Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Kidney Int ; 103(2): 320-330, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36341730

RESUMO

IgA nephropathy (IgAN) is the most common glomerulonephritis, characterized by the presence of predominant IgA deposits in the mesangium. Deposition of pathogenic IgA in kidney tissue is a fundamental initiating process that has not been fully studied. Here, we employed optical imaging to directly visualize kidney deposition of IgA with optimized spatial and temporal resolution in BALB/c nude mice. Real-time fluorescence imaging revealed that IgA isolated from patients with IgAN preferentially accumulated in the kidneys, compared with IgA purified from healthy individuals. There was no difference in the distribution of either IgA preparation by the liver. Photoacoustic computed tomography dynamically demonstrated and quantified the enhanced retention of pathological IgA in the kidney cortex. Photoacoustic microscopy tracked IgA deposition in the glomeruli with a resolution down to three microns in a mouse model. Notably, longitudinal fluorescent imaging revealed that galactose-deficient IgA (Gd-IgA), which was elevated in the circulation of patients with IgAN, persisted in the kidney for longer than two weeks, and stable deposition of Gd-IgA induced kidney impairment, including albuminuria and mesangial proliferation. Thus, our study highlights that the aberrant kidney depositional kinetics of Gd-IgA is involved in the pathogenesis of IgAN. Hence, cross-scale optical imaging has potential applications in assessing immune-mediated kidney diseases and uncovering underlying mechanisms of disease.


Assuntos
Glomerulonefrite por IGA , Animais , Camundongos , Glomerulonefrite por IGA/diagnóstico por imagem , Glomerulonefrite por IGA/patologia , Galactose , Camundongos Nus , Imunoglobulina A , Imagem Óptica
2.
Biochem Biophys Res Commun ; 643: 61-68, 2023 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-36586160

RESUMO

Nicotinamide adenine dinucleotide (NAD) is an essential coenzyme in the kidney. The first step in de novo NAD synthesis is regulated by indoleamine 2,3-dioxygenase (IDO), a tryptophan-catabolizing enzyme. Here, we investigated NAD synthetic flux and NAD levels in podocytes under diabetic conditions. We also studied the effects of IDO overexpression on NAD synthetic flux and high glucose (HG)-induced podocyte injury. NAD synthetases in the de novo, Preiss-Handler and salvage pathways were analyzed using in vivo single-nucleus RNA sequencing datasets (GSE131882) of control and diabetic kidney disease (DKD). The mRNA levels of these NAD synthetases were measured in vitro in HG-treated podocytes. The effects of IDO on NAD synthesis were examined by transducing cultured podocytes with an adenovirus encoding IDO, and apoptosis, podocyte markers and mobility were investigated. Cellular transcriptome analysis revealed that control podocytes had relatively low levels of NAD synthetases. In DKD podocytes, de novo NAD synthetase levels were further downregulated. IDO levels were virtually undetectable and did not increase in DKD. In vitro experiments confirmed aberrant de novo NAD synthetic flux and decreased IDO levels in HG-treated podocytes. Overexpression of IDO promoted NAD de novo synthesis, reduced NAD-bypass metabolic enzyme, increased NAD content and recovered podocyte injury markers under diabetic conditions. Taken together, our findings suggest that the de novo NAD synthetic flux is aberrant in DKD, and IDO promotes de novo NAD synthesis and NAD levels, as well as alleviates injury in HG-treated podocytes.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Podócitos , Humanos , NAD/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Podócitos/metabolismo , Ligases
3.
Nat Commun ; 14(1): 6581, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37853001

RESUMO

A comprehensive atlas of cis-regulatory elements and their dynamic activity is necessary to understand the transcriptional basis of cellular structure maintenance, metabolism, and responses to the environment. Here we show, using matched single-nucleus chromatin accessibility and RNA-sequencing from juvenile male C57BL6 mice, an atlas of accessible chromatin regions in both normal and denervated skeletal muscles. We identified cell-type-specific cis-regulatory networks, highlighting the dynamic regulatory circuits mediating transitions between myonuclear types. Through comparison of normal and perturbed muscle, we delineated the reprogramming of cis-regulatory networks in response to denervation, described the interplay of promoters/enhancers and target genes. We further unveil a hierarchical structure of transcription factors that delineate a regulatory network in atrophic muscle, identifying ELK4 as a key atrophy-related transcription factor that instigates muscle atrophy through TGF-ß1 regulation. This study furnishes a rich genomic resource, essential for decoding the regulatory dynamics of skeletal muscle in both physiological and pathological states.


Assuntos
Músculo Esquelético , Atrofia Muscular , Camundongos , Masculino , Animais , Camundongos Endogâmicos C57BL , Atrofia Muscular/patologia , Músculo Esquelético/metabolismo , Cromatina/genética , Cromatina/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
bioRxiv ; 2023 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-37905139

RESUMO

Chronic kidney disease (CKD) is often associated with protein-energy wasting (PEW), which is characterized by a reduction in muscle mass and strength. Although mitochondrial dysfunction and oxidative stress have been implicated to play a role in the pathogenesis of muscle wasting, the underlying mechanisms remain unclear. In this study, we used transcriptomics, metabolomics analyses and mouse gene manipulating approaches to investigate the effects of mitochondrial plasticity and oxidative stress on muscle wasting in mouse CKD models. Our results showed that the expression of oxidative stress response genes was increased, and that of oxidative phosphorylation genes was decreased in the muscles of mice with CKD. This was accompanied by reduced oxygen consumption rates, decreased levels of mitochondrial electron transport chain proteins, and increased cellular oxidative damage. Excessive mitochondrial fission was also observed, and we found that the activation of ROCK1 was responsible for this process. Inducible expression of muscle-specific constitutively active ROCK1(mROCK1ca)exacerbated mitochondrial fragmentation and muscle wasting in CKD mice. Conversely, ROCK1 depletion (ROCK1-/-) alleviated these phenomena. Mechanistically, ROCK1 activation promoted the recruitment of Drp1 to mitochondria, thereby facilitating fragmentation. Notably, the pharmacological inhibition of ROCK1 mitigated muscle wasting by suppressing mitochondrial fission and oxidative stress. Our findings demonstrate that ROCK1 participates in CKD-induced muscle wasting by promoting mitochondrial fission and oxidative stress, and pharmacological suppression of ROCK1 could be a therapeutic strategy for combating muscle wasting in CKD conditions.

5.
Front Immunol ; 13: 901465, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35983053

RESUMO

IgA nephropathy (IgAN) is the most common primary glomerulonephritis, characterized by glomerular deposition of IgA immune complexes, mainly produced by B cells under the regulation of CD4+T cells. However, the alterations of specific CD4+T cell subsets and the mechanism of B cells activation in IgAN remain unclear. Therefore, we aimed to investigate the landscape characteristics and role of CD4+T cells in the progression of IgAN. We identified that the proportion of Th2, Th17 and Tfh (follicular helper T) cells in patients with IgAN was significantly higher than that of healthy controls (P < 0.05). Single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) showed that Th cells and B cells in patients with IgAN were more activated. Correspondingly, multiplex immunohistochemistry staining of renal biopsy showed increased infiltration of CD4+T and B cells in the kidneys of patients with IgAN. The degree of infiltration was positively correlated with the degree of renal damage. Interestingly, the proportion of Tfh cells in peripheral blood was positively correlated with the severity of proteinuria. Moreover, the proximity position of Tfh cells and B cells suggested that cell-cell interactions between Tfh and B cells were happening in situ. Intercellular communication analysis also showed enhanced interaction between Tfh cells and B cells in IgAN. Our findings suggested that Tfh cells of patients possibly contributed to the progression of IgAN by activating B cells via cell-cell interactions and TNFSF14-TNFRSF14 may be an underlying signaling pathway.


Assuntos
Glomerulonefrite por IGA , Humanos , Leucócitos Mononucleares/patologia , Índice de Gravidade de Doença , Células T Auxiliares Foliculares , Células Th17
6.
Signal Transduct Target Ther ; 7(1): 7, 2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34983926

RESUMO

Activation-induced cytidine deaminase (AID) initiates class-switch recombination and somatic hypermutation (SHM) in antibody genes. Protein expression and activity are tightly controlled by various mechanisms. However, it remains unknown whether a signal from the extracellular environment directly affects the AID activity in the nucleus where it works. Here, we demonstrated that a deubiquitinase USP10, which specifically stabilizes nuclear AID protein, can translocate into the nucleus after AKT-mediated phosphorylation at its T674 within the NLS domain. Interestingly, the signals from BCR and TLR1/2 synergistically promoted this phosphorylation. The deficiency of USP10 in B cells significantly decreased AID protein levels, subsequently reducing neutralizing antibody production after immunization with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or human immunodeficiency virus type 1 (HIV-1) nanoparticle vaccines. Collectively, we demonstrated that USP10 functions as an integrator for both BCR and TLR signals and directly regulates nuclear AID activity. Its manipulation could be used for the development of vaccines and adjuvants.


Assuntos
Vacinas contra a AIDS/imunologia , Fator Ativador de Células B/imunologia , Vacinas contra COVID-19/imunologia , Citidina Desaminase/imunologia , HIV-1/imunologia , Nanopartículas , SARS-CoV-2/imunologia , Transdução de Sinais/imunologia , Ubiquitina Tiolesterase/imunologia , Ubiquitinação/imunologia , Vacinas contra a AIDS/genética , Animais , Fator Ativador de Células B/genética , Vacinas contra COVID-19/genética , Citidina Desaminase/genética , Células HEK293 , HIV-1/genética , Humanos , Camundongos , Camundongos Knockout , SARS-CoV-2/genética , Transdução de Sinais/genética , Ubiquitina Tiolesterase/genética
7.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 28(6): 1148-53, 2011 Dec.
Artigo em Zh | MEDLINE | ID: mdl-22295704

RESUMO

The aim of this study is to investigate the growth and proliferation of bone marrow mesenchymal stem cells (BMSCs) three-dimensionally cultured in Pluronic F-127 gel, in order to explore the cellular compatibility of gel and to investigate the feasibility of BMSCs differentiating into adipocytes in gel. Rat BMSCs were isolated from adult bone marrow, and then cultured and amplified in vitro. The BMSCs derived from the 4th passage were seeded on the scaffolds and incubated in adipogenic stimuli culture to differentiate into adipocytes. BMSCs were dispersed into gel and cultured in vitro for two weeks then the status of adhesion, growth and proliferation of the cells were observed. The edipogenic differentiation of the BMSCs was assessed by cellular morphology and further confirmed by Oil Red O staining. BMSCs were able to attach, grow and proliferate well in Pluronic F-127 gel. The BMSCs differentiated into adipocytes in gel in the presence of adipogenic stimuli over a period of 2 weeks. After only 4 days of adipogenic induction, small lipid droplets were observed within BMSCs in gel wells treated with differentiation media. At the end of 14 days, in the presence of differentiation media in gel, the size of the lipid droplets increased to occupy most of the cytoplasm, consistent with differentiation of BMSCs into adipocytes. Lipid droplets in differentiating BMSCs were positively stained with Oil Red O in the presence of differentiation media in the Pluronic F-127 treatment. We demostrated BMSCs incubated in the 3D Pluronic F-127 gel scaffolds could be induced and differentiated into adipocytes. The system for inducing differentiation of BMSCs into adipocytes is promising to apply in the construction of tissue engineering adipose tissue and the repair of fat injury, and Pluronic F-127 gel may be a suitable scaffold for cellular therapy of BMSCs.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular/efeitos dos fármacos , Hidrogéis/química , Células-Tronco Mesenquimais/citologia , Poloxâmero/química , Adipócitos/citologia , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Ratos , Ratos Sprague-Dawley , Engenharia Tecidual
8.
Sci Transl Med ; 11(495)2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31167927

RESUMO

Progressive peritoneal fibrosis affects patients receiving peritoneal dialysis (PD) and has no reliable treatment. The mechanisms that initiate and sustain peritoneal fibrosis remain incompletely elucidated. To overcome these problems, we developed a strategy that prevents peritoneal fibrosis by suppressing PD-stimulated mesothelial-to-mesenchymal transition (MMT). We evaluated single-cell transcriptomes of mesothelial cells obtained from normal peritoneal biopsy and effluent from PD-treated patients. In cells undergoing MMT, we found cellular heterogeneity and intermediate transition states associated with up-regulation of enzymes involved in glycolysis. The expression of glycolytic enzymes was correlated with the development of MMT. Using gene expression profiling and metabolomics analyses, we confirmed that PD fluid induces metabolic reprogramming, characterized as hyperglycolysis, in mouse peritoneum. We found that transforming growth factor ß1 (TGF-ß1) can substitute for PD fluid to stimulate hyperglycolysis, suppressing mitochondrial respiration in mesothelial cells. Blockade of hyperglycolysis with 2-deoxyglucose (2-DG) inhibited TGF-ß1-induced profibrotic cellular phenotype and peritoneal fibrosis in mice. We developed a triad of adeno-associated viruses that overexpressed microRNA-26a and microRNA-200a while inhibiting microRNA-21a to target hyperglycolysis and fibrotic signaling. Intraperitoneal injection of the viral triad inhibited the development of peritoneal fibrosis induced by PD fluid in mice. We conclude that hyperglycolysis is responsible for MMT and peritoneal fibrogenesis, and this aberrant metabolic state can be corrected by modulating microRNAs in the peritoneum. These results could provide a therapeutic strategy to combat peritoneal fibrosis.


Assuntos
Glicólise/efeitos dos fármacos , Fibrose Peritoneal/tratamento farmacológico , Fibrose Peritoneal/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/uso terapêutico , Animais , Células Cultivadas , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
BMJ Case Rep ; 20182018 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-29776936

RESUMO

Ethambutol (EMB) and isoniazid (INH) are the first-line antituberculosis (anti-TB) drugs. However, their neurotoxicity could cause adverse effect and the patients with end-stage renal disease are especially vulnerable due to the reduction in renal drug clearance. Here, we report a 36-year-old man receiving peritoneal dialysis developed progressive paralysis in lower extremities, vision loss and hoarseness 4 months after anti-TB treatment with INH, EMB and rifapentine because of concomitant pulmonary tuberculosis. A diagnosis of EMB/INH-induced peripheral neuropathy, retrobulbar neuritis and laryngoparalysis was made. The patient's neuropathy gradually improved 2 years after discontinuation of EMB/INH. Since EMB and INH may cause simultaneously severe and complex multineuropathy in dialysis patients, their adverse effects should be closely supervised in dialysis patients.


Assuntos
Antituberculosos/efeitos adversos , Etambutol/efeitos adversos , Isoniazida/efeitos adversos , Falência Renal Crônica/terapia , Neurite Óptica/induzido quimicamente , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Diálise Peritoneal Ambulatorial Contínua/efeitos adversos , Paralisia das Pregas Vocais/induzido quimicamente , Adulto , Humanos , Falência Renal Crônica/microbiologia , Masculino , Diálise Peritoneal Ambulatorial Contínua/métodos , Tuberculose Pulmonar/tratamento farmacológico
10.
J Cachexia Sarcopenia Muscle ; 9(5): 962-974, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30043444

RESUMO

BACKGROUND: Chronic kidney disease (CKD) is commonly associated with cachexia, a condition that causes skeletal muscle wasting and an unfavourable prognosis. Although mechanisms leading to cachexia have been intensively studied, the advance of biological knowledges and technologies encourages us to make progress in understanding the pathogenesis of this disorder. Long noncoding RNAs (lncRNAs) are defined as >200 nucleotides RNAs but lack the protein-coding potential. LncRNAs are involved in the pathogenesis of many diseases, but whether they functionally involve in muscle protein loss has not been investigated. METHODS: We performed lncRNA array and identified an lncRNA, which we named Atrolnc-1, remarkably elevated in atrophying muscles from mice with cachexia. We examined how overexpression or knockdown of Atrolnc-1 could influence muscle protein synthesis and degradation. We also examined whether inhibition of Atrolnc-1 ameliorates muscle wasting in mice with CKD. RESULTS: We documented that Atrolnc-1 expression is continuously increased in muscles of mice with fasting (5.4 fold), cancer (2.0 fold), or CKD (5.1 fold). We found that depressed insulin signalling stimulates the transcription factor C/EBP-α binding to the promoter of Atrolnc-1 and promotes the expression of Atrolnc-1. In cultured C2C12 myotubes, overexpression of Atrolnc-1 increases protein degradation (0.45±0.03 vs. 0.64±0.02, *p<0.05); Atrolnc-1 knockdown significantly reduces the rate of protein degradation stimulated by serum depletion (0.61±0.03 vs. 0.47±0.02, *p<0.05). Using mass spectrometry and a lncRNA pull-down assay, we identified that Atrolnc-1 interacts with A20 binding inhibitor of NF-κB-1 (ABIN-1). The interaction impairs function, resulting in enhanced NF-κB activity plus MuRF-1 transcription. This response is counteracted by CRISPR/dCas9 mediated overexpression. In muscles from normal mice, overexpression of Atrolnc-1 stimulates a 2.7-fold increase in MuRF-1 expression leading to myofibers atrophy. In contrast, Atrolnc-1 knockdown attenuates muscle wasting by 42% in mice with CKD via suppression of NF-κB activity and MuRF-1 expression. CONCLUSIONS: Our findings provide evidence that lncRNAs initiates the pathophysiological process of muscle wasting. The interaction between Atrolnc-1 and NF-κB signalling modulates muscle mass and proteolysis in CKD and perhaps other catabolic conditions.


Assuntos
Caquexia/etiologia , Caquexia/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , RNA Longo não Codificante/genética , Insuficiência Renal Crônica/complicações , Animais , Biomarcadores , Caquexia/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Expressão Gênica , Perfilação da Expressão Gênica , Masculino , Camundongos , Modelos Biológicos , Fibras Musculares Esqueléticas/metabolismo , NF-kappa B/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Transdução de Sinais , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA