Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29.295
Filter
1.
Nat Commun ; 15(1): 8175, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39289391

ABSTRACT

Cerebral dopamine neurotrophic factor (CDNF) is an unconventional neurotrophic factor that is a disease-modifying drug candidate for Parkinson's disease. CDNF has pleiotropic protective effects on stressed cells, but its mechanism of action remains incompletely understood. Here, we use state-of-the-art advanced structural techniques to resolve the structural basis of CDNF interaction with GRP78, the master regulator of the unfolded protein response (UPR) pathway. Subsequent binding studies confirm the obtained structural model of the complex, eventually revealing the interaction site of CDNF and GRP78. Finally, mutating the key residues of CDNF mediating its interaction with GRP78 not only results in impaired binding of CDNF but also abolishes the neuroprotective activity of CDNF-derived peptides in mesencephalic neuron cultures. These results suggest that the molecular interaction with GRP78 mediates the neuroprotective actions of CDNF and provide a structural basis for development of next generation CDNF-based therapeutic compounds against neurodegenerative diseases.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins , Unfolded Protein Response , Endoplasmic Reticulum Chaperone BiP/metabolism , Humans , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Animals , Protein Binding , Nerve Growth Factors/metabolism , Nerve Growth Factors/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neurons/metabolism , Models, Molecular , Binding Sites
2.
J Hematol Oncol ; 17(1): 81, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232809

ABSTRACT

Heat shock proteins are essential molecular chaperones that play crucial roles in stabilizing protein structures, facilitating the repair or degradation of damaged proteins, and maintaining proteostasis and cellular functions. Extensive research has demonstrated that heat shock proteins are highly expressed in cancers and closely associated with tumorigenesis and progression. The "Hallmarks of Cancer" are the core features of cancer biology that collectively define a series of functional characteristics acquired by cells as they transition from a normal state to a state of tumor growth, including sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, enabled replicative immortality, the induction of angiogenesis, and the activation of invasion and metastasis. The pivotal roles of heat shock proteins in modulating the hallmarks of cancer through the activation or inhibition of various signaling pathways has been well documented. Therefore, this review provides an overview of the roles of heat shock proteins in vital biological processes from the perspective of the hallmarks of cancer and summarizes the small-molecule inhibitors that target heat shock proteins to regulate various cancer hallmarks. Moreover, we further discuss combination therapy strategies involving heat shock proteins and promising dual-target inhibitors to highlight the potential of targeting heat shock proteins for cancer treatment. In summary, this review highlights how targeting heat shock proteins could regulate the hallmarks of cancer, which will provide valuable information to better elucidate and understand the roles of heat shock proteins in oncology and the mechanisms of cancer occurrence and development and aid in the development of more efficacious and less toxic novel anticancer agents.


Subject(s)
Heat-Shock Proteins , Neoplasms , Humans , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/physiology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Signal Transduction , Neovascularization, Pathologic/metabolism , Molecular Targeted Therapy/methods
3.
J Cell Mol Med ; 28(17): e70018, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39223962

ABSTRACT

Ferroptosis, an iron-dependent form of cell death, plays a crucial role in the progression of liver injury in Wilson's disease (WD). Gandouling (GDL) has emerged as a potential therapeutic agent for preventing and treating liver injury in WD. However, the precise mechanisms by which GDL mitigates ferroptosis in WD liver injury remain unclear. In this study, we discovered that treating Toxic Milk (TX) mice with GDL effectively decreased liver copper content, corrected iron homeostasis imbalances, and lowered lipid peroxidation levels, thereby preventing ferroptosis and improving liver injury. Bioinformatics analysis and machine learning algorithms identified Hspb1 as a pivotal regulator of ferroptosis. GDL treatment significantly upregulated the expression of HSPB1 and its upstream regulatory factor HSF1, thereby activating the HSF1/HSPB1 pathway. Importantly, inhibition of this pathway by NXP800 reversed the protective effects of GDL on ferroptosis in the liver of TX mice. In conclusion, GDL shows promise in alleviating liver injury in WD by inhibiting ferroptosis through modulation of the HSF1/HSPB1 pathway, suggesting its potential as a novel therapeutic agent for treating liver ferroptosis in WD.


Subject(s)
Ferroptosis , Heat Shock Transcription Factors , Hepatolenticular Degeneration , Liver , Molecular Chaperones , Signal Transduction , Ferroptosis/drug effects , Animals , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Mice , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Hepatolenticular Degeneration/pathology , Molecular Chaperones/metabolism , Liver/metabolism , Liver/drug effects , Liver/pathology , Signal Transduction/drug effects , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Disease Models, Animal , Male , Iron/metabolism , Copper/metabolism , Mice, Inbred C57BL , Humans
4.
Protein Sci ; 33(10): e5173, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39291732

ABSTRACT

Almost all types of cellular stress induce post-translational O-GlcNAc modifications of proteins, and this increase promotes cell survival. We previously demonstrated that O-GlcNAc on certain small heat shock proteins (sHSPs), including HSP27, directly increases their chaperone activity as one potential protective mechanism. Here, we furthered our use of synthetic proteins to prepare biotinylated sHSPs and show that O-GlcNAc modification of HSP27 also changes how it interacts within the sHSP system and the broader HSP network. Specifically, we show that O-GlcNAc modified HSP27 binds more strongly to the co-chaperone protein BAG3, which then promotes refolding of a model substrate by HSP70. We use proteomics to identify other potential HSP27 interactions that are changed by O-GlcNAc, including one that we confirm with another sHSP, αB-crystallin. These findings add additional evidence for O-GlcNAc as a switch for regulating protein-protein interactions and for modifications of chaperones as one mechanism by which O-GlcNAc protects against protein aggregation.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Molecular Chaperones , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/chemistry , HSP27 Heat-Shock Proteins/metabolism , HSP27 Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/chemistry , Acetylglucosamine/metabolism , Acetylglucosamine/chemistry , Protein Refolding , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , Protein Binding , alpha-Crystallin B Chain/chemistry , alpha-Crystallin B Chain/metabolism , Protein Processing, Post-Translational
5.
Sci Rep ; 14(1): 21361, 2024 09 12.
Article in English | MEDLINE | ID: mdl-39266731

ABSTRACT

The heat shock response (HSR) is a universal mechanism of cellular adaptation to elevated temperatures and is regulated by heat shock transcription factor 1 (HSF1) or HSF3 in vertebrate endotherms, such as humans, mice, and chickens. We here showed that HSF1 and HSF3 from egg-laying mammals (monotremes), with a low homeothermic capacity, equally possess a potential to maximally induce the HSR, whereas either HSF1 or HSF3 from birds have this potential. Therefore, we focused on cellular adaptation to daily temperature fluctuations and found that HSF1 was required for the proliferation and survival of human cells under daily temperature fluctuations. The ectopic expression of vertebrate HSF1 proteins, but not HSF3 proteins, restored the resistance in HSF1-null cells, regardless of the induction of heat shock proteins. This function was associated with the up-regulation of specific HSF1-target genes. These results indicate the distinct role of HSF1 in adaptation to thermally fluctuating environments and suggest association of homeothermic capacity with functional diversification of vertebrate HSF genes.


Subject(s)
Adaptation, Physiological , Heat Shock Transcription Factors , Heat-Shock Response , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Animals , Humans , Heat-Shock Response/genetics , Heat-Shock Response/physiology , Adaptation, Physiological/genetics , Temperature , Mice , Cell Proliferation , Chickens/genetics , Cell Survival/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 121(38): e2400781121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39259589

ABSTRACT

During homeostasis, the endoplasmic reticulum (ER) maintains productive transmembrane and secretory protein folding that is vital for proper cellular function. The ER-resident HSP70 chaperone, binding immunoglobulin protein (BiP), plays a pivotal role in sensing ER stress to activate the unfolded protein response (UPR). BiP function is regulated by the bifunctional enzyme filamentation induced by cyclic-AMP domain protein (FicD) that mediates AMPylation and deAMPylation of BiP in response to changes in ER stress. AMPylated BiP acts as a molecular rheostat to regulate UPR signaling, yet little is known about the molecular consequences of FicD loss. In this study, we investigate the role of FicD in mouse embryonic fibroblast (MEF) response to pharmacologically and metabolically induced ER stress. We find differential BiP AMPylation signatures when comparing robust chemical ER stress inducers to physiological glucose starvation stress and recovery. Wildtype MEFs respond to pharmacological ER stress by down-regulating BiP AMPylation. Conversely, BiP AMPylation in wildtype MEFs increases upon metabolic stress induced by glucose starvation. Deletion of FicD results in widespread gene expression changes under baseline growth conditions. In addition, FicD null MEFs exhibit dampened UPR signaling, altered cell stress recovery response, and unconstrained protein secretion. Taken together, our findings indicate that FicD is important for tampering UPR signaling, stress recovery, and the maintenance of secretory protein homeostasis.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Fibroblasts , Glucose , Unfolded Protein Response , Animals , Mice , Embryo, Mammalian/metabolism , Embryo, Mammalian/cytology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Fibroblasts/metabolism , Glucose/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Mice, Knockout , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Signal Transduction
7.
Theranostics ; 14(12): 4643-4666, 2024.
Article in English | MEDLINE | ID: mdl-39239519

ABSTRACT

Rationale: Ferroptosis-driven loss of dopaminergic neurons plays a pivotal role in the pathogenesis of Parkinson's disease (PD). In PD patients, Hspb1 is commonly observed at abnormally high levels in the substantia nigra. The precise consequences of Hspb1 overexpression in PD, however, have yet to be fully elucidated. Methods: We used human iPSC-derived dopaminergic neurons and Coniferaldehyde (CFA)-an Nrf2 agonist known for its ability to cross the blood-brain barrier-to investigate the role of Hspb1 in PD. We examined the correlation between Hspb1 overexpression and Nrf2 activation and explored the transcriptional regulation of Hspb1 by Nrf2. Gene deletion techniques were employed to determine the necessity of Nrf2 and Hspb1 for CFA's neuroprotective effects. Results: Our research demonstrated that Nrf2 can upregulate the transcription of Hspb1 by directly binding to its promoter. Deletion of either Nrf2 or Hspb1 gene abolished the neuroprotective effects of CFA. The Nrf2-Hspb1 pathway, newly identified as a defense mechanism against ferroptosis, was shown to be essential for preventing neurodegeneration progression. Additionally, we discovered that prolonged overexpression of Hspb1 leads to neuronal death and that Hspb1 released from ruptured cells can trigger secondary cell death in neighboring cells, exacerbating neuroinflammatory responses. Conclusions: These findings highlight a biphasic role of Hspb1 in PD, where it initially provides neuroprotection through the Nrf2-Hspb1 pathway but ultimately contributes to neurodegeneration and inflammation when overexpressed. Understanding this dual role is crucial for developing therapeutic strategies targeting Hspb1 and Nrf2 in PD.


Subject(s)
Dopaminergic Neurons , Ferroptosis , Molecular Chaperones , NF-E2-Related Factor 2 , Parkinson Disease , Humans , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Ferroptosis/genetics , Induced Pluripotent Stem Cells/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Animals , Mice , HSP27 Heat-Shock Proteins/metabolism , HSP27 Heat-Shock Proteins/genetics , Cell Death
8.
BMC Pediatr ; 24(1): 568, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39243091

ABSTRACT

BACKGROUND: Newborns are exposed to varying degrees of stressful interventions due to procedures such as heel lancing used in routine metabolic screenings. It is an examination of the effects of white noise and kangaroo care on some physiological parameters and stress markers (cortisol and glucose-regulated protein 78-GRP78) in heel lancing in newborns. METHODS: Randomized controlled study was conducted at a gynecology service of a hospital between January and September 2023. 90 babies were divided into three groups: 30 babies in the Kangaroo Care Group (KCG), 30 babies in the White Music Group (WMG), and 30 babies in the Control Group (CG). All babies were randomly divided into groups. Stress parameters were measured by saliva collection method and physiological parameters by saturation device. RESULTS: A statistically significant difference was determined between the total crying time, pulse and saturation values ​​according to the groups (p < 0.001; p = 0.001). A statistically significant difference was determined between the mean values ​​of cortisol and GRP78 measurements according to group and time interaction (p < 0.001). KCG was more effective in reducing total crying time and stabilizing pulse, saturation, salivary cortisol, GRP-78 values compared to WNG and CG. CONCLUSION: It was concluded that white noise and kangaroo care help reduce newborns' stress in the case of heel lancing. PRACTICAL IMPLICATIONS: The practice of kangaroo care and the use of white noise methods may assist healthcare professionals as supportive methods in stress management during invasive procedures. TRIAL REGISTRATION: NCT06278441, registered on 19/02/2024.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Hydrocortisone , Kangaroo-Mother Care Method , Noise , Saliva , Stress, Physiological , Humans , Infant, Newborn , Hydrocortisone/analysis , Hydrocortisone/metabolism , Female , Saliva/chemistry , Saliva/metabolism , Male , Noise/adverse effects , Heat-Shock Proteins/metabolism , Heel , Crying
9.
Cell Death Dis ; 15(9): 646, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39227586

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that commonly affects children and adolescents with a poor prognosis. The terminal unfolded protein response (UPR) is an emerging anti-cancer approach, although its role in pediatric T-ALL remains unclear. In our pediatric T-ALL cohort from different centers, a lower QRICH1 expression was found associated with a worse prognosis of pediatric T-ALL. Overexpression of QRICH1 significantly inhibited cell proliferation and stimulated apoptosis of T-ALL both in vitro and in vivo. Upregulation of QRICH1 significantly downregulated 78 KDa glucose-regulated protein (GRP78) and upregulated CHOP, thus activating the terminal UPR. Co-overexpression of GRP78 in T-ALL cells overexpressing QRICH1 partially reverted the inhibited proliferation and stimulated apoptosis. QRICH1 bound to the residues Asp212 and Glu155 of the nucleotide-binding domain (NBD) of GRP78, thereby inhibiting its ATP hydrolysis activity. In addition, QRICH1 was associated with endoplasmic reticulum (ER) stress in T-ALL, and overexpression of QRICH1 reversed drug resistance. Overall, low QRICH1 expression is an independent risk factor for a poor prognosis of pediatric T-ALL. By inhibiting GRP78, QRICH1 suppresses pediatric T-ALL.


Subject(s)
Apoptosis , Cell Proliferation , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Endoplasmic Reticulum Chaperone BiP/metabolism , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Child , Cell Proliferation/drug effects , Animals , Apoptosis/drug effects , Male , Cell Line, Tumor , Female , Mice , Endoplasmic Reticulum Stress/drug effects , Unfolded Protein Response/drug effects , Adolescent , Child, Preschool , Prognosis
10.
J Exp Clin Cancer Res ; 43(1): 261, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39267084

ABSTRACT

BACKGROUND: Long-term accumulation of misfolded proteins leads to endoplasmic reticulum (ER) stress in colorectal cancer (CRC). However, the precise pathways controlling the decision between survival and apoptosis in CRC are unclear. Therefore, in this study, we investigated the function and molecular mechanism of glucosidase I (GCS1) in regulating ER stress in CRC. METHODS: A public database was used to confirm the expression level of GCS1 in CRC and normal tissues. Clinical samples from our center were used to confirm the mRNA and protein expression levels of GCS1. Cell proliferation, migration, invasion, and apoptosis assays revealed the biological role of GCS1. Immunohistochemical techniques were used to evaluate the expression of key proteins in subcutaneous implanted tumors in nude mice, which provided further evidence for the biological function of GCS1 in promoting cancer in vivo. The results of coimmunoprecipitation-mass spectrometry analysis and immunofluorescence colocalization analysis the interaction between GCS1 and GRP78. In addition, the mechanism of action of USP10, GRP78, and GCS1 at the post- translational level was investigated. Finally, a tissue microarray was used to examine the connection between GCS1 and GRP78 expression and intracellular localization of these proteins using immunohistochemistry and immunofluorescence. RESULTS: The experimental results revealed that GCS1 was substantially expressed in CRC, with higher expression indicating a worse prognosis. Thus, GCS1 can enhance the proliferation and metastasis while inhibiting the apoptosis of CRC cells both in vivo and in vitro. Mechanistically, GCS1 binds to GRP78, recruits USP10 for deubiquitination of GRP78 to promote its degradation, and decreases ER stress-mediated apoptosis, increasing CRC cell proliferation and metastasis. CONCLUSIONS: In summary, GCS1 stimulates CRC growth and migration and reduces ER stress-mediated apoptosis via USP10-mediated deubiquitination of GRP78. Our findings identify a possible therapeutic target for CRC.


Subject(s)
Colorectal Neoplasms , Disease Progression , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Heat-Shock Proteins , Ubiquitin Thiolesterase , Ubiquitination , Humans , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Endoplasmic Reticulum Chaperone BiP/metabolism , Animals , Mice , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Mice, Nude , Cell Proliferation , Male , Cell Line, Tumor , Apoptosis , Female , Cell Movement
11.
Part Fibre Toxicol ; 21(1): 36, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261835

ABSTRACT

Microplastics (MPs), a brand-new class of worldwide environmental pollutant, have received a lot of attention. MPs are consumed by both humans and animals through water, food chain and other ways, which may cause potential health risks. However, the effects of MPs on embryonic development, especially placental function, and its related mechanisms still need to be further studied. We investigated the impact on fetal development and placental physiological function of pregnant mice by consecutive gavages of MPs at 0, 25, 50, 100 mg/kg body weight during gestational days (GDs 0-14). The results showed that continuous exposure to high concentrations of MP significantly reduced daily weight gain and impaired reproductive performance of pregnant mice. In addition, MPs could significantly induce oxidative stress and placental dysfunction in pregnant mice. On the other hand, MPs exposure significantly decreased placental barrier function and induced placental inflammation. Specifically, MPs treatment significantly reduced the expression of tight junction proteins in placentas, accompanied by inflammatory cell infiltration and increased mRNA levels of pro-inflammatory cytokines and chemokines in placentas. Finally, we found that MPs induced placental apoptosis and endoplasmic reticulum (ER) stress through the GRP78/IRE1α/JNK axis, leading to placental dysfunction and decreased reproductive performance in pregnant mice. We revealed for the first time that the effects of MPs on placental dysfunction in pregnant animals. Blocking the targets of MPs mediated ER stress will provide potential therapeutic ideas for the toxic effects of MPs on maternal pregnancy.


Subject(s)
Apoptosis , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Microplastics , Placenta , Animals , Female , Pregnancy , Endoplasmic Reticulum Chaperone BiP/metabolism , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Placenta/drug effects , Placenta/metabolism , Microplastics/toxicity , Mice , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Fetal Growth Retardation/chemically induced , Mice, Inbred ICR
12.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273100

ABSTRACT

Autophagy is the primary intracellular degradation system, and it plays an important role in many biological and pathological processes. Studies of autophagy involvement in developmental processes are important for understanding various processes. Among them are fibrosis, degenerative diseases, cancer development, and metastasis formation. Diabetic kidney disease is one of the main causes of chronic kidney disease and end-stage renal failure. The aim of this study was to investigate the immunohistochemical expression patterns of LC3B, LAMP2A, and GRP78 during different developmental stages of early-developing human kidneys and in samples from patients with type II diabetes mellitus. During the 7/8th DW, moderate expression of LC3B and LAMP2A and strong expression of GRP78 were found in the mesonephric glomeruli and tubules. In the 9/10th DW, the expression of LC3B and LAMP2A was even more pronounced in the mesonephric tubules. LC3B, LAMP2A, and GRP78 immunoreactivity was also found in the paramesonephric and mesonephric ducts and was stronger in the 9/10th DW compared with the 7/8th DW. In addition, the expression of LC3B, LAMP2A, and GRP78 also appeared in the mesenchyme surrounding the paramesonephric duct in the 9/10th DW. In the 15/16th DW, the expression of LC3B in the glomeruli was weak, that of LAMP2A was moderate, and that of GRP78 was strong. In the tubuli, the expression of LC3B was moderate, while the expression of LAMP2A and GRP78 was strong. The strongest expression of LC3B, LAMP2A, and GRP78 was observed in the renal medullary structures, including developing blood vessels. In postnatal human kidneys, the most extensive LC3B, LAMP2A, and GRP78 expression in the cortex was found in the epithelium of the proximal convoluted tubules, with weak to moderate expression in the glomeruli. The medullary expression of LC3B was weak, but the expression of LAMP2A and GRP78 was the strongest in the medullary tubular structures. Significantly lower expression of LC3B was found in the glomeruli of the diabetic patients in comparison with the nondiabetic patients, but there was no difference in the expression of LC3B in the tubule-interstitial compartment. The expression of LAMP2A was significantly higher in the tubule-interstitial compartments of the diabetic patients in comparison with the nondiabetic patients, while its expression did not differ in the glomeruli. Extensive expression of GRP78 was found in the glomeruli and the tubule-interstitial compartments, but there was no difference in the expression between the two groups of patients. These data give us new information about the expression of LC3B, LAMP2A, and GRP78 during embryonic, fetal, and early postnatal development. The spatiotemporal expression of LC3B, LAMP2A, and GRP78 indicates the important role of autophagy during the early stages of renal development. In addition, our data suggest a disturbance in autophagy processes in the glomeruli and tubuli of diabetic kidneys as an important factor in the pathogenesis of diabetic kidney disease.


Subject(s)
Autophagy , Diabetic Nephropathies , Endoplasmic Reticulum Chaperone BiP , Kidney , Lysosomal-Associated Membrane Protein 2 , Microtubule-Associated Proteins , Humans , Endoplasmic Reticulum Chaperone BiP/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Kidney/metabolism , Kidney/pathology , Microtubule-Associated Proteins/metabolism , Biomarkers/metabolism , Female , Male , Heat-Shock Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology
13.
Drug Dev Res ; 85(7): e22256, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39285641

ABSTRACT

Severe acute pancreatitis (SAP) is characterized by acute inflammation of the pancreas. The transcription factor BTB and CNC homology 1 (BACH1) has been implicated in various biological processes, including oxidative stress, apoptosis, and cell cycle regulation. However, its involvement in the pathogenesis of SAP remains relatively understudied. In the present work, our data demonstrated that BACH1 level was significantly increased in SAP patients, cellular, and animal models, while heat shock protein B1 (HSPB1) expression was weakened. Mechanistic assays validated that BACH1 acted as a transcriptional inhibitor of HSPB1. Moreover, HPDE6-C7 cells were stimulated with cerulein (Cer) and LPS to mimic the pathological stages of SAP in vitro. Depletion of BACH1 remarkably improved cell survival and alleviated the oxidative stress, ferroptosis, and inflammatory responses in SAP cell models. However, these changes were dramatically reversed upon co-inhibition of HSPB1. Animal findings confirmed that loss of BACH1 decreased pancreatic injury, inflammatory responses, and ferroptosis, but these effects were weakened by HSPB1 silence. Overall, these findings elucidate that the overexpression of BACH1 favors the ferroptosis and inflammation by transcriptionally inhibiting HSBP1, thereby exacerbating SAP progression.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Ferroptosis , Pancreatitis , Ferroptosis/drug effects , Humans , Animals , Pancreatitis/genetics , Pancreatitis/metabolism , Pancreatitis/chemically induced , Mice , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Male , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Epigenesis, Genetic , Molecular Chaperones/genetics , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Mice, Inbred C57BL , Cell Line , Disease Models, Animal
14.
Cancer Biol Ther ; 25(1): 2396694, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39215616

ABSTRACT

The incidence of intrahepatic cholangiocarcinoma (ICC) is steadily rising, and it is associated with a high mortality rate. Clinical samples were collected to detect the expression of HSPB8 and BAG3 in ICC tissues. ICC cells were cultured and transfected with plasmids that overexpressed or silenced specific genes to investigate the impact of gene expression alterations on cell function. qPCR and Western blot techniques were utilized to measure gene and protein expression levels. A wound healing assay was conducted to assess cell migration ability. The Transwell assay was used to assess cell invasion ability. Co-IP was used to verify the binding relationship between HSPB8 and BAG3. The effects of HSPB8 and BAG3 on lung metastasis of tumors in vivo were verified by constructing a metastatic tumor model. Through the above experiments, we discovered that the expressions of HSPB8 and BAG3 were up-regulated in ICC tissues and cells, and their expressions were positively correlated. The metastatic ability of ICC cells could be promoted or inhibited by upregulating or downregulating the expression of BAG3. Furthermore, the HSPB8-BAG3 chaperone complex resulted in the abnormal degradation of Filamin A by activating autophagy. Increased expression of Filamin A inhibits the migration and invasion of ICC cells. Overexpression of HSPB8 and BAG3 in vivo promoted the lung metastasis ability of ICC cells. The HSPB8-BAG3 chaperone complex promotes ICC cell migration and invasion by regulating CASA-mediated degradation of Filamin A, offering insights for enhancing ICC therapeutic strategies.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Bile Duct Neoplasms , Cholangiocarcinoma , Filamins , Molecular Chaperones , Neoplasm Invasiveness , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Humans , Filamins/metabolism , Filamins/genetics , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Animals , Mice , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Cell Movement , Cell Line, Tumor , Male , Female , Mice, Nude , Gene Expression Regulation, Neoplastic
15.
Int Immunopharmacol ; 140: 112815, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39088921

ABSTRACT

OBJECTIVE: The purpose of the present study was to potential effects of forsythiaside A (FA) on Sjogren's syndrome (SS). METHODS: Enzyme linked immunosorbent assay for detecting cytokines and Western blotting was used for detecting related protein expression. RESULTS: FA effectively reduced the secretion of inflammatory cytokines, the expression of Caspase-1 and NLRP3 proteins and the expression of p65 in SS. FA also effectively inhibited the high expression of Grp78 in SS. When Grp78 expression was silenced, it effectively reduced the secretion of inflammatory cytokines, the expression of Caspase-1 and NLRP3 proteins and the expression of p65 in the nucleus in SS. FA effectively inhibit the secretion of inflammatory cytokines induced by overexpression of Grp78, the expression of Caspase-1 and NLRP3 proteins and the expression of p65 in the nucleus in SS. CONCLUSION: FA induces the degradation of Grp78 protein, regulates the NF-κB signaling pathway in SS and inhibited NLRP3 inflammasome activation and reduced the release of inflammatory cytokines to alleviate SS.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Sjogren's Syndrome , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Humans , Endoplasmic Reticulum Chaperone BiP/metabolism , Inflammasomes/metabolism , Sjogren's Syndrome/immunology , Sjogren's Syndrome/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Female , Signal Transduction , Cytokines/metabolism , Caspase 1/metabolism , Middle Aged , Male , Transcription Factor RelA/metabolism , NF-kappa B/metabolism
16.
Int Immunopharmacol ; 140: 112836, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39094362

ABSTRACT

Excessive oxidative stress triggers cerebrovascular and neurodegenerative diseases resulting in acute and chronic brain injury. However, the underlying mechanisms remain unknown. Levels of small heat shock protein B8 (HSPB8), which is highly expressed in the brain, are known to be significantly elevated in cerebral injury models. Exogenous HSPB8 protects the brain against mitochondrial damage. One potential mechanism underlying this protection is that HSPB8 overexpression alleviates the mitochondria-dependent pathways of apoptosis; mitochondrial biogenesis, fission, and mitophagy. Overexpression of HSPB8 may therefore have potential as a clinical therapy for cerebrovascular and neurodegenerative diseases. This review provides an overview of advances in the protective effects of HSPB8 against excessive cerebral oxidative stress, including the modulation of mitochondrial dysfunction and potent signaling pathways.


Subject(s)
Heat-Shock Proteins , Mitochondria , Neurons , Oxidative Stress , Humans , Animals , Mitochondria/metabolism , Neurons/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Neurodegenerative Diseases/metabolism , Molecular Chaperones/metabolism , Apoptosis , Signal Transduction , Brain/metabolism , Brain/pathology
17.
Gen Comp Endocrinol ; 357: 114598, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39122124

ABSTRACT

Environmental cues such as temperature induce macroscopic changes in the molting cycle of crustaceans, however, the physiological mechanisms behind these changes remain unclearWe aimed to investigate the regulatory mechanisms in the intermolt and premolt stages of the Callinectes sapidus molt cycle in response to thermal stimuli. The concentration of ecdysteroids and lipids in the hemolymph, and the expression of heat shock proteins (HSPs) and molt key genes were assessed at 19 °C, 24 °C and 29 °C. The premolt animals exhibited a much larger response to the colder temperature than intermolt animals. Ecdysteroids decreased drastically in premolt animals, whereas the expression of their hepatopancreas receptor (CasEcR) increased, possibly compensating for the low hemolymphatic levels at 19 °C. This decrease might be due to increased HSPs and inhibited ecdysteroidogenesis in the Y-organ. In addition, the molting-inhibiting hormone expression in the X-organ/sinus gland (XO/SG) remained constant between temperatures and stages, suggesting it is constitutive in this species. Lipid concentration in the hemolymph, and the expression of CasEcR and CasHSP90 in the XO/SG were influenced by the molting stage, not temperature. On the other hand, the expression of HSPs in the hepatopancreas is the result of the interaction between the two factors evaluated in the study. Our results demonstrated that temperature is an effective modulator of responses related to the molting cycle at the endocrine level and that temperature below the control condition caused a greater effect on the evaluated responses compared to the thermostable condition, especially when the animal was in the premolt stage.


Subject(s)
Brachyura , Ecdysteroids , Hemolymph , Molting , Temperature , Animals , Brachyura/metabolism , Brachyura/physiology , Brachyura/growth & development , Molting/physiology , Hemolymph/metabolism , Ecdysteroids/metabolism , Neurosecretory Systems/metabolism , Neurosecretory Systems/physiology , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Hepatopancreas/metabolism
18.
Animal ; 18(9): 101264, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39116469

ABSTRACT

Heat stress is a significant challenge in dairy cattle herds, affecting milk production and quality, and generating important changes at the cellular level. Most in vitro research on heat shock (HS) effects on dairy cow mammary cells was focused on medium-long-term effects. In recent years, Fourier transform-infrared (FT-IR) micro-spectroscopy has been increasingly used to study the effects of several external stresses on different cell lines, down to the level of single cellular components, such as DNA/RNA, lipids, and proteins. In this study, the possible changes at the biochemical and molecular level induced by acute (30 min-2 h) HS in bovine mammary epithelial (BME-UV1) cells were investigated. The cells were exposed to different temperatures, thermoneutral (TN, 37 °C) and HS (42 °C), and FT-IR spectra were acquired to analyse the effects of HS on biochemical characteristics of BME-UV1 cellular components (proteins, lipids, and DNA/RNA). Moreover, cell viability assay, reactive oxygen species production, and mRNA expression of heat shock proteins (HSPA1A, HSP90AA1, GRP78, GRP94) and antioxidant genes (SOD1, SOD2) by RT-qPCR were also analysed. The FT-IR results showed a change already at 30 min of HS exposure, in the content of long-chain fatty acids, which probably acted as a response to a modification of membrane fluidity in HS cells compared with TN cells. After 2 h of HS exposure, modification of DNA/RNA activity and accumulation of aggregated proteins was highlighted in HS cells. The gene expression analyses showed the overexpression of HSPA1A and HSP90AA1 starting from 30 min up to 2 h in HS cells compared with TN cells. At 2 h of HS exposure, also the overexpression of GRP94 was observed in HS cells. Acute HS did not affect cell viability, reactive oxygen species level, and SOD1 and SOD2 gene expression of BME-UV1 cells. According to the results obtained, cells initiate early defence mechanisms in case of acute HS and probably this efficient response capacity may be decisive for tolerance to heat stress of dairy cattle.


Subject(s)
Epithelial Cells , Heat-Shock Proteins , Heat-Shock Response , Mammary Glands, Animal , Reactive Oxygen Species , Animals , Cattle , Female , Spectroscopy, Fourier Transform Infrared/veterinary , Reactive Oxygen Species/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Cell Survival , Cell Line
19.
EMBO J ; 43(18): 4020-4048, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39134659

ABSTRACT

Sex determination in animals is not only determined by karyotype but can also be modulated by environmental cues like temperature via unclear transduction mechanisms. Moreover, in contrast to earlier views that sex may exclusively be determined by either karyotype or temperature, recent observations suggest that these factors rather co-regulate sex, posing another mechanistic mystery. Here, we discovered that certain wild-isolated and mutant C. elegans strains displayed genotypic germline sex determination (GGSD), but with a temperature-override mechanism. Further, we found that BiP, an ER chaperone, transduces temperature information into a germline sex-governing signal, thereby enabling the coexistence of GGSD and temperature-dependent germline sex determination (TGSD). At the molecular level, increased ER protein-folding requirements upon increased temperatures lead to BiP sequestration, resulting in ERAD-dependent degradation of the oocyte fate-driving factor, TRA-2, thus promoting male germline fate. Remarkably, experimentally manipulating BiP or TRA-2 expression allows to switch between GGSD and TGSD. Physiologically, TGSD allows C. elegans hermaphrodites to maintain brood size at warmer temperatures. Moreover, BiP can also influence germline sex determination in a different, non-hermaphroditic nematode species. Collectively, our findings identify thermosensitive BiP as a conserved temperature sensor in TGSD, and provide mechanistic insights into the transition between GGSD and TGSD.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Germ Cells , Sex Determination Processes , Temperature , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Male , Germ Cells/metabolism , Female , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics
20.
Int J Pharm ; 664: 124583, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39153642

ABSTRACT

Photothermal therapy (PTT) is a prospective therapeutic method for breast cancer. However, excess inflammatory response induced by PTT may aggravate tumor metastasis. Meanwhile, the overexpressed heat shock proteins (HSPs) by cancer cells can protect them from hyperthermia during PTT. Therefore, to attenuate the PTT-induced inflammation and inhibit tumor metastasis, a folate receptor-targeted thermo-sensitive liposome (BI-FA-LP) co-loading Berberine (BBR) and Indocyanine green (ICG) was developed. BI-FA-LP utilized enhanced permeability and retention (EPR) effect and FA receptor-mediated endocytosis to selectively accumulate at tumor, reducing off-target toxicity during the treatment. After targeting to the tumor site, BBR and ICG were released from BI-FA-LP upon laser irradiation, and ICG showed good photothermal performance, while BBR inhibited HSP70 and HSP90 expression during PTT, exerting chemo-photothermal synergetic anti-tumor effect. Moreover, BBR could suppress the PTT induced inflammation, thus inhibiting tumor metastasis and ameliorating tissue injury. Thus, this versatile liposome provided a new strategy to enhance PTT and anti-inflammatory effects for breast cancer treatment.


Subject(s)
Berberine , Breast Neoplasms , Indocyanine Green , Liposomes , Female , Animals , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Indocyanine Green/administration & dosage , Berberine/administration & dosage , Berberine/pharmacology , Mice , Humans , Mice, Inbred BALB C , Cell Line, Tumor , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Photothermal Therapy/methods , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Inflammation/drug therapy , Neoplasm Metastasis/prevention & control , Folate Receptors, GPI-Anchored/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL