albumin endocytosis
Recently Published Documents


TOTAL DOCUMENTS

36
(FIVE YEARS 13)

H-INDEX

13
(FIVE YEARS 0)

2022 ◽  
Vol 23 (2) ◽  
pp. 848
Author(s):  
Rodrigo P. Silva-Aguiar ◽  
Diogo B. Peruchetti ◽  
Lucas S. Florentino ◽  
Christina M. Takiya ◽  
María-Paz Marzolo ◽  
...  

Renal proximal tubule cells (PTECs) act as urine gatekeepers, constantly and efficiently avoiding urinary protein waste through receptor-mediated endocytosis. Despite its importance, little is known about how this process is modulated in physiologic conditions. Data suggest that the phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT) pathway regulates PTEC protein reabsorption. Here, we worked on the hypothesis that the physiologic albumin concentration and PI3K/AKT pathway form a positive feedback loop to expand endocytic capacity. Using LLC-PK1 cells, a model of PTECs, we showed that the PI3K/AKT pathway is required for megalin recycling and surface expression, affecting albumin uptake. Inhibition of this pathway stalls megalin at EEA1+ endosomes. Physiologic albumin concentration (0.01 mg/mL) activated AKT; this depends on megalin-mediated albumin endocytosis and requires previous activation of PI3K/mTORC2. This effect is correlated to the increase in albumin endocytosis, a phenomenon that we refer to as “albumin-induced albumin endocytosis”. Mice treated with L-lysine present decreased albumin endocytosis leading to proteinuria and albuminuria associated with inhibition of AKT activity. Renal cortex explants obtained from control mice treated with MK-2206 decreased albumin uptake and promoted megalin internalization. Our data highlight the mechanism behind the capacity of PTECs to adapt albumin reabsorption to physiologic fluctuations in its filtration, avoiding urinary excretion.


Peptides ◽  
2021 ◽  
Vol 146 ◽  
pp. 170646
Author(s):  
Sarah A.S. Alves ◽  
Lucas S. Florentino ◽  
Douglas E. Teixeira ◽  
Rodrigo P. Silva-Aguiar ◽  
Diogo B. Peruchetti ◽  
...  

2021 ◽  
Vol 35 (S1) ◽  
Author(s):  
Rodrigo Silva‐Aguiar ◽  
Diogo Peruchetti ◽  
Christina Takiya ◽  
Maria Marzolo ◽  
Ana Acacia Pinheiro ◽  
...  

2020 ◽  
Vol 529 (3) ◽  
pp. 740-746
Author(s):  
Mark A. Bryniarski ◽  
Benjamin M. Yee ◽  
Lee D. Chaves ◽  
Corrine M. Stahura ◽  
Rabi Yacoub ◽  
...  

2020 ◽  
Vol 133 (13) ◽  
pp. jcs242859
Author(s):  
Seiya Urae ◽  
Yutaka Harita ◽  
Tomohiro Udagawa ◽  
Koji L. Ode ◽  
Masami Nagahama ◽  
...  

PeerJ ◽  
2020 ◽  
Vol 8 ◽  
pp. e8599
Author(s):  
Huijun Shen ◽  
Yu Bao ◽  
Chunyue Feng ◽  
Haidong Fu ◽  
Jianhua Mao

Background As a fundamental process internalizing molecules from the plasma membrane, endocytosis plays a crucial role in podocyte biology. Our previous study has identified that overexpression of Myole may enhance podocyte endocytosis. However, its potential mechanism has been not well understand. Thus, we aimed to analyze whether albumin endocytosis by mouse glomerular podocytes is dependent on Myo1e expression. Also, we aimed to elucidate whether the underlying mechanism is mediated by Dynamin. Methods Firstly, mouse podocyte cells (MPC5) were treated with different concentrations of FITC-bovine serum albumin (BSA). The fluorescence intensity and cell viability were detected by flow cytometry and MTT assays, respectively. Afterwards, the optimal concentration of FITC-BSA was determined. Secondly, MPC5 cells were treated with Myole overexpression or knockdown. Cell morphology was observed under microscope. Immunofluorescence assay was used to determine the expression of F-actin. The protein expression of nephrin and podocin was detected by western blot. Flow cytometry was used to detect MPC5 cell apoptosis with annexin V. Finally, MPC5 cells were treated with Myole overexpression and/or Dynasore (a GTPase inhibitor of Dynamin). The fluorescence intensity was detected using flow cytometry assay. Results MPC5 endocytosis BSA was elevated with a concentration-dependent manner. MTT results showed that MPC5 cell viability was inhibited with a concentration-dependent manner. Myo1e overexpression promoted podocyte endocytic FITC-BSA, which was contrary to its knockdown. Under microscope, after inhibition of Myo1e, podocyte foot process fusion was observed. Myo1e overexpression promoted the expression of cytoskeleton F-actin and podocyte-specific molecules (nephrin and podocin) in podocyte endocytic FITC-BSA. Furthermore, we found that Myo1e promoted the apoptosis of podocytes. Dynasore attenuated the increase in endocytosis of FITC-BSA induced by Myo1e overexpression, suggesting that podocytes might mediate albumin endocytosis via Myo1e-Dynamin-Albumin. Conclusion Our findings revealed that overexpression of Myo1e promotes albumin endocytosis in mouse glomerular podocyte endocytic albumin mediated by Dynamin.


Sign in / Sign up

Export Citation Format

Share Document