Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
Add more filters

Publication year range
1.
Mol Cell ; 69(2): 238-252.e7, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29351844

ABSTRACT

Maintenance of endoplasmic reticulum (ER) proteostasis is controlled by a dynamic signaling network known as the unfolded protein response (UPR). IRE1α is a major UPR transducer, determining cell fate under ER stress. We used an interactome screening to unveil several regulators of the UPR, highlighting the ER chaperone Hsp47 as the major hit. Cellular and biochemical analysis indicated that Hsp47 instigates IRE1α signaling through a physical interaction. Hsp47 directly binds to the ER luminal domain of IRE1α with high affinity, displacing the negative regulator BiP from the complex to facilitate IRE1α oligomerization. The regulation of IRE1α signaling by Hsp47 is evolutionarily conserved as validated using fly and mouse models of ER stress. Hsp47 deficiency sensitized cells and animals to experimental ER stress, revealing the significance of Hsp47 to global proteostasis maintenance. We conclude that Hsp47 adjusts IRE1α signaling by fine-tuning the threshold to engage an adaptive UPR.


Subject(s)
Endoribonucleases/metabolism , HSP47 Heat-Shock Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , HSP47 Heat-Shock Proteins/physiology , Humans , Mice , Molecular Chaperones/metabolism , Signal Transduction , Stress, Physiological , Transcription Factors/metabolism , Unfolded Protein Response
2.
Trends Biochem Sci ; 45(9): 723-725, 2020 09.
Article in English | MEDLINE | ID: mdl-32616332

ABSTRACT

The endoplasmic reticulum-associated degradation (ERAD) pathway eliminates misfolded proteins. The Hrd1 complex represents the main gate mediating retrotranslocation of ER luminal misfolded (ERAD-L) substrates to the cytosol. A recent cryo-electron microscopy (cryo-EM) study by Wu et al. unveils the structural features of active Hrd1, providing mechanistic insights into the movement of proteins directed for degradation across ER membranes.


Subject(s)
Cryoelectron Microscopy , Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism
3.
Int J Mol Sci ; 25(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39273210

ABSTRACT

The infiltration of immune cells into the central nervous system mediates the development of autoimmune neuroinflammatory diseases. We previously showed that the loss of either Fabp5 or calnexin causes resistance to the induction of experimental autoimmune encephalomyelitis (EAE) in mice, an animal model of multiple sclerosis (MS). Here we show that brain endothelial cells lacking either Fabp5 or calnexin have an increased abundance of cell surface CD200 and soluble CD200 (sCD200) as well as decreased T-cell adhesion. In a tissue culture model of the blood-brain barrier, antagonizing the interaction of CD200 and sCD200 with T-cell CD200 receptor (CD200R1) via anti-CD200 blocking antibodies or the RNAi-mediated inhibition of CD200 production by endothelial cells increased T-cell adhesion and transmigration across monolayers of endothelial cells. Our findings demonstrate that sCD200 produced by brain endothelial cells regulates immune cell trafficking through the blood-brain barrier and is primarily responsible for preventing activated T-cells from entering the brain.


Subject(s)
Antigens, CD , Blood-Brain Barrier , Cell Adhesion , Endothelial Cells , T-Lymphocytes , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/immunology , Animals , Antigens, CD/metabolism , Antigens, CD/genetics , Endothelial Cells/metabolism , Endothelial Cells/immunology , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice, Inbred C57BL , Humans , Brain/metabolism , Brain/immunology
4.
Mol Pharmacol ; 103(3): 158-165, 2023 03.
Article in English | MEDLINE | ID: mdl-36460345

ABSTRACT

Cisplatin is an effective chemotherapeutic agent, yet its use is limited by several adverse drug reactions, known as cisplatin-induced toxicities (CITs). We recently demonstrated that cisplatin could elicit proinflammatory responses associated with CITs through Toll-like receptor 4 (TLR4). TLR4 is best recognized for binding bacterial lipopolysaccharide (LPS) via its coreceptor, MD-2. TLR4 is also proposed to directly bind transition metals, such as nickel. Little is known about the nature of the cisplatin-TLR4 interaction. Here, we show that soluble TLR4 was capable of blocking cisplatin-induced, but not LPS-induced, TLR4 activation. Cisplatin and nickel, but not LPS, were able to directly bind soluble TLR4 in a microscale thermophoresis binding assay. Interestingly, TLR4 histidine variants that abolish nickel binding reduced, but did not eliminate, cisplatin-induced TLR4 activation. This was corroborated by binding data that showed cisplatin, but not nickel, could directly bind mouse TLR4 that lacks these histidine residues. Altogether, our findings suggest that TLR4 can directly bind cisplatin in a manner that is enhanced by, but not dependent on, histidine residues that facilitate binding to transition metals. SIGNIFICANCE STATEMENT: This work describes how the xenobiotic cisplatin interacts with Toll-like receptor 4 (TLR4) to initiate proinflammatory signaling that underlies cisplatin toxicities, which are severe adverse outcomes in cisplatin treatment. Here, this study provides a mechanistic bridge between cisplatin extracellular interactions with TLR4 and previous observations that genetic and chemical inhibition of TLR4 mitigates cisplatin-induced toxicity.


Subject(s)
Cisplatin , Toll-Like Receptor 4 , Animals , Mice , Allergens , Cisplatin/toxicity , Histidine , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/metabolism , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism
5.
J Cell Mol Med ; 28(5): e17839, 2023 Jul 09.
Article in English | MEDLINE | ID: mdl-37424156

ABSTRACT

Endoplasmic reticulum (ER) luminal Ca2+ is vital for the function of the ER and regulates many cellular processes. Calreticulin is a highly conserved, ER-resident Ca2+ binding protein and lectin-like chaperone. Over four decades of studying calreticulin demonstrate that this protein plays a crucial role in maintaining Ca2+ supply under different physiological conditions, in managing access to Ca2+ and how Ca2+ is used depending on the environmental events and in making sure that Ca2+ is not misused. Calreticulin plays a role of ER luminal Ca2+ sensor to manage Ca2+ -dependent ER luminal events including maintaining interaction with its partners, Ca2+ handling molecules, substrates and stress sensors. The protein is strategically positioned in the lumen of the ER from where the protein manages access to and distribution of Ca2+ for many cellular Ca2+ -signalling events. The importance of calreticulin Ca2+ pool extends beyond the ER and includes influence of cellular processes involved in many aspects of cellular pathophysiology. Abnormal handling of the ER Ca2+ contributes to many pathologies from heart failure to neurodegeneration and metabolic diseases.

6.
Cancer Immunol Immunother ; 71(7): 1655-1669, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34800147

ABSTRACT

BACKGROUND: Calreticulin (CRT) is an endoplasmic reticulum (ER) chaperone, but can appear surface bound on cancers cells, including ovarian cancers (OC). We investigated at what stage of cell viability, CRT appeared associated with surface of human OC cells. CRT on pre-apoptotic tumour cells is thought to initiate their eradication via a process termed immunogenic cell death (ICD). METHODS: We treated OC cells with the chemotherapeutic-doxorubicin (DX) known to induce translocation of CRT to some tumour cell surfaces, with and without the ER stressor-thapsigargin (TG)-and/or an ER stress inhibitor-TUDCA. We monitored translocation/release of CRT in pre-apoptotic cells by flow cytometry, immunoblotting and ELISA. We investigated the difference in binding of FITC-CRT to pre-apoptotic, apoptotic and necrotic cells and the ability of extracellular CRT to generate immature dendritic cells from THP-1 monocytes. RESULTS: Dx-treatment increased endogenously released CRT and extracellular FITC_CRT binding to human pre-apoptotic OC cells. DX and TG also promoted cell death in OC cells which also increased CRT release. These cellular responses were significantly inhibited by TUDCA, suggesting that ER stress is partially responsible for the changes in CRT cellular distribution. Extracellular CRT induces maturation of THP-1 towards a imDC phenotype, an important component of ICD. CONCLUSION: Collectively, these cellular responses suggest that ER stress is partially responsible for the changes in CRT cellular distribution. ER-stress regulates in part the release and binding of CRT to human OC cells where it may play a role in ICD.


Subject(s)
Calreticulin , Endoplasmic Reticulum Stress , Ovarian Neoplasms , Apoptosis , Calreticulin/metabolism , Carcinoma, Ovarian Epithelial , Female , Fluorescein-5-isothiocyanate , Humans , Thapsigargin/pharmacology
7.
Molecules ; 27(13)2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35807231

ABSTRACT

Scoulerine is a natural compound that is known to bind to tubulin and has anti-mitotic properties demonstrated in various cancer cells. Its molecular mode of action has not been precisely known. In this work, we perform computational prediction and experimental validation of the mode of action of scoulerine. Based on the existing data in the Protein Data Bank (PDB) and using homology modeling, we create human tubulin structures corresponding to both free tubulin dimers and tubulin in a microtubule. We then perform docking of the optimized structure of scoulerine and find the highest affinity binding sites located in both the free tubulin and in a microtubule. We conclude that binding in the vicinity of the colchicine binding site and near the laulimalide binding site are the most likely locations for scoulerine interacting with tubulin. Thermophoresis assays using scoulerine and tubulin in both free and polymerized form confirm these computational predictions. We conclude that scoulerine exhibits a unique property of a dual mode of action with both microtubule stabilization and tubulin polymerization inhibition, both of which have similar affinity values.


Subject(s)
Antineoplastic Agents , Berberine Alkaloids , Antineoplastic Agents/pharmacology , Berberine Alkaloids/analysis , Binding Sites , Colchicine/chemistry , Humans , Microtubules/metabolism , Molecular Docking Simulation , Tubulin/metabolism , Tubulin Modulators/pharmacology
8.
Prog Mol Subcell Biol ; 59: 1-11, 2021.
Article in English | MEDLINE | ID: mdl-34050859

ABSTRACT

Calreticulin is well known as an ER-resident protein that serves as the major endoplasmic reticulum (ER) Ca2+ binding protein. This protein has been the major topic of discussion in an international workshop that has been meeting for a quarter of a century. In sharing information about this protein, the field also witnessed remarkable insights into the importance of the ER as an organelle and the role of ER Ca2+ in coordinating ER and cellular functions. Recent technological advances have helped to uncover the contributions of calreticulin in maintaining Ca2+ homeostasis in the ER and to unravel its involvement in a multitude of cellular processes as highlighted in this collection of articles. The continuing revelations of unexpected involvement of calreticulin and Ca2+ in many critical aspects of cellular function promises to further improve insights into the significance of this protein in the promotion of physiology as well as prevention of pathology.


Subject(s)
Calreticulin , Endoplasmic Reticulum , Calreticulin/genetics , Calreticulin/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Homeostasis , Lens, Crystalline/metabolism
9.
J Cell Physiol ; 236(4): 2934-2949, 2021 04.
Article in English | MEDLINE | ID: mdl-33037615

ABSTRACT

Transient receptor potential melastatin member 8 (TRPM8), a Ca2+ -permeable nonselective cation channel activated by cold and cooling agents, mediates allodynia. Dysfunction or abnormal expression of TRPM8 has been found in several human cancers. The role of ubiquitination in the regulation of TRPM8 function remains poorly understood. Here, we identified the ubiquitin (Ub)-ligase E3, tripartite motif-containing 4 (TRIM4), as a novel interaction partner of TRPM8 and confirmed that the TRIM4-TRPM8 interaction was mediated through the SPRY domain of TRIM4. Patch-clamp assays showed that TRIM4 negatively regulates TRPM8-mediated currents in HEK293 cells. Moreover, TRIM4 reduced the expression of TRPM8 on the cell surface by promoting the K63-linked ubiquitination of TRPM8. Further analyses revealed that the TRPM8 N-terminal lysine residue at 423 was the major ubiquitination site that mediates its functional regulation by TRIM4. A Ub-activating enzyme E1, Ub-like modifier-activating enzyme 1 (UBA1), was also found to interact with TRPM8, thereby regulating its channel function and ubiquitination. In addition, knockdown of UBA1 impaired the regulation of TRPM8 ubiquitination and function by TRIM4. Thus, this study demonstrates that TRIM4 downregulates TRPM8 via K423-mediated TRPM8 ubiquitination and requires UBA1 to regulate TRPM8.


Subject(s)
Lysine/metabolism , TRPM Cation Channels/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitination , Amino Acid Sequence , Animals , HEK293 Cells , Humans , MCF-7 Cells , Protein Binding , Protein Domains , Rats , Sequence Deletion , Tripartite Motif Proteins/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/metabolism
10.
Biol Reprod ; 105(1): 76-86, 2021 07 02.
Article in English | MEDLINE | ID: mdl-33889948

ABSTRACT

Conditions of impaired energy and nutrient homeostasis, such as diabetes and obesity, are associated with infertility. Hyperglycemia increases endoplasmic reticulum stress as well as oxidative stress and reduces embryo development and quality. Oxidative stress also causes deoxyribonucleic acid damage, which impairs embryo quality and development. The natural bile acid tauroursodeoxycholic acid reduces endoplasmic reticulum stress and rescues developmentally incompetent late-cleaving embryos, as well as embryos subjected to nuclear stress, suggesting the endoplasmic reticulum stress response, or unfolded protein response, and the genome damage response are linked. Tauroursodeoxycholic acid acts via the Takeda-G-protein-receptor-5 to alleviate nuclear stress in embryos. To evaluate the role of tauroursodeoxycholic acid/Takeda-G-protein-receptor-5 signaling in embryo unfolded protein response, we used a model of glucose-induced endoplasmic reticulum stress. Embryo development was impaired by direct injection of tauroursodeoxycholic acid into parthenogenetically activated oocytes, whereas it was improved when tauroursodeoxycholic acid was added to the culture medium. Attenuation of the Takeda-G-protein-receptor-5 precluded the positive effect of tauroursodeoxycholic acid supplementation on development of parthenogenetically activated and fertilized embryos cultured under standard conditions and parthenogenetically activated embryos cultured with excess glucose. Moreover, attenuation of tauroursodeoxycholic acid/Takeda-G-protein-receptor-5 signaling induced endoplasmic reticulum stress, oxidative stress and cell survival genes, but decreased expression of pluripotency genes in parthenogenetically activated embryos cultured under excess glucose conditions. These data suggest that Takeda-G-protein-receptor-5 signaling pathways link the unfolded protein response and genome damage response. Furthermore, this study identifies Takeda-G-protein-receptor-5 signaling as a potential target for mitigating fertility issues caused by nutrient excess-associated blastomere stress and embryo death.


Subject(s)
Cholagogues and Choleretics/pharmacology , Endoplasmic Reticulum Stress/physiology , Oxidative Stress/physiology , Receptors, G-Protein-Coupled/genetics , Sus scrofa/embryology , Taurochenodeoxycholic Acid/pharmacology , Animals , Blastomeres/physiology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Development/physiology , Glucose/adverse effects , Receptors, G-Protein-Coupled/metabolism , Unfolded Protein Response/physiology
11.
FASEB J ; 34(12): 16662-16675, 2020 12.
Article in English | MEDLINE | ID: mdl-33124722

ABSTRACT

We previously showed that calnexin (Canx)-deficient mice are desensitized to experimental autoimmune encephalomyelitis (EAE) induction, a model that is frequently used to study inflammatory demyelinating diseases, due to increased resistance of the blood-brain barrier to immune cell transmigration. We also discovered that Fabp5, an abundant cytoplasmic lipid-binding protein found in brain endothelial cells, makes protein-protein contact with the cytoplasmic C-tail domain of Canx. Remarkably, both Canx-deficient and Fabp5-deficient mice commonly manifest resistance to EAE induction. Here, we evaluated the importance of Fabp5/Canx interactions on EAE pathogenesis and on the patency of a model blood-brain barrier to T-cell transcellular migration. The results demonstrate that formation of a complex comprised of Fabp5 and the C-tail domain of Canx dictates the permeability of the model blood-brain barrier to immune cells and is also a prerequisite for EAE pathogenesis.


Subject(s)
Calnexin/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fatty Acid-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Line , Cell Movement/physiology , Disease Models, Animal , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Permeability
12.
Mol Cancer ; 19(1): 118, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32727463

ABSTRACT

BACKGROUND: Pancreatic cancer is one of the most lethal malignancies and has an extremely poor diagnosis and prognosis. The development of resistance to gemcitabine is still a major challenge. The long noncoding RNA PVT1 was reported to be involved in carcinogenesis and chemoresistance; however, the mechanism by which PVT1 regulates the sensitivity of pancreatic cancer to gemcitabine remains poorly understood. METHODS: The viability of pancreatic cancer cells was assessed by MTT assay in vitro and xenograft tumor formation assay in vivo. The expression levels of PVT1 and miR-619-5p were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Western blotting analysis and qRT-PCR were performed to assess the protein and mRNA levels of Pygo2 and ATG14, respectively. Autophagy was explored via autophagic flux detection under confocal microscopy and autophagic vacuole investigation under transmission electron microscopy (TEM). The functional role and mechanism of PVT1 were further investigated by gain- and loss-of-function assays in vitro. RESULTS: In the present study, we demonstrated that PVT1 was up-regulated in gemcitabine-resistant pancreatic cancer cell lines. Gain- and loss-of-function assays revealed that PVT1 impaired sensitivity to gemcitabine in vitro and in vivo. We further found that PVT1 up-regulated the expression of both Pygo2 and ATG14 and thus regulated Wnt/ß-catenin signaling and autophagic activity to overcome gemcitabine resistance through sponging miR-619-5p. Moreover, we discovered three TCF/LEF binding elements (TBEs) in the promoter region of PVT1, and activation of Wnt/ß-catenin signaling mediated by the up-regulation of Pygo2 increased PVT1 expression by direct binding to the TBE region. Furthermore, PVT1 was discovered to interact with ATG14, thus promoting assembly of the autophagy specific complex I (PtdIns3K-C1) and ATG14-dependent class III PtdIns3K activity. CONCLUSIONS: These data indicate that PVT1 plays a critical role in the sensitivity of pancreatic cancer to gemcitabine and highlight its potential as a valuable target for pancreatic cancer therapy.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Autophagy-Related Proteins/genetics , Autophagy/genetics , Drug Resistance, Neoplasm/genetics , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Wnt Signaling Pathway , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Humans , Mice , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Binding , RNA Interference , Xenograft Model Antitumor Assays , Gemcitabine
13.
Mol Reprod Dev ; 87(1): 161-173, 2020 01.
Article in English | MEDLINE | ID: mdl-31793725

ABSTRACT

DNA damage associated with assisted reproductive technologies is an important factor affecting gamete fertility and embryo development. Activation of the TGR5 receptor by tauroursodeoxycholic acid (TUDCA) has been shown to reduce endoplasmic reticulum (ER) stress in embryos; however, its effect on genome damage responses (GDR) activation to facilitate DNA damage repair has not been examined. This study aimed to investigate the effect of TUDCA on DNA damage repair and embryo development. In a porcine model of ultraviolet light (UV)-induced nuclear stress, TUDCA reduced DNA damage and ER stress in developing embryos, as measured by γH2AX and glucose-regulated protein 78 immunofluorescence, respectively. TUDCA was equally able to rescue early embryo development. No difference in total cell number, DNA damage, or percentage of apoptotic cells, measured by cleaved caspase 3 immunofluorescence, was noted in embryos that reached the blastocyst stage. Interestingly, Dicer-substrate short interfering RNA-mediated disruption of TGR5 signaling abrogated the beneficial effects of TUDCA on UV-treated embryos. Quantitative PCR analysis revealed activation of the GDR, through increased messenger RNA abundance of DNAPK, 53BP1, and DNA ligase IV, as well as the ER stress response, through increased spliced XBP1 and X-linked inhibitor of apoptosis. Results from this study demonstrated that TUDCA activates TGR5-mediated signaling to reduce DNA damage and improve embryo development after UV exposure.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , Embryonic Development/drug effects , Receptors, G-Protein-Coupled/metabolism , Swine/embryology , Taurochenodeoxycholic Acid/pharmacology , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Blastocyst/cytology , Blastocyst/radiation effects , Cells, Cultured , DNA Damage/genetics , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Embryonic Development/genetics , Embryonic Development/radiation effects , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/radiation effects , Female , Fertilization in Vitro/methods , Gene Knockdown Techniques , In Vitro Oocyte Maturation Techniques/methods , Oocyte Retrieval/methods , Ovary/cytology , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Ultraviolet Rays , Unfolded Protein Response/genetics , Unfolded Protein Response/radiation effects , Zygote/radiation effects
14.
FASEB J ; 33(8): 8892-8904, 2019 08.
Article in English | MEDLINE | ID: mdl-31051095

ABSTRACT

The endoplasmic reticulum (ER) plays a central role in cellular stress responses via mobilization of ER stress coping responses, such as the unfolded protein response (UPR). The inositol-requiring 1α (IRE1α) is an ER stress sensor and component of the UPR. Muscle cells also have a well-developed and highly subspecialized membrane network of smooth ER called the sarcoplasmic reticulum (SR) surrounding myofibrils and specialized for Ca2+ storage, release, and uptake to control muscle excitation-contraction coupling. Here, we describe 2 distinct pools of IRE1α in cardiac and skeletal muscle cells, one localized at the perinuclear ER and the other at the junctional SR. We discovered that, at the junctional SR, calsequestrin binds to the ER luminal domain of IRE1α, inhibiting its dimerization. This novel interaction of IRE1α with calsequestrin, one of the highly abundant Ca2+ handling proteins at the junctional SR, provides new insights into the regulation of stress coping responses in muscle cells.-Wang, Q., Groenendyk, J., Paskevicius, T., Qin, W., Kor, K. C., Liu, Y., Hiess, F., Knollmann, B. C., Chen, S. R. W., Tang, J., Chen, X.-Z., Agellon, L. B., Michalak, M. Two pools of IRE1α in cardiac and skeletal muscle cells.


Subject(s)
Endoribonucleases/metabolism , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Binding Sites , COS Cells , Calcium Signaling , Calsequestrin/metabolism , Cells, Cultured , Chlorocebus aethiops , Endoribonucleases/chemistry , Mice , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Rabbits , Sarcoplasmic Reticulum/metabolism
15.
Can J Physiol Pharmacol ; 97(6): 515-527, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31063413

ABSTRACT

Stress responses are important to human physiology and pathology, and the inability to adapt to cellular stress leads to cell death. To mitigate cellular stress and re-establish homeostasis, cells, including those in the cardiovascular system, activate stress coping response mechanisms. The endoplasmic reticulum, a component of the cellular reticular network in cardiac cells, mobilizes so-called endoplasmic reticulum stress coping responses, such as the unfolded protein response. MicroRNAs play an important part in the maintenance of cellular and tissue homeostasis, perform a central role in the biology of the cardiac myocyte, and are involved in pathological cardiac function and remodeling. In this paper, we review a link between endoplasmic reticulum homeostasis and microRNA with an emphasis on the impact on stress responses in the cardiovascular system.


Subject(s)
Cardiovascular System/cytology , Cardiovascular System/metabolism , Endoplasmic Reticulum/metabolism , MicroRNAs/genetics , Animals , Base Sequence , Endoplasmic Reticulum Stress , Homeostasis , Humans , Up-Regulation
16.
Yale J Biol Med ; 91(2): 95-103, 2018 06.
Article in English | MEDLINE | ID: mdl-29955217

ABSTRACT

Nutrition transition, which includes a change from consumption of traditional to modern diets that feature high-energy density and low nutrient diversity, is associated with acquired metabolic syndromes. The human diet is comprised of diverse components which include both nutrients, supplying the raw materials that drive multiple metabolic processes in every cell of the body, and non-nutrients. These components and their metabolites can also regulate gene expression and cellular function via a variety of mechanisms. Some of these components are beneficial while others have toxic effects. Studies have found that persistent disturbance of nutrient metabolism and/or energy homeostasis, caused by either nutrient deficiency or excess, induces cellular stress leading to metabolic dysregulation and tissue damage, and eventually to development of acquired metabolic syndromes. It is now evident that metabolism is influenced by extrinsic factors (e.g., food, xenobiotics, environment), intrinsic factors (e.g., sex, age, gene variations) as well as host/microbiota interaction, that together modify the risk for developing various acquired metabolic diseases. It is also becoming apparent that intake of diets with low-energy density but high in nutrient diversity may be the key to promoting and maintaining optimal health.


Subject(s)
Food , Energy Metabolism , Humans , Metabolic Syndrome
17.
J Cell Mol Med ; 21(12): 3141-3149, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29160038

ABSTRACT

Starting from 1994, every 2 years, an international workshop is organized focused on calreticulin and other endoplasmic reticulum chaperones. In 2017, the workshop took place at Delphi Greece. Participants from North and South America, Europe, Asia and Australia presented their recent data and discussed them extensively with their colleagues. Presentations dealt with structural aspects of calreticulin and calnexin, the role of Ca2+ in cellular signalling and in autophagy, the endoplasmic reticulum stress and the unfolded protein response, the role of calreticulin in immune responses. Several presentations focused on the role of calreticulin and other ER chaperones in a variety of disease states, including haemophilia, obesity, diabetes, Sjogren's syndrome, Chagas diseases, multiple sclerosis, amyotrophic lateral sclerosis, neurological malignancies (especially glioblastoma), haematological malignancies (especially essential thrombocythemia and myelofibrosis), lung adenocarcinoma, renal pathology with emphasis in fibrosis and drug toxicity. In addition, the role of calreticulin and calnexin in growth and wound healing was discussed, as well as the possible use of extracellular calreticulin as a marker for certain diseases. It was agreed that the 13th International Calreticulin Workshop will be organized in 2019 in Montreal, Quebec, Canada.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Calreticulin/genetics , Endoplasmic Reticulum/genetics , Hemophilia A/genetics , Neoplasms/genetics , Obesity/genetics , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy , Calcium/metabolism , Calnexin/genetics , Calnexin/isolation & purification , Calreticulin/immunology , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress , Gene Expression Regulation , Hemophilia A/immunology , Hemophilia A/pathology , Humans , Immunity, Innate , Molecular Chaperones/genetics , Molecular Chaperones/immunology , Neoplasms/immunology , Neoplasms/pathology , Obesity/immunology , Obesity/pathology , Signal Transduction , Unfolded Protein Response , Wound Healing/genetics , Wound Healing/immunology
18.
J Biol Chem ; 291(13): 7045-59, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26861875

ABSTRACT

Within the family of NADPH oxidases, NOX4 is unique as it is predominantly localized in the endoplasmic reticulum, has constitutive activity, and generates hydrogen peroxide (H2O2). We hypothesize that these features are consequences of a so far unidentified NOX4-interacting protein. Two-dimensional blue native (BN) electrophorese combined with SDS-PAGE yielded NOX4 to reside in macromolecular complexes. Interacting proteins were screened by quantitative SILAC (stable isotope labeling of amino acids in cell culture) co-immunoprecipitation (Co-IP) in HEK293 cells stably overexpressing NOX4. By this technique, several interacting proteins were identified with calnexin showing the most robust interaction. Calnexin also resided in NOX4-containing complexes as demonstrated by complexome profiling from BN-PAGE. The calnexin NOX4 interaction could be confirmed by reverse Co-IP and proximity ligation assay, whereas NOX1, NOX2, or NOX5 did not interact with calnexin. Calnexin deficiency as studied in mouse embryonic fibroblasts from calnexin(-/-)mice or in response to calnexin shRNA reduced cellular NOX4 protein expression and reactive oxygen species formation. Our results suggest that endogenous NOX4 forms macromolecular complexes with calnexin, which are needed for the proper maturation, processing, and function of NOX4 in the endoplasmic reticulum.


Subject(s)
Calnexin/genetics , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , NADPH Oxidases/genetics , Animals , Calnexin/antagonists & inhibitors , Calnexin/metabolism , Cell Line , Endoplasmic Reticulum/chemistry , Fibroblasts/cytology , Gene Expression , HEK293 Cells , Humans , Immunoprecipitation , Isotope Labeling , Mice , Mice, Knockout , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
19.
Biochem Biophys Res Commun ; 493(1): 202-206, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28911862

ABSTRACT

Calnexin is a type 1 integral endoplasmic reticulum membrane molecular chaperone with an endoplasmic reticulum luminal chaperone domain and a highly conserved C-terminal domain oriented to the cytoplasm. Fabp5 is a cytoplasmic protein that binds long-chain fatty acids and other lipophilic ligands. Using a yeast two-hybrid screen, immunoprecipitation, microscale thermophoresis analysis and cellular fractionation, we discovered that Fabp5 interacts with the calnexin cytoplasmic C-tail domain at the endoplasmic reticulum. These observations identify Fabp5 as a previously unrecognized calnexin binding partner.


Subject(s)
Calnexin/chemistry , Calnexin/metabolism , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Fatty Acid-Binding Proteins/metabolism , Fibroblasts/metabolism , Neoplasm Proteins/metabolism , Animals , Binding Sites , Cells, Cultured , Cytoplasm/chemistry , Endoplasmic Reticulum/chemistry , Fatty Acid-Binding Proteins/chemistry , Fibroblasts/chemistry , Mice , Neoplasm Proteins/chemistry , Protein Binding , Protein Domains
20.
Adv Exp Med Biol ; 981: 61-76, 2017.
Article in English | MEDLINE | ID: mdl-29594858

ABSTRACT

The endoplasmic reticulum and the other organelles of the eukaryotic cell are membrane-bound structures that carry out specialized functions. In this chapter, we discuss strategies that the cell has adopted to link and coordinate the different activities occurring within its various organelles as the cell carries out its physiological role.


Subject(s)
Endoplasmic Reticulum/physiology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL