Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Mol Cell ; 81(2): 370-385.e7, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33271062

ABSTRACT

The mechanisms of cellular energy sensing and AMPK-mediated mTORC1 inhibition are not fully delineated. Here, we discover that RIPK1 promotes mTORC1 inhibition during energetic stress. RIPK1 is involved in mediating the interaction between AMPK and TSC2 and facilitate TSC2 phosphorylation at Ser1387. RIPK1 loss results in a high basal mTORC1 activity that drives defective lysosomes in cells and mice, leading to accumulation of RIPK3 and CASP8 and sensitization to cell death. RIPK1-deficient cells are unable to cope with energetic stress and are vulnerable to low glucose levels and metformin. Inhibition of mTORC1 rescues the lysosomal defects and vulnerability to energetic stress and prolongs the survival of RIPK1-deficient neonatal mice. Thus, RIPK1 plays an important role in the cellular response to low energy levels and mediates AMPK-mTORC1 signaling. These findings shed light on the regulation of mTORC1 during energetic stress and unveil a point of crosstalk between pro-survival and pro-death pathways.


Subject(s)
Autophagy-Related Protein 5/genetics , Fas-Associated Death Domain Protein/genetics , Intestine, Large/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Autophagy-Related Protein 5/deficiency , Caspase 8/genetics , Caspase 8/metabolism , Cell Death/genetics , Fas-Associated Death Domain Protein/deficiency , Gene Expression Regulation , Glucose/antagonists & inhibitors , Glucose/pharmacology , HEK293 Cells , HT29 Cells , Humans , Intestine, Large/drug effects , Intestine, Large/pathology , Jurkat Cells , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Metformin/antagonists & inhibitors , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Signal Transduction , Sirolimus/pharmacology , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
2.
EMBO J ; 42(19): e112814, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37635626

ABSTRACT

The regulation of autophagy initiation is a key step in autophagosome biogenesis. However, our understanding of the molecular mechanisms underlying the stepwise assembly of ATG proteins during this process remains incomplete. The Rab GTPase Ypt1/Rab1 is recognized as an essential autophagy regulator. Here, we identify Atg23 and Atg17 as binding partners of Ypt1, with their direct interaction proving crucial for the stepwise assembly of autophagy initiation complexes. Disruption of Ypt1-Atg23 binding results in significantly reduced Atg9 interactions with Atg11, Atg13, and Atg17, thus preventing the recruitment of Atg9 vesicles to the phagophore assembly site (PAS). Likewise, Ypt1-Atg17 binding contributes to the PAS recruitment of Ypt1 and Atg1. Importantly, we found that Ypt1 is phosphorylated by TOR at the Ser174 residue. Converting this residue to alanine blocks Ypt1 phosphorylation by TOR and enhances autophagy. Conversely, the Ypt1S174D phosphorylation mimic impairs both PAS recruitment and activation of Atg1, thus inhibiting subsequent autophagy. Thus, we propose TOR-mediated Ypt1 as a multifunctional assembly factor that controls autophagy initiation via its regulation of the stepwise assembly of ATG proteins.


Subject(s)
Saccharomyces cerevisiae Proteins , Autophagy/physiology , Autophagy-Related Proteins/metabolism , Phagosomes/metabolism , Phosphorylation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
3.
Cell ; 147(1): 223-34, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21962518

ABSTRACT

Autophagy is an important intracellular catabolic mechanism that mediates the degradation of cytoplasmic proteins and organelles. We report a potent small molecule inhibitor of autophagy named "spautin-1" for specific and potent autophagy inhibitor-1. Spautin-1 promotes the degradation of Vps34 PI3 kinase complexes by inhibiting two ubiquitin-specific peptidases, USP10 and USP13, that target the Beclin1 subunit of Vps34 complexes. Beclin1 is a tumor suppressor and frequently monoallelically lost in human cancers. Interestingly, Beclin1 also controls the protein stabilities of USP10 and USP13 by regulating their deubiquitinating activities. Since USP10 mediates the deubiquitination of p53, regulating deubiquitination activity of USP10 and USP13 by Beclin1 provides a mechanism for Beclin1 to control the levels of p53. Our study provides a molecular mechanism involving protein deubiquitination that connects two important tumor suppressors, p53 and Beclin1, and a potent small molecule inhibitor of autophagy as a possible lead compound for developing anticancer drugs.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Benzylamines/pharmacology , Endopeptidases/metabolism , Quinazolines/pharmacology , Tumor Suppressor Protein p53/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Autophagy , Beclin-1 , Class III Phosphatidylinositol 3-Kinases/metabolism , Humans , Mice , Ubiquitin-Specific Proteases , Ubiquitination
4.
Mol Cell ; 68(2): 323-335.e6, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29033323

ABSTRACT

Acetylation is increasingly recognized as one of the major post-translational mechanisms for the regulation of multiple cellular functions in mammalian cells. Acetyltransferase p300, which acetylates histone and non-histone proteins, has been intensively studied in its role in cell growth and metabolism. However, the mechanism underlying the activation of p300 in cells remains largely unknown. Here, we identify the homeostatic sensor mTORC1 as a direct activator of p300. Activated mTORC1 interacts with p300 and phosphorylates p300 at 4 serine residues in the C-terminal domain. Mechanistically, phosphorylation of p300 by mTORC1 prevents the catalytic HAT domain from binding to the RING domain, thereby eliminating intra-molecular inhibition. Functionally, mTORC1-dependent phosphorylation of p300 suppresses cell-starvation-induced autophagy and activates cell lipogenesis. These results uncover p300 as a direct target of mTORC1 and suggest that the mTORC1-p300 pathway plays a pivotal role in cell metabolism by coordinately controlling cell anabolism and catabolism.


Subject(s)
Autophagy , Lipogenesis , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , p300-CBP Transcription Factors/metabolism , Animals , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Phosphorylation/genetics , Protein Domains , TOR Serine-Threonine Kinases/genetics , p300-CBP Transcription Factors/genetics
5.
Kidney Int ; 105(4): 759-774, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38296028

ABSTRACT

Lupus nephritis (LN) is one of the most severe manifestations of systemic lupus erythematosus (SLE), but its mechanism of onset remains unclear. Since impaired mitophagy has been implicated in multiple organs in SLE, we hypothesized that mitophagy dysfunction is critical in the development of LN and that pharmacologically targeting mitophagy would ameliorate this disease. Therefore, lupus-prone MRL/MpJ-Faslpr (MRL/lpr) and NZBWF1/J mice were treated with a novel mitophagy inducer, UMI-77, during their onset of LN. This treatment effectively mitigated kidney inflammation and damage as assessed by histology and flow cytometry. Furthermore, dendritic cell (DC)-T-cell coculture assay indicated that UMI-77 treatment attenuated DC function that would drive T-cell proliferation but did not directly influence the potent T-cell proliferation in lupus mice. UMI-77 also restored mitochondrial function and attenuated proinflammatory phenotypes in lupus DCs. Adoptive transfer of DCs from MRL/lpr mice augmented serum anti-dsDNA IgG, urine protein and T-cell infiltration of the kidney in MRL/MpJ mice, which could be prevented by either treating lupus donors in vivo or lupus DCs directly with UMI-77. UMI-77 also restored mitochondrial function in myeloid cells from patients with LN in vitro as evidenced by increased ATP levels. Thus, enhancing mitophagy in SLE restrains autoimmunity and limits kidney inflammation for LN development. Hence, our findings suggest targeting mitophagy as a tangible pathway to treat LN.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Sulfonamides , Thioglycolates , Humans , Mice , Animals , Lupus Nephritis/pathology , Autoantigens , Mitophagy , Mice, Inbred MRL lpr , Kidney/pathology , Myeloid Cells , Inflammation/pathology
6.
Genes Dev ; 30(7): 870, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27036968

ABSTRACT

In the above-mentioned article, it has come to the authors' attention that, during the preparation of Figure 5C and Supplemental Figure S2C for the final version of this article, the authors unintentionally assembled incorrect tubulin immunoblots due to similarities in the markings or names, such as FLT3 versus FT, between two similar experiments. The amended versions of these figures are shown below. Neither the quantitative determinations nor the conclusions of this article are altered. The authors apologize for these errors.

7.
FASEB J ; 36(6): e22342, 2022 06.
Article in English | MEDLINE | ID: mdl-35524750

ABSTRACT

Renal fibrosis is the final common outcome of chronic kidney disease (CKD), which remains a huge challenge due to a lack of targeted treatment. Growing evidence suggests that during the process of CKD, the integrity and function of mitochondria in renal tubular epithelial cells (TECs) are generally impaired and strongly connected with the progression of renal fibrosis. Mitophagy, a selective form of autophagy, could remove aberrant mitochondria to maintain mitochondrial homeostasis. Deficiency of mitophagy has been reported to aggravate renal fibrosis. However, whether induction of mitophagy could alleviate renal fibrosis has not been stated. In this study, we explored the effect of mitophagy activation by UMI-77, a compound recently verified to induce mitophagy, on murine CKD model of unilateral ureteral obstruction (UUO) in vivo and TECs in vitro. In UUO mice, we found the changes of mitochondrial damage, ROS production, transforming growth factor (TGF)-ß1/Smad pathway activation, as well as epithelial-mesenchymal transition phenotype and renal fibrosis, and these changes were ameliorated by mitophagy enhancement using UMI-77. Moreover, TEC apoptosis, nuclear factor (NF)-κB signaling activation, and interstitial inflammation after UUO were significantly mitigated by augmented mitophagy. Then, we found UMI-77 could effectively and safely induce mitophagy in TECs in vitro, and reduced TGF-ß1/Smad signaling and downstream profibrotic responses in TGF-ß1-treated TECs. These changes were restored by a mitophagy inhibitor. In conclusion, we demonstrated that mitophagy activation protected against renal fibrosis through improving mitochondrial fitness, downregulating TGF-ß1/Smad signaling and alleviating TEC injuries and inflammatory infiltration in kidneys.


Subject(s)
Renal Insufficiency, Chronic , Animals , Epithelial Cells/metabolism , Fibrosis , Kidney/metabolism , Mice , Mitochondria/metabolism , Mitophagy , NF-kappa B/metabolism , Renal Insufficiency, Chronic/metabolism , Sulfonamides , Thioglycolates , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/metabolism
8.
Org Biomol Chem ; 21(45): 9021-9028, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37927060

ABSTRACT

A three-component reaction of cyclobutanone oxime esters, DABCO·(SO2)2 and N-alkyl-N-methacryloyl benzamides is described. This reaction proceeds without the addition of any oxidant or transition metal, affording sulfonyl-containing isoquinoline-1,3-(2H,4H)-diones in moderate to good yields. Various functional groups are tolerated well in this transformation. Mechanistic studies suggest that a radical pathway is involved, including ß-scission, sulfur dioxide insertion, and intramolecular cyclization processes.

9.
J Chem Inf Model ; 63(16): 5232-5243, 2023 08 28.
Article in English | MEDLINE | ID: mdl-37574904

ABSTRACT

Fatty acids (FAs) are one of the essential energy sources for physiological processes, and they play a vital role in regulating immune and inflammatory responses, promoting cell differentiation and apoptosis, and inhibiting tumor growth. These functions are carried out by FA binding proteins (FABPs) that recognize and transport FAs. Although the crystal structure of the FA-FABPs complex has long been characterized, the mechanism behind FA binding and dissociation from FABP remains unclear. This study employed conventional MD simulations and enhanced sampling technologies to investigate the atomic-scale complexes of heart fatty acid binding proteins and stearic acid (SA). The results revealed two primary pathways for the binding or dissociation of the flexible long-chain ligand, with the orientation of the SA carboxyl head during dissociation determining the chosen path. Conformational changes in the portal region of FABP during the ligand binding/unbinding were found to be trivial, and the overturn of the ″cap″ or the unfolding of the α2 helix was not required. This study resolves the long-standing debate on the binding mechanism of SA with the long-flexible tail to FABP, which significantly improves the understanding of the transport mechanism of FABPs and the development of related therapeutic agents.


Subject(s)
Fatty Acid-Binding Proteins , Neoplasm Proteins , Fatty Acid-Binding Proteins/chemistry , Ligands , Neoplasm Proteins/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Protein Binding
10.
Acta Pharmacol Sin ; 44(7): 1500-1518, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36639570

ABSTRACT

As a major class of medicine for treating the lethal type of castration-resistant prostate cancer (PCa), long-term use of androgen receptor (AR) antagonists commonly leads to antiandrogen resistance. When AR signaling pathway is blocked by AR-targeted therapy, glucocorticoid receptor (GR) could compensate for AR function especially at the late stage of PCa. AR-GR dual antagonist is expected to be a good solution for this situation. Nevertheless, no effective non-steroidal AR-GR dual antagonist has been reported so far. In this study, an AR-GR dual binder H18 was first discovered by combining structure-based virtual screening and biological evaluation. Then with the aid of computationally guided design, the AR-GR dual antagonist HD57 was finally identified with antagonistic activity towards both AR (IC50 = 0.394 µM) and GR (IC50 = 17.81 µM). Moreover, HD57 could effectively antagonize various clinically relevant AR mutants. Further molecular dynamics simulation provided more atomic insights into the mode of action of HD57. Our research presents an efficient and rational strategy for discovering novel AR-GR dual antagonists, and the new scaffold provides important clues for the development of novel therapeutics for castration-resistant PCa.


Subject(s)
Androgen Antagonists , Prostatic Neoplasms , Male , Humans , Androgen Antagonists/pharmacology , Receptors, Glucocorticoid/metabolism , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/pharmacology , Prostatic Neoplasms/metabolism , Cell Line, Tumor
11.
Cytokine ; 138: 155402, 2021 02.
Article in English | MEDLINE | ID: mdl-33352397

ABSTRACT

Alcoholic fatty liver disease (AFLD) is a disease that causes liver damage due to chronic heavy drinking. AFLD is related to lipid accumulation in liver cells caused by alcohol intake. Interleukin-8 (IL-8) is an inflammatory cytokine associated with chemotaxis (deletion in mice) that has robust effects on the occurrence and development of disease by activating related signal transduction pathways to promote inflammation and cell proliferation. There is significantly increased IL-8 expression in liver disease, which may be related to the pathogenesis of AFLD. In this study, we used hydrodynamic injection to deliver the liver-specific expression vector pLIVE-hIL-8 into mice. We found that hIL-8 can exacerbate alcohol-induced fatty liver disease via the Akt/HIF-1α pathway. Exacerbated liver lipid degeneration in mice, which is characterized by excessive accumulation of triglycerides, and liver damage markers were significantly increased. Moreover, hIL-8 could increase the alcohol-induced release of ROS in fatty liver caused by alcohol and exacerbate fatty liver disease. The expression of liver lipid metabolism-related gene sterol regulatory element-binding protein-1c (SREBP-1c) was increased. Furthermore, the expression of peroxisome proliferator-activated receptor alpha (PPARα), which is related to liver fatty acid oxidation, was decreased. The findings obtained in this study of hIL-8 will help identify a potential target for the clinical treatment of AFLD.


Subject(s)
Fatty Liver, Alcoholic/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-8/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Hepatocytes/metabolism , Humans , Inflammation , Lipids/chemistry , Male , Mice , Neutrophils/metabolism , Oxidative Stress , Reactive Oxygen Species , Signal Transduction
12.
Genes Dev ; 27(15): 1718-30, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23913924

ABSTRACT

Missense mutations in the gene TP53, which encodes p53, one of the most important tumor suppressors, are common in human cancers. Accumulated mutant p53 proteins are known to actively contribute to tumor development and metastasis. Thus, promoting the removal of mutant p53 proteins in cancer cells may have therapeutic significance. Here we investigated the mechanisms that govern the turnover of mutant p53 in nonproliferating tumor cells using a combination of pharmacological and genetic approaches. We show that suppression of macroautophagy by multiple means promotes the degradation of mutant p53 through chaperone-mediated autophagy in a lysosome-dependent fashion. In addition, depletion of mutant p53 expression due to macroautophagy inhibition sensitizes the death of dormant cancer cells under nonproliferating conditions. Taken together, our results delineate a novel strategy for killing tumor cells that depend on mutant p53 expression by the activation of chaperone-mediated autophagy and potential pharmacological means to reduce the levels of accumulated mutant p53 without the restriction of mutant p53 conformation in quiescent tumor cells.


Subject(s)
Autophagy/genetics , Molecular Chaperones/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leupeptins/pharmacology , Lysosomes/metabolism , Mutation , Proteasome Endopeptidase Complex/drug effects , Proteolysis/drug effects , Pyrazines/pharmacology , Ubiquitination
13.
Cancer Sci ; 111(10): 3503-3515, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32726482

ABSTRACT

Hepatocellular carcinoma (HCC) represents the majority of liver cancer and is the fourth most common cause of cancer-related death. Although advances in molecular targeted therapy have shown promise, none of these agents has yet demonstrated significant clinical benefit. Bromo- and extraterminal domain (BET) protein inhibitors have been considered potential therapeutic drugs for HCC but the biological activity remains unclear. This study found that BET protein inhibition did not effectively suppress the progression of HCC, using a transgenic HCC mouse model. Mechanistically, the BET protein inhibitor JQ1 upregulated the expression of programmed cell death-ligand 1 (PD-L1) on the plasma membrane in vivo and in vitro. Moreover, JQ1 enhanced the expression of Rab8A, which upregulated the expression of PD-L1 on the plasma membrane. This study also showed that JQ1 combined with anti-PD-L1 Ab effectively suppressed HCC progression, and this benefit was obtained by enhancing the activation and cytotoxic capabilities of CD8 T cells. These results revealed the crucial role and regulation of BET protein inhibition on the expression of PD-L1 in HCC. Thus, combining BET protein inhibition with immune checkpoint blockade offers an efficient therapeutic approach for HCC.


Subject(s)
Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Disease Progression , Hep G2 Cells , Humans , Immunotherapy/methods , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Up-Regulation/drug effects , Up-Regulation/immunology
14.
Chem Pharm Bull (Tokyo) ; 67(8): 786-794, 2019.
Article in English | MEDLINE | ID: mdl-31366828

ABSTRACT

Teriflunomide (TEF, A771726) is the active metabolite of leflunomide (LEF), a disease-modifying anti-rheumatic drug. The main purpose of this study was to develop and evaluate water-in-oil (W/O) microemulsion formulation of TEF. The W/O microemulsion was optimized formula is the physical and chemical stability of lecithin, ethanol, isopropyl myristate (IPM) and water (20.65/20.78/41.52/17.05 w/w) by using the pseudo-ternary phase diagram and the average droplet size is about 40 nm. The permeability of TEF microemulsion is about 6 times higher than control group in vitro penetration test. The results of anti-inflammatory effect showed that compared with the control group, the external TEF microemulsion group could significantly inhibit swelling of paw in rats, and no significant difference compared with oral LEF group. The results of hepatotoxicity test show that there were normal content of alanine aminotransferase (ALT)/aspartate aminotransferase (AST) and no obvious inflammatory infiltration of TEF microemulsion group compared with LEF group. The plasma concentration curve showed that compared with LEF group, the peak concentration of TEF microemulsion group was decreased, the half-life (t1/2) was prolonged, and the relative bioavailability of TEF microemulsion was 75.35%. These results suggest that TEF W/O microemulsion can be used as a promising preparation to play an anti-inflammatory role while significantly reducing hepatotoxicity.


Subject(s)
Antirheumatic Agents/pharmacology , Crotonates/pharmacology , Drug Delivery Systems , Edema/drug therapy , Toluidines/pharmacology , Animals , Antirheumatic Agents/chemistry , Crotonates/chemistry , Drug Compounding , Edema/pathology , Emulsions/chemical synthesis , Emulsions/chemistry , Hydroxybutyrates , Molecular Structure , Nitriles , Oils/chemistry , Pain Measurement , Rats , Rats, Sprague-Dawley , Toluidines/chemistry , Water/chemistry
15.
Org Biomol Chem ; 16(8): 1227-1241, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29431815

ABSTRACT

This review is focused on the recent advances in the C(sp2)-H functionalization of aldehyde-derived hydrazones via a radical process. Diverse substituted hydrazones including N-heterocycles are afforded under mild conditions with excellent selectivities. In general, an aminyl radical as the key intermediate is involved during the reaction process, and a broad reaction scope with good functional group tolerance can be observed. In some cases, a photocatalyst under visible light irradiation is involved, which initiates the radical reaction.

16.
Org Biomol Chem ; 14(5): 1665-9, 2016 Feb 07.
Article in English | MEDLINE | ID: mdl-26700192

ABSTRACT

A facile route to synthesise N-aminosulfonamides through a palladium-catalyzed coupling reaction of aryl nonaflates, sulfur dioxide, and hydrazines is reported. This transformation proceeds in the presence of Pd(OAc)2/XantPhos, and TBAB in 1,4-dioxane at 80 °C, leading to the corresponding N-aminosulfonamides in moderate to good yields. The reaction scope has been demonstrated, and good functional tolerance is observed. A plausible mechanism is proposed through the insertion of sulfur dioxide.

17.
BMC Cancer ; 14: 248, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24716791

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) has been associated with diabetes and obesity, but a possible connection with the metabolic syndrome (MetS) and its potential interaction with hepatitis and cirrhosis are open to discussion. Our previous investigations have shown that GGPPS1 plays a critical role during hyperinsulinism. In this report, the expression and distribution of GGPPS1 in liver cancer, and its clinical significance were investigated. METHODS: 70 patients with hepatocellular carcinoma (HCC) were included in this study. Three different types of tissues from each HCC patient were assembled immediately after surgical resection: tumor-free tissue >5 cm far from tumor edge (TF), adjacent nonmalignant tissue within 2 cm (AT), and tissue from the tumor (TT). Normal liver tissues from 10 liver transplant donors served as healthy control (HC) while 10 patients with liver cirrhosis as cirrhosis control (CC). The expression and distribution of GGPPS1 were detected by immunohistochemistry, western blots, or real-time PCR. The relationship between the expression of GGPPS1 and clinic pathologic index were analyzed. RESULTS: We found that GGPPS1 was intensified mainly in the cytoplasm of liver tumor cells. Both the expression of GGPPS1 mRNA and protein were upregulated in TT comparing to AT or TF. Meanwhile, HCC patients with cirrhosis had relative higher expression of GGPPS1. In addition, many pathologic characters show close correlation with GGPPS1, such as tumor stage, vessel invasion, and early recurrence. CONCLUSION: GGPPS1 may play a critical role during the development of HCC from cirrhosis and is of clinical significance for predicting biological character of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Dimethylallyltranstransferase/genetics , Farnesyltranstransferase/genetics , Geranyltranstransferase/genetics , Liver Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Adult , Aged , Biomarkers, Tumor , Carcinoma, Hepatocellular/pathology , Dimethylallyltranstransferase/biosynthesis , Farnesyltranstransferase/biosynthesis , Female , Fibrosis/complications , Fibrosis/genetics , Fibrosis/pathology , Gene Expression Regulation, Neoplastic , Geranyltranstransferase/biosynthesis , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , RNA, Messenger/biosynthesis
18.
Lupus Sci Med ; 11(1)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38906550

ABSTRACT

OBJECTIVE: Systemic lupus erythematosus (SLE) is a type of autoimmune disease that involves multiple organs involved as well as cytokine dysregulation. The treatment of SLE is still challenging due to the side effects of the different drugs used. Receptor-interacting protein kinase 1 (RIPK1) is a kinase involved in T cell homeostasis and autoinflammation. Although clinical trials have shown that RIPK1 inhibition exhibits significant efficacy in different autoimmune diseases, its role in SLE remains unclear. METHODS: MRL/lpr lupus-prone mice received RIPK1 inhibitor ZJU37 or vehicle intraperitoneally for 10 weeks. A BM12-induced chronic graft-versus-host-disease (cGVHD) lupus-like model was introduced in RIPK1 D138N mice or C57BL/6 mice. Nephritis, serum autoantibody levels, dysregulation of adaptive immune response and cytokines were compared in treated and untreated mice. RESULTS: ZJU37 alleviated the clinical features of the MRL/lpr mice including nephritis and anti-dsDNA antibody production. In addition, ZJU37 treatment reduced the proportion of double-negative T cells in the spleen and the cytokines of TNFα, IFN-γ, IL-6, IL-17 and IL-1ß in the serum. Moreover, RIPK1 D138N mice were able to prevent the cGVHD lupus-like model from SLE attack, manifesting as anti-dsDNA antibody production, the proliferation of germinal centre B cells, plasma cells, and T follicular helper cells as well as IgG and C3 deposits in kidneys. CONCLUSION: RIPK1 inhibition has a protective effect in the mouse model of SLE and can potentially become a new therapeutic target for SLE in humans.


Subject(s)
Cytokines , Disease Models, Animal , Graft vs Host Disease , Lupus Erythematosus, Systemic , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Mice , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , Cytokines/metabolism , Female , Antibodies, Antinuclear/blood , Lupus Nephritis/drug therapy , Lupus Nephritis/immunology , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Spleen/immunology , Spleen/drug effects
19.
CNS Neurosci Ther ; 30(6): e14800, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38887162

ABSTRACT

BACKGROUND: Impaired mitochondrial dynamics have been identified as a significant contributing factor to reduced neurogenesis under pathological conditions. However, the relationship among mitochondrial dynamics, neurogenesis, and spatial memory during normal development remains unclear. This study aims to elucidate the role of mitophagy in spatial memory mediated by neurogenesis during development. METHODS: Adolescent and adult male mice were used to assess spatial memory performance. Immunofluorescence staining was employed to evaluate levels of neurogenesis, and mitochondrial dynamics were assessed through western blotting and transmission electron microscopy. Pharmacological interventions further validated the causal relationship among mitophagy, neurogenesis, and behavioral performance during development. RESULTS: The study revealed differences in spatial memory between adolescent and adult mice. Diminished neurogenesis, accompanied by reduced mitophagy, was observed in the hippocampus of adult mice compared to adolescent subjects. Pharmacological induction of mitophagy in adult mice with UMI-77 resulted in enhanced neurogenesis and prolonged spatial memory retention. Conversely, inhibition of mitophagy with Mdivi-1 in adolescent mice led to reduced hippocampal neurogenesis and impaired spatial memory. CONCLUSION: The observed decline in spatial memory in adult mice is associated with decreased mitophagy, which affects neurogenesis in the dentate gyrus. This underscores the therapeutic potential of enhancing mitophagy to counteract age- or disease-related cognitive decline.


Subject(s)
Hippocampus , Mitophagy , Neurogenesis , Spatial Memory , Animals , Neurogenesis/physiology , Neurogenesis/drug effects , Mitophagy/physiology , Mitophagy/drug effects , Spatial Memory/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondrial Dynamics/physiology , Quinazolinones
20.
Aging Cell ; 23(6): e14143, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38482753

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal disease manifested by premature aging and aging-related phenotypes, making it a disease model for aging. The cellular machinery mediating age-associated phenotypes in HGPS remains largely unknown, resulting in limited therapeutic targets for HGPS. In this study, we showed that mitophagy defects impaired mitochondrial function and contributed to cellular markers associated with aging in mesenchymal stem cells derived from HGPS patients (HGPS-MSCs). Mechanistically, we discovered that mitophagy affected the aging-associated phenotypes of HGPS-MSCs by inhibiting the STING-NF-ĸB pathway and the downstream transcription of senescence-associated secretory phenotypes (SASPs). Furthermore, by utilizing UMI-77, an effective mitophagy inducer, we showed that mitophagy induction alleviated aging-associated phenotypes in HGPS and naturally aged mice. Collectively, our results uncovered that mitophagy defects mediated the aging-associated markers in HGPS, highlighted the function of mitochondrial homeostasis in HGPS progression, and suggested mitophagy as an intervention target for HGPS and aging.


Subject(s)
Mitophagy , Progeria , Progeria/metabolism , Progeria/genetics , Progeria/pathology , Mitophagy/genetics , Humans , Mice , Animals , Aging/metabolism , Cellular Senescence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL