Network


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

Hotspot


Dive into the research topics where Yiming Xing is active.

Publication


Featured researches published by Yiming Xing.


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.


BMC Molecular Biology | 2008

Ror2 modulates the canonical Wnt signaling in lung epithelial cells through cooperation with Fzd2

Changgong Li; Hongyan Chen; Lingyan Hu; Yiming Xing; Tomoyo Sasaki; Maria F Villosis; John T. Li; Michiru Nishita; Yasuhiro Minami; Parviz Minoo

BackgroundWnt signaling is mediated through 1) the beta-catenin dependent canonical pathway and, 2) the beta-catenin independent pathways. Multiple receptors, including Fzds, Lrps, Ror2 and Ryk, are involved in Wnt signaling. Ror2 is a single-span transmembrane receptor-tyrosine kinase (RTK). The functions of Ror2 in mediating the non-canonical Wnt signaling have been well established. The role of Ror2 in canonical Wnt signaling is not fully understood.ResultsHere we report that Ror2 also positively modulates Wnt3a-activated canonical signaling in a lung carcinoma, H441 cell line. This activity of Ror2 is dependent on cooperative interactions with Fzd2 but not Fzd7. In addition, Ror2-mediated enhancement of canonical signaling requires the extracellular CRD, but not the intracellular PRD domain of Ror2. We further provide evidence that the positive effect of Ror2 on canonical Wnt signaling is inhibited by Dkk1 and Krm1 suggesting that Ror2 enhances an Lrp-dependent STF response.ConclusionThe current study demonstrates the function of Ror2 in modulating canonical Wnt signaling. These findings support a functional scheme whereby regulation of Wnt signaling is achieved by cooperative functions of multiple mediators.


Molecular and Cellular Biology | 2007

Physical and Functional Interactions between Homeodomain NKX2.1 and Winged Helix/Forkhead FOXA1 in Lung Epithelial Cells

Parviz Minoo; Lingyan Hu; Yiming Xing; Nian Ling Zhu; Hongyan Chen; Min Li; Zea Borok; Changgong Li

ABSTRACT NKX2.1 is a homeodomain transcription factor that controls development of the brain, lung, and thyroid. In the lung, Nkx2.1 is expressed in a proximo-distal gradient and activates specific genes in phenotypically distinct epithelial cells located along this axis. The mechanisms by which NKX2.1 controls its target genes may involve interactions with other transcription factors. We examined whether NKX2.1 interacts with members of the winged-helix/forkhead family of FOXA transcription factors to regulate two spatially and cell type-specific genes, SpC and Ccsp. The results show that NKX2.1 interacts physically and functionally with FOXA1. The nature of the interaction is inhibitory and occurs through the NKX2.1 homeodomain in a DNA-independent manner. On SpC, which lacks a FOXA1 binding site, FOXA1 attenuates NKX2.1-dependent transcription. Inhibition of FOXA1 by small interfering RNA increased SpC mRNA, demonstrating the in vivo relevance of this finding. In contrast, FOXA1 and NKX2.1 additively activate transcription from Ccsp, which includes both NKX2.1 and FOXA1 binding sites. In electrophoretic mobility shift assays, the GST-FOXA1 fusion protein interferes with the formation of NKX2.1 transcriptional complexes by potentially masking the latters homeodomain DNA binding function. These findings suggest a novel mode of selective gene regulation by proximo-distal gradient distribution of and functional interactions between forkhead and homeodomain transcription factors.


Stem Cells | 2015

Progenitors of Secondary Crest Myofibroblasts Are Developmentally Committed in Early Lung Mesoderm

Changgong Li; Min Li; Sha Li; Yiming Xing; Chang–Yo Yang; Aimin Li; Zea Borok; Stijn De Langhe; Parviz Minoo

Development of the mammalian lung is predicated on cross‐communications between two highly interactive tissues, the endodermally derived epithelium and the mesodermally derived pulmonary mesenchyme. While much attention has been paid for the lung epithelium, the pulmonary mesenchyme, partly due to lack of specific tractable markers remains under‐investigated. The lung mesenchyme is derived from the lateral plate mesoderm and is the principal recipient of Hedgehog (Hh) signaling, a morphogenetic network that regulates multiple aspects of embryonic development. Using the Hh‐responsive Gli1‐creERT2 mouse line, we identified the mesodermal targets of Hh signaling at various time points during embryonic and postnatal lung development. Cell lineage analysis showed these cells serve as progenitors to contribute to multiple lineages of mesodermally derived differentiated cell types that include parenchymal or interstitial myofibroblasts, peribronchial and perivascular smooth muscle as well as rare populations of cells within the mesothelium. Most importantly, Gli1‐creERT2 identified the progenitors of secondary crest myofibroblasts, a hitherto intractable cell type that plays a key role in alveolar formation, a vital process about which little is currently known. Transcriptome analysis of Hh‐targeted progenitor cells transitioning from the pseudoglandular to the saccular phase of lung development revealed important modulations of key signaling pathways. Among these, there was significant downregulation of canonical WNT signaling. Ectopic stabilization of β‐catenin via inactivation of Apc by Gli1‐creERT2 expanded the Hh‐targeted progenitor pools, which caused the formation of fibroblastic masses within the lung parenchyma. The Gli1‐creERT2 mouse line represents a novel tool in the analysis of mesenchymal cell biology and alveolar formation during lung development. Stem Cells 2015;33:999–1012


Stem Cells | 2012

NOTCH1 is Required for Regeneration of Clara Cells During Repair of Airway Injury

Yiming Xing; Aimin Li; Zea Borok; Changgong Li; Parviz Minoo

The airways of the mammalian lung are lined with highly specialized epithelial cell types that are the targets of airborne toxicants and injury. Notch signaling plays an important role in the ontogeny of airway epithelial cells, but its contributions to recruitment, expansion or differentiation of resident progenitor/stem cells, and repair and re‐establishment of the normal composition of airway epithelium following injury have not been addressed. In this study, the role of a specific Notch receptor, Notch1, was investigated by targeted inactivation in the embryonic lung epithelium using the epithelial‐specific Gata5‐Cre driver line. Notch1‐deficient mice are viable without discernible defects in pulmonary epithelial cell‐fate determination and differentiation. However, in an experimental model of airway injury, activity of Notch1 is found to be required for normal repair of the airway epithelium. Absence of Notch1 reduced the ability of a population of cells distinguished by expression of PGP9.5, otherwise a marker of pulmonary neuroendocrine cells, which appears to serve as a reservoir for regeneration of Clara cells. Hairy/enhancer of split‐5 (Hes5) and paired‐box‐containing gene 6 (Pax6) were found to be downstream targets of Notch1. Both Hes5 and Pax6 expressions were significantly increased in association with Clara cell regeneration in wild‐type lungs. Ablation of Notch1 reduced Hes5 and Pax6 and inhibited airway epithelial repair. Thus, although dispensable in developmental ontogeny of airway epithelial cells, normal activity of Notch1 is required for repair of the airway epithelium. The signaling pathway by which Notch1 regulates the repair process includes stimulation of Hes5 and Pax6 gene expression. STEM CELLS 2012;30:946–955


American Journal of Respiratory and Critical Care Medicine | 2009

Deletion of Pten expands lung epithelial progenitor pools and confers resistance to airway injury.

Caterina Tiozzo; Stijn De Langhe; Mingke Yu; Vedang A. Londhe; Gianni Carraro; Min Li; Changgong Li; Yiming Xing; Stewart A. Anderson; Zea Borok; Saverio Bellusci; Parviz Minoo

RATIONALE Pten is a tumor-suppressor gene involved in stem cell homeostasis and tumorigenesis. In mouse, Pten expression is ubiquitous and begins as early as 7 days of gestation. Pten(-/-) mouse embryos die early during gestation indicating a critical role for Pten in embryonic development. OBJECTIVES To test the role of Pten in lung development and injury. METHODS We conditionally deleted Pten throughout the lung epithelium by crossing Pten(flox/flox) with Nkx2.1-cre driver mice. The resulting Pten(Nkx2.1-cre) mutants were analyzed for lung defects and response to injury. MEASUREMENTS AND MAIN RESULTS Pten(Nkx2.1-cre) embryonic lungs showed airway epithelial hyperplasia with no branching abnormalities. In adult mice, Pten(Nkx2.1-cre) lungs exhibit increased progenitor cell pools composed of basal cells in the trachea, CGRP/CC10 double-positive neuroendocrine cells in the bronchi, and CC10/SPC double-positive cells at the bronchioalveolar duct junctions. Pten deletion affected differentiation of various lung epithelial cell lineages, with a decreased number of terminally differentiated cells. Over time, Pten(Nxk2.1-cre) epithelial cells residing in the bronchioalveolar duct junctions underwent proliferation and formed uniform masses, supporting the concept that the cells residing in this distal niche may also be the source of procarcinogenic stem cells. Finally, increased progenitor cells in all the lung compartments conferred an overall selective advantage to naphthalene injury compared with wild-type control mice. CONCLUSIONS Pten has a pivotal role in lung stem cell homeostasis, cell differentiation, and consequently resistance to lung injury.


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


Journal of Biological Chemistry | 2008

Mesodermal Deletion of Transforming Growth Factor-β Receptor II Disrupts Lung Epithelial Morphogenesis: CROSS-TALK BETWEEN TGF-β AND SONIC HEDGEHOG PATHWAYS*

Min Li; Changgong Li; Yi-hsin Liu; Yiming Xing; Lingyan Hu; Zea Borok; Kenny Y.C. Kwong; Parviz Minoo

^{\rm{flox/KO}}


Developmental Biology | 2008

Mechanisms of TGFβ Inhibition of Lung Endodermal Morphogenesis: The role of TβRII, Smads, Nkx2.1 and Pten

Yiming Xing; Changgong Li; Lingyan Hu; Caterina Tiozzo; Min Li; Yang Chai; Saverio Bellusci; Stewart A. Anderson; Parviz Minoo

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


PLOS ONE | 2013

Tissue-Dependent Consequences of Apc Inactivation on Proliferation and Differentiation of Ciliated Cell Progenitors via Wnt and Notch Signaling

Aimin Li; Belinda Chan; Juan C. Felix; Yiming Xing; Min Li; Steven L. Brody; Zea Borok; Changgong Li; Parviz Minoo

In vertebrates, Sonic hedgehog (Shh) and transforming growth factor-beta (TGF-beta) signaling pathways occur in an overlapping manner in many morphogenetic processes. In vitro data indicate that the two pathways may interact. Whether such interactions occur during embryonic development remains unknown. Using embryonic lung morphogenesis as a model, we generated transgenic mice in which exon 2 of the TbetaRII gene, which encodes the type II TGF-beta receptor, was deleted via a mesodermal-specific Cre. Mesodermal-specific deletion of TbetaRII (TbetaRII(Delta/Delta)) resulted in embryonic lethality. The lungs showed abnormalities in both number and shape of cartilage in trachea and bronchi. In the lung parenchyma, where epithelial-mesenchymal interactions are critical for normal development, deletion of mesenchymal TbetaRII caused abnormalities in epithelial morphogenesis. Failure in normal epithelial branching morphogenesis in the TbetaRII(Delta/Delta) lungs caused cystic airway malformations. Interruption of the TbetaRII locus in the lung mesenchyme increased mRNA for Patched and Gli-1, two downstream targets of Shh signaling, without alterations in Shh ligand levels produced in the epithelium. Therefore, we conclude that TbetaRII-mediated signaling in the lung mesenchyme modulates transduction of Shh signaling that originates from the epithelium. To our knowledge, this is the first in vivo evidence for a reciprocal and novel mode of cross-communication between Shh and TGF-beta pathways during embryonic development.In vertebrates, Sonic hedgehog (Shh) and transforming growth factor-β (TGF-β) signaling pathways occur in an overlapping manner in many morphogenetic processes. In vitro data indicate that the two pathways may interact. Whether such interactions occur during embryonic development remains unknown. Using embryonic lung morphogenesis as a model, we generated transgenic mice in which exon 2 of the TβRII gene, which encodes the type II TGF-β receptor, was deleted via a mesodermal-specific Cre. Mesodermal-specific deletion of TβRII (TβRIIΔ/Δ) resulted in embryonic lethality. The lungs showed abnormalities in both number and shape of cartilage in trachea and bronchi. In the lung parenchyma, where epithelial-mesenchymal interactions are critical for normal development, deletion of mesenchymal TβRII caused abnormalities in epithelial morphogenesis. Failure in normal epithelial branching morphogenesis in the TβRIIΔ/Δ lungs caused cystic airway malformations. Interruption of the TβRII locus in the lung mesenchyme increased mRNA for Patched and Gli-1, two downstream targets of Shh signaling, without alterations in Shh ligand levels produced in the epithelium. Therefore, we conclude that TβRII-mediated signaling in the lung mesenchyme modulates transduction of Shh signaling that originates from the epithelium. To our knowledge, this is the first in vivo evidence for a reciprocal and novel mode of cross-communication between Shh and TGF-β pathways during embryonic development.

Collaboration


Dive into the Yiming Xing's collaboration.

Top Co-Authors

Avatar

Parviz Minoo

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Changgong Li

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Zea Borok

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Aimin Li

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Min Li

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Belinda Chan

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Caterina Tiozzo

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Lingyan Hu

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge