Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Biol Sci ; 20(6): 2310-2322, 2024.
Article in English | MEDLINE | ID: mdl-38617540

ABSTRACT

Wnt/ß-catenin signaling plays a pivotal role in the pathogenesis of chronic kidney diseases (CKD), which is associated with macrophage activation and polarization. However, the relative contribution of macrophage-derived Wnts in the evolution of CKD is poorly understood. Here we demonstrate a critical role of Wnts secreted by macrophages in regulating renal inflammation and fibrosis after various injuries. In mouse model of kidney fibrosis induced by unilateral ureteral obstruction (UUO), macrophages were activated and polarized to M1 and M2 subtypes, which coincided with the activation of Wnt/ß-catenin signaling. In vitro, multiple Wnts were induced in primary cultured bone marrow-derived macrophages (BMDMs) after polarization. Conversely, Wnt proteins also stimulated the activation and polarization of BMDMs to M1 and M2 subtype. Blockade of Wnt secretion from macrophages in mice with myeloid-specific ablation of Wntless (Wls), a cargo receptor that is obligatory for Wnt trafficking and secretion, blunted macrophage infiltration and activation and inhibited the expression of inflammatory cytokines. Inhibition of Wnt secretion by macrophages also abolished ß-catenin activation in tubular epithelium, repressed myofibroblast activation and reduced kidney fibrosis after either obstructive or ischemic injury. Furthermore, conditioned medium from Wls-deficient BMDMs exhibited less potency to stimulate fibroblast proliferation and activation, compared to the controls. These results underscore an indispensable role of macrophage-derived Wnts in promoting renal inflammation, fibroblasts activation and kidney fibrosis.


Subject(s)
Renal Insufficiency, Chronic , beta Catenin , Animals , Mice , Macrophages , Myofibroblasts , Inflammation , Kidney
2.
BMC Psychiatry ; 23(1): 849, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37974123

ABSTRACT

OBJECTIVES: To investigate the method of resting EEG assessment of depressive symptoms in college students and to clarify the relationship between physical activity level and depressive symptoms in college students. METHODS: Using a cross-sectional study design, 140 current full-time college students were recruited to complete the Self-Rating Depression Scale and the International Physical Activity Questionnaire, and 10-min resting EEGs were obtained. RESULTS: 1) The power values of δ and α2 in the central (C3, C4) and parietal (P3, P4) regions of depressed college students were significantly higher than those of normal college students. And the degree of lateralization of δ, θ, α1, and α2 in the prefrontal regions (F3, F4) of depressed college students was significantly higher than that of normal college students (all P < 0. 008). 2) The recall rate of the depression recognition model for college students based on resting EEG was 66.67%, the precision was 65.05%, and the AUCs of the training group and validation group were 0.791 and 0.786, respectively, with better detection effects. 3) The two indicators, δ (C3 + C4) and α1 (F4-F3), are significantly correlated with IPAQ scores, and among college students who engage in ball games most commonly, those with a higher level of physical activity have lower δ (C3 + C4) and higher α1 (F4-F3), while among those who engage in resistance training most commonly, higher levels of physical activity are associated with lower δ (C3 + C4). CONCLUSION: The resting EEG of depressed college students has a certain specificity that can objectively assess the risk of developing depressive symptoms in college students. Physical activity is associated with abnormal EEG signals of depressive symptoms. Different types of physical activity may modulate the relationship between physical activity levels and EEG indicators.


Subject(s)
Depression , Electroencephalography , Humans , Depression/diagnosis , Cross-Sectional Studies , Electroencephalography/methods , Exercise , Students
3.
Theranostics ; 13(11): 3897-3913, 2023.
Article in English | MEDLINE | ID: mdl-37441594

ABSTRACT

Background: Renal infiltration of inflammatory cells including macrophages is a crucial event in kidney fibrogenesis. However, how macrophage regulates fibroblast activation in the fibrotic kidney remains elusive. In this study, we show that macrophages promoted fibroblast activation by assembling a vitronectin (Vtn)-enriched, extracellular microenvironment. Methods: We prepared decellularized kidney tissue scaffold (KTS) from normal and fibrotic kidney after unilateral ischemia-reperfusion injury (UIRI) and carried out an unbiased quantitative proteomics analysis. NRK-49F cells were seeded on macrophage-derived extracellular matrix (ECM) scaffold. Genetic Vtn knockout (Vtn-/-) mice and chronic kidney disease (CKD) model with overexpression of Vtn were used to corroborate a role of Vtn/integrin αvß5/Src in kidney fibrosis. Results: Vtn was identified as one of the most upregulated proteins in the decellularized kidney tissue scaffold from fibrotic kidney by mass spectrometry. Furthermore, Vtn was upregulated in the kidney of mouse models of CKD and primarily expressed and secreted by activated macrophages. Urinary Vtn levels were elevated in CKD patients and inversely correlated with kidney function. Genetic ablation or knockdown of Vtn protected mice from developing kidney fibrosis after injury. Conversely, overexpression of Vtn exacerbated renal fibrotic lesions and aggravated renal insufficiency. We found that macrophage-derived, Vtn-enriched extracellular matrix scaffold promoted fibroblast activation and proliferation. In vitro, Vtn triggered fibroblast activation by stimulating integrin αvß5 and Src kinase signaling. Either blockade of αvß5 with neutralizing antibody or pharmacological inhibition of Src by Saracatinib abolished Vtn-induced fibroblast activation. Moreover, Saracatinib dose-dependently ameliorated Vtn-induced kidney fibrosis in vivo. These results demonstrate that macrophage induces fibroblast activation by assembling a Vtn-enriched extracellular microenvironment, which triggers integrin αvß5 and Src kinase signaling. Conclusion: Our findings uncover a novel mechanism by which macrophages contribute to kidney fibrosis via assembling a Vtn-enriched extracellular niche and suggest that disrupting fibrogenic microenvironment could be a therapeutic strategy for fibrotic CKD.


Subject(s)
Renal Insufficiency, Chronic , Vitronectin , Mice , Animals , Vitronectin/metabolism , Kidney/pathology , Renal Insufficiency, Chronic/metabolism , src-Family Kinases/metabolism , Macrophages/metabolism , Fibroblasts/metabolism , Fibrosis
4.
Sci Adv ; 7(5)2021 01.
Article in English | MEDLINE | ID: mdl-33571112

ABSTRACT

Endothelial cell injury leading to microvascular rarefaction is a characteristic feature of chronic kidney disease (CKD). However, the mechanism underlying endothelial cell dropout is poorly defined. Here, we show a central role of the extracellular microenvironment in controlling endothelial cell survival and proliferation in CKD. When cultured on a decellularized kidney tissue scaffold (KTS) from fibrotic kidney, endothelial cells increased the expression of proapoptotic proteins. Proteomics profiling identified fibrillin-1 (FBN1) as a key component of the fibrotic KTS, which was up-regulated in animal models and patients with CKD. FBN1 induced apoptosis of endothelial cells and inhibited their proliferation in vitro. RNA sequencing uncovered activated integrin αvß6/transforming growth factor-ß signaling, and blocking this pathway abolished FBN1-triggered endothelial injury. In a mouse model of CKD, depletion of FBN1 ameliorated renal fibrotic lesions and mitigated vascular rarefaction. These studies illustrate that FBN1 plays a role in mediating vascular rarefaction by orchestrating a hostile microenvironment for endothelial cells.


Subject(s)
Endothelial Cells , Fibrillin-1 , Microvascular Rarefaction , Renal Insufficiency, Chronic , Animals , Cellular Microenvironment/genetics , Cellular Microenvironment/physiology , Endothelial Cells/metabolism , Female , Fibrillin-1/genetics , Fibrillin-1/metabolism , Fibrosis , Humans , Kidney/pathology , Male , Mice , Microvascular Rarefaction/metabolism , Microvascular Rarefaction/pathology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology
5.
Cell Death Dis ; 12(1): 70, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436543

ABSTRACT

Matrix metalloproteinase-10 (MMP-10) is a zinc-dependent endopeptidase involved in regulating a wide range of biologic processes, such as apoptosis, cell proliferation, and tissue remodeling. However, the role of MMP-10 in the pathogenesis of acute kidney injury (AKI) is unknown. In this study, we show that MMP-10 was upregulated in the kidneys and predominantly localized in the tubular epithelium in various models of AKI induced by ischemia/reperfusion (IR) or cisplatin. Overexpression of exogenous MMP-10 ameliorated AKI, manifested by decreased serum creatinine, blood urea nitrogen, tubular injury and apoptosis, and increased tubular regeneration. Conversely, knockdown of endogenous MMP-10 expression aggravated kidney injury. Interestingly, alleviation of AKI by MMP-10 in vivo was associated with the activation of epidermal growth factor receptor (EGFR) and its downstream AKT and extracellular signal-regulated kinase-1 and 2 (ERK1/2) signaling. Blockade of EGFR signaling by erlotinib abolished the MMP-10-mediated renal protection after AKI. In vitro, MMP-10 potentiated EGFR activation and protected kidney tubular cells against apoptosis induced by hypoxia/reoxygenation or cisplatin. MMP-10 was colocalized with heparin-binding EGF-like growth factor (HB-EGF) in vivo and activated it by a process of proteolytical cleavage in vitro. These studies identify HB-EGF as a previously unrecognized substrate of MMP-10. Our findings also underscore that MMP-10 can protect against AKI by augmenting EGFR signaling, leading to promotion of tubular cell survival and proliferation after injury.


Subject(s)
Acute Kidney Injury/metabolism , ErbB Receptors/metabolism , Matrix Metalloproteinase 10/metabolism , Humans , Signal Transduction
6.
Kidney Int ; 97(5): 1017-1031, 2020 05.
Article in English | MEDLINE | ID: mdl-32245660

ABSTRACT

Tenascin-C is an extracellular matrix glycoprotein that plays a critical role in kidney fibrosis by orchestrating a fibrogenic niche. Here, we demonstrate that tenascin-C is a biomarker and a mediator of kidney fibrogenesis by impairing tubular integrity. Tenascin-C was found to be increased in kidney biopsies from patients with chronic kidney disease (CKD). In a cohort of 225 patients with CKD, the urinary tenascin-C level was markedly elevated, compared to 39 healthy individuals. Moreover, the level of urinary tenascin-C in CKD was correlated with the severity of kidney dysfunction and fibrosis. In mouse model of acute kidney injury-to-CKD induced by ischemia/reperfusion, depletion of tenascin-C preserved tubular integrity and ameliorated renal fibrotic lesions. In vitro, tenascin-C impaired tubular cell integrity by inducing partial epithelial-mesenchymal transition. Using decellularized kidney tissue scaffolds, we found that tenascin-C-enriched scaffolds facilitated tubular epithelial-mesenchymal transition ex vivo. Mechanistically, tenascin-C specifically induced integrins αvß6 in tubular cells and activated focal adhesion kinase (FAK). Blocking αvß6 integrins or inhibition of FAK restored tubular integrity by repressing epithelial-mesenchymal transition and alleviated kidney fibrosis. Thus, our studies underscore that tenascin-C is a noninvasive biomarker of kidney fibrogenesis and a pathogenic mediator that impairs tubular integrity. Hence, blockade of the tenascin-C/αvß6 integrin/FAK signal cascade may be a novel strategy for therapeutic intervention of kidney fibrosis.


Subject(s)
Acute Kidney Injury , Renal Insufficiency, Chronic , Animals , Antigens, Neoplasm , Epithelial-Mesenchymal Transition , Extracellular Matrix , Fibrosis , Humans , Integrins , Mice , Tenascin
7.
Kidney Int ; 95(5): 1167-1180, 2019 05.
Article in English | MEDLINE | ID: mdl-30878215

ABSTRACT

Matrix metalloproteinase-7 (MMP-7) is a secreted endopeptidase that degrades a broad range of substrates. Recent studies have identified MMP-7 as an early biomarker to predict severe acute kidney injury (AKI) and poor outcomes after cardiac surgery; however, the role of MMP-7 in the pathogenesis of AKI is unknown. In this study, we investigated the expression of MMP-7 and the impact of MMP-7 deficiency in several models of AKI. MMP-7 was induced in renal tubules following ischemia/ reperfusion injury or cisplatin administration, and in folic acid-induced AKI. MMP-7 knockout mice experienced higher mortality, elevated serum creatinine, and more severe histologic lesions after ischemic or toxic insults. Tubular apoptosis and interstitial inflammation were more prominent in MMP-7 knockout kidneys. These histologic changes were accompanied by increased expression of FasL and other components of the extrinsic apoptotic pathway, as well as increased expression of pro-inflammatory chemokines. In a rescue experiment, exogenous MMP-7 ameliorated kidney injury in MMP-7 knockout mice after ischemia/reperfusion. In vitro, MMP-7 protected tubular epithelial cells against apoptosis by directly degrading FasL. In isolated tubules ex vivo, MMP-7 promoted cell proliferation by degrading E-cadherin and thereby liberating ß-catenin, priming renal tubules for regeneration. Taken together, these results suggest that induction of MMP-7 is protective in AKI by degrading FasL and mobilizing ß-catenin, thereby priming kidney tubules for survival and regeneration.


Subject(s)
Acute Kidney Injury/pathology , Kidney Tubules/pathology , Matrix Metalloproteinase 7/metabolism , Regeneration/physiology , Reperfusion Injury/pathology , Acute Kidney Injury/chemically induced , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Disease Models, Animal , Epithelial Cells/metabolism , Fas Ligand Protein/metabolism , Folic Acid/toxicity , Humans , Kidney Tubules/blood supply , Kidney Tubules/drug effects , Matrix Metalloproteinase 7/genetics , Mice , Mice, Knockout , Proteolysis , Signal Transduction/physiology , beta Catenin/metabolism
8.
Kidney Int ; 95(1): 62-74, 2019 01.
Article in English | MEDLINE | ID: mdl-30409456

ABSTRACT

The development of acute kidney injury (AKI) is a complex process involving tubular, inflammatory, and vascular components, but less is known about the role of the interstitial microenvironment. We have previously shown that the extracellular matrix glycoprotein tenascin-C (TNC) is induced in fibrotic kidneys. In mouse models of AKI induced by ischemia-reperfusion injury (IRI) or cisplatin, TNC was induced de novo in the injured sites and localized to the renal interstitium. The circulating level of TNC protein was also elevated in AKI patients after cardiac surgery. Knockdown of TNC by shRNA in vivo aggravated AKI after ischemic or toxic injury. This effect was associated with reduced renal ß-catenin expression, suggesting an impact on Wnt signaling. In vitro, TNC protected tubular epithelial cells against apoptosis and augmented Wnt1-mediated ß-catenin activation. Co-immunoprecipitation revealed that TNC physically interacts with Wnt ligands. Furthermore, a TNC-enriched kidney tissue scaffold prepared from IRI mice was able to recruit and concentrate Wnt ligands from the surrounding milieu ex vivo. The ability to recruit Wnt ligands in this ex vivo model diminished after TNC depletion. These studies indicate that TNC is specifically induced at sites of injury and recruits Wnt ligands, thereby creating a favorable microenvironment for tubular repair and regeneration after AKI.


Subject(s)
Acute Kidney Injury/pathology , Tenascin/metabolism , Wnt Signaling Pathway , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Adult , Animals , Apoptosis , Cell Line , Cisplatin/toxicity , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/pathology , Extracellular Matrix/metabolism , Female , Gene Knockdown Techniques , Healthy Volunteers , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/pathology , Ligands , Male , Mice , Middle Aged , RNA, Small Interfering/metabolism , Regeneration , Reperfusion Injury/complications , Tenascin/blood , Tenascin/genetics , Wnt1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...