesis and progression in TLR2-deficient mice. These findings may be used to prevent the development of liver cancer.


Subject(s)
Carcinoma, Hepatocellular/immunology , Cell Transformation, Neoplastic , Cellular Senescence , Liver Neoplasms/immunology , Toll-Like Receptor 2/metabolism , Alkylating Agents , Animals , Autophagy , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Diethylnitrosamine , Disease Progression , Female , Interferon-gamma/metabolism , Liver/immunology , Liver/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Transcription Factor TFIIH , Transcription Factors/metabolism
15.
Hepatology ; 57(5): 1869-81, 2013 May.
Article in English | MEDLINE | ID: mdl-23299825

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is a devastating consequence of chronic inflammatory liver diseases. The goal of this study was to investigate whether Toll-like receptor 4 (TLR4) activity contributes to HCC initiation and progression in mice. A mouse model of diethylnitrosamine (DEN)-induced HCC was generated with wild-type and TLR4 mutant mice, and the development and progression of HCC and senescent responses were assessed using morphologic, immunological, and biochemical criteria. We found that genetic or pharmacologic blocking of TLR4 increased susceptibility to DEN-induced HCC carcinogenesis and progression, which was indicated by increases in number of tumor nodules, tumor volume, and animal death. The enhanced HCC was associated with a broad-spectrum reduction of immune response to DEN liver injury, as indicated by decreases in the liver-infiltrating F4/80+ macrophages, the apoptosis signal-regulating kinase 1/p38 mitogen-activated protein kinase/NF-κB and IRF3 signaling activities, and the expression of inflammatory cytokines. Suppressed immune networks resulted in a halt of cellular senescence induction in TLR4 mutant liver tissue, which promoted proliferation and suppressed programmed cell death. Moreover, TLR4 mutation resulted in a suppressed capacity of DNA repair due to a decrease in TLR4-medicated expression of DNA repair proteins Ku70/80 in liver tissue and cells. Isotopic expression of Ku70 in TLR4 mutant mice restored senescence and interrupted the positive feedback loop of DNA damage and oxidative stress, which reversed TLR4 mutation-deteriorated HCC carcinogenesis and progression. CONCLUSION: TLR4 plays an integrated defense role against HCC carcinogenesis by enhancing the expression and function of DNA repair protein Ku70. Our studies provide novel insight into TLR4 activity in the regulation of HCC tumorigenesis, which may be useful for the prevention of HCC development.


Subject(s)
Antigens, Nuclear/physiology , Carcinoma, Hepatocellular/prevention & control , Cell Transformation, Neoplastic , DNA-Binding Proteins/physiology , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms/prevention & control , Toll-Like Receptor 4/physiology , Animals , Antigens, Nuclear/genetics , Apoptosis/physiology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/physiopathology , Cell Transformation, Neoplastic/chemically induced , DNA Repair/genetics , DNA Repair/physiology , DNA-Binding Proteins/genetics , Diethylnitrosamine/adverse effects , Disease Models, Animal , Female , Ku Autoantigen , Liver Neoplasms/pathology , Liver Neoplasms/physiopathology , Male , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Mutation/genetics , Signal Transduction/physiology , Toll-Like Receptor 4/genetics
16.
Yao Xue Xue Bao ; 49(5): 615-21, 2014 May.
Article in Zh | MEDLINE | ID: mdl-25151730

ABSTRACT

To investigate the protective effects and possible mechanism of Mycelium of Hirsutella hepiali Chen et Shen (MHCS) on metabolic syndromes, free fatty acid and MHCS-treated hepatocytes were used for detecting autophagy-related LC3, p62 and lipid accumulation. Moreover, high fat diet fed mice were used to establish metabolic syndromes model. 50-weeks age mice were randomly divided into: control group, model group and MHCS group. At 80-weeks age, 15 mice were randomly chosen from each group separately for examining oral glucose tolerance, serum insulin, insulin-like growth factor 1 (IGF-1), hepatic LC3, p62, p-NF-kappaB p65, NF-kappaB p65, IL-6 and CXCL-8. Moreover, insulin resistance index (IRI) was calculated. Hepatic pathological changes, including vacuoles, lipids accumulation and fibrosis were observed. Remaining mice were fed with diet separately to 110 weeks-age for statistics of mortality. MHCS promoted autophagy of free fatty acid treated hepatocytes. Mice fed with high fat plus MHCS diet exhibited improved oral glucose tolerance, insulin resistance, hepatic pathology, inflammation, mortality and activated autophagy. The protective effects of MHCS against metabolic syndroms might be through the activation of hepatic autophagy.


Subject(s)
Autophagy , Hepatocytes/pathology , Liver/pathology , Metabolic Syndrome/pathology , Mycelium/physiology , Animals , Diet, High-Fat/adverse effects , Glucose Tolerance Test , Hepatocytes/metabolism , Hypocreales , Insulin/blood , Insulin Resistance , Insulin-Like Growth Factor I/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Liver/metabolism , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Random Allocation , Transcription Factor RelA/metabolism , Transcription Factor TFIIH , Transcription Factors/metabolism
17.
Yao Xue Xue Bao ; 49(6): 764-73, 2014 Jun.
Article in Zh | MEDLINE | ID: mdl-25212019

ABSTRACT

Autophagy is an important homeostatic cellular recycling mechanism responsible for degrading injured or dysfunctional cellular organelles and proteins in all living cells. Aging is a universal phenomenon characterized by progressive deterioration of cells and organs due to accumulation of macromolecular and organelle damage. Growing evidences indicate that the rate of autophagosome formation and maturation and the efficiency of autophagosome/lysosome fusion decline with age. Dysfunctional autophagy has also been observed in age-related diseases. Autophagy disruption resulted accumulation of mutated or misfolded proteins is the essential feature of neurodegenerative disorders. However, in cancers, fibroproliferative diseases or cardiovascular diseases, autophagy can play either a protective or destructive role in different types of disease, and even in different stages of the same disease. The review will discuss the cellular and molecular mechanisms of autophagy and its important role in the pathogenesis of aging and age-related diseases, and the ongoing drug discovery strategies for therapeutic intervention.


Subject(s)
Aging , Autophagy , Drug Discovery , Humans , Lysosomes/metabolism , Neurodegenerative Diseases , Phagosomes/metabolism , Protein Folding
18.
Nat Commun ; 15(1): 203, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38172124

ABSTRACT

Dysregulated hematopoietic niches remodeled by leukemia cells lead to imbalances in immunological mediators that support leukemogenesis and drug resistance. Targeting immune niches may ameliorate disease progression and tyrosine kinase inhibitor (TKI) resistance in Philadelphia chromosome-positive B-ALL (Ph+ B-ALL). Here, we show that T helper type 17 (Th17) cells and IL-17A expression are distinctively elevated in Ph+ B-ALL patients. IL-17A promotes the progression of Ph+ B-ALL. Mechanistically, IL-17A activates BCR-ABL, IL6/JAK/STAT3, and NF-kB signalling pathways in Ph+ B-ALL cells, resulting in robust cell proliferation and survival. In addition, IL-17A-activated Ph+ B-ALL cells secrete the chemokine CXCL16, which in turn promotes Th17 differentiation, attracts Th17 cells and forms a positive feedback loop supporting leukemia progression. These data demonstrate an involvement of Th17 cells in Ph+ B-ALL progression and suggest potential therapeutic options for Ph+ B-ALL with Th17-enriched niches.


Subject(s)
Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Fusion Proteins, bcr-abl/genetics , Interleukin-17/genetics , Drug Resistance, Neoplasm/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Acute Disease
19.
Am J Pathol ; 180(1): 275-92, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22062220

ABSTRACT

Pulmonary fibrosis is an inflammation-driven lung disease with a poor prognosis and no cure. Here we report that basal toll-like receptor 4 (TLR4) activity is critical for the resolution of acute and chronic inflammation and pulmonary fibrosis in mouse models of lung injury. We found that genetic or pharmacologic inhibition of TLR4 exacerbates bleomycin-induced pulmonary inflammation, fibrosis, dysfunction, and animal death through promoting formation of an immunosuppressive tissue microenvironment and attenuating autophagy-associated degradation of collagen and cell death in the fibrotic lung tissues. In contrast, pharmacologic activation of TLR4 resulted in a quick resolution of acute inflammation, reversed the established pulmonary fibrosis, improved lung function, and rescued mice from death. Similarly, blocking TLR4 impaired the resolution of silica-induced chronic inflammation and fibrosis. Importantly, altering autophagic activity could reverse the TLR4-regulated lung inflammation, fibrosis, dysfunction, and animal death. Rapamycin, an autophagy activator, reversed the effects of TLR4 antagonism. In contrast, inhibition of autophagy by 3-methyladenine reversed the proresolving and antifibrotic roles of TLR4 agonists and increased animal death. These results not only highlight a pivotal role for TLR4-mediated basal immunity, particularly autophagic activity, in the proresolution of inflammation and fibrosis after chemical-induced lung injury but also provide proof for the concept for activating TLR4 signaling, particularly TLR4-mediated autophagy, as a novel therapeutic strategy against chronic fibroproliferative diseases that are unresponsive to current therapy.


Subject(s)
Acute Lung Injury/physiopathology , Idiopathic Pulmonary Fibrosis/physiopathology , Lung Injury/physiopathology , Pneumonia/physiopathology , Toll-Like Receptor 4/physiology , Acute Lung Injury/pathology , Animals , Apoptosis/physiology , Autophagy/physiology , Idiopathic Pulmonary Fibrosis/pathology , Lung Injury/pathology , MAP Kinase Signaling System/physiology , Mice , Pneumonia/pathology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/deficiency
20.
J Immunol ; 187(6): 3003-14, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21841134

ABSTRACT

Pulmonary fibrosis is the pathologic basis for a variety of incurable human chronic lung diseases. IL-17A, a glycoprotein secreted from IL-17-producing cells, has recently been shown to be a proinflammatory cytokine involved in chronic inflammation and autoimmune disease. In this study, we report that IL-17A increased the synthesis and secretion of collagen and promoted the epithelial-mesenchymal transition in alveolar epithelial cells in a TGF-ß1-dependent manner. Using in vivo fibrotic models, we found IL-17A expression to be elevated and IL-17A-associated signaling pathways to be activated in fibrotic lung tissues. Neutralization of IL-17A in vivo promoted the resolution of bleomycin-induced acute inflammation, attenuated pulmonary fibrosis, and increased survival. Additionally, IL-17A antagonism inhibited silica-induced chronic inflammation and pulmonary fibrosis. Targeting IL-17A resulted in a shift of the suppressive immune response in fibrotic lung tissue toward a Th1-type immune response, and it effectively induced autophagy, which promoted the autophagic degradation of collagen and autophagy-associated cell death. Moreover, IL-17A was found to attenuate the starvation-induced autophagy, and autophagy modulators regulated collagen degradation in the alveolar epithelial cells in a TGF-ß1-independent manner. Administration of 3-methylamphetamine, an autophagy inhibitor, reversed the therapeutic efficacy of IL-17A antagonism in pulmonary fibrosis. Our studies indicate that IL-17A participates in the development and progression of pulmonary fibrosis in both TGF-ß1-dependent and -independent manners and that the components of the IL-17A signaling pathway are potential therapeutic targets for the treatment of fibroproliferative lung diseases.


Subject(s)
Interleukin-17/metabolism , Pneumonia/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction/immunology , Transforming Growth Factor beta1/metabolism , Animals , Autophagy , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Cell Separation , Collagen/biosynthesis , Epithelial-Mesenchymal Transition/immunology , Flow Cytometry , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL