Network


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

Hotspot


Dive into the research topics where Mallika Valapala is active.

Publication


Featured researches published by Mallika Valapala.


Autophagy | 2014

Lysosomal-mediated waste clearance in retinal pigment epithelial cells is regulated by CRYBA1/βA3/A1-crystallin via V-ATPase-MTORC1 signaling

Mallika Valapala; Christine Wilson; Stacey Hose; Imran Bhutto; Rhonda Grebe; Aling Dong; Seth Greenbaum; Limin Gu; Samhita Sengupta; Marisol Cano; Sean F. Hackett; Guo-Tong Xu; Gerard A. Lutty; Lijin Dong; Yuri V. Sergeev; James T. Handa; Peter A. Campochiaro; Eric F. Wawrousek; J. Samuel Zigler; Debasish Sinha

In phagocytic cells, including the retinal pigment epithelium (RPE), acidic compartments of the endolysosomal system are regulators of both phagocytosis and autophagy, thereby helping to maintain cellular homeostasis. The acidification of the endolysosomal system is modulated by a proton pump, the V-ATPase, but the mechanisms that direct the activity of the V-ATPase remain elusive. We found that in RPE cells, CRYBA1/βA3/A1-crystallin, a lens protein also expressed in RPE, is localized to lysosomes, where it regulates endolysosomal acidification by modulating the V-ATPase, thereby controlling both phagocytosis and autophagy. We demonstrated that CRYBA1 coimmunoprecipitates with the ATP6V0A1/V0-ATPase a1 subunit. Interestingly, in mice when Cryba1 (the gene encoding both the βA3- and βA1-crystallin forms) is knocked out specifically in RPE, V-ATPase activity is decreased and lysosomal pH is elevated, while cathepsin D (CTSD) activity is decreased. Fundus photographs of these Cryba1 conditional knockout (cKO) mice showed scattered lesions by 4 months of age that increased in older mice, with accumulation of lipid-droplets as determined by immunohistochemistry. Transmission electron microscopy (TEM) of cryba1 cKO mice revealed vacuole-like structures with partially degraded cellular organelles, undigested photoreceptor outer segments and accumulation of autophagosomes. Further, following autophagy induction both in vivo and in vitro, phospho-AKT and phospho-RPTOR/Raptor decrease, while pMTOR increases in RPE cells, inhibiting autophagy and AKT-MTORC1 signaling. Impaired lysosomal clearance in the RPE of the cryba1 cKO mice also resulted in abnormalities in retinal function that increased with age, as demonstrated by electroretinography. Our findings suggest that loss of CRYBA1 causes lysosomal dysregulation leading to the impairment of both autophagy and phagocytosis.


Nature Communications | 2013

Impaired endolysosomal function disrupts Notch signalling in optic nerve astrocytes

Mallika Valapala; Stacey Hose; Céline Gongora; Lijin Dong; Eric F. Wawrousek; J. Samuel Zigler; Debasish Sinha

Astrocytes migrate from the optic nerve into the inner retina, forming a template upon which retinal vessels develop. In the Nuc1 rat, mutation in the gene encoding βA3/A1-crystallin disrupts both Notch signalling in astrocytes and formation of the astrocyte template. Here we show that loss of βA3/A1-crystallin in astrocytes does not impede Notch ligand binding or extracellular cleavages. However, it affects vacuolar-type proton ATPase (V-ATPase) activity, thereby compromising acidification of the endolysosomal compartments, leading to reduced γ-secretase-mediated processing and release of the Notch intracellular domain (NICD). Lysosomal-mediated degradation of Notch is also impaired. These defects decrease the level of NICD in the nucleus, inhibiting the expression of Notch target genes. Overexpression of βA3/A1-crystallin in those same astrocytes restored V-ATPase activity and normal endolysosomal acidification, thereby increasing the levels of γ-secretase to facilitate optimal Notch signalling. We postulate that βA3/A1-crystallin is essential for normal endolysosomal acidification, and thereby, normal activation of Notch signalling in astrocytes.


Aging Cell | 2014

Increased Lipocalin-2 in the retinal pigment epithelium of Cryba1 cKO mice is associated with a chronic inflammatory response.

Mallika Valapala; Malia M. Edwards; Stacey Hose; Rhonda Grebe; Imran Bhutto; Marisol Cano; Thorsten Berger; Tak W. Mak; Eric F. Wawrousek; James T. Handa; Gerard A. Lutty; J. Samuel Zigler; Debasish Sinha

Although chronic inflammation is believed to contribute to the pathology of age‐related macular degeneration (AMD), knowledge regarding the events that elicit the change from para‐inflammation to chronic inflammation in the pathogenesis of AMD is lacking. We propose here that lipocalin‐2 (LCN2), a mammalian innate immunity protein that is trafficked to the lysosomes, may contribute to this process. It accumulates significantly with age in retinal pigment epithelial (RPE) cells of Cryba1 conditional knockout (cKO) mice, but not in control mice. We have recently shown that these mice, which lack βA3/A1‐crystallin specifically in RPE, have defective lysosomal clearance. The age‐related increase in LCN2 in the cKO mice is accompanied by increases in chemokine (C‐C motif) ligand 2 (CCL2), reactive gliosis, and immune cell infiltration. LCN2 may contribute to induction of a chronic inflammatory response in this mouse model with AMD‐like pathology.


Transgenic Research | 2012

βA3/A1-crystallin is required for proper astrocyte template formation and vascular remodeling in the retina

Debasish Sinha; Mallika Valapala; Imran Bhutto; Bonnie Patek; Cheng Zhang; Stacey Hose; Fang Yang; Marisol Cano; Walter J. Stark; Gerard A. Lutty; J. Samuel Zigler; Eric F. Wawrousek

Nuc1 is a spontaneous rat mutant resulting from a mutation in the Cryba1 gene, coding for βA3/A1-crystallin. Our earlier studies with Nuc1 provided novel evidence that astrocytes, which express βA3/A1-crystallin, have a pivotal role in retinal remodeling. The role of astrocytes in the retina is only beginning to be explored. One of the limitations in the field is the lack of appropriate animal models to better investigate the function of astrocytes in retinal health and disease. We have now established transgenic mice that overexpress the Nuc1 mutant form of Cryba1, specifically in astrocytes. Astrocytes in wild type mice show normal compact stellate structure, producing a honeycomb-like network. In contrast, in transgenics over-expressing the mutant (Nuc1) Cryba1 in astrocytes, bundle-like structures with abnormal patterns and morphology were observed. In the nerve fiber layer of the transgenic mice, an additional layer of astrocytes adjacent to the vitreous is evident. This abnormal organization of astrocytes affects both the superficial and deep retinal vascular density and remodeling. Fluorescein angiography showed increased venous dilation and tortuosity of branches in the transgenic retina, as compared to wild type. Moreover, there appear to be fewer interactions between astrocytes and endothelial cells in the transgenic retina than in normal mouse retina. Further, astrocytes overexpressing the mutant βA3/A1-crystallin migrate into the vitreous, and ensheath the hyaloid artery, in a manner similar to that seen in the Nuc1 rat. Together, these data demonstrate that developmental abnormalities of astrocytes can affect the normal remodeling process of both fetal and retinal vessels of the eye and that βA3/A1-crystallin is essential for normal astrocyte function in the retina.


Experimental Eye Research | 2016

Lysosomes: Regulators of autophagy in the retinal pigmented epithelium.

Debasish Sinha; Mallika Valapala; Peng Shang; Stacey Hose; Rhonda Grebe; Gerard A. Lutty; J. Samuel Zigler; Kai Kaarniranta; James T. Handa

The retinal pigmented epithelium (RPE) is critically important to retinal homeostasis, in part due to its very active processes of phagocytosis and autophagy. Both of these processes depend upon the normal functioning of lysosomes, organelles which must fuse with (auto)phagosomes to deliver the hydrolases that effect degradation of cargo. It has become clear that signaling through mTOR complex 1 (mTORC1), is very important in the regulation of lysosomal function. This signaling pathway is becoming a target for therapeutic intervention in diseases, including age-related macular degeneration (AMD), where lysosomal function is defective. In addition, our laboratory has been studying animal models in which the gene (Cryba1) for βA3/A1-crystallin is deficient. These animals exhibit impaired lysosomal clearance in the RPE and pathological signs that are similar to some of those seen in AMD patients. The data demonstrate that βA3/A1-crystallin localizes to lysosomes in the RPE and that it is a binding partner of V-ATPase, the proton pump that acidifies the lysosomal lumen. This suggests that βA3/A1-crystallin may also be a potential target for therapeutic intervention in AMD. In this review, we focus on effector molecules that impact the lysosomal-autophagic pathway in RPE cells.


Aging Cell | 2017

The amino acid transporter SLC36A4 regulates the amino acid pool in retinal pigmented epithelial cells and mediates the mechanistic target of rapamycin, complex 1 signaling

Peng Shang; Mallika Valapala; Rhonda Grebe; Stacey Hose; Sayan Ghosh; Imran Bhutto; James T. Handa; Gerard A. Lutty; Lixia Lu; Jun Wan; Jiang Qian; Yuri V. Sergeev; Rosa Puertollano; J. Samuel Zigler; Guo Tong Xu; Debasish Sinha

The dry (nonneovascular) form of age‐related macular degeneration (AMD), a leading cause of blindness in the elderly, has few, if any, treatment options at present. It is characterized by early accumulation of cellular waste products in the retinal pigmented epithelium (RPE); rejuvenating impaired lysosome function in RPE is a well‐justified target for treatment. It is now clear that amino acids and vacuolar‐type H+‐ATPase (V‐ATPase) regulate the mechanistic target of rapamycin, complex 1 (mTORC1) signaling in lysosomes. Here, we provide evidence for the first time that the amino acid transporter SLC36A4/proton‐dependent amino acid transporter (PAT4) regulates the amino acid pool in the lysosomes of RPE. In Cryba1 (gene encoding βA3/A1‐crystallin) KO (knockout) mice, where PAT4 and amino acid levels are increased in the RPE, the transcription factors EB (TFEB) and E3 (TFE3) are retained in the cytoplasm, even after 24 h of fasting. Consequently, genes in the coordinated lysosomal expression and regulation (CLEAR) network are not activated, and lysosomal function remains low. As these mice age, expression of RPE65 and lecithin retinol acyltransferase (LRAT), two vital visual cycle proteins, decreases in the RPE. A defective visual cycle would possibly slow down the regeneration of new photoreceptor outer segments (POS). Further, photoreceptor degeneration also becomes obvious during aging, reminiscent of human dry AMD disease. Electron microscopy shows basal laminar deposits in Bruchs membrane, a hallmark of development of AMD. For dry AMD patients, targeting PAT4/V‐ATPase in the lysosomes of RPE cells may be an effective means of preventing or delaying disease progression.


Biochimica et Biophysica Acta | 2016

βA3/A1-crystallin and persistent fetal vasculature (PFV) disease of the eye ☆

J. Samuel Zigler; Mallika Valapala; Peng Shang; Stacey Hose; Morton F. Goldberg; Debasish Sinha

BACKGROUND Persistent fetal vasculature (PFV) is a human disease in which the fetal vasculature of the eye fails to regress normally. The fetal, or hyaloid, vasculature nourishes the lens and retina during ocular development, subsequently regressing after formation of the retinal vessels. PFV causes serious congenital pathologies and is responsible for as much as 5% of blindness in the United States. SCOPE OF REVIEW The causes of PFV are poorly understood, however there are a number of animal models in which aspects of the disease are present. One such model results from mutation or elimination of the gene (Cryba1) encoding βA3/A1-crystallin. In this review we focus on the possible mechanisms whereby loss of functional βA3/A1-crystallin might lead to PFV. MAJOR CONCLUSIONS Cryba1 is abundantly expressed in the lens, but is also expressed in certain other ocular cells, including astrocytes. In animal models lacking βA3/A1-crystallin, astrocyte numbers are increased and they migrate abnormally from the retina to ensheath the persistent hyaloid artery. Evidence is presented that the absence of functional βA3/A1-crystallin causes failure of the normal acidification of endolysosomal compartments in the astrocytes, leading to impairment of certain critical signaling pathways, including mTOR and Notch/STAT3. GENERAL SIGNIFICANCE The findings suggest that impaired endolysosomal signaling in ocular astrocytes can cause PFV disease, by adversely affecting the vascular remodeling processes essential to ocular development, including regression of the fetal vasculature. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.


Advances in Experimental Medicine and Biology | 2016

Modulation of V-ATPase by βA3/A1-Crystallin in Retinal Pigment Epithelial Cells

Mallika Valapala; Yuri V. Sergeev; Eric F. Wawrousek; Stacey Hose; J. Samuel Zigler; Debasish Sinha

We have previously demonstrated that βA3/A1-crystallin, a member of the β/γ-crystallin superfamily, is expressed in the astrocytes and retinal pigment epithelial (RPE) cells of the eye. In order to understand the physiological functions of βA3/A1-crystallin in RPE cells, we generated conditional knockout (cKO) mice where Cryba1, the gene encoding βA3/A1-crystallin, is deleted specifically from the RPE using the Cre-loxP system. By utilizing the cKO model, we have shown that this protein is required by RPE cells for proper lysosomal degradation of photoreceptor outer segments (OS) that have been internalized in phagosomes and also for the proper functioning of the autophagy process. We also reported that βA3/A1-crystallin is trafficked to lysosomes, where it regulates endolysosomal acidification by modulating the activity of the lysosomal V-ATPase complex. Our results show that the V-ATPase activity in cKO RPE is significantly lower than WT RPE. Since, V-ATPase is important for regulating lysosomal pH, we noticed that endolysosomal pH was higher in the cKO cells compared to the WT cells. Increased lysosomal pH in cKO RPE is also associated with reduced Cathepsin D activity. Cathepsin D is a major lysosomal aspartic protease involved in the degradation of the OS and hence we believe that reduced proteolytic activity contributes to impaired degradation of OS in the cKO RPE. Reduced lysosomal activity in the cKO RPE also contributes to the incomplete degradation of the autophagosomes. Our results also suggest that βA3/A1-crystallin regulates V-ATPase activity by binding to the V0 subunit of the V-ATPase complex. Taken together, these results suggest a novel mechanism by which βA3/A1-crystallin regulates lysosomal function by modulating the activity of V-ATPase.


Investigative Ophthalmology & Visual Science | 2015

Loss of βA3/A1-crystallin in astrocytes affects cell migration by downregulating Rho family of proteins

Debasish Sinha; Mallika Valapala; Stacey Hose; Shang Peng; J. Samuel Zigler


Investigative Ophthalmology & Visual Science | 2015

Activation of mTORC1 in RPE leads to RPE cell death by modulating PTEN and BAD

Mallika Valapala; Shang Peng; Stacey Hose; J. Samuel Zigler; Debasish Sinha

Collaboration


Dive into the Mallika Valapala's collaboration.

Top Co-Authors

Avatar

Stacey Hose

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Debasish Sinha

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

J. Samuel Zigler

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Eric F. Wawrousek

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rhonda Grebe

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Imran Bhutto

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

James T. Handa

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Lijin Dong

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peng Shang

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge