Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Gabriel C. Baltazar is active.

Publication


Featured researches published by Gabriel C. Baltazar.


PLOS ONE | 2012

Acidic Nanoparticles Are Trafficked to Lysosomes and Restore an Acidic Lysosomal pH and Degradative Function to Compromised ARPE-19 Cells

Gabriel C. Baltazar; Sonia Guha; Wennan Lu; Jason C. Lim; Kathleen Boesze-Battaglia; Alan M. Laties; Puneet Tyagi; Uday B. Kompella; Claire H. Mitchell

Lysosomal enzymes function optimally in acidic environments, and elevation of lysosomal pH can impede their ability to degrade material delivered to lysosomes through autophagy or phagocytosis. We hypothesize that abnormal lysosomal pH is a key aspect in diseases of accumulation and that restoring lysosomal pH will improve cell function. The propensity of nanoparticles to end up in the lysosome makes them an ideal method of delivering drugs to lysosomes. This study asked whether acidic nanoparticles could traffic to lysosomes, lower lysosomal pH and enhance lysosomal degradation by the cultured human retinal pigmented epithelial cell line ARPE-19. Acidic nanoparticles composed of poly (DL-lactide-co-glycolide) (PLGA) 502 H, PLGA 503 H and poly (DL-lactide) (PLA) colocalized to lysosomes of ARPE-19 cells within 60 min. PLGA 503 H and PLA lowered lysosomal pH in cells compromised by the alkalinizing agent chloroquine when measured 1 hr. after treatment, with acidification still observed 12 days later. PLA enhanced binding of Bodipy-pepstatin-A to the active site of cathepsin D in compromised cells. PLA also reduced the cellular levels of opsin and the lipofuscin-like autofluorescence associated with photoreceptor outer segments. These observations suggest the acidification produced by the nanoparticles was functionally effective. In summary, acid nanoparticles lead to a rapid and sustained lowering of lysosomal pH and improved degradative activity.


American Journal of Physiology-cell Physiology | 2012

Cystic fibrosis transmembrane conductance regulator contributes to reacidification of alkalinized lysosomes in RPE cells

Ji Liu; Wennan Lu; Sonia Guha; Gabriel C. Baltazar; Erin E. Coffey; Alan M. Laties; Ronald C. Rubenstein; William W. Reenstra; Claire H. Mitchell

The role of the cystic fibrosis transmembrane conductance regulator (CFTR) in lysosomal acidification has been difficult to determine. We demonstrate here that CFTR contributes more to the reacidification of lysosomes from an elevated pH than to baseline pH maintenance. Lysosomal alkalinization is increasingly recognized as a factor in diseases of accumulation, and we previously showed that cAMP reacidified alkalinized lysosomes in retinal pigmented epithelial (RPE) cells. As the influx of anions to electrically balance proton accumulation may enhance lysosomal acidification, the contribution of the cAMP-activated anion channel CFTR to lysosomal reacidification was probed. The antagonist CFTR(inh)-172 had little effect on baseline levels of lysosomal pH in cultured human RPE cells but substantially reduced the reacidification of compromised lysosomes by cAMP. Likewise, CFTR activators had a bigger impact on cells whose lysosomes had been alkalinized. Knockdown of CFTR with small interfering RNA had a larger effect on alkalinized lysosomes than on baseline levels. Inhibition of CFTR in isolated lysosomes altered pH. While CFTR and Lamp1 were colocalized, treatment with cAMP did not increase targeting of CFTR to the lysosome. The inhibition of CFTR slowed lysosomal degradation of photoreceptor outer segments while activation of CFTR enhanced their clearance from compromised lysosomes. Activation of CFTR acidified RPE lysosomes from the ABCA4(-/-) mouse model of recessive Stargardts disease, whose lysosomes are considerably alkalinized. In summary, CFTR contributes more to reducing lysosomal pH from alkalinized levels than to maintaining baseline pH. Treatment to activate CFTR may thus be of benefit in disorders of accumulation associated with lysosomal alkalinization.


Journal of Neurochemistry | 2012

Stimulation of the D5 dopamine receptor acidifies the lysosomal pH of retinal pigmented epithelial cells and decreases accumulation of autofluorescent photoreceptor debris.

Sonia Guha; Gabriel C. Baltazar; Leigh-Anne Tu; Ji Liu; Jason C. Lim; Wennan Lu; Arthur J. Argall; Kathleen Boesze-Battaglia; Alan M. Laties; Claire H. Mitchell

J. Neurochem. (2012) 122, 823–833.


The FASEB Journal | 2013

Lysosomal alkalinization, lipid oxidation, and reduced phagosome clearance triggered by activation of the P2X7 receptor

Sonia Guha; Gabriel C. Baltazar; Erin E. Coffey; Leigh-Anne Tu; Jason C. Lim; Jonathan M. Beckel; Shaun Patel; Thor Eysteinsson; Wennan Lu; Ann O'Brien-Jenkins; Alan M. Laties; Claire H. Mitchell


The FASEB Journal | 2011

P2X7 receptor stimulation raises lysosomal pH and triggers cytokine release from RPE cells

Sonia Guha; Gabriel C. Baltazar; Leigh-Anne Tu; Jason C. Lim; Thor Eysteinsson; Alan M. Laties; Claire H. Mitchell


Investigative Ophthalmology & Visual Science | 2011

Acidic Nanoparticles Enhance Degradative Lysosomal Enzyme Activity In Compromised Rpe Cells

Gabriel C. Baltazar; Sonia Guha; Alan M. Laties; Puneet Tyagi; Uday B. Kompella; Claire H. Mitchell


Investigative Ophthalmology & Visual Science | 2010

Reduction of Lipofuscin-Like Autofluorescence in RPE Cells by Sustained D1/D5 Receptor Stimulation

Sonia Guha; Leigh-Anne Tu; Gabriel C. Baltazar; A. Argalla; Alan M. Laties; Claire H. Mitchell


Investigative Ophthalmology & Visual Science | 2010

Acidification of RPE Lysosomes by Nanoparticles

Gabriel C. Baltazar; Sonia Guha; Alan M. Laties; Uday B. Kompella; Claire H. Mitchell


Archive | 2016

RPE cells contributes to reacidification of alkalinized lysosomes in Cystic fibrosis transmembrane conductance regulator

Ronald C. Rubenstein; William W. Reenstra; Claire H. Mitchell; Ji Liu; Wennan Lu; Sonia Guha; Gabriel C. Baltazar; Erin E. Coffey; Alan M. Laties


Investigative Ophthalmology & Visual Science | 2012

Multiple Autofluorescent Peaks In RPE Of Control And ABCA4-/- Mice: A Role For Perturbed Autophagy?

Ann O'Brien-Jenkins; Sonia Guha; Wennan Lu; Bardia Nabet; Gabriel C. Baltazar; Alan M. Laties; Claire H. Mitchell

Collaboration


Dive into the Gabriel C. Baltazar's collaboration.

Top Co-Authors

Avatar

Alan M. Laties

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sonia Guha

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Wennan Lu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jason C. Lim

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Leigh-Anne Tu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Erin E. Coffey

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Uday B. Kompella

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge