Network


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

Hotspot


Dive into the research topics where Joyce Christina F. Ibe is active.

Publication


Featured researches published by Joyce Christina F. Ibe.


American Journal of Respiratory Cell and Molecular Biology | 2013

Adenosine monophosphate-activated protein kinase is required for pulmonary artery smooth muscle cell survival and the development of hypoxic pulmonary hypertension.

Joyce Christina F. Ibe; Qiyuan Zhou; Tianji Chen; Haiyang Tang; Jason X.-J. Yuan; J. Usha Raj; Guofei Zhou

Human pulmonary artery smooth muscle cells (HPASMCs) express both adenosine monophosphate-activated protein kinase (AMPK) α1 and α2. We investigated the distinct roles of AMPK α1 and α2 in the survival of HPASMCs during hypoxia and hypoxia-induced pulmonary hypertension (PH). The exposure of HPASMCs to hypoxia (3% O2) increased AMPK activation and phosphorylation, and the inhibition of AMPK with Compound C during hypoxia decreased their viability and increased lactate dehydrogenase activity and apoptosis. Although the suppression of either AMPK α1 or α2 expression led to increased cell death, the suppression of AMPK α2 alone increased caspase-3 activity and apoptosis in HPASMCs exposed to hypoxia. It also resulted in the decreased expression of myeloid cell leukemia sequence 1 (MCL-1). The knockdown of MCL-1 or MCL-1 inhibitors increased caspase-3 activity and apoptosis in HPASMCs exposed to hypoxia. On the other hand, the suppression of AMPK α1 expression alone prevented hypoxia-mediated autophagy. The inhibition of autophagy induced cell death in HPASMCs. Our results suggest that AMPK α1 and AMPK α2 play differential roles in the survival of HPASMCs during hypoxia. The activation of AMPK α2 maintains the expression of MCL-1 and prevents apoptosis, whereas the activation of AMPK α1 stimulates autophagy, promoting HPASMC survival. Moreover, treatment with Compound C, which inhibits both isoforms of AMPK, prevented and partly reversed hypoxia-induced PH in mice. Taking these results together, our study suggests that AMPK plays a key role in the pathogenesis of pulmonary arterial hypertension, and AMPK may represent a novel therapeutic target for the treatment of pulmonary arterial hypertension.


Cellular Signalling | 2012

cAMP-dependent protein kinase is essential for hypoxia-mediated epithelial-mesenchymal transition, migration, and invasion in lung cancer cells.

Dooniya Shaikh; Qiyuan Zhou; Tianji Chen; Joyce Christina F. Ibe; J. Usha Raj; Guofei Zhou

Lung cancer is the leading cause of cancer-related death worldwide. Hypoxia is known to increase cancer cell migration and invasion. We have previously reported that hypoxia induces epithelial-mesenchymal transition (EMT) in lung cancer cells. However, it is unknown whether hypoxia promotes lung cancer cell migration and invasion via EMT and whether cyclic AMP (cAMP) dependent protein kinase (PKA) plays a role in this process. We found that hypoxia increased PKA activity and induced mRNA and protein expression of PKA catalytic subunit α (PKACA), and regulatory subunits R1A and R1B. Knockdown of HIF-1/2α prevented hypoxia-mediated induction of PKACA mRNA expression and PKA activity. Inhibition of PKA activity with chemical inhibitors prevented EMT induced by hypoxia and tumor growth factor β1. However, activation of PKA by forskolin and 8-Br-cAMP did not induce EMT. Furthermore, treatment with H89 and knockdown of PKACA prevented hypoxia-mediated, EMT, cell migration, and invasion, whereas overexpression of mouse PKACA rescued hypoxia-mediated migration and invasion in PKACA deficient cancer cells. Our results suggest that hypoxia enhances PKA activity by upregulating PKA gene expression in a HIF dependent mechanism and that PKA plays a key role in hypoxia-mediated EMT, migration, and invasion in lung cancer cells.


American Journal of Respiratory and Critical Care Medicine | 2015

Loss of MicroRNA-17∼92 in Smooth Muscle Cells Attenuates Experimental Pulmonary Hypertension via Induction of PDZ and LIM Domain 5

Tianji Chen; Guofei Zhou; Qiyuan Zhou; Haiyang Tang; Joyce Christina F. Ibe; Hongqiang Cheng; Deming Gou; Ju Chen; Jason X.-J. Yuan; J. Usha Raj

RATIONALE Recent studies suggest that microRNAs (miRNAs) play important roles in regulation of pulmonary artery smooth muscle cell (PASMC) phenotype and are implicated in pulmonary arterial hypertension (PAH). However, the underlying molecular mechanisms remain elusive. OBJECTIVES This study aims to understand the mechanisms regulating PASMC proliferation and differentiation by microRNA-17∼92 (miR-17∼92) and to elucidate its implication in PAH. METHODS We generated smooth muscle cell (SMC)-specific miR-17∼92 and PDZ and LIM domain 5 (PDLIM5) knockout mice and overexpressed miR-17∼92 and PDLIM5 by injection of miR-17∼92 mimics or PDLIM5-V5-His plasmids and measured their responses to hypoxia. We used miR-17∼92 mimics, inhibitors, overexpression vectors, small interfering RNAs against PDLIM5, Smad, and transforming growth factor (TGF)-β to determine the role of miR-17∼92 and its downstream targets in PASMC proliferation and differentiation. MEASUREMENTS AND MAIN RESULTS We found that human PASMC (HPASMC) from patients with PAH expressed decreased levels of the miR-17∼92 cluster, TGF-β, and SMC markers. Overexpression of miR-17∼92 increased and restored the expression of TGF-β3, Smad3, and SMC markers in HPASMC of normal subjects and patients with idiopathic PAH, respectively. Knockdown of Smad3 but not Smad2 prevented miR-17∼92-induced expression of SMC markers. SMC-specific knockout of miR-17∼92 attenuated hypoxia-induced pulmonary hypertension (PH) in mice, whereas reconstitution of miR-17∼92 restored hypoxia-induced PH in these mice. We also found that PDLIM5 is a direct target of miR-17/20a, and hypertensive HPASMC and mouse PASMC expressed elevated PDLIM5 levels. Suppression of PDLIM5 increased expression of SMC markers and enhanced TGF-β/Smad2/3 activity in vitro and enhanced hypoxia-induced PH in vivo, whereas overexpression of PDLIM5 attenuated hypoxia-induced PH. CONCLUSIONS We provided the first evidence that miR-17∼92 inhibits PDLIM5 to induce the TGF-β3/SMAD3 pathway, contributing to the pathogenesis of PAH.


American Journal of Respiratory Cell and Molecular Biology | 2012

Hypoxia-Induced Pulmonary Arterial Smooth Muscle Cell Proliferation Is Controlled by Forkhead Box M1

Aarti Raghavan; Guofei Zhou; Qiyuan Zhou; Joyce Christina F. Ibe; Ramaswamy Ramchandran; Qiwei Yang; Harini Racherla; Pradip Raychaudhuri; J. Usha Raj

Pulmonary arterial hypertension (PAH) is a devastating disease, and no effective treatments are available. Hypoxia-induced pulmonary artery remodeling, including smooth muscle cell proliferation, contributes to PAH, but the exact mechanisms underlying this abnormal process are largely undefined. The forkhead box M1 (FoxM1) transcription factor regulates cancer cell growth by modulating gene expression critical for cell cycle progression. Here, we report for the first time, to the best of our knowledge, a novel function of FoxM1 in the hypoxia-stimulated proliferation of human pulmonary artery smooth muscle cells (HPASMCs). Exposure to hypoxia caused a marked up-regulation of FoxM1 gene expression, mainly at the transcription level, and this induction correlated with HPASMC cell proliferation. The knockdown of FoxM1 inhibited the hypoxia-stimulated proliferation of HPASMCs. We found that the knockdown of HIF-2α, but not HIF-1α, diminished FoxM1 induction in response to hypoxia. However, the knockdown of FoxM1 did not alter expression levels of HIF-2α or HIF-1α, suggesting that HIF-2α is an upstream regulator of FoxM1. Furthermore, the knockdown of FoxM1 prevented the hypoxia-induced expression of aurora A kinase and cyclin D1. Collectively, our results suggest that hypoxia induces FoxM1 gene expression in an HIF-2α-dependent pathway, thereby promoting HPASMC proliferation.


FEBS Letters | 2012

Knockdown of von Hippel–Lindau protein decreases lung cancer cell proliferation and colonization

Qiyuan Zhou; Tianji Chen; Joyce Christina F. Ibe; J. Usha Raj; Guofei Zhou

Although von Hippel–Lindau protein (pVHL) is known as a tumor suppressor in kidney and other organs, it remains unclear whether pVHL plays a role in lung cancer development. We investigated the role of pVHL in lung cancer cell proliferation, migration, and colonization using stable A549 cells with knockdown of pVHL. We found that knockdown of pVHL promotes epithelial‐mesenchymal transition (EMT) in lung cancer cells. Knockdown of pVHL decreased tumor colonization in a tail‐vein injection model and decreased cell proliferation, whereas overexpression of constitutive active HIF increased tumor colonization, suggesting a HIF‐independent function of pVHL in lung. Knockdown of pVHL decreased phosphorylation of FAK and expression of integrin, suggesting that pVHL regulates lung cancer development via integrin/FAK signaling pathway.


PLOS ONE | 2014

Intratracheal instillation of high dose adenoviral vectors is sufficient to induce lung injury and fibrosis in mice.

Qiyuan Zhou; Tianji Chen; Melike Bozkanat; Joyce Christina F. Ibe; John W. Christman; J. Usha Raj; Guofei Zhou

Rationale Replication deficient adenoviruses (Ad) vectors are common tools in gene therapy. Since Ad vectors are known to activate innate and adaptive immunity, we investigated whether intratracheal administration of Ad vectors alone is sufficient to induce lung injury and pulmonary fibrosis. Methods We instilled Ad viruses ranging from 107 to 1.625×109 ifu/mouse as well as the same volume of PBS and bleomycin. 14 and 21 days after administration, we collected bronchoalveolar lavage fluid (BALF) and mouse lung tissues. We measured the protein concentration, total and differential cell counts, and TGF-β1 production, performed Trichrome staining and Sircol assay, determined gene and protein levels of profibrotic cytokines, MMPs, and Wnt signaling proteins, and conducted TUNEL staining and co-immunofluorescence for GFP and α-SMA staining. Results Instillation of high dose Ad vectors (1.625×109 ifu/mouse) into mouse lungs induced high levels of protein content, inflammatory cells, and TGF-β1 in BALF, comparable to those in bleomycin-instilled lungs. The collagen content and mRNA levels of Col1a1, Col1a2, PCNA, and α-SMA were also increased in the lungs. Instillation of both bleomycin and Ad vectors increased expression levels of TNFα and IL-1β but not IL-10. Instillation of bleomycin but not Ad increased the expression of IL-1α, IL-13 and IL-16. Treatment with bleomycin or Ad vectors increased expression levels of integrin α1, α5, and αv, MMP9, whereas treatment with bleomycin but not Ad vectors induced MMP2 expression levels. Both bleomycin and Ad vectors induced mRNA levels of Wnt2, 2b, 5b, and Lrp6. Intratracheal instillation of Ad viruses also induced DNA damages and Ad viral infection-mediated fibrosis is not limited to the infection sites. Conclusions Our results suggest that administration of Ad vectors induces an inflammatory response, lung injury, and pulmonary fibrosis in a dose dependent manner.


Cancer Research | 2012

Abstract 329: Downregulation of PKC∈/par3/par6 polarity complex is critical for hypoxia-induced epithelial-mesenchymal transition, migration, and invasion in lung cancer cells

Qiyuan Zhou; Tianji Chen; Joyce Christina F. Ibe; J. Usha Raj; Guofei Zhou

Rationale: We have previously reported that hypoxia induces epithelial-mesenchymal transition (EMT) in lung cancer cells. EMT has been known to contribute to tumor cell invasion and metastasis. Recent evidence suggests that loss of cell polarity complex, which consists of atypical protein kinase C ≤ (PKC∈), Par (partitioning defective) 3, and Par6, is strongly correlated with malignancy. However, it is unknown whether hypoxia-induced cancer cell migration, invasion, and metastasis are caused by the loss of PKC∈/Par3/Par6 polarity complex and subsequent EMT. Methods: We incubated A549 in hypoxic conditions (1.5% O 2 ) for a period of time and assessed the protein abundance of α-SMA, vimentin, E-cadherin, PKC∈, Par3, and Par6 as well as PKC∈ ubiquitination. Next we knocked down PKC∈/Par3/Par6 by siRNAs and determined the abundance of α-SMA, vimentin, and E-cadherin. We also measured cell migration and invasion after knockdown of PKC∈/Par3/Par6 in vitro. In addition, we established A549 cells with knockdown of Par3 and determined cell proliferation rate and lung colonization in a mouse tail-vein injection model, in which lung cancer cells were injected intravenously into the tail vein of mice and lung metastatic foci were quantified in five weeks. Furthermore, we obtained human lung adenocarcinoma tissue samples and self-matched adjacent normal tissues as well as unmatched normal tissues and detected the expression levels of PKC∈/Par3/Par6 polarity complex. Results: We found that A549 cells exposed to hypoxia expressed decreased PKC∈/Par3/Par6 polarity complex and E-cadherin and increased expression of α-SMA and vimentin, suggesting that loss of PKC∈/Par3/Par6 polarity complex is associated with the induction of EMT. We also showed that hypoxia increased ubiquitination of PKC∈. Our results also showed that loss of either of PKC∈, Par3, or Par6 resulted in EMT, elevated migration and invasion, and increased the numbers of metastatic foci in the lungs. Moreover, we show that human lung adenocarcinoma tissues express less Par6 protein than the adjacent normal tissues. Conclusions: These results suggest that in lung cancer cells hypoxia downregulates PKC∈/Par3/par6, leading to EMT and increased cancer cell migration, invasion, and colonization. Support: UIC Faculty Scholarship Support Program and the University of Illinois Cancer Center Pilot Subsidy Program. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 329. doi:1538-7445.AM2012-329


Cell Cycle | 2011

Loss of either hypoxia inducible factor 1 or 2 promotes lung cancer cell colonization

Qiyuan Zhou; Tianji Chen; Joyce Christina F. Ibe; J. Usha Raj; Guofei Zhou


american thoracic society international conference | 2012

Adenosine Monophosphate-Activated Protein Kinase ±2 Is Critical For The Survival Of Human Pulmonary Artery Smooth Muscle Cells During Hypoxia

Joyce Christina F. Ibe; Guofei Zhou; Qiyuan Zhou; Tianji Chen; J. Usha Raj


american thoracic society international conference | 2012

MiR-17~92 Regulates Differentiation Of Pulmonary Arterial Smooth Muscle Cells Via A TGF-² Dependent Pathway

Tianji Chen; Guofei Zhou; Qiyuan Zhou; Joyce Christina F. Ibe; Suzy Comhair; Serpil C. Erzurum; J. U. Raj

Collaboration


Dive into the Joyce Christina F. Ibe's collaboration.

Top Co-Authors

Avatar

Guofei Zhou

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Qiyuan Zhou

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

J. Usha Raj

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Tianji Chen

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Aarti Raghavan

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harini Racherla

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pradip Raychaudhuri

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Ramaswamy Ramchandran

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge