Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
EMBO Rep ; 24(6): e56128, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37042626

ABSTRACT

Surgery-induced renal ischemia and reperfusion (I/R) injury and nephrotoxic drugs like cisplatin can cause acute kidney injury (AKI), for which there is no effective therapy. Lipid accumulation is evident following AKI in renal tubules although the mechanisms and pathological effects are unclear. Here, we report that Ehmt2-encoded histone methyltransferase G9a is upregulated in patients and mouse kidneys after AKI. Renal tubular specific knockout of G9a (Ehmt2Ksp ) or pharmacological inhibition of G9a alleviates lipid accumulation associated with AKI. Mechanistically, G9a suppresses transcription of the lipolytic enzyme Ces1; moreover, G9a and farnesoid X receptor (FXR) competitively bind to the same promoter regions of Ces1. Ces1 is consistently observed to be downregulated in the kidney of AKI patients. Pharmacological inhibition of Ces1 increases lipid accumulation, exacerbates renal I/R-injury and eliminates the beneficial effects on AKI observed in Ehmt2Ksp mice. Furthermore, lipid-lowering atorvastatin and an FXR agonist alleviate AKI by activating Ces1 and reducing renal lipid accumulation. Together, our results reveal a G9a/FXR-Ces1 axis that affects the AKI outcome via regulating renal lipid accumulation.


Subject(s)
Acute Kidney Injury , Kidney Tubules , Mice , Animals , Kidney Tubules/metabolism , Kidney Tubules/pathology , Acute Kidney Injury/genetics , Acute Kidney Injury/chemically induced , Lipids , Kidney/pathology , Mice, Inbred C57BL
2.
J Cell Physiol ; 237(1): 98-117, 2022 01.
Article in English | MEDLINE | ID: mdl-34289108

ABSTRACT

Angiopoietin-like proteins (ANGPTLs), a family of eight secreted glycoproteins termed ANGTPL1-8, are involved in angiogenesis, lipid metabolism, cancer progression, and inflammation. Their roles in regulating lipid metabolism have been intensively studied, as some ANGPTLs are promising pharmacological targets for hypertriglyceridemia and associated cardiovascular disease. Recently, the emerging roles of ANGPTLs in inflammation have attracted great attention. First, elevated levels of multiple circulating ANGPTLs in inflammatory diseases make them potential disease biomarkers. Second, multiple ANGPTLs regulate acute or chronic inflammation via various mechanisms, including triggering inflammatory signaling through their action as ligands for integrin or forming homo- /hetero-oligomers to regulate signal transduction via extra- or intracellular mechanisms. As dysregulation of the inflammatory response is a critical trigger in many diseases, understanding the roles of ANGPTLs in inflammation will aid in drug/therapy development. Here, we summarize the roles, mechanisms, and potential therapeutic values for ANGPTLs in inflammation and inflammatory diseases.


Subject(s)
Angiopoietins , Inflammation , Angiopoietin-like Proteins/genetics , Angiopoietin-like Proteins/metabolism , Angiopoietins/metabolism , Humans , Inflammation/drug therapy , Lipid Metabolism , Neovascularization, Pathologic/drug therapy
3.
Arch Biochem Biophys ; 700: 108768, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33485848

ABSTRACT

Numerous neurological and non-neurological disorders are associated with dysfunction of epigenetic modulators, and methyl CpG binding protein 2 (MeCP2) is one of such proteins. Initially identified as a transcriptional repressor, MeCP2 specifically binds to methylated DNA, and mutations of MeCP2 have been shown to cause Rett syndrome (RTT), a severe neurological disorder. Recently, accumulating evidence suggests that ubiquitously expressed MeCP2 also plays a central role in non-neurological disorders including cardiac dysfunction, liver injury, respiratory disorders, urological dysfunction, adipose tissue metabolism disorders, movement abnormality and inflammatory responses in a DNA methylation dependent or independent manner. Despite significant progresses in our understanding of MeCP2 over the last few decades, there is still a considerable knowledge gap to translate the in vitro and in vivo experimental findings into therapeutic interventions. In this review, we provide a synopsis of the role of MeCP2 in the pathophysiology of non-neurological disorders, MeCP2-based research directions and therapeutic strategies for non-neurological disorders are also discussed.


Subject(s)
DNA Methylation , DNA/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Animals , DNA/genetics , Humans , Methyl-CpG-Binding Protein 2/genetics , Rett Syndrome/metabolism
4.
FASEB J ; 34(3): 4732-4748, 2020 03.
Article in English | MEDLINE | ID: mdl-32030835

ABSTRACT

Repeated cycles of weight loss and regain, known as weight cycling, is often seen when people try to lose weight. The exact pathophysiological effects and the underlying mechanisms of weight cycling remain largely unclear. Here, we report that weight cycling induced by alternating feeding mice with a low-fat diet or a high-fat diet in a 1-week switch protocol caused further increased epididymal white adipose tissue (eWAT) weight, preadipocyte proliferation, hepatic inflammation, fasting blood glucose level, and glucose intolerance, compared with the continuously HF-fed mice. Combining the secretory protein database with RNA-sequencing and quantitative PCR (qPCR) results in eWAT, the mRNA levels of several adipokines, including Retn (encoding resistin), were found altered by weight cycling. A transcriptional co-factor Lmo4 was found regulated by weight cycling; Lmo4 enhanced preadipocyte proliferation, in vitro adipogenesis, transcription of Retn, and resistin secretion in 3T3-L1 cells. Primary mouse hepatocytes administrated with recombinant mouse resistin (rm-resistin), or exposed to media from Lmo4-overexpressed 3T3-L1 cells, showed increased inflammatory responses and gluconeogenesis. Furthermore, rm-resistin-injected normal chow-fed mice showed upregulated blood glucose level by increasing gluconeogenesis, and upregulated the hepatic inflammatory responses. Together, our results suggest a regulatory role of Lmo4-resistin signaling in weight cycling, indicating a crosstalk between the adipose tissue and liver.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/metabolism , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/metabolism , LIM Domain Proteins/metabolism , 3T3-L1 Cells , Adaptor Proteins, Signal Transducing/blood , Adaptor Proteins, Signal Transducing/genetics , Adipokines/blood , Adipokines/genetics , Adipokines/metabolism , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Immunohistochemistry , Inflammation/immunology , Inflammation/metabolism , LIM Domain Proteins/blood , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Resistin/blood , Resistin/genetics , Resistin/metabolism , Sequence Analysis, RNA , Weight Gain/genetics , Weight Gain/physiology
5.
J Hepatol ; 73(3): 603-615, 2020 09.
Article in English | MEDLINE | ID: mdl-32593682

ABSTRACT

BACKGROUND & AIMS: Obesity is an independent risk factor for malignancies, including hepatocellular carcinoma (HCC). However, it remains unknown whether maternal obesity affects the incidence of HCC in offspring. Thus, we aimed to investigate this association and its underlying mechanisms. METHODS: Diethylnitrosamine (DEN) was used to induce HCC in a high-fat diet (HFD)-induced multigenerational obesity model. RNA-sequencing was performed to identify the genes and microRNAs (miRNAs) that were altered over generations. The role of the miR-27a-3p-Acsl1/Aldh2 axis in HCC was evaluated in cell lines and HCC-bearing nude mice, and its intergenerational impact was studied in pregnant mice and their offspring. RESULTS: Under HFD stress, maternal obesity caused susceptibility of offspring to DEN-induced HCC, and such susceptibility was cumulative over generations. We identified that Acsl1 and Aldh2, direct targets of miR-27a-3p, were gradually changed over generations. Under hyperlipidemic conditions, downregulation of Acsl1 and Aldh2 increased cell proliferation (in vitro) or tumor growth (in vivo) in synergy. Intratumor injection of an miR-27a-3p agomir exacerbated tumor growth by downregulating Acsl1 and Aldh2; while intratumor injection of an miR-27a-3p antagomir had the opposite effect. Moreover, serum miR-27a-3p levels gradually increased in the HFD-fed maternal lineage over generations. Injecting pregnant mice with an miR-27a-3p agomir not only upregulated hepatic miR-27a-3p and downregulated Acsl1/Aldh2 in offspring (fetus, young and adult stages), but also exacerbated HCC development in DEN-treated offspring. In human HCC, upregulated miR-27a-3p and downregulated Acsl1/Aldh2 were negatively correlated with survival on TCGA analysis; while, hepatic miR-27a-3p was negatively correlated with Acsl1/Aldh2 expression in tumor/non-tumor tissues from fatty/non-fatty livers. CONCLUSIONS: Maternal obesity plays a role in regulating cumulative susceptibility to HCC development in offspring over multiple generations through the miR-27a-3p-Acsl1/Aldh2 axis. LAY SUMMARY: It is not currently known how maternal obesity affects the incidence of liver cancer in offspring. In this study, we identified a microRNA (miR-27a-3p) that was upregulated in obese mothers and could be passed on to their offspring. This microRNA enhanced the risk of liver cancer in offspring by regulating 2 genes (Acsl1 and Aldh2). This mechanism could be a future therapeutic target.


Subject(s)
Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/epidemiology , Liver Neoplasms/chemically induced , Liver Neoplasms/epidemiology , MicroRNAs/metabolism , Obesity, Maternal/complications , Obesity, Maternal/metabolism , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Diet, High-Fat/adverse effects , Diethylnitrosamine/adverse effects , Disease Models, Animal , Female , Gene Knockdown Techniques , Hep G2 Cells , Humans , Incidence , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , MicroRNAs/genetics , Obesity, Maternal/etiology , Pregnancy , Transfection , Tumor Burden/genetics , Xenograft Model Antitumor Assays
6.
Biochim Biophys Acta Gen Subj ; 1862(10): 2226-2235, 2018 10.
Article in English | MEDLINE | ID: mdl-30036601

ABSTRACT

Protein misfolding and aggregation are associated with amyloidosis. The toxic aggregation of amyloid-ß 1-42 (Aß42) may disrupt cell membranes and lead to cell death and is thus regarded as a contributing factor in Alzheimer's disease (AD). 1,4-naphthoquinone (NQ) has been shown to exhibit strong anti-aggregation effects on amyloidogenic proteins such as insulin and α-synuclein; however, its high toxicity and poor solubility limit its clinical application. Menadione sodium bisulfite (MSB, also known as vitamin K3), is used clinically in China to treat hemorrhagic diseases caused by vitamin K deficiency and globally as a vitamin K supplement. We hypothesized that MSB could inhibit amyloid formation since its backbone structure is similar to NQ. To test our hypothesis, we first investigated the effects of MSB on Aß42 amyloid formation in vitro. We found that MSB inhibited Aß42 amyloid formation in a dose dependent manner, delayed the secondary structural conversion of Aß42 from random coil to ordered ß-sheet, and attenuated the ability of Aß42 aggregates to disrupt membranes; moreover, the quinone backbone rather than lipophilicity is esstial for the inhibitory effects of MSB. Next, in cells expressing a pathogenic APP mutation (Osaka mutation) that results in the formation of intraneuronal Aß oligomers, MSB inhibited the intracellular aggregation of Aß. Moreover, MSB treatment significantly extended the life span of Caenorhabditis elegans CL2120, a strain that expresses human Aß42. Together, these results suggest that MSB and its derivatives may be further explored as potential therapeutic agents for the prevention or treatment of AD.


Subject(s)
Amyloid beta-Peptides/chemistry , Animals, Genetically Modified/growth & development , Caenorhabditis elegans/growth & development , Peptide Fragments/chemistry , Protein Aggregation, Pathological/prevention & control , Vitamin K 3/pharmacology , Vitamins/pharmacology , Amyloid beta-Peptides/drug effects , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Humans , Longevity , Peptide Fragments/drug effects
7.
Mol Ther ; 25(1): 127-139, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28129109

ABSTRACT

Dominant missense mutations in TAR DNA-binding protein 43 (TDP-43) cause amyotrophic lateral sclerosis (ALS), and the cytoplasmic accumulation of TDP-43 represents a pathological hallmark in ALS and frontotemporal lobar degeneration (FTD). Behavioral investigation of the transgenic mouse model expressing the disease-causing human TDP-43 M337V mutant (TDP-43M337V mice) is encumbered by premature death in homozygous transgenic mice and a reported lack of phenotype assessed by tail elevation and footprint in hemizygous transgenic mice. Here, using a battery of motor-coordinative and cognitive tests, we report robust motor-coordinative and cognitive deficits in hemizygous TDP-43M337V mice by 8 months of age. After 12 months of age, cortical neurons are significantly affected by the mild expression of mutant TDP-43, characterized by cytoplasmic TDP-43 mislocalization, mitochondrial dysfunction, and neuronal loss. Compared with age-matched non-transgenic mice, TDP-43M337V mice demonstrate a similar expression of total TDP-43 but higher levels of TDP-43 in mitochondria. Interestingly, a TDP-43 mitochondrial localization inhibitory peptide abolishes cytoplasmic TDP-43 accumulation, restores mitochondrial function, prevents neuronal loss, and alleviates motor-coordinative and cognitive deficits in adult hemizygous TDP-43M337V mice. Thus, this study suggests hemizygous TDP-43M337V mice as a useful animal model to study TDP-43 toxicity and further consolidates mitochondrial TDP-43 as a novel therapeutic target for TDP-43-linked neurodegenerative diseases.


Subject(s)
Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Psychomotor Performance , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Brain/cytology , Brain/metabolism , Brain/pathology , Locomotion , Mice , Mice, Transgenic , Motor Activity , Muscle Strength , Neurons/metabolism , Peptide Fragments , Protein Transport
8.
Biochim Biophys Acta ; 1860(4): 719-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26777153

ABSTRACT

BACKGROUND: The misfolding of human islet amyloid polypeptide (hIAPP) is an important pathological factor on the onset of type 2 diabetes. A number of studies have been focused on His(18), the only histidine of hIAPP, whose imidazole ring and the protonation state might impact hIAPP amyloid formation, but the exact mechanism remains unclear. METHODS: We used diethylpyrocarbonate (DEPC) to specifically modify His(18) and obtained mono-ethyloxyformylated hIAPP (DMI). Thioflavin T based fluorescence, transmission electronic microscopy, circular dichroism spectroscopy, fluorescence dye leakage, Fourier transform infrared spectroscopy and replica-exchange molecular dynamics (REMD) simulation were applied to study the impact of DEPC-modification on hIAPP amyloid formation. RESULTS: After an ethyl-acetate group was introduced to the His(18) of hIAPP by diethylpyrocarbonate (DEPC) modification, the pH dependent hIAPP fibrillation went to the opposite order and the number of intra-molecular hydrogen bonds decreased, while the possibility of His(18) participating in the formation of α-helical structures increased. Furthermore, the membrane-peptide interaction and ion-peptide interaction were both impaired. CONCLUSIONS: The intramolecular hydrogen bond formation by His(18) and the possibility of His(18) participating in the formation of α-helical structures greatly modulated the manner of hIAPP amyloid formation. The imidazole ring directly participates in the hIAPP-membrane/ion interaction. GENERAL SIGNIFICANCE: DEPC modification is an alternative approach to investigate the role of the imidazole ring during amyloid formation.


Subject(s)
Diethyl Pyrocarbonate/chemistry , Imidazoles/chemistry , Islet Amyloid Polypeptide/chemistry , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Thiazoles/chemistry , Benzothiazoles , Humans , Islet Amyloid Polypeptide/genetics , Islet Amyloid Polypeptide/metabolism , Protein Structure, Secondary
9.
Biochim Biophys Acta ; 1852(7): 1278-87, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25748499

ABSTRACT

Renal ischemia/reperfusion (I/R) injury is the most common cause of acute kidney injury, having a high rate of mortality and no effective therapy currently available. Apelin-13, a bioactive peptide, has been shown to inhibit the early lesions of diabetic nephropathy in several mouse models by us and others. To test whether apelin-13 protects against renal I/R induced injury, male rats were exposed to renal I/R injury with or without apelin-13 treatment for 3 days. Apelin-13 treatment markedly reduced the injury-induced tubular lesions, renal cell apoptosis, and normalized the injury induced renal dysfunction. Apelin-13 treatment inhibited the injury-induced elevation of inflammatory factors and Tgf-ß1, as well as apoptosis. Apelin-13 treatment also inhibited the injury-induced elevation of histone methylation and Kmt2d, a histone methyltransferase of H3K4me2, following renal I/R injury. Furthermore, in cultured renal mesangial and tubular cells, apelin-13 suppressed the injury-induced elevation of Tgf-ß1, apoptosis, H3K4me2 and Kmt2d under the in vitro hypoxia/reperfusion (H/R) conditions. Consistently, over-expression of apelin significantly inhibited H/R-induced elevation of TGF-ß1, apoptosis, H3K4me2 and Kmt2d. The present study therefore suggests apelin-13 may be a therapeutic candidate for treating acute kidney injury.


Subject(s)
Intercellular Signaling Peptides and Proteins/pharmacology , Kidney/blood supply , Reperfusion Injury/prevention & control , Transforming Growth Factor beta1/metabolism , Animals , Apoptosis , Cell Line , Histones/metabolism , Intercellular Signaling Peptides and Proteins/therapeutic use , Kidney/drug effects , Kidney/metabolism , Male , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Transforming Growth Factor beta1/genetics
10.
FASEB J ; 29(7): 2749-58, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25805836

ABSTRACT

Ischemia and reperfusion (I/R) injury is a common cause of many vascular and neuronal diseases. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) has been found down-regulated or dysfunctional in several tissues upon I/R injury. To investigate the role of GAPDH in retinal I/R injury-induced neurovascular degeneration, the injured retinas of GAPDH transgenic (Tg) mice and wild-type (WT) littermates were analyzed. I/R injury induced neurovascular degeneration, energy failure, DNA damage, and necroptosis in the retinas of WT mice. In contrast, the GAPDH Tg mice showed resistance to all of these injury-induced abnormalities. In addition, I/R-induced effects were further examined in a neuroblastoma cell line and an endothelial cell line, which were transfected with a vector encoding human GAPDH or a control vector. After I/R challenge, energy failure, DNA damage, and elevation of receptor-interacting serine/threonine-protein kinase (RIP) 1/3 were observed in the cells transfected with the control vector. However, overexpression of GAPDH in these cells prevented the injury-induced RIP3 up-regulation by restoring energy production and preventing DNA damage. Together, the protective role of GAPDH in retinal neurovascular degeneration after I/R injury provides a better understanding of the underlying mechanism of I/R injury and a potential therapeutic target to attenuate I/R injury-related diseases.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Retina/injuries , Retinal Degeneration/prevention & control , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Cell Line , DNA Damage , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Necrosis , Oxidative Stress , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Retina/enzymology , Retina/pathology , Retinal Degeneration/enzymology , Retinal Degeneration/genetics , Retinal Vessels/enzymology , Retinal Vessels/pathology , Up-Regulation
11.
J Physiol ; 592(3): 505-21, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24247978

ABSTRACT

Diabetic nephropathy is the primary cause of end-stage renal disease. Increasing numbers of patients are suffering from this disease and therefore novel medications and therapeutic approaches are urgently needed. Here, we investigated whether apelin-13, the most active member of the adipokine apelin group, could effectively suppress the development of nephropathy in Akita mouse, a spontaneous type 1 diabetic model. Apelin-13 treatment decreased diabetes-induced glomerular filtration rate, proteinuria, glomerular hypertrophy, mesangial expansion and renal inflammation. The inflammatory factors, activation of NF-κB, histone acetylation and the enzymes involved in histone acetylation were further examined in diabetic kidneys and high glucose- or sodium butyrate-treated mesangial cells in the presence or absence of apelin-13. Apelin-13 treatment inhibited diabetes-, high glucose- and NaB-induced elevation of inflammatory factors, and histone hyperacetylation by upregulation of histone deacetylase 1. Furthermore, overexpression of apelin in mesangial cells induced histone deacetylation under high glucose condition. Thus, apelin-13 may be a novel therapeutic candidate for treatment of diabetic nephropathy via regulation of histone acetylation.


Subject(s)
Diabetic Nephropathies/drug therapy , Histones/metabolism , Intercellular Signaling Peptides and Proteins/therapeutic use , Protein Processing, Post-Translational/drug effects , Acetylation , Animals , Diabetic Nephropathies/metabolism , Glomerular Filtration Rate , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , NF-kappa B/metabolism
12.
Biochim Biophys Acta ; 1830(10): 4860-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23820032

ABSTRACT

BACKGROUND: The deposition of self-assembled amyloidogenic proteins is associated with multiple diseases, including Alzheimer's disease, Parkinson's disease and type 2 diabetes mellitus. The toxic misfolding and self-assembling of amyloidogenic proteins are believed to underlie protein misfolding diseases. Novel drug candidates targeting self-assembled amyloidogenic proteins represent a potential therapeutic approach for protein misfolding diseases. SCOPE OF REVIEW: In this perspective review, we provide an overview of the recent progress in identifying inhibitors that block the aggregation of amyloidogenic proteins and the clinical applications thereof. MAJOR CONCLUSIONS: Compounds such as polyphenols, certain short peptides, and monomer- or oligomer-specific antibodies, can interfere with the self-assembly of amyloidogenic proteins, prevent the formation of oligomers, amyloid fibrils and the consequent cytotoxicity. GENERAL SIGNIFICANCE: Some inhibitors have been tested in clinical trials for treating protein misfolding diseases. Inhibitors that target the aggregation of amyloidogenic proteins bring new hope to therapy for protein misfolding diseases.


Subject(s)
Amyloidogenic Proteins/toxicity , Protein Folding , Proteostasis Deficiencies/metabolism , Amyloidogenic Proteins/metabolism , Humans , Oxidative Stress
13.
Hum Mol Genet ; 21(5): 1138-44, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22100763

ABSTRACT

In Alzheimer disease (AD), amyloid-ß (Aß) oligomer is suggested to play a critical role in imitating neurodegeneration, although its pathogenic mechanism remains to be determined. Recently, the cellular prion protein (PrP(C)) has been reported to be an essential co-factor in mediating the neurotoxic effect of Aß oligomer. However, these previous studies focused on the synaptic plasticity in either the presence or the absence of PrP(C) and no study to date has reported whether PrP(C) is required for the neuronal cell death, the most critical element of neurodegeneration in AD. Here, we show that Prnp(-/-) mice are resistant to the neurotoxic effect of Aß oligomer in vivo and in vitro. Furthermore, application of an anti-PrP(C) antibody or PrP(C) peptide prevents Aß oligomer-induced neurotoxicity. These findings are the first to demonstrate that PrP(C) is required for Aß oligomer-induced neuronal cell death, the pathology essential to cognitive loss.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Death , Neurons/physiology , PrPC Proteins/metabolism , Prions/metabolism , Amyloid beta-Peptides/chemistry , Animals , Antibodies/immunology , Mice , PrPC Proteins/genetics , PrPC Proteins/immunology , Prion Proteins , Prions/genetics , Tissue Culture Techniques
14.
Cell Mol Neurobiol ; 34(7): 925-49, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24927694

ABSTRACT

The thesis of this review is that oxidative stress is the central factor in major depressive disorder (MDD) and Alzheimer's disease (AD). The major elements involved are inflammatory cytokines, the hypothalamic-pituitary axis, the hypothalamic-pituitary gonadal, and arginine vasopressin systems, which induce glucocorticoid and "oxidopamatergic" cascades when triggered by psychosocial stress, severe life-threatening events, and mental-affective and somatic diseases. In individuals with a genomic vulnerability to depression, these cascades may result in chronic depression-anxiety-stress spectra, resulting in MDD and other known depressive syndromes. In contrast, in subjects with genomic vulnerability to AD, oxidative stress-induced brain damage triggers specific antioxidant defenses, i.e., increased levels of amyloid-ß (Aß) and aggregation of hyper-phosphorylated tau, resulting in paired helical filaments and impaired functions related to the ApoEε4 isoform, leading to complex pathological cascades culminating in AD. Surprisingly, all the AD-associated molecular pathways mentioned in this review have been shown to be similar or analogous to those found in depression, including structural damage, i.e., hippocampal and frontal cortex atrophy. Other interacting molecular signals, i.e., GSK-3ß, convergent survival factors (brain-derived neurotrophic factor and heat shock proteins), and transition redox metals are also mentioned to emphasize the vast array of intermediates that could interact via comparable mechanisms in both MDD and AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Depressive Disorder, Major/genetics , Depressive Disorder, Major/pathology , Oxidative Stress , Alzheimer Disease/therapy , Chemokines/metabolism , Depressive Disorder, Major/therapy , Hippocampus/pathology , Humans , Stress, Psychological/complications
15.
Biochem Biophys Res Commun ; 441(1): 133-8, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24129189

ABSTRACT

Endoplasmic reticulum (ER) stress is associated with the development of diabetes. The present study sought to investigate the effect of Liraglutide, a glucagon like peptide 1 analogue, on ER stress in ß-cells. We found that Liraglutide protected the pancreatic INS-1 cells from thapsigargin-induced ER stress and the ER stress associated cell apoptosis, mainly by suppressing the PERK and IRE1 pathways. We further tested the effects of Liraglutide in the Akita mouse, an ER-stress induced type 1 diabetes model. After administration of Liraglutide for 8weeks, p-eIF2α and p-JNK were significantly decreased in the pancreas of the Akita mouse, while the treatment showed no significant impact on the levels of insulin of INS-cells. Taken together, our findings suggest that Liraglutide may protect pancreatic cells from ER stress and its related cell death.


Subject(s)
Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Endoplasmic Reticulum Stress/drug effects , Glucagon-Like Peptide 1/analogs & derivatives , Animals , Cell Death/drug effects , Cytoprotection/drug effects , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Liraglutide , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Rats , Signal Transduction/drug effects , Thapsigargin/pharmacology
16.
Biophys Chem ; 296: 106984, 2023 05.
Article in English | MEDLINE | ID: mdl-36889133

ABSTRACT

Protein misfolding diseases (PMDs) in humans are characterized by the deposition of protein aggregates in tissues, including Alzheimer's disease, Parkinson's disease, type 2 diabetes, and amyotrophic lateral sclerosis. Misfolding and aggregation of amyloidogenic proteins play a central role in the onset and progression of PMDs, and these processes are regulated by multiple factors, especially the interaction between proteins and bio-membranes. Bio-membranes induce conformational changes in amyloidogenic proteins and affect their aggregation; on the other hand, the aggregates of amyloidogenic proteins may cause membrane damage or dysfunction leading to cytotoxicity. In this review, we summarize the factors that affect the binding of amyloidogenic proteins and membranes, the effects of bio-membranes on the aggregation of amyloidogenic proteins, mechanisms of membrane disruption by amyloidogenic aggregates, technical approaches for detecting these interactions, and finally therapeutic strategies targeting membrane damage caused by amyloidogenic proteins.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Parkinson Disease , Humans , Amyloidogenic Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Alzheimer Disease/metabolism
17.
Theranostics ; 13(15): 5348-5364, 2023.
Article in English | MEDLINE | ID: mdl-37908721

ABSTRACT

Rationale: Increased methylation of key genes has been observed in kidney diseases, suggesting that the ten-eleven translocation (Tet) methyl-cytosine dioxygenase family as well as 5mC oxidation may play important roles. As a member of the Tet family, the role of Tet1 in acute kidney injury (AKI) remains unclear. Methods: Tet1 knockout mice, with or without tempol treatment, a scavenger of reactive oxygen species (ROS), were challenged with ischemia and reperfusion (I/R) injury or unilateral ureteral obstruction (UUO) injury. RNA-sequencing, Western blotting, qRT-PCR, bisulfite sequencing, chromatin immunoprecipitation, immunohistochemical staining, and dot blot assays were performed. Results: Tet1 expression was rapidly upregulated following I/R or UUO injury. Moreover, Tet1 knockout mice showed increased renal injury and renal cell death, increased ROS accumulation, G2/M cell cycle arrest, inflammation, and fibrosis. Severe renal damage in injured Tet1 knockout mice was alleviated by tempol treatment. Mechanistically, Tet1 reduced the 5mC levels in an enzymatic activity-dependent manner on the promoters of Sod1 and Sod2 to promote their expression, thus lowering injury-induced excessive ROS and reducing I/R or UUO injury. Conclusions: Tet1 plays an important role in the development of AKI by promoting SOD expression through a DNA demethylase-dependent mechanism.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Ureteral Obstruction , Animals , Mice , Acute Kidney Injury/metabolism , Kidney/metabolism , Mice, Knockout , Oxidative Stress , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Superoxide Dismutase/metabolism , Ureteral Obstruction/metabolism
18.
Nat Commun ; 14(1): 4261, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37460623

ABSTRACT

Acute kidney injury (AKI) exhibits high morbidity and mortality. Kidney injury molecule-1 (KIM1) is dramatically upregulated in renal tubules upon injury, and acts as a biomarker for various renal diseases. However, the exact role and underlying mechanism of KIM1 in the progression of AKI remain elusive. Herein, we report that renal tubular specific knockout of Kim1 attenuates cisplatin- or ischemia/reperfusion-induced AKI in male mice. Mechanistically, transcription factor Yin Yang 1 (YY1), which is downregulated upon AKI, binds to the promoter of KIM1 and represses its expression. Injury-induced KIM1 binds to the ECD domain of death receptor 5 (DR5), which activates DR5 and the following caspase cascade by promoting its multimerization, thus induces renal cell apoptosis and exacerbates AKI. Blocking the KIM1-DR5 interaction with rationally designed peptides exhibit reno-protective effects against AKI. Here, we reveal a YY1-KIM1-DR5 axis in the progression of AKI, which warrants future exploration as therapeutic targets.


Subject(s)
Acute Kidney Injury , Kidney , Animals , Male , Mice , Acute Kidney Injury/metabolism , Apoptosis , Cisplatin/adverse effects , Kidney/metabolism , Kidney Tubules/metabolism , Mice, Inbred C57BL , Receptors, TNF-Related Apoptosis-Inducing Ligand
19.
Theranostics ; 13(10): 3387-3401, 2023.
Article in English | MEDLINE | ID: mdl-37351176

ABSTRACT

Rationale: Ischemia-reperfusion injury (I/R) is a common cause of acute kidney injury (AKI). Post-ischemic recovery of renal blood supply plays an important role in attenuating injury. Exogenous application of elabela (ELA) peptides has been demonstrated by us and others to alleviate AKI, partly through its receptor APJ. However, the endogenous role of ELA in renal I/R remains unclear. Methods: Renal tubule specific ELA knockout (ApelaKsp KO) mice challenged with bilateral or unilateral I/R were used to investigate the role of endogenous ELA in renal I/R. RNA-sequencing analysis was performed to unbiasedly investigate altered genes in kidneys of ApelaKsp KO mice. Injured mice were treated with ELA32 peptide, Nω-hydroxy-nor-L-arginine (nor-NOHA), prostaglandin E2 (PGE2), Paricalcitol, ML221 or respective vehicles, individually or in combination. Results: ELA is mostly expressed in renal tubules. Aggravated pathological injury and further reduction of renal microvascular blood flow were observed in ApelaKsp KO mice during AKI and the following transition to chronic kidney disease (AKI-CKD). RNA-seq analysis suggested that two blood flow regulators, arginine metabolizing enzyme arginase 2 (ARG2) and PGE2 metabolizing enzyme carbonyl reductases 1 and 3 (CBR1/3), were altered in injured ApelaKsp KO mice. Notably, combination application of an ARG2 inhibitor nor-NOHA, and Paricalcitol, a clinically used activator for PGE2 synthesis, alleviated injury-induced AKI/AKI-CKD stages and eliminated the worst outcomes observed in ApelaKsp KO mice. Moreover, while the APJ inhibitor ML221 blocked the beneficial effects of ELA32 peptide on AKI, it showed no effect on combination treatment of nor-NOHA and Paricalcitol. Conclusions: An endogenous tubular ELA-APJ axis regulates renal microvascular blood flow that plays a pivotal role in I/R-induced AKI. Furthermore, improving renal blood flow by inhibiting ARG2 and activating PGE2 is an effective treatment for AKI and prevents the subsequent AKI-CKD transition.


Subject(s)
Acute Kidney Injury , Peptide Hormones , Renal Insufficiency, Chronic , Reperfusion Injury , Mice , Animals , Microcirculation , Dinoprostone/pharmacology , Kidney/pathology , Acute Kidney Injury/pathology , Renal Insufficiency, Chronic/etiology , Reperfusion Injury/pathology , Ischemia/pathology , Peptide Hormones/adverse effects , Peptide Hormones/genetics , Reperfusion/adverse effects
20.
J Biol Chem ; 286(17): 15095-105, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21393248

ABSTRACT

The prion protein (PrP) is best known for its association with prion diseases. However, a controversial new role for PrP in Alzheimer disease (AD) has recently emerged. In vitro studies and mouse models of AD suggest that PrP may be involved in AD pathogenesis through a highly specific interaction with amyloid-ß (Aß42) oligomers. Immobilized recombinant human PrP (huPrP) also exhibited high affinity and specificity for Aß42 oligomers. Here we report the novel finding that aggregated forms of huPrP and Aß42 are co-purified from AD brain extracts. Moreover, an anti-PrP antibody and an agent that specifically binds to insoluble PrP (iPrP) co-precipitate insoluble Aß from human AD brain. Finally, using peptide membrane arrays of 99 13-mer peptides that span the entire sequence of mature huPrP, two distinct types of Aß binding sites on huPrP are identified in vitro. One specifically binds to Aß42 and the other binds to both Aß42 and Aß40. Notably, Aß42-specific binding sites are localized predominantly in the octapeptide repeat region, whereas sites that bind both Aß40 and Aß42 are mainly in the extreme N-terminal or C-terminal domains of PrP. Our study suggests that iPrP is the major PrP species that interacts with insoluble Aß42 in vivo. Although this work indicated the interaction of Aß42 with huPrP in the AD brain, the pathophysiological relevance of the iPrP/Aß42 interaction remains to be established.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Prions/metabolism , Aged , Aged, 80 and over , Binding Sites , Brain/metabolism , Case-Control Studies , Humans , Middle Aged , Protein Binding , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL