Network


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

Hotspot


Dive into the research topics where Yelin Wu is active.

Publication


Featured researches published by Yelin Wu.


Immunity | 2012

The antimicrobial protein REG3A regulates keratinocyte proliferation and differentiation after skin injury.

Yuping Lai; Dongqing Li; Changwei Li; Beda Muehleisen; Katherine A. Radek; Hyun Jeong Park; Ziwei Jiang; Zhiheng Li; Hu Lei; Yanchun Quan; Tian Zhang; Yelin Wu; Paul Kotol; Shin Morizane; Tissa Hata; Keiji Iwatsuki; Ce Tang; Richard L. Gallo

Epithelial keratinocyte proliferation is an essential element of wound repair, and abnormal epithelial proliferation is an intrinsic element in the skin disorder psoriasis. The factors that trigger epithelial proliferation in these inflammatory processes are incompletely understood. Here we have shown that regenerating islet-derived protein 3-alpha (REG3A) is highly expressed in keratinocytes during psoriasis and wound repair and in imiquimod-induced psoriatic skin lesions. The expression of REG3A by keratinocytes is induced by interleukin-17 (IL-17) via activation of keratinocyte-encoded IL-17 receptor A (IL-17RA) and feeds back on keratinocytes to inhibit terminal differentiation and increase cell proliferation by binding to exostosin-like 3 (EXTL3) followed by activation of phosphatidylinositol 3 kinase (PI3K) and the kinase AKT. These findings reveal that REG3A, a secreted intestinal antimicrobial protein, can promote skin keratinocyte proliferation and can be induced by IL-17. This observation suggests that REG3A may mediate the epidermal hyperproliferation observed in normal wound repair and in psoriasis.


Cancer Research | 2011

Steroid Receptor Coactivator-1 Upregulates Integrin α5 Expression to Promote Breast Cancer Cell Adhesion and Migration

Li Qin; Xian Chen; Yelin Wu; Zhen Feng; Tao He; Li Wang; Lan Liao; Jianming Xu

Metastatic breast cancer remains a lethal disease with poorly understood molecular mechanisms. Steroid receptor coactivator-1 (SRC-1 or NCOA1) is overexpressed in a subset of breast cancers with poor prognosis. It potentiates gene expression by serving as a coactivator for nuclear receptors and other transcription factors. We previously reported that SRC-1 promotes breast cancer metastasis without affecting primary mammary tumor formation. Herein, we found that SRC-1 deficiency in mouse and human breast cancer cells substantially reduced cell adhesion and migration capabilities on fibronectin and significantly extended the time of focal adhesion disassembly and reassembly. In agreement with this phenotype, SRC-1 expression positively correlated with integrin α(5) (ITGA5) expression in estrogen receptor-negative breast tumors whereas SRC-1 deficiency decreased ITGA5 expression. Furthermore, ITGA5 reduction in SRC-1-deficient/insufficient breast cancer cells or knockdown of ITGA5 in SRC-1-expressing breast cancer cells was associated with a disturbed integrin-mediated signaling. Critical downstream changes included reduced phosphorylation and/or dampened activation of focal adhesion kinase, paxillin, Rac1, and Erk1/2 during cell adhesion. Finally, we found that SRC-1 enhanced ITGA5 promoter activity through an AP-1 (activator protein)-binding site proximal to the transcriptional initiation site; both SRC-1 and c-Jun were recruited to this promoter region in breast cancer cells. These results show that SRC-1 can promote breast cancer metastasis by directly enhancing ITGA5 expression and thus promoting ITGA5-mediated cell adhesion and migration. Therefore, targeting ITGA5 in SRC-1-positive breast cancers may result in inhibition of SRC-1-promoted breast cancer metastasis.


PLOS Pathogens | 2014

Interleukin-33 Increases Antibacterial Defense by Activation of Inducible Nitric Oxide Synthase in Skin

Changwei Li; Hongquan Li; Ziwei Jiang; Tian Zhang; Yue Wang; Zhiheng Li; Yelin Wu; Shizhao Ji; Shichu Xiao; Bernhard Ryffel; Katherine A. Radek; Zhaofan Xia; Yuping Lai

Interleukin-33 (IL-33) is associated with multiple diseases, including asthma, rheumatoid arthritis, tissue injuries and infections. Although IL-33 has been indicated to be involved in Staphylococcus aureus (S. aureus) wound infection, little is known about how IL-33 is regulated as a mechanism to increase host defense against skin bacterial infections. To explore the underlying intricate mechanism we first evaluated the expression of IL-33 in skin from S. aureus-infected human patients. Compared to normal controls, IL-33 was abundantly increased in skin of S. aureus-infected patients. We next developed a S. aureus cutaneous infection mouse model and found that IL-33 was significantly increased in dermal macrophages of infected mouse skin. The expression of IL-33 by macrophages was induced by staphylococcal peptidoglycan (PGN) and lipoteichoic acid (LTA) via activation of toll-like receptor 2(TLR2) –mitogen-activated protein kinase (MAPK)-AKT-signal transducer and activator of transcription 3(STAT3) signaling pathway as PGN and LTA failed to induce IL-33 in Tlr2-deficient peritoneal macrophages, and MAPK,AKT, STAT3 inhibitors significantly decreased PGN- or LTA-induced IL-33. IL-33, in turn, acted on macrophages to induce microbicidal nitric oxygen (NO) release. This induction was dependent on inducible nitric oxide synthase (iNOS) activation, as treatment of macrophages with an inhibitor of iNOS, aminoguanidine, significantly decreased IL-33-induced NO release. Moreover, aminoguanidine significantly blocked the capacity of IL-33 to inhibit the growth of S. aureus, and IL-33 silencing in macrophages significantly increased the survival of S. aureus in macrophages. Furthermore, the administration of IL-33-neutralizing antibody into mouse skin decreased iNOS production but increased the survival of S. aureus in skin. These findings reveal that IL-33 can promote antimicrobial capacity of dermal macrophages, thus enhancing antimicrobial defense against skin bacterial infections.


Cancer Research | 2013

The Steroid Receptor Coactivator-3 Is Required for the Development of Castration-Resistant Prostate Cancer

Jean Ching-Yi Tien; Zhaoliang Liu; Lan Liao; Fen Wang; Yixiang Xu; Yelin Wu; Niya Zhou; Michael Ittmann; Jianming Xu

The transcriptional coactivator SRC-3 plays a key role in enhancing prostate cancer cell proliferation. Although SRC-3 is highly expressed in advanced prostate cancer, its role in castration-resistant prostate cancer (CRPC) driven by PTEN mutation is unknown. We documented elevated SRC-3 in human CRPC and in PTEN-negative human prostate cancer. Patients with high SRC-3 and undetectable PTEN exhibited decreased recurrence-free survival. To explore the causal relationship in these observations, we generated mice in which both Pten and SRC-3 were inactivated in prostate epithelial cells (Pten3CKO mice), comparing them with mice in which only Pten was inactivated in these cells (PtenCKO mice). SRC-3 deletion impaired cellular proliferation and reduced tumor size. Notably, while castration of PtenCKO control mice increased the aggressiveness of prostate tumors relative to noncastrated counterparts, deletion of SRC-3 in Pten3CKO mice reversed all these changes. In support of this finding, castrated Pten3CKO mice also exhibited decreased levels of phospho-Akt, S6 kinase (RPS6KB1), and phosphorylated S6 protein (RPS6), all of which mediate cell growth and proliferation. Moreover, these tumors appeared to be more differentiated as evidenced by higher levels of Fkbp5, an AR-responsive gene that inhibits Akt signaling. Lastly, these tumors also displayed lower levels of certain androgen-repressed genes such as cyclin E2 and MMP10. Together, our results show that SRC-3 drives CRPC formation and offer preclinical proof of concept for a transcriptional coactivator as a therapeutic target to abrogate CRPC progression.


Cancer Research | 2014

NCOA1 Directly Targets M-CSF1 Expression to Promote Breast Cancer Metastasis

Li Qin; Yelin Wu; Michael J. Toneff; Dabing Li; Lan Liao; Xiuhua Gao; Fiona Bane; Jean C.-Y. Tien; Yixiang Xu; Zhen Feng; Zhihui Yang; Yan Xu; Sarah M. Theissen; Yi Li; Leonie Young; Jianming Xu

In breast cancer, overexpression of the nuclear coactivator NCOA1 (SRC-1) is associated with disease recurrence and resistance to endocrine therapy. To examine the impact of NCOA1 overexpression on morphogenesis and carcinogenesis in the mammary gland (MG), we generated MMTV-hNCOA1 transgenic [Tg(NCOA1)] mice. In the context of two distinct transgenic models of breast cancer, NCOA1 overexpression did not affect the morphology or tumor-forming capability of MG epithelial cells. However, NCOA1 overexpression increased the number of circulating breast cancer cells and the efficiency of lung metastasis. Mechanistic investigations showed that NCOA1 and c-Fos were recruited to a functional AP-1 site in the macrophage attractant CSF1 promoter, directly upregulating colony-simulating factor 1 (CSF1) expression to enhance macrophage recruitment and metastasis. Conversely, silencing NCOA1 reduced CSF1 expression and decreased macrophage recruitment and breast cancer cell metastasis. In a cohort of 453 human breast tumors, NCOA1 and CSF1 levels correlated positively with disease recurrence, higher tumor grade, and poor prognosis. Together, our results define an NCOA1/AP-1/CSF1 regulatory axis that promotes breast cancer metastasis, offering a novel therapeutic target for impeding this process.


Regulatory Peptides | 2010

Addition of a cysteine to glucagon-like peptide-1 (GLP-1) conjugates GLP-1 to albumin in serum and prolongs GLP-1 action in vivo

Yelin Wu; Jing Huang; Jin Xu; Jian Liu; Zhen Feng; Yang Wang; Yuping Lai; Zirong Wu

Glucagon-like peptide-1 (GLP-1) is a promising new therapeutic agent for the treatment of type 2 diabetes. However, GLP-1 has a short half-life (t(1/)(2)<2min) due to rapid degradation by dipeptidyl peptidase IV in vivo. To circumvent this problem, a recombinant mGLP-1 with a cysteine at the C-terminus of GLP-1 was expressed in Escherichia coli and purified by affinity and reverse-phase chromatography. This addition of a cysteine facilitates mGLP-1 binding to serum albumin both in vitro and in vivo, thus protecting mGLP-1 from protease degradation. Similar to GLP-1, mGLP-1 stimulated cAMP production in PC12 cells and exhibited insulinotropic activity in MIN6 cells under in vitro culture conditions. Importantly, in glucose tolerance tests mice treated with mGLP-1 exhibited much lower glucose levels and much higher insulin levels versus that in mice treated with unmodified GLP-1. Furthermore, the effects of mGLP-1 on reduction of blood glucose levels lasted for 6-7h, while the effects of unmodified GLP-1 only lasted for 0.5-1h after injection. These results demonstrate that mGLP-1 is biologically active and its pharmaceutical efficacy is largely enhanced by the cysteine-mediated covalent conjugation with albumin in the serum after injection. Therefore, the mGLP-1 with a cysteine may be a better potential therapeutic drug than the unmodified GLP-1 for treating type 2 diabetes.


Nature Communications | 2016

Hyperglycaemia inhibits REG3A expression to exacerbate TLR3-mediated skin inflammation in diabetes.

Yelin Wu; Yanchun Quan; Yuanqi Liu; Keiwei Liu; Hongquan Li; Ziwei Jiang; Tian Zhang; Hu Lei; Katherine A. Radek; Dongqing Li; Zhenhua Wang; Jilong Lu; Wang Wang; Shizhao Ji; Zhaofan Xia; Yuping Lai

Dysregulated inflammatory responses are known to impair wound healing in diabetes, but the underlying mechanisms are poorly understood. Here we show that the antimicrobial protein REG3A controls TLR3-mediated inflammation after skin injury. This control is mediated by REG3A-induced SHP-1 protein, and acts selectively on TLR3-activated JNK2. In diabetic mouse skin, hyperglycaemia inhibits the expression of IL-17-induced IL-33 via glucose glycation. The decrease in cutaneous IL-33 reduces REG3A expression in epidermal keratinocytes. The reduction in REG3A is associated with lower levels of SHP-1, which normally inhibits TLR3-induced JNK2 phosphorylation, thereby increasing inflammation in skin wounds. To our knowledge, these findings show for the first time that REG3A can modulate specific cutaneous inflammatory responses and that the decrease in cutaneous REG3A exacerbates inflammation in diabetic skin wounds.


Journal of Peptide Science | 2011

Protective effect of recombinant human glucagon-like peptide-1 (rhGLP-1) pretreatment in STZ-induced diabetic mice.

Yelin Wu; Jing Huang; Jian Liu; Mingfei Jin; Mei Gu; Yiguo Hong; Zirong Wu

Human glucagon‐like peptide‐1 (hGLP‐1) and its mimetics have emerged as therapies for type 2 diabetes. However, clinical treatment of diabetes with hGLP‐1 is ineffective because of rapid DPPIV‐mediated hGLP‐1 degradation in the circulation. In this study, we investigated the protective effect of recombinant human glucagon‐like peptide‐1 (rhGLP‐1) treatment on STZ‐induced diabetic mice. Mice were treated daily with rhGLP‐1 (24 nmol/kg body weight) starting before or after STZ injection (40 mg/kg body weight) to induce diabetes. Mice pretreated with rhGLP‐1 before but not after STZ showed significantly reduced blood glucose levels (P < 0.05), increased oral glucose tolerance (area under the curve, 1740 ± 71.18 vs 2416 ± 205.6, P < 0.05). Furthermore, the bioproduct of lipid peroxidation, MDA, was reduced and SOD and GSH‐PX activities were enhanced globally and in pancreas of mice that received rhGLP‐1 pretreatment before STZ, when comparing with STZ‐treated mice. Finally, STZ‐induced pancreatic islet damage was rescued by rhGLP‐1 pretreatment. Taken together, the results of this study demonstrate that rhGLP‐1 pretreatment has a protective effect against STZ‐induced diabetes in mice. These findings suggest that the GLP‐1 pretreatment may be a new therapeutic strategy in the preventive and protective treatment during diabetes initiation and progression. Copyright


Immunity | 2018

An Interleukin-25-Mediated Autoregulatory Circuit in Keratinocytes Plays a Pivotal Role in Psoriatic Skin Inflammation

Miao Xu; Huiping Lu; Young Hee Lee; Yelin Wu; Kewei Liu; Yuling Shi; Haoran An; Jingren Zhang; Xiaohu Wang; Yuping Lai; Chen Dong

SUMMARY Psoriasis is a chronic autoinflammatory skin disease. Although interleukin‐17, derived from lymphocytes, has been shown to be critical in psoriasis, the initiation and maintenance of chronic skin inflammation has not been well understood. IL‐25 (also called IL‐17E), another IL‐17 family cytokine, is well known to regulate allergic responses and type 2 immunity. Here we have shown that IL‐25, also highly expressed in the lesional skin of psoriasis patients, was regulated by IL‐17 in murine skin of a imiquimod (IMQ)‐induced psoriasis model. IL‐25 injection induced skin inflammation, whereas germline or keratinocyte‐specific deletion of IL‐25 caused resistance to IMQ‐induced psoriasis. Via IL‐17RB expression in keratinocytes, IL‐25 stimulated the proliferation of keratinocytes and induced the production of inflammatory cytokines and chemokines, via activation of the STAT3 transcription factor. Thus, our data demonstrate that an IL‐17‐induced autoregulatory circuit in keratinocytes is mediated by IL‐25 and suggest that this circuit could be targeted in the treatment of psoriasis patients. Graphical Abstract Figure. No caption available. HighlightsIL‐25 is highly expressed in psoriasisIL‐25 treatment induces psoriasis‐like skin inflammationIL‐25 in keratinocytes is necessary for psoriasis‐like skin inflammationIL‐25 promotes keratinocyte proliferation and pro‐inflammatory response In Brief The inflammatory mechanism of psoriasis remains incompletely understood. In this issue, Xu et al. identified IL‐25 as a key pathogenic factor regulating the proliferation of keratinocytes and psoriasis development in an autocrine expression manner.


Science China-life Sciences | 2017

TLR3 activation induces S100A7 to regulate keratinocyte differentiation after skin injury.

Hu Lei; Yue Wang; Tian Zhang; Leilei Chang; Yelin Wu; Yuping Lai

Human S100A7 (psoriasin) is highly expressed in psoriasis and other inflammatory diseases; however, the function of S100A7 in wound repair remains largely unknown. Here we demonstrated that skin injury increased the expression of S100A7. Damaged cells from wounded skin induced the expression of S100A7 via the activation of Toll-like receptor 3 (TLR3) followed by the activation of p38 MAPK. S100A7, in turn, acted on keratinocytes to induce the expression of terminal differentiation marker gene loricrin through the activation of p38 MAPK and caspase-1. The differentiation of keratinocytes induced by S100A7 resulted in skin stratification, thus efficiently promoting wound closure. Taken together, our results demonstrate that the activation of TLR3 accelerates wound closure via the induction of S100A7 to induce keratinocyte differentiation. These findings also provide new insights into the development of different forms of treatment with skin wounds.

Collaboration


Dive into the Yelin Wu's collaboration.

Top Co-Authors

Avatar

Yuping Lai

East China Normal University

View shared research outputs
Top Co-Authors

Avatar

Jianming Xu

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lan Liao

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ziwei Jiang

East China Normal University

View shared research outputs
Top Co-Authors

Avatar

Li Qin

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Zhen Feng

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tian Zhang

East China Normal University

View shared research outputs
Top Co-Authors

Avatar

Yixiang Xu

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hongquan Li

East China Normal University

View shared research outputs
Top Co-Authors

Avatar

Hu Lei

East China Normal University

View shared research outputs
Researchain Logo
Decentralizing Knowledge