Network


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

Hotspot


Dive into the research topics where Hanning You is active.

Publication


Featured researches published by Hanning You.


Hepatology | 2010

Epigenetic Regulation of Cancer Stem Cell Marker CD133 by Transforming Growth Factor-β

Hanning You; Wei Ding; C. Bart Rountree

Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality worldwide. CD133, a transmembrane glycoprotein, is an important cell surface marker for both stem cells and cancer stem cells in various tissues including liver. CD133 expression has been recently linked to poor prognosis in HCC patients. CD133+ liver cancer cells are characterized by resistance to chemotherapy, self‐renewal, multilineage potential, increased colony formation, and in vivo cancer initiation at limited dilution. Recent studies demonstrate that CD133 expression is regulated by DNA methylation. In this study, we explored the role of transforming growth factor β (TGFβ), a multifunctional cytokine that plays a critical role in chronic liver injury, in the regulation of CD133 expression. TGFβ1 is capable of up‐regulating CD133 expression specifically within the Huh7 HCC cell line in a time‐ and dose‐dependent manner. Most important, TGFβ1‐induced CD133+ Huh7 cells demonstrate increased tumor initiation in vivo. Forced expression of inhibitory Smads, including Smad6 and Smad7, attenuated TGFβ1‐induced CD133 expression. Within CD133− Huh7 cells, TGFβ1 stimulation inhibited the expression of DNA methyltransferases (DNMT) 1 and DNMT3β, which are critical in the maintenance of regional DNA methylation, and global DNMT activity in CD133− Huh7 cells was inhibited by TGFβ1. DNMT3β inhibition by TGFβ1 was partially rescued with overexpression of inhibitory Smads. Lastly, TGFβ1 treatment led to significant demethylation in CD133 promoter‐1 in CD133− Huh7 cells. Conclusion: TGFβ1 is able to regulate CD133 expression through inhibition of DNMT1 and DNMT3β expression and subsequent demethylation of promoter‐1. TGFβ1‐induced CD133+ Huh7 cells are tumorigenic. The mechanism by which TGFβ induces CD133 expression is partially dependent on the Smads pathway. HEPATOLOGY 2010


Hepatology | 2011

c‐Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinoma

Hanning You; Wei Ding; Hien Dang; Yixing Jiang; C. Bart Rountree

c‐Met, a high‐affinity receptor for hepatocyte growth factor (HGF), plays a critical role in cancer growth, invasion, and metastasis. Hepatocellular carcinoma (HCC) patients with an active HGF/c‐Met signaling pathway have a significantly worse prognosis. Although targeting the HGF/c‐Met pathway has been proposed for the treatment of multiple cancers, the effect of c‐Met inhibition in HCC remains unclear. The human HCC cell lines Huh7, Hep3B, MHCC97‐L, and MHCC97‐H were used in this study to investigate the effect of c‐Met inhibition using the small molecule selective c‐Met tyrosine kinase inhibitor PHA665752. MHCC97‐L and MHCC97‐H cells demonstrate a mesenchymal phenotype with decreased expression of E‐cadherin and increased expression of c‐Met, fibronectin, and Zeb2 compared with Huh7 and Hep3B cells, which have an epithelial phenotype. PHA665752 treatment blocked phosphorylation of c‐Met and downstream phosphoinositide 3‐kinase/Akt and mitogen‐activated protein kinase/Erk pathways, inhibited cell proliferation, and induced apoptosis in c‐Met–positive MHCC97‐L and MHCC97‐H cells. In xenograft models, administration of PHA665752 significantly inhibited c‐Met–positive MHCC97‐L and MHCC97‐H tumor growth, and PHA665752‐treated tumors demonstrated marked reduction of both c‐Met phosphorylation and cell proliferation. c‐Met–negative Huh7 and Hep3B cells were not affected by c‐Met inhibitor treatment in vitro or in vivo. In addition, c‐Met–positive MHCC97‐L and MHCC97‐H cells demonstrated cancer stem cell–like characteristics, such as resistance to chemotherapy, tumor sphere formation, and increased expression of CD44 and ABCG2, and PHA665752 treatment suppressed tumor sphere formation and inhibited CD44 expression. Conclusion: c‐Met represents a potential target of personalized treatment for HCC with an active HGF/c‐Met pathway. (HEPATOLOGY 2011;)


Hepatology | 2010

Epithelial-to-mesenchymal transition of murine liver tumor cells promotes invasion†‡

Wei Ding; Hanning You; Hien Dang; Francis LeBlanc; Vivian Galicia; Shelly C. Lu; Bangyan L. Stiles; C. Bart Rountree

Epithelial‐to‐mesenchymal transition (EMT) is predicted to play a critical role in metastatic disease in hepatocellular carcinoma. In this study, we used a novel murine model of EMT to elucidate a mechanism of tumor progression and metastasis. A total of 2 × 106 liver cells isolated from Ptenloxp/loxp/Alb‐Cre+ mice, expanded from a single CD133+CD45− cell clone, passage 0 (P0), were sequentially transplanted to obtain two passages of tumor cells, P1 and P2. Cells were analyzed for gene expression using microarray and real‐time polymerase chain reaction. Functional analysis included cell proliferation, migration, and invasion in vitro and orthotopic tumor metastasis assays in vivo. Although P0, P1, and P2 each formed tumors consistent with mixed liver epithelium, within the P2 cells, two distinct cell types were clearly visible: cells with epithelial morphology similar to P0 cells and cells with fibroblastoid morphology. These P2 mesenchymal cells demonstrated increased locomotion on wound healing; increased cell invasion on Matrigel basement membrane; increased EMT‐associated gene expression of Snail1, Zeb1, and Zeb2; and down‐regulated E‐cadherin. P2 mesenchymal cells demonstrated significantly faster tumor growth in vivo compared with P2 epithelial counterparts, with invasion of intestine, pancreas, spleen, and lymph nodes. Furthermore, P2 mesenchymal cells secreted high levels of hepatocyte growth factor (HGF), which we propose acts in a paracrine fashion to drive epithelial cells to undergo EMT. In addition, a second murine liver cancer stem cell line with methionine adenosyltransferase 1a deficiency acquired EMT after sequential transplantations, indicating that EMT was not restricted to Pten‐deleted tumors. Conclusion: EMT is associated with a high rate of liver tumor proliferation, invasion, and metastasis in vivo, which is driven by HGF secreted from mesenchymal tumor cells in a feed‐forward mechanism. (HEPATOLOGY 2010)


Hepatology | 2009

CD133+ liver cancer stem cells from methionine adenosyl transferase 1A–deficient mice demonstrate resistance to transforming growth factor (TGF)‐β–induced apoptosis

Wei Ding; Marialena Mouzaki; Hanning You; Joshua C. Laird; José M. Mato; Shelly C. Lu; C. Bart Rountree

Methionine adenosyltransferase (MAT) is an essential enzyme required for S‐adenosylmethionine biosynthesis. Hepatic MAT activity falls during chronic liver injury, and mice lacking Mat1a develop spontaneous hepatocellular carcinoma by 18 months. We have previously demonstrated that CD133+CD45− oval cells isolated from 16‐month‐old Mat1a−/− mice represent a liver cancer stem cell population. The transforming growth factor β (TGF‐β) pathway constitutes a central signaling network in proliferation, apoptosis, and tumorigenesis. In this study, we tested the response of tumorigenic liver stem cells to TGF‐β. CD133+CD45− oval cells were isolated from premalignant 16‐month‐old Mat1a−/− mice by flow cytometry and expanded as five clone lines derived from a single cell. All clone lines demonstrated expression of both hepatocyte and cholangiocyte markers and maintained a small population (0.5% to 2%) of CD133+ cells in vitro, and three of five clone lines produced tumors. Although TGF‐β1 inhibited cell growth equally in CD133− and CD133+ cells from each clone line, the CD133+ population demonstrated significant resistance to TGF‐β–induced apoptosis compared with CD133− cells. Furthermore, CD133+ cells demonstrated a substantial increase in mitogen‐activated protein kinase (MAPK) pathway activation, as demonstrated by phosphorylated extracellular signal‐regulated kinase levels before and after TGF‐β stimulation. MAPK inhibition using mitogen‐activated protein kinase kinase 1 (MEK1) inhibitor PD98059 led to a significant increase in TGF‐β–induced apoptosis in CD133+ cells. Conversely, a constitutively active form of MEK1 blocked the apoptotic effects of TGF‐β in CD133− cells. Conclusion: CD133+ liver cancer stem cells exhibit relative resistance to TGF‐β–induced apoptosis. One mechanism of resistance to TGF‐β–induced apoptosis in CD133+ cancer stem cells is an activated mitogen‐activated protein kinase/extracellular signal‐regulated kinase pathway. (HEPATOLOGY 2009.)


American Journal of Physiology-renal Physiology | 2013

Macrophages directly mediate diabetic renal injury

Hanning You; Ting Gao; Timothy K. Cooper; W. Brian Reeves; Alaa S. Awad

Monocyte/macrophage recruitment correlates strongly with the progression of renal impairment in diabetic nephropathy (DN), yet their direct role is not clear. We hypothesized that macrophages contribute to direct podocyte injury and/or an abnormal podocyte niche leading to DN. Experiments were conducted in CD11b-DTR mice treated with diphtheria toxin (DT) to deplete macrophages after streptozotocin-induced diabetes. Additional experiments were conducted in bone marrow chimeric (CD11b-DTR→ C57BL6/J) mice. Diabetes was associated with an increase in the M1-to-M2 ratio by 6 wk after the induction of diabetes. Macrophage depletion in diabetic CD11b-DTR mice significantly attenuated albuminuria, kidney macrophage recruitment, and glomerular histological changes and preserved kidney nephrin and podocin expression compared with diabetic CD11b-DTR mice treated with mutant DT. These data were confirmed in chimeric mice indicating a direct role of bone marrow-derived macrophages in DN. In vitro, podocytes grown in high-glucose media significantly increased macrophage migration compared with podocytes grown in normal glucose media. In addition, classically activated M1 macrophages, but not M2 macrophages, induced podocyte permeability. These findings provide evidence showing that macrophages directly contribute to kidney injury in DN, perhaps by altering podocyte integrity through the proinflammatory M1 subset of macrophages. Attenuating the deleterious effects of macrophages on podocytes could provide a new therapeutic approach to the treatment of DN.


Kidney International | 2015

Macrophage-derived tumor necrosis factor-α mediates diabetic renal injury.

Alaa S. Awad; Hanning You; Ting Gao; Timothy K. Cooper; Sergei A. Nedospasov; Jean Vacher; Patrick Wilkinson; Francis X. Farrell; W. Brian Reeves

Monocyte/macrophage recruitment correlates strongly with the progression of diabetic nephropathy. Tumor necrosis factor-alpha (TNF-α) is produced by monocytes/macrophages but the direct role of TNF-α and/or macrophage-derived TNF-α in the progression of diabetic nephropathy remains unclear. Here we tested whether inhibition of TNF-α confers kidney protection in diabetic nephropathy via a macrophage-derived TNF-α dependent pathway. Compared to vehicle-treated mice, blockade of TNF-α with a murine anti-TNF-α antibody conferred kidney protection in Ins2Akita mice as indicated by reductions in albuminuria, plasma creatinine, histopathologic changes, kidney macrophage recruitment and plasma inflammatory cytokine levels at 18 weeks of age. To assess the direct role of macrophage-derived TNF-α in diabetic nephropathy, we generated macrophage specific TNF-α deficient mice (CD11bCre/TNF-αFlox/Flox). Conditional ablation of TNF-α in macrophages significantly reduced albuminuria, the increase in plasma creatinine and BUN, histopathologic changes and kidney macrophage recruitment compared to diabetic TNF-αFlox/Flox control mice after 12 weeks of streptozotocin-induced diabetes. Thus, production of TNF-α by macrophages plays a major role in diabetic renal injury. Hence, blocking TNF-α could be a novel therapeutic approach for treatment of diabetic nephropathy.


Proteome Science | 2010

Clinical application for the preservation of phospho- proteins through in-situ tissue stabilization

C. Bart Rountree; Colleen A. Van Kirk; Hanning You; Wei Ding; Hien Dang; Heather D. VanGuilder; Willard M. Freeman

BackgroundProtein biomarkers will play a pivotal role in the future of personalized medicine for both diagnosis and treatment decision-making. While the results of several pre-clinical and small-scale clinical studies have demonstrated the value of protein biomarkers, there have been significant challenges to translating these findings into routine clinical care. Challenges to the use of protein biomarkers include inter-sample variability introduced by differences in post-collection handling and ex vivo degradation of proteins and protein modifications.ResultsIn this report, we re-create laboratory and clinical scenarios for sample collection and test the utility of a new tissue stabilization technique in preserving proteins and protein modifications. In the laboratory setting, tissue stabilization with the Denator Stabilizor T1 resulted in a significantly higher yield of phospho-protein when compared to standard snap freeze preservation. Furthermore, in a clinical scenario, tissue stabilization at collection resulted in a higher yield of total phospho-protein, total phospho-tyrosine, pErkT202/Y204 and pAktS473 when compared to standard methods. Tissue stabilization did not have a significant effect on other post-translational modifications such as acetylation and glycosylation, which are more stable ex-vivo. Tissue stabilization did decrease total RNA quantity and quality.ConclusionStabilization at the time of collection offers the potential to better preserve tissue protein and protein modification levels, as well as reduce the variability related to tissue processing delays that are often associated with clinical samples.


Oncogenesis | 2012

miR-200b restoration and DNA methyltransferase inhibitor block lung metastasis of mesenchymal-phenotype hepatocellular carcinoma

Wen-Xing Ding; Hien Dang; Hanning You; Steven Nathaniel Steinway; Yoshinori Takahashi; Hong-Gang Wang; Jason Liao; Stiles B; Albert R; Carl B. Rountree

Epithelial-to-mesenchymal transition (EMT) is associated with poor prognosis and metastasis in hepatocellular carcinoma. We have previously demonstrated an in vivo model of liver cancer in which mesenchymal cells post-EMT demonstrate a high rate of invasive growth and metastasis. Here, we investigate the role of microRNA 200 (miR-200) family members and epigenetic modifications on the maintenance of mesenchymal/metastatic phenotype after EMT. Mesenchymal cells post-EMT demonstrates high levels of E-box repressors Zeb1 and Zeb2 and downregulation of four miR-200 family members (miR-200a, miR-200b, miR-200c and miR-429). In addition, DNA sequencing after bisulfite modification demonstrates that several CpG sites within the E-cadherin promoter are methylated in mesenchymal cells. In mesenchymal cells, forced expression of miR-200b results in a significant increase in E-cadherin and a reduction in cell migration/invasion. Despite these mesenchymal-to-epithelial transition (MET) changes in vitro, there is no significant change in metastatic potential after miR-200b upregulation in vivo. After the mesenchymal cells were treated with combination of DNA methyltransferase (DNMT) inhibitor and upregulation of miR-200b, invasive phenotype was significantly reduced and metastatic potential was eliminated. Direct targeting of E-cadherin with short hairpin RNA does not restore metastatic potential after DNMT inhibition and miR-200b re-expression. In addition, restoration of E-cadherin alone was unable to block metastatic potential in primary mesenchymal cells. In conclusion, targeting mesenchymal liver cancer cells with miR-200b and DNMT inhibitor reduces metastatic potential irrespective of E-cadherin expression. Thus, the broader differentiation and MET effects of DNMT inhibition and miR-200b must be considered in terms of rescuing metastatic potential.


American Journal of Physiology-renal Physiology | 2014

Diabetic nephropathy is resistant to oral l-arginine or l-citrulline supplementation

Hanning You; Ting Gao; Timothy K. Cooper; Sidney M. Morris; Alaa S. Awad

Our recent publication showed that pharmacological blockade of arginases confers kidney protection in diabetic nephropathy via a nitric oxide (NO) synthase (NOS)3-dependent mechanism. Arginase competes with endothelial NOS (eNOS) for the common substrate L-arginine. Lack of L-arginine results in reduced NO production and eNOS uncoupling, which lead to endothelial dysfunction. Therefore, we hypothesized that L-arginine or L-citrulline supplementation would ameliorate diabetic nephropathy. DBA mice injected with multiple low doses of vehicle or streptozotocin (50 mg/kg ip for 5 days) were provided drinking water with or without L-arginine (1.5%, 6.05 g·kg(-1)·day(-1)) or L-citrulline (1.66%, 5.73 g·kg(-1)·day(-1)) for 9 wk. Nonsupplemented diabetic mice showed significant increases in albuminuria, blood urea nitrogen, glomerular histopathological changes, kidney macrophage recruitment, kidney TNF-α and fibronectin mRNA expression, kidney arginase activity, kidney arginase-2 protein expression, and urinary oxidative stress along with a significant reduction of nephrin and eNOS protein expression and kidney nitrite + nitrate compared with normal mice after 9 wk of diabetes. Surprisingly, L-arginine or L-citrulline supplementation in diabetic mice did not affect any of these parameters despite greatly increasing kidney and plasma arginine levels. These findings demonstrate that chronic L-arginine or L-citrulline supplementation does not prevent or reduce renal injury in a model of type 1 diabetes.


American Journal of Physiology-renal Physiology | 2013

Protective role of small pigment epithelium-derived factor (PEDF) peptide in diabetic renal injury

Alaa S. Awad; Ting Gao; Anzor Gvritishvili; Hanning You; Yanling Liu; Timothy K. Cooper; W. Brian Reeves; Joyce Tombran-Tink

Pigment epithelium-derived factor (PEDF) is a multifunctional protein with antiangiogenic, antioxidative, and anti-inflammatory properties. PEDF is involved in the pathogenesis of diabetic retinopathy, but its direct role in the kidneys remains unclear. We hypothesize that a PEDF fragment (P78-PEDF) confers kidney protection in diabetic nephropathy (DN). The localization of the full-length PEDF protein were determined in DBA mice following multiple low doses of streptozotocin. Using immunohistochemistry, PEDF was localized in the kidney vasculature, interstitial space, glomeruli, tubules, and renal medulla. Kidney PEDF protein and mRNA expression were significantly reduced in diabetic mice. Continuous infusion of P78-PEDF for 6 wk resulted in protection from diabetic neuropathy as indicated by reduced albuminuria and blood urea nitrogen, increased nephrin expression, decreased kidney macrophage recruitment and inflammatory cytokines, and reduced histological changes compared with vehicle-treated diabetic mice. In vitro, P78-PEDF blocked the increase in podocyte permeability to albumin and disruption of the actin cytoskeleton induced by puromycin aminonucleoside treatment. These findings highlight the importance of P78-PEDF peptide as a potential therapeutic modality in early phase diabetic renal injury.

Collaboration


Dive into the Hanning You's collaboration.

Top Co-Authors

Avatar

Alaa S. Awad

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Timothy K. Cooper

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Ting Gao

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Wei Ding

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Hien Dang

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

C. Bart Rountree

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

W. Brian Reeves

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Carl B. Rountree

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge