Network


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

Hotspot


Dive into the research topics where Manda S. Krishnaveni is active.

Publication


Featured researches published by Manda S. Krishnaveni.


Journal of Biological Chemistry | 2012

Interactions Between β-Catenin and Transforming Growth Factor-β Signaling Pathways Mediate Epithelial-Mesenchymal Transition and Are Dependent on the Transcriptional Co-activator cAMP-response Element-binding Protein (CREB)-binding Protein (CBP)

Beiyun Zhou; Yixin Liu; Michael Kahn; David K. Ann; Arum Han; Hongjun Wang; Cu Nguyen; Per Flodby; Qian Zhong; Manda S. Krishnaveni; Janice M. Liebler; Parviz Minoo; Edward D. Crandall; Zea Borok

Background: Direct evidence for molecular interdependence between transforming growth factor-β (TGF-β) and Wnt pathways in mesenchymal gene regulation during epithelial-mesenchymal transition (EMT) is limited. Results: TGF-β induction of α-smooth muscle actin (α-SMA) involves ternary complex formation among Smad3, β-catenin, and CBP. Conclusion: TGF-β and β-catenin/CBP-dependent pathways coordinately regulate α-SMA induction. Significance: Inhibition of β-catenin/CBP-dependent effects of TGF-β suggests a novel therapeutic approach to EMT/fibrosis. Interactions between transforming growth factor-β (TGF-β) and Wnt are crucial to many biological processes, although specific targets, rationale for divergent outcomes (differentiation versus block of epithelial proliferation versus epithelial-mesenchymal transition (EMT)) and precise mechanisms in many cases remain unknown. We investigated β-catenin-dependent and transforming growth factor-β1 (TGF-β1) interactions in pulmonary alveolar epithelial cells (AEC) in the context of EMT and pulmonary fibrosis. We previously demonstrated that ICG-001, a small molecule specific inhibitor of the β-catenin/CBP (but not β-catenin/p300) interaction, ameliorates and reverses pulmonary fibrosis and inhibits TGF-β1-mediated α-smooth muscle actin (α-SMA) and collagen induction in AEC. We now demonstrate that TGF-β1 induces LEF/TCF TOPFLASH reporter activation and nuclear β-catenin accumulation, while LiCl augments TGF-β-induced α-SMA expression, further confirming co-operation between β-catenin- and TGF-β-dependent signaling pathways. Inhibition and knockdown of Smad3, knockdown of β-catenin and overexpression of ICAT abrogated effects of TGF-β1 on α-SMA transcription/expression, indicating a requirement for β-catenin in these Smad3-dependent effects. Following TGF-β treatment, co-immunoprecipitation demonstrated direct interaction between endogenous Smad3 and β-catenin, while chromatin immunoprecipitation (ChIP)-re-ChIP identified spatial and temporal regulation of α-SMA via complex formation among Smad3, β-catenin, and CBP. ICG-001 inhibited α-SMA expression/transcription in response to TGF-β as well as α-SMA promoter occupancy by β-catenin and CBP, demonstrating a previously unknown requisite TGF-β1/β-catenin/CBP-mediated pro-EMT signaling pathway. Clinical relevance was shown by β-catenin/Smad3 co-localization and CBP expression in AEC of IPF patients. These findings suggest a new therapeutic approach to pulmonary fibrosis by specifically uncoupling CBP/catenin-dependent signaling downstream of TGF-β.


Journal of Clinical Investigation | 2011

Epithelium-specific deletion of TGF-β receptor type II protects mice from bleomycin-induced pulmonary fibrosis

Min Li; Manda S. Krishnaveni; Changgong Li; Beiyun Zhou; Yiming Xing; Agnes Banfalvi; Aimin Li; Vincent Lombardi; Omid Akbari; Zea Borok; Parviz Minoo

Idiopathic pulmonary fibrosis (IPF) is a chronic fibroproliferative pulmonary disorder for which there are currently no treatments. Although the etiology of IPF is unknown, dysregulated TGF-β signaling has been implicated in its pathogenesis. Recent studies also suggest a central role for abnormal epithelial repair. In this study, we sought to elucidate the function of epithelial TGF-β signaling via TGF-β receptor II (TβRII) and its contribution to fibrosis by generating mice in which TβRII was specifically inactivated in mouse lung epithelium. These mice, which are referred to herein as TβRIINkx2.1-cre mice, were used to determine the impact of TβRII inactivation on (a) embryonic lung morphogenesis in vivo; and (b) the epithelial cell response to TGF-β signaling in vitro and in a bleomycin-induced, TGF-β-mediated mouse model of pulmonary fibrosis. Although postnatally viable with no discernible abnormalities in lung morphogenesis and epithelial cell differentiation, TβRIINkx2.1-cre mice developed emphysema, suggesting a requirement for epithelial TβRII in alveolar homeostasis. Absence of TβRII increased phosphorylation of Smad2 and decreased, but did not entirely block, phosphorylation of Smad3 in response to endogenous/physiologic TGF-β. However, TβRIINkx2.1-cre mice exhibited increased survival and resistance to bleomycin-induced pulmonary fibrosis. To our knowledge, these findings are the first to demonstrate a specific role for TGF-β signaling in the lung epithelium in the pathogenesis of pulmonary fibrosis.


American Journal of Respiratory Cell and Molecular Biology | 2011

Role of Endoplasmic Reticulum Stress in Epithelial–Mesenchymal Transition of Alveolar Epithelial Cells: Effects of Misfolded Surfactant Protein

Qian Zhong; Beiyun Zhou; David K. Ann; Parviz Minoo; Yixin Liu; Agnes Banfalvi; Manda S. Krishnaveni; Mickael Dubourd; Lucas DeMaio; Brigham C. Willis; Kwang-Jin Kim; Roland M. duBois; Edward D. Crandall; Michael F. Beers; Zea Borok

Endoplasmic reticulum (ER) stress has been implicated in alveolar epithelial type II (AT2) cell apoptosis in idiopathic pulmonary fibrosis. We hypothesized that ER stress (either chemically induced or due to accumulation of misfolded proteins) is also associated with epithelial-mesenchymal transition (EMT) in alveolar epithelial cells (AECs). ER stress inducers, thapsigargin (TG) or tunicamycin (TN), increased expression of ER chaperone, Grp78, and spliced X-box binding protein 1, decreased epithelial markers, E-cadherin and zonula occludens-1 (ZO-1), increased the myofibroblast marker, α-smooth muscle actin (α-SMA), and induced fibroblast-like morphology in both primary AECs and the AT2 cell line, RLE-6TN, consistent with EMT. Overexpression of the surfactant protein (SP)-C BRICHOS mutant SP-C(ΔExon4) in A549 cells increased Grp78 and α-SMA and disrupted ZO-1 distribution, and, in primary AECs, SP-C(ΔExon4) induced fibroblastic-like morphology, decreased ZO-1 and E-cadherin and increased α-SMA, mechanistically linking ER stress associated with mutant SP to fibrosis through EMT. Whereas EMT was evident at lower concentrations of TG or TN, higher concentrations caused apoptosis. The Src inhibitor, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4]pyramidine) (PP2), abrogated EMT associated with TN or TG in primary AECs, whereas overexpression of SP-C(ΔExon4) increased Src phosphorylation, suggesting a common mechanism. Furthermore, increased Grp78 immunoreactivity was observed in AT2 cells of mice after bleomycin injury, supporting a role for ER stress in epithelial abnormalities in fibrosis in vivo. These results demonstrate that ER stress induces EMT in AECs, at least in part through Src-dependent pathways, suggesting a novel role for ER stress in fibroblast accumulation in pulmonary fibrosis.


The Journal of Pathology | 2012

Ligand-independent transforming growth factor-β type I receptor signalling mediates type I collagen-induced epithelial-mesenchymal transition.

Lucas DeMaio; Stephen T. Buckley; Manda S. Krishnaveni; Per Flodby; Mickael Dubourd; Agnes Banfalvi; Yiming Xing; Carsten Ehrhardt; Parviz Minoo; Beiyun Zhou; Edward D. Crandall; Zea Borok

Evidence suggests epithelial–mesenchymal transition (EMT) as one potential source of fibroblasts in idiopathic pulmonary fibrosis. To assess the contribution of alveolar epithelial cell (AEC) EMT to fibroblast accumulation in vivo following lung injury and the influence of extracellular matrix on AEC phenotype in vitro, Nkx2.1‐Cre;mT/mG mice were generated in which AECs permanently express green fluorescent protein (GFP). On days 17–21 following intratracheal bleomycin administration, ∼4% of GFP‐positive epithelial‐derived cells expressed vimentin or α‐smooth muscle actin (α‐SMA). Primary AECs from Nkx2.1‐Cre;mT/mG mice cultured on laminin‐5 or fibronectin maintained an epithelial phenotype. In contrast, on type I collagen, cells of epithelial origin displayed nuclear localization of Smad3, acquired spindle‐shaped morphology, expressed α‐SMA and phospho‐Smad3, consistent with activation of the transforming growth factor‐β (TGFβ) signalling pathway and EMT. α‐SMA induction and Smad3 nuclear localization were blocked by the TGFβ type I receptor (TβRI, otherwise known as Alk5) inhibitor SB431542, while AEC derived from Nkx2.1‐Cre;Alk5


PLOS ONE | 2012

Troglitazone Attenuates TGF-β1-Induced EMT in Alveolar Epithelial Cells via a PPARγ-Independent Mechanism

Beiyun Zhou; Stephen T. Buckley; Vipul Patel; Yixin Liu; Jiao Luo; Manda S. Krishnaveni; Mihaela Ivan; Lucas DeMaio; Kwang-Jin Kim; Carsten Ehrhardt; Edward D. Crandall; Zea Borok

^{\rm{flox/KO}}


American Journal of Physiology-lung Cellular and Molecular Physiology | 2017

Region-specific role for Pten in maintenance of epithelial phenotype and integrity

Per Flodby; Janice M. Liebler; Mitsuhiro Sunohara; Dan R. Castillo; Alicia M. McConnell; Manda S. Krishnaveni; Agnes Banfalvi; Min Li; Barry R. Stripp; Beiyun Zhou; Edward D. Crandall; Parviz Minoo; Zea Borok

mice did not undergo EMT on collagen, consistent with a requirement for signalling via Alk5 in collagen‐induced EMT. Inability of a pan‐specific TGFβ neutralizing antibody to inhibit effects of collagen together with absence of active TGFβ in culture supernatants is consistent with TGFβ ligand‐independent activation of Smad signalling. These results support the notion that AECs can acquire a mesenchymal phenotype following injury in vivo and implicate type I collagen as a key regulator of EMT in AECs through signalling via Alk5, likely in a TGFβ ligand‐independent manner. Copyright


publisher | None

title

author

Peroxisome proliferator activated receptor γ (PPARγ) agonists are effective antifibrotic agents in a number of tissues. Effects of these agents on epithelial-mesenchymal transition (EMT) of primary alveolar epithelial cells (AEC) and potential mechanisms underlying effects on EMT have not been well delineated. We examined effects of troglitazone, a synthetic PPARγ agonist, on transforming growth factor (TGF)-β1-induced EMT in primary rat AEC and an alveolar epithelial type II (AT2) cell line (RLE-6TN). TGF-β1 (2.5 ng/mL) induced EMT in both cell types, as evidenced by acquisition of spindle-like morphology, increased expression of the mesenchymal marker α-smooth muscle actin (α-SMA) and downregulation of the tight junctional protein zonula occludens-1 (ZO-1). Concurrent treatment with troglitazone (or rosiglitazone), ameliorated effects of TGF-β1. Furthermore, following stimulation with TGF-β1 for 6 days, troglitazone reversed EMT-related morphological changes and restored both epithelial and mesenchymal markers to control levels. Treatment with GW9662 (an irreversible PPARγ antagonist), or overexpression of a PPARγ dominant negative construct, failed to inhibit these effects of troglitazone in AEC. Troglitazone not only attenuated TGF-β1-induced phosphorylation of Akt and glycogen synthase kinase (GSK)-3β, but also inhibited nuclear translocation of β-catenin, phosphorylation of Smad2 and Smad3 and upregulation of the EMT-associated transcription factor SNAI1. These results demonstrate inhibitory actions of troglitazone on TGF-β1-induced EMT in AEC via a PPARγ-independent mechanism likely through inhibition of β-catenin-dependent signaling downstream of TGF-β1, supporting a role for interactions between TGF-β and Wnt/β-catenin signaling pathways in EMT.


american thoracic society international conference | 2012

Wnt/²-Catenin Signaling Induces Epithelial-Mesenchymal Transition (EMT) In Alveolar Epithelial Cells (AEC) Through Activation Of Transforming Growth Factor-² (TGF-²)/Smad Signaling

Yixin Liu; Beiyun Zhou; Michael Kahn; David K. Ann; Arum Han; Manda S. Krishnaveni; Aimin Li; Edward D. Crandall; Zea Borok

Previous studies have demonstrated resistance to naphthalene-induced injury in proximal airways of mice with lung epithelial-specific deletion of the tumor-suppressor gene Pten, attributed to increased proliferation of airway progenitors. We tested effects of Pten loss following bleomycin injury, a model typically used to study distal lung epithelial injury, in conditional PtenSFTPC-cre knockout mice. Pten-deficient airway epithelium exhibited marked hyperplasia, particularly in small bronchioles and at bronchoalveolar duct junctions, with reduced E-cadherin and β-catenin expression between cells toward the luminal aspect of the hyperplastic epithelium. Bronchiolar epithelial and alveolar epithelial type II (AT2) cells in PtenSFTPC-cre mice showed decreased expression of epithelial markers and increased expression of mesenchymal markers, suggesting at least partial epithelial-mesenchymal transition at baseline. Surprisingly, and in contrast to previous studies, mutant mice were exquisitely sensitive to bleomycin, manifesting rapid weight loss, respiratory distress, increased early mortality (by day 5), and reduced dynamic lung compliance. This was accompanied by sloughing of the hyperplastic airway epithelium with occlusion of small bronchioles by cellular debris, without evidence of increased parenchymal lung injury. Increased airway epithelial cell apoptosis due to loss of antioxidant defenses, reflected by decreased expression of superoxide dismutase 3, in combination with deficient intercellular adhesion, likely predisposed to airway sloughing in knockout mice. These findings demonstrate an important role for Pten in maintenance of airway epithelial phenotype integrity and indicate that responses to Pten deletion in respiratory epithelium following acute lung injury are highly context-dependent and region-specific.


american thoracic society international conference | 2011

Induction Of Epithelial-Mesenchymal Transition (EMT) In Alveolar Epithelial Cells (AEC) By Type I Collagen (Col1) Involves Signaling Via Transforming Growth Factor-ß Type I Receptor (TßRI)

Lucas DeMaio; Stephen T. Buckley; Per Flodby; Agnes Banfalvi; Manda S. Krishnaveni; Yiming Xing; Carsten Ehrhardt; Parviz Minoo; Edward D. Crandall; Zea Borok


american thoracic society international conference | 2011

Transforming Growth Factor-Beta1 (TGF-Beta1)-Induced Epithelial Mesenchymal Transition (EMT) Is Nf-kB Dependent In Alveolar Epithelial Cells (AEC)

Manda S. Krishnaveni; Beiyun Zhou; Lucas DeMaio; Edward D. Crandall; Vedang A. Londhe; Parviz Minoo; Zea Borok

Collaboration


Dive into the Manda S. Krishnaveni's collaboration.

Top Co-Authors

Avatar

Zea Borok

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Edward D. Crandall

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Beiyun Zhou

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Parviz Minoo

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Lucas DeMaio

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Agnes Banfalvi

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Per Flodby

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Yixin Liu

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

David K. Ann

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Min Li

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge