Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 901
Filter
1.
Bull Math Biol ; 86(7): 82, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38837083

ABSTRACT

Many neurodegenerative diseases (NDs) are characterized by the slow spatial spread of toxic protein species in the brain. The toxic proteins can induce neuronal stress, triggering the Unfolded Protein Response (UPR), which slows or stops protein translation and can indirectly reduce the toxic load. However, the UPR may also trigger processes leading to apoptotic cell death and the UPR is implicated in the progression of several NDs. In this paper, we develop a novel mathematical model to describe the spatiotemporal dynamics of the UPR mechanism for prion diseases. Our model is centered around a single neuron, with representative proteins P (healthy) and S (toxic) interacting with heterodimer dynamics (S interacts with P to form two S's). The model takes the form of a coupled system of nonlinear reaction-diffusion equations with a delayed, nonlinear flux for P (delay from the UPR). Through the delay, we find parameter regimes that exhibit oscillations in the P- and S-protein levels. We find that oscillations are more pronounced when the S-clearance rate and S-diffusivity are small in comparison to the P-clearance rate and P-diffusivity, respectively. The oscillations become more pronounced as delays in initiating the UPR increase. We also consider quasi-realistic clinical parameters to understand how possible drug therapies can alter the course of a prion disease. We find that decreasing the production of P, decreasing the recruitment rate, increasing the diffusivity of S, increasing the UPR S-threshold, and increasing the S clearance rate appear to be the most powerful modifications to reduce the mean UPR intensity and potentially moderate the disease progression.


Subject(s)
Mathematical Concepts , Models, Neurological , Neurons , Prion Diseases , Unfolded Protein Response , Unfolded Protein Response/physiology , Prion Diseases/metabolism , Prion Diseases/pathology , Prion Diseases/physiopathology , Neurons/metabolism , Humans , Animals , Nonlinear Dynamics , Computer Simulation , Prions/metabolism , Spatio-Temporal Analysis , Apoptosis
2.
Biomed Pharmacother ; 175: 116812, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38781866

ABSTRACT

The endoplasmic reticulum (ER) is important to cells because of its essential functions, including synthesizing three major nutrients and ion transport. When cellular homeostasis is disrupted, ER quality control (ERQC) system is activated effectively to remove misfolded and unfolded proteins through ER-phagy, ER-related degradation (ERAD), and molecular chaperones. When unfolded protein response (UPR) and ER stress are activated, the cell may be suffering a huge blow, and the most probable consequence is apoptosis. The membrane contact points between the ER and sub-organelles contribute to communication between the organelles. The decrease in oxygen concentration affects the morphology and structure of the ER, thereby affecting its function and further disrupting the stable state of cells, leading to the occurrence of disease. In this study, we describe the functions of ER-, ERQC-, and ER-related membrane contact points and their changes under hypoxia, which will help us further understand ER and treat ER-related diseases.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Unfolded Protein Response , Endoplasmic Reticulum/metabolism , Humans , Animals , Endoplasmic Reticulum Stress/physiology , Unfolded Protein Response/physiology , Hypoxia/metabolism , Apoptosis/physiology , Cell Hypoxia/physiology , Endoplasmic Reticulum-Associated Degradation
3.
Acta Neuropathol Commun ; 12(1): 68, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664739

ABSTRACT

Some individuals show a discrepancy between cognition and the amount of neuropathological changes characteristic for Alzheimer's disease (AD). This phenomenon has been referred to as 'resilience'. The molecular and cellular underpinnings of resilience remain poorly understood. To obtain an unbiased understanding of the molecular changes underlying resilience, we investigated global changes in gene expression in the superior frontal gyrus of a cohort of cognitively and pathologically well-defined AD patients, resilient individuals and age-matched controls (n = 11-12 per group). 897 genes were significantly altered between AD and control, 1121 between resilient and control and 6 between resilient and AD. Gene set enrichment analysis (GSEA) revealed that the expression of metallothionein (MT) and of genes related to mitochondrial processes was higher in the resilient donors. Weighted gene co-expression network analysis (WGCNA) identified gene modules related to the unfolded protein response, mitochondrial processes and synaptic signaling to be differentially associated with resilience or dementia. As changes in MT, mitochondria, heat shock proteins and the unfolded protein response (UPR) were the most pronounced changes in the GSEA and/or WGCNA, immunohistochemistry was used to further validate these processes. MT was significantly increased in astrocytes in resilient individuals. A higher proportion of the mitochondrial gene MT-CO1 was detected outside the cell body versus inside the cell body in the resilient compared to the control group and there were higher levels of heat shock protein 70 (HSP70) and X-box-binding protein 1 spliced (XBP1s), two proteins related to heat shock proteins and the UPR, in the AD donors. Finally, we show evidence for putative sex-specific alterations in resilience, including gene expression differences related to autophagy in females compared to males. Taken together, these results show possible mechanisms involving MTs, mitochondrial processes and the UPR by which individuals might maintain cognition despite the presence of AD pathology.


Subject(s)
Alzheimer Disease , Gene Expression Profiling , Metallothionein , Mitochondria , Unfolded Protein Response , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Metallothionein/genetics , Metallothionein/metabolism , Female , Male , Aged , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/pathology , Aged, 80 and over , Resilience, Psychological
4.
Nature ; 618(7966): 849-854, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37286597

ABSTRACT

The mitochondrial unfolded protein response (UPRmt) is essential to safeguard mitochondria from proteotoxic damage by activating a dedicated transcriptional response in the nucleus to restore proteostasis1,2. Yet, it remains unclear how the information on mitochondria misfolding stress (MMS) is signalled to the nucleus as part of the human UPRmt (refs. 3,4). Here, we show that UPRmt signalling is driven by the release of two individual signals in the cytosol-mitochondrial reactive oxygen species (mtROS) and accumulation of mitochondrial protein precursors in the cytosol (c-mtProt). Combining proteomics and genetic approaches, we identified that MMS causes the release of mtROS into the cytosol. In parallel, MMS leads to mitochondrial protein import defects causing c-mtProt accumulation. Both signals integrate to activate the UPRmt; released mtROS oxidize the cytosolic HSP40 protein DNAJA1, which leads to enhanced recruitment of cytosolic HSP70 to c-mtProt. Consequently, HSP70 releases HSF1, which translocates to the nucleus and activates transcription of UPRmt genes. Together, we identify a highly controlled cytosolic surveillance mechanism that integrates independent mitochondrial stress signals to initiate the UPRmt. These observations reveal a link between mitochondrial and cytosolic proteostasis and provide molecular insight into UPRmt signalling in human cells.


Subject(s)
Cytosol , Mitochondria , Proteotoxic Stress , Unfolded Protein Response , Humans , Cell Nucleus/metabolism , Cytosol/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Unfolded Protein Response/physiology , Reactive Oxygen Species/metabolism , Transcriptional Activation , Proteostasis , Proteotoxic Stress/physiology
5.
Circulation ; 147(1): 66-82, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36317534

ABSTRACT

BACKGROUND: Cardiac hypertrophy increases demands on protein folding, which causes an accumulation of misfolded proteins in the endoplasmic reticulum (ER). These misfolded proteins can be removed by the adaptive retrotranslocation, polyubiquitylation, and a proteasome-mediated degradation process, ER-associated degradation (ERAD), which, as a biological process and rate, has not been studied in vivo. To investigate a role for ERAD in a pathophysiological model, we examined the function of the functional initiator of ERAD, valosin-containing protein-interacting membrane protein (VIMP), positing that VIMP would be adaptive in pathological cardiac hypertrophy in mice. METHODS: We developed a new method involving cardiac myocyte-specific adeno-associated virus serovar 9-mediated expression of the canonical ERAD substrate, TCRα, to measure the rate of ERAD, ie, ERAD flux, in the heart in vivo. Adeno-associated virus serovar 9 was also used to either knock down or overexpress VIMP in the heart. Then mice were subjected to transverse aortic constriction to induce pressure overload-induced cardiac hypertrophy. RESULTS: ERAD flux was slowed in both human heart failure and mice after transverse aortic constriction. Surprisingly, although VIMP adaptively contributes to ERAD in model cell lines, in the heart, VIMP knockdown increased ERAD and ameliorated transverse aortic constriction-induced cardiac hypertrophy. Coordinately, VIMP overexpression exacerbated cardiac hypertrophy, which was dependent on VIMP engaging in ERAD. Mechanistically, we found that the cytosolic protein kinase SGK1 (serum/glucocorticoid regulated kinase 1) is a major driver of pathological cardiac hypertrophy in mice subjected to transverse aortic constriction, and that VIMP knockdown decreased the levels of SGK1, which subsequently decreased cardiac pathology. We went on to show that although it is not an ER protein, and resides outside of the ER, SGK1 is degraded by ERAD in a noncanonical process we call ERAD-Out. Despite never having been in the ER, SGK1 is recognized as an ERAD substrate by the ERAD component DERLIN1, and uniquely in cardiac myocytes, VIMP displaces DERLIN1 from initiating ERAD, which decreased SGK1 degradation and promoted cardiac hypertrophy. CONCLUSIONS: ERAD-Out is a new preferentially favored noncanonical form of ERAD that mediates the degradation of SGK1 in cardiac myocytes, and in so doing is therefore an important determinant of how the heart responds to pathological stimuli, such as pressure overload.


Subject(s)
Cardiomegaly , Endoplasmic Reticulum-Associated Degradation , Animals , Humans , Mice , Cardiomegaly/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation/physiology , Myocytes, Cardiac/metabolism , Unfolded Protein Response/physiology
6.
Cancer Treat Res Commun ; 33: 100644, 2022.
Article in English | MEDLINE | ID: mdl-36368296

ABSTRACT

BACKGROUND: Previous studies showed that proline-rich polypeptide (PRP-1) is a ligand for innate immunity toll-like receptors (TLR), and an inhibitor of the mammalian target of rapamycin complex 1 (mTORC1) which induces the death of chondrosarcoma cancer stem cells (CSC). The aim of this study was to investigate the effect of PRP-1 on the regulation of unfolded protein response (UPR) in human chondrosarcoma cells. MATERIALS AND METHODS: Lysates were prepared from a monolayer (bulk or ALDHhigh population), or spheroids chondrosarcoma cell cultures and treated with PRP-1 or control, followed by protein levels quantification by western blotting and mRNA expression by RT-qPCR of protein-RNA-like endoplasmic reticulum kinase (PERK), eukaryotic translation initiation factor 2α (eIF2α), activating transcription factor 4 (ATF4), CCAAT-enhancer-binding protein homologous protein (CHOP), activating transcription factor 6 (ATF6), inositol-requiring enzyme 1 (IRE1α), and X-box binding protein (XBP1). RESULTS: The PRP-1 has been shown to increase the expression of PERK, eIF2α, ATF4, CHOP, ATF6, IRE1α, and XBP1, on both protein and mRNA levels. CONCLUSION: PRP-1 activated UPR branches in monolayer, spheroid, and stem cell populations of human chondrosarcoma.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Toll-Like Receptors , Unfolded Protein Response , Humans , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Ligands , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology
7.
Theranostics ; 12(17): 7289-7306, 2022.
Article in English | MEDLINE | ID: mdl-36438488

ABSTRACT

Rationale: A C9orf72 hexanucleotide repeat expansion (GGGGCC) is the most common genetic origin of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Haploinsufficiency of C9orf72 has been proposed as a possible disease mechanism (loss-of-function mechanism). Additionally, the aberrantly activated unfolded protein response (UPR) and stress granule (SG) formation are associated with the etiopathology of both ALS and FTD. However, the molecular determinants in this pathogenesis are not well characterized. Methods: We performed an immunoprecipitation-mass spectrometry (IP-MS) assay to identify potential proteins interacting with the human C9orf72 protein. We used C9orf72 knockout cell and rat models to determine the roles of C9orf72 in translation initiation and the stress response. Results: Here, we show that C9orf72, which is genetically and pathologically related to ALS and FTD, interacts with eukaryotic initiation factor 2 subunit alpha (eIF2α) and regulates its function in translation initiation. C9orf72 knockout weakens the interaction between eIF2α and eIF2B5, leading to global translation inhibition. Moreover, the loss of C9orf72 results in primary ER stress with activated UPR in rat spleens, which is one of the causes of splenomegaly with inflammation in C9orf72 -/- rats. Finally, C9orf72 delays SG formation by interacting with eIF2α in stressed cells. Conclusions: In summary, these data reveal that C9orf72 modulates translation initiation, the UPR and SG formation, which have implications for understanding ALS/FTD pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein , Frontotemporal Dementia , Animals , Humans , Rats , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , DNA Repeat Expansion , Eukaryotic Initiation Factor-2/genetics , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Stress Granules/genetics , Stress Granules/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology
8.
Adv Sci (Weinh) ; 9(21): e2105469, 2022 07.
Article in English | MEDLINE | ID: mdl-35619328

ABSTRACT

Targeting the G2/M checkpoint mediator WEE1 has been explored as a novel treatment strategy in ovarian cancer, but mechanisms underlying its efficacy and resistance remains to be understood. Here, it is demonstrated that the WEE1 inhibitor AZD1775 induces endoplasmic reticulum stress and activates the protein kinase RNA-like ER kinase (PERK) and inositol-required enzyme 1α (IRE1α) branches of the unfolded protein response (UPR) in TP53 mutant (mtTP53) ovarian cancer models. This is facilitated through NF-κB mediated senescence-associated secretory phenotype. Upon AZD1775 treatment, activated PERK promotes apoptotic signaling via C/EBP-homologous protein (CHOP), while IRE1α-induced splicing of XBP1 (XBP1s) maintains cell survival by repressing apoptosis. This leads to an encouraging synergistic antitumor effect of combining AZD1775 and an IRE1α inhibitor MKC8866 in multiple cell lines and preclinical models of ovarian cancers. Taken together, the data reveal an important dual role of the UPR signaling network in mtTP53 ovarian cancer models in response to AZD1775 and suggest that inhibition of the IRE1α-XBP1s pathway may enhance the efficacy of AZD1775 in the clinics.


Subject(s)
Endoribonucleases , Ovarian Neoplasms , Protein Serine-Threonine Kinases , Benzopyrans , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/metabolism , Female , Humans , Inositol/metabolism , Morpholines , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidinones/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology
9.
Int J Mol Sci ; 23(4)2022 Feb 11.
Article in English | MEDLINE | ID: mdl-35216136

ABSTRACT

Dementia is a global medical and societal challenge; it has devastating personal, social and economic costs, which will increase rapidly as the world's population ages. Despite this, there are no disease-modifying treatments for dementia; current therapy modestly improves symptoms but does not change the outcome. Therefore, new treatments are urgently needed-particularly any that can slow down the disease's progression. Many of the neurodegenerative diseases that lead to dementia are characterised by common pathological responses to abnormal protein production and misfolding in brain cells, raising the possibility of the broad application of therapeutics that target these common processes. The unfolded protein response (UPR) is one such mechanism. The UPR is a highly conserved cellular stress response to abnormal protein folding and is widely dysregulated in neurodegenerative diseases. In this review, we describe the basic machinery of the UPR, as well as the evidence for its overactivation and pathogenicity in dementia, and for the marked neuroprotective effects of its therapeutic manipulation in murine models of these disorders. We discuss drugs identified as potential UPR-modifying therapeutic agents-in particular the licensed antidepressant trazodone-and we review epidemiological and trial data from their use in human populations. Finally, we explore future directions for investigating the potential benefit of using trazodone or similar UPR-modulating compounds for disease modification in patients with dementia.


Subject(s)
Dementia/pathology , Unfolded Protein Response/physiology , Animals , Brain/drug effects , Brain/pathology , Dementia/drug therapy , Humans , Trazodone/pharmacology , Unfolded Protein Response/drug effects
10.
Cell Mol Life Sci ; 79(2): 133, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35165763

ABSTRACT

BACKGROUND AND AIMS: Sec62 is a membrane protein of the endoplasmic reticulum that facilitates protein transport. Its role in cancer is increasingly recognised, but remains largely unknown. We investigated the functional role of Sec62 in gastric cancer (GC) and its underlying mechanism. METHODS: Bioinformatics, tissue microarray, immunohistochemistry (IHC), western blotting (WB), quantitative polymerase chain reaction (qPCR), and immunofluorescence were used to examine the expression of target genes. Transwell, scratch healing assays, and xenograft models were used to evaluate cell migration and invasion. Transmission electron microscopy and mRFP-GFP-LC3 double-labeled adenoviruses were used to monitor autophagy. Co-immunoprecipitation (CO-IP) was performed to evaluate the binding activity between the proteins. RESULTS: Sec62 expression was upregulated in GC, and Sec62 upregulation was an independent predictor of poor prognosis. Sec62 overexpression promoted GC cell migration and invasion both in vitro and in vivo. Sec62 promoted migration and invasion by affecting TIMP-1 and MMP2/9 balance. Moreover, Sec62 could activate autophagy by upregulating PERK/ATF4 expression and binding to LC3II with concomitant FIP200/Beclin-1/Atg5 activation. Furthermore, autophagy blockage impaired the promotive effects of Sec62 on GC cell migration and invasion, whereas autophagy activation rescued the inhibitory effect of Sec62 knockdown on GC metastasis. Notably, Sec62 inhibition combined with autophagy blockage exerted a synergetic anti-metastatic effect in vitro and in vivo. CONCLUSION: Sec62 promotes GC metastasis by activating autophagy and subsequently regulating TIMP-1 and MMP2/9 balance. The activation of autophagy by Sec62 may involve the unfolded protein response (UPR)-related PERK/ATF4 pathway and binding of LC3II during UPR recovery involving FIP200/Beclin-1/Atg5 upregulation. Specifically, the dual inhibition of Sec62 and autophagy may provide a promising therapeutic strategy for GC metastasis.


Subject(s)
Autophagy/physiology , Membrane Transport Proteins/physiology , Stomach Neoplasms/pathology , Unfolded Protein Response/physiology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , Humans , Hydroxychloroquine/pharmacology , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Stomach Neoplasms/mortality , Tissue Inhibitor of Metalloproteinase-1/physiology , eIF-2 Kinase/genetics
11.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35115407

ABSTRACT

Plant root growth is indeterminate but continuously responds to environmental changes. We previously reported on the severe root growth defect of a double mutant in bZIP17 and bZIP28 (bz1728) modulating the unfolded protein response (UPR). To elucidate the mechanism by which bz1728 seedlings develop a short root, we obtained a series of bz1728 suppressor mutants, called nobiro, for rescued root growth. We focused here on nobiro6, which is defective in the general transcription factor component TBP-ASSOCIATED FACTOR 12b (TAF12b). The expression of hundreds of genes, including the bZIP60-UPR regulon, was induced in the bz1728 mutant, but these inductions were markedly attenuated in the bz1728nobiro6 mutant. In view of this, we assigned transcriptional cofactor activity via physical interaction with bZIP60 to NOBIRO6/TAF12b. The single nobiro6/taf12b mutant also showed an altered sensitivity to endoplasmic reticulum stress for both UPR and root growth responses, demonstrating that NOBIRO6/TAF12b contributes to environment-responsive root growth control through UPR.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Factor XII/metabolism , Plant Roots/metabolism , TATA-Binding Protein Associated Factors/metabolism , Unfolded Protein Response/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , Gene Expression Regulation, Plant/physiology , Seedlings/metabolism , Signal Transduction/physiology
12.
Cell Rep ; 38(2): 110206, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021096

ABSTRACT

Mitochondria are known as the powerhouse of the cell. Dysfunction of mitochondria homeostasis induces the mitochondrial unfolded protein response (UPRmt), altering cellular metabolism. How cells sense the UPRmt to rewire metabolism is largely unknown. Here, we show that inactivation of either the citric/tricarboxylic acid (TCA) cycle enzymes aco-2 or idha-1, which encode aconitase and isocitrate dehydrogenase respectively, leads to citrate accumulation. In Caenorhabditis elegans, both in vitro and in vivo, citrate accumulation consequently triggers the UPRmt and also promotes lipid accumulation. The transcription factor DVE-1 binds to the promoter of the nuclear hormone receptor nhr-80 to transactivate its expression. NHR-80 then upregulates lipogenesis and lipid accumulation, shifting excess citrate for use in lipogenesis and for storage as triacylglycerol in lipid droplets. Inactivation of DVE-1 or NHR-80 fully abolishes the citrate-induced lipid accumulation. Therefore, our work uncovers a DVE-1-NHR-80-lipogenesis axis linking the transmission of the mitochondrial stress signal to lipid metabolism.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Citric Acid/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Unfolded Protein Response/physiology , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/physiology , Gene Expression/genetics , Gene Expression Regulation/genetics , Homeostasis , Lipid Metabolism/physiology , Lipids/physiology , Mitochondria/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction , Transcription Factors/metabolism
13.
Cell Signal ; 91: 110241, 2022 03.
Article in English | MEDLINE | ID: mdl-34998932

ABSTRACT

The hyperglycemic microenvironment induced by diabetes mellitus aggravates the inflammatory response, in which the IRE1α signal transduction pathway of the unfolded protein response (UPR) participates. However, the mechanism by which hyperglycemia regulates the IRE1α signaling pathway and affects endoplasmic reticulum (ER) homeostasis in human gingival epithelium in periodontitis with diabetes mellitus remains unknown. Our current data provide evidence that diabetes mellitus causes a hyperinflammatory response in the gingival epithelium, which accelerates periodontal inflammation. Next, we assessed UPR-IRE1α signaling in periodontitis with diabetes mellitus by examining human clinical gingival epithelium samples from healthy subjects, subjects with periodontitis and subjects with periodontitis with diabetes mellitus and by in vitro challenge of human epithelial cells with a hyperglycemic microenvironment. The results showed that a hyperglycemic microenvironment inhibited the IRE1α/XBP1 axis, decreased the expression of a UPR target gene (GRP78), and ultimately impaired the UPR, causing ER stress to be prolonged or more severe in human gingival epithelium. Subsequently, RNA sequencing (RNA-seq) data was analyzed to investigate the expression of ER-related genes in human gingival epithelium. Experiments verified that the mechanism by which periodontitis is aggravated in individuals with diabetes mellitus may involve decreased SERPINH1 expression. Furthermore, experiments in SERPINH1-knockdown and SERPINH1-overexpression models established in vitro indicated that SERPINH1 might act as an activator of IRE1α, maintaining human gingival epithelium homeostasis and reducing proinflammatory cytokine expression by preventing prolonged ER stress induced by high-glucose conditions. In conclusion, regulation of the UPR transducer IRE1α by SERPINH1 alleviates periodontitis with diabetes mellitus by mitigating prolonged ER stress. This finding provides evidence for the further study of periodontitis with diabetes mellitus.


Subject(s)
Diabetes Mellitus , Endoribonucleases , HSP47 Heat-Shock Proteins , Periodontitis , Protein Serine-Threonine Kinases , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/genetics , Endoribonucleases/metabolism , HSP47 Heat-Shock Proteins/genetics , HSP47 Heat-Shock Proteins/metabolism , Humans , Periodontitis/complications , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/physiology
14.
Cell Rep ; 38(1): 110197, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986346

ABSTRACT

AMP-activated protein kinase (AMPK) regulates the balance between cellular anabolism and catabolism dependent on energy resources to maintain proliferation and survival. Small-compound AMPK activators show anti-cancer activity in preclinical models. Using the direct AMPK activator GSK621, we show that the unfolded protein response (UPR) is activated by AMPK in acute myeloid leukemia (AML) cells. Mechanistically, the UPR effector protein kinase RNA-like ER kinase (PERK) represses oxidative phosphorylation, tricarboxylic acid (TCA) cycle, and pyrimidine biosynthesis and primes the mitochondrial membrane to apoptotic signals in an AMPK-dependent manner. Accordingly, in vitro and in vivo studies reveal synergy between the direct AMPK activator GSK621 and the Bcl-2 inhibitor venetoclax. Thus, selective AMPK-activating compounds kill AML cells by rewiring mitochondrial metabolism that primes mitochondria to apoptosis by BH3 mimetics, holding therapeutic promise in AML.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , Unfolded Protein Response/physiology , eIF-2 Kinase/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Line, Tumor , Citric Acid Cycle/drug effects , Drug Evaluation, Preclinical , Female , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Middle Aged , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , THP-1 Cells , U937 Cells , Young Adult
15.
Brain Res Bull ; 181: 97-108, 2022 04.
Article in English | MEDLINE | ID: mdl-35093469

ABSTRACT

Subarachnoid hemorrhage (SAH) is a hemorrhagic stroke disease with high mortality and disability rates. Neurological recovery in early brain injury (EBI) after SAH is a crucial stage to reduce complications and improve the prognosis of patients. The mitochondrial unfolded protein response (UPRmt) is an essential mitochondrial damage repair process, that degrades aggresomes formed by misfolded proteins. UPRmt is a response to cellular stress and enhances mitochondrial homeostasis. GrpEL1 is a nucleotide exchange factor that assists mtHSP70 in nonnative folding proteins in mitochondria. However, the role of UPRmt and GrpEL1 after SAH is unclear. Western blot, Immunofluorescence, Aggresome staining, JC-1 staining were conducted to detect UPRmt after SAH in vivo and in vitro. The results showed that the UPRmt-related proteins HSP60 and mtHSP70 did not change in the EBI after SAH in vivo and in vitro but increased in the isolated mitochondria. In vitro primary neurons treated with oxyhemoglobin (OxyHb) achieved the same result as MG132 induction, increasing neuron protein aggresomes. The expression of GRPEL1 was unchanged in total protein and mitochondrial protein by Western blot. Co-immunoprecipitation (Co-IP) experiments showed that the GRPEL1-mtHSP70 complex decreased after OxyHb treatment. After GRPEL1 overexpression, the GRPEL1-mtHSP70 complex increased, while aggresome in neurons decreased. JC-1 showed an increased mitochondrial membrane potential, ATP content increased, and Western blot analysis revealed decreased cleaved-Caspase 9, suggesting improved mitochondrial function. In conclusion, the reduced GrpEL1-mtHSP70 complex is an essential factor affecting UPRmt in EBI after SAH. Increasing GrpEL1 promotes GrpEL1 and mtHSP70 binding, promoting the neuronal mitochondrial homeostasis, and might be an essential clinical intervention target for EBI after SAH.


Subject(s)
Chaperonin 60/metabolism , HSP70 Heat-Shock Proteins/metabolism , Mitochondrial Proteins/metabolism , Molecular Chaperones/metabolism , Neurons/metabolism , Subarachnoid Hemorrhage/metabolism , Unfolded Protein Response/physiology , Animals , Cells, Cultured , Disease Models, Animal , Mice
16.
Eur J Pharmacol ; 914: 174665, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34861208

ABSTRACT

INTRODUCTION: The function of endoplasmic reticulum (ER), a Ca2+ storage compartment and site of protein folding, is altered by disruption of intracellular homeostasis. Misfolded proteins accumulated in the ER lead to ER stress (ERS), unfolded protein response (UPR) activation and ER Ca2+ loss. Myocardial stunning is a temporary contractile dysfunction, which occurs after brief ischemic periods with minimal or no cell death, being oxidative stress and Ca2+ overload potential underlying mechanisms. Myocardial stunning induces ERS response with negatively impact on the post-ischemic mechanical performance through an unknown mechanism. AIMS: In this study, we explored whether ER Ca2+ efflux through the translocon, a major Ca2+ leak channel, contributes to Ca2+ mishandling and the consequent contractile abnormalities of the stunned myocardium. METHODS: Mechanical performance, cytosolic Ca2+, UPR markers and oxidative state were evaluated in perfused rat/mouse hearts subjected to a brief ischemia followed by reperfusion (I/R) in absence or presence of the translocon inhibitor, emetine (1 µM), comparing its effects with those of the chaperones TUDCA (30 µM) and 4-PBA (3 mM). RESULTS: Emetine treatment precluded the I/R-induced increase in UPR signaling markers and improved the contractile recovery together with a remarkable attenuation in myocardial stiffness when compared to I/R hearts with no drug. This alleviation of I/R-induced mechanical abnormalities was more effective than that obtained with the chemical chaperones, TUDCA and 4-PBA. Moreover, emetine treatment produced a striking improvement in diastolic Ca2+ handling with a partial recovery of the I/R-induced oxidative stress. CONCLUSION: Blocking ER Ca2+ store depletion via translocon suppressed ER stress and improved mechanical performance and diastolic Ca2+ handling of stunned myocardium. Modulation of translocon permeability emerges as a therapeutic approach to face dysfunctional consequences of the I/R injury.


Subject(s)
Calcium/metabolism , Emetine/pharmacology , Endoplasmic Reticulum Stress/drug effects , Myocardial Contraction , Myocardial Stunning , SEC Translocation Channels/antagonists & inhibitors , Unfolded Protein Response , Animals , Calcium Signaling , Mice , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocardial Stunning/drug therapy , Myocardial Stunning/metabolism , Oxidative Stress/drug effects , Protein Synthesis Inhibitors/pharmacology , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Unfolded Protein Response/drug effects , Unfolded Protein Response/physiology
17.
Endocrinology ; 163(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34614512

ABSTRACT

Estrogen and estrogen receptor (ER) play a fundamental role in breast cancer. To support the rapid proliferation of ER+ breast cancer cells, estrogen increases glucose uptake and reprograms glucose metabolism. Meanwhile, estrogen/ER activates the anticipatory unfolded protein response (UPR) preparing cancer cells for the increased protein production required for subsequent cell proliferation. Here, we report that thioredoxin-interacting protein (TXNIP) is an important regulator of glucose metabolism in ER+ breast cancer cells, and estrogen/ER increases glucose uptake and reprograms glucose metabolism via activating anticipatory UPR and subsequently repressing TXNIP expression. In 2 widely used ER+ breast cancer cell lines, MCF7 and T47D, we showed that MCF7 cells express high TXNIP levels and exhibit mitochondrial oxidative phosphorylation (OXPHOS) phenotype, while T47D cells express low TXNIP levels and display aerobic glycolysis (Warburg effect) phenotype. Knockdown of TXNIP promoted glucose uptake and Warburg effect, while forced overexpression of TXNIP inhibited glucose uptake and Warburg effect. We further showed that estrogen represses TXNIP expression and activates UPR sensor inositol-requiring enzyme 1 (IRE1) via ER in the breast cancer cells, and IRE1 activity is required for estrogen suppression of TXNIP expression and estrogen-induced cell proliferation. Our study suggests that TXNIP is involved in estrogen-induced glucose uptake and metabolic reprogramming in ER+ breast cancer cells and links anticipatory UPR to estrogen reprogramming glucose metabolism.


Subject(s)
Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Estrogens/metabolism , Glucose/metabolism , Unfolded Protein Response/physiology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Endoribonucleases/metabolism , Female , Glycolysis , Humans , Lipid Metabolism , MCF-7 Cells , Mitochondria/metabolism , Organoids/metabolism , Oxidative Phosphorylation , Phenotype , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism
18.
Life Sci ; 290: 120226, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34953889

ABSTRACT

The multi-factorial Parkinson's disease (PD) is known to be associated with mitochondrial dysfunction, endoplasmic reticulum stress, alpha synuclein aggregation and dopaminergic neuronal death, with oxidative stress being a common denominator to these underlying processes. The perception of mitochondria being 'just ATP producing compartments' have been counterpoised as studies, particularly related to PD, have underlined their strong role in cause and progression of the disease. During PD pathogenesis, neurons encounter chronic stress conditions mainly due to failure of Mitochondrial Quality Control (MQC) machinery. To dissect the regulatory understanding of mitochondrial dysfunction during neurological disease progression, we endeavored to identify key regulatory endpoints that control multiple facets of MQC machinery. Our studies, employing transgenic C. elegans strain expressing human α-synuclein, led us to identification of mitochondrial genes nuo-5 (involved in oxidative phosphorylation), F25B4.7 (exhibits ATP transmembrane transporter activity) and C05D11.9 (having ribonuclease activity), which form predicted downstream targets of most elevated and down-regulated mi-RNA molecules. RNAi mediated silencing, gene ontology and functional genomics analysis studies demonstrated their role in modulating major MQC pathways. The attenuated MQC pathways mainly affected clearance of misfolded and aggregated proteins, redox homeostasis and longevity with compromised dopaminergic functions. Overexpression of the mitochondrial genes by 3 beta-hydroxyl steroid, Tomatidine, was found to curtail the redox imbalance thus leading to amelioration of effects associated with PD and an increase in the lifespan of treated nematodes. Therefore, this study unveils the regulatory role of mitochondrial genes as critical modulators of stress control involved in effects associated with PD pathogenesis.


Subject(s)
Caenorhabditis elegans/genetics , DNA, Mitochondrial/genetics , Stress, Physiological/genetics , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , DNA, Mitochondrial/metabolism , Dopaminergic Neurons/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Stress/genetics , Oxidative Stress/physiology , Parkinson Disease/metabolism , Stress, Physiological/physiology , Unfolded Protein Response/physiology , alpha-Synuclein/metabolism
19.
Neurochem Int ; 151: 105218, 2021 12.
Article in English | MEDLINE | ID: mdl-34732355

ABSTRACT

After ischemic stroke or cardiac arrest, brain ischemia occurs. Currently, no pharmacologic intervention that targets cellular processes has proven effective in improving neurologic outcome in patients after brain ischemia. Recent experimental research has identified the crucial role of proteostasis in survival and recovery of cells after ischemia. In particular, the unfolded protein response (UPR), a key signaling pathway that safeguards cellular proteostasis, is emerging as a promising therapeutic target for brain ischemia. For some time, the UPR has been known to play a critical role in the pathophysiology of brain ischemia; however, only in the recent years has the field grown substantially, largely due to the extensive use of UPR-specific mouse genetic models and the rapidly expanding availability of pharmacologic tools that target the UPR. In this review, we provide a timely update on the progress in our understanding of the UPR in experimental brain ischemia, and discuss the therapeutic implications of targeting the UPR in ischemic stroke and cardiac arrest.


Subject(s)
Brain Ischemia/drug therapy , Heart Arrest/metabolism , Proteostasis/drug effects , Stroke/drug therapy , Unfolded Protein Response/drug effects , Animals , Brain Ischemia/metabolism , Heart Arrest/drug therapy , Humans , Proteostasis/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Stroke/metabolism , Unfolded Protein Response/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...