Imperial College London

DrWeiCui

Faculty of MedicineDepartment of Metabolism, Digestion and Reproduction

Senior Lecturer
 
 
 
//

Contact

 

+44 (0)20 7594 2124wei.cui Website

 
 
//

Location

 

1010Institute of Reproductive and Developmental BiologyHammersmith Campus

//

Summary

 

Publications

Publication Type
Year
to

78 results found

De Vas M, Boulet F, Joshi SS, Garstang MG, Khan TN, Atla G, Parry D, Moore D, Cebola I, Zhang S, Cui W, Lampe AK, Lam WW, Genomics England Research Consortium, Ferrer J, Madapura PM, Atanur SSet al., 2023, Regulatory de novo mutations underlying intellectual disability, Life Science Alliance, Vol: 6, Pages: 1-16, ISSN: 2575-1077

The genetic aetiology of a major fraction of patients with intellectual disability (ID) remains unknown. De novo mutations (DNMs) in protein-coding genes explain up to 40% of cases, but the potential role of regulatory DNMs is still poorly understood. We sequenced 63 whole genomes from 21 ID probands and their unaffected parents. In addition, we analysed 30 previously sequenced genomes from exome-negative ID probands. We found that regulatory DNMs were selectively enriched in fetal brain-specific enhancers as compared with adult brain enhancers. DNM-containing enhancers were associated with genes that show preferential expression in the prefrontal cortex. Furthermore, we identified recurrently mutated enhancer clusters that regulate genes involved in nervous system development (CSMD1, OLFM1, and POU3F3). Most of the DNMs from ID probands showed allele-specific enhancer activity when tested using luciferase assay. Using CRISPR-mediated mutation and editing of epigenomic marks, we show that DNMs at regulatory elements affect the expression of putative target genes. Our results, therefore, provide new evidence to indicate that DNMs in fetal brain-specific enhancers play an essential role in the aetiology of ID.

Journal article

Patel V, Cui W, Cobben JM, 2022, SHORT syndrome with microcephaly and developmental delay, AMERICAN JOURNAL OF MEDICAL GENETICS PART A, ISSN: 1552-4825

Journal article

De Vas MG, Boulet F, Joshi SS, Garstang MG, Khan TN, Atla G, Parry D, Moore D, Cebola I, Zhang S, Cui W, Lampe AK, Lam WW, Ferrer J, Pradeepa MM, Atanur SSet al., 2022, Regulatory<i>de novo</i>mutations underlying intellectual disability

<jats:title>Abstract</jats:title><jats:p>The genetic aetiology of a major fraction of patients with intellectual disability (ID) remains unknown.<jats:italic>De novo</jats:italic>mutations (DNMs) in protein-coding genes explain up to 40% of cases, but the potential role of regulatory DNMs is still poorly understood. We sequenced 63 whole genomes from 21 ID probands and their unaffected parents (trio). Additionally, we analysed 30 previously sequenced genomes from exome-negative ID probands. We found that regulatory DNMs were selectively enriched in fetal brain-specific and human-gained enhancers. DNM-containing enhancers were associated with genes that show preferential expression in the pre-frontal cortex, have been previously implicated in ID or related disorders, and exhibit intolerance to loss of function mutations. Moreover, we found that highly interacting regulatory regions from intermediate progenitor cells of the developing human cortex were strongly enriched for ID DNMs. Furthermore, we identified recurrently mutated enhancer clusters that regulate genes involved in nervous system development (<jats:italic>CSMD1, OLFM1</jats:italic>, and<jats:italic>POU3F3)</jats:italic>. The majority of the DNMs from ID probands showed allele-specific enhancer activity when tested using luciferase assay. Using CRISPR-mediated mutation and editing of epigenomic marks, we show that regulatory elements harbouring DNMs indeed function as enhancers and DNMs at regulatory elements affect the expression of putative target genes. Our results, therefore, provide new evidence to indicate that DNMs in fetal brain-specific enhancers play an essential role in the aetiology of ID.</jats:p>

Journal article

Thornton JM, Dehbi H-M, Liney T, Shah N, Cui W, Johnson M, Singh Net al., 2022, Gestational diabetes during the pandemic: screening methods and outcomes in first-time GDM, RCOG World Congress 2022, Publisher: Wiley, Pages: 114-115, ISSN: 1470-0328

Conference paper

Thornton JM, Dehbi H-M, Shah N, Cui W, Johnson M, Singh Net al., 2022, Effect of SARS-CoV-2 pandemic on patient characteristics and outcomes in gestational diabetes, RCOG World Congress 2022, Publisher: Wiley, Pages: 116-117, ISSN: 1470-0328

Conference paper

Imran SAM, Yazid MD, Cui W, Lokanathan Yet al., 2021, The intra- and extra-telomeric role of TRF2 in the DNA damage response, International Journal of Molecular Sciences, Vol: 22, ISSN: 1422-0067

Telomere repeat binding factor 2 (TRF2) has a well-known function at the telomeres, which acts to protect the telomere end from being recognized as a DNA break or from unwanted recombination. This protection mechanism prevents DNA instability from mutation and subsequent severe diseases caused by the changes in DNA, such as cancer. Since TRF2 actively inhibits the DNA damage response factors from recognizing the telomere end as a DNA break, many more studies have also shown its interactions outside of the telomeres. However, very little has been discovered on the mechanisms involved in these interactions. This review aims to discuss the known function of TRF2 and its interaction with the DNA damage response (DDR) factors at both telomeric and non-telomeric regions. In this review, we will summarize recent progress and findings on the interactions between TRF2 and DDR factors at telomeres and outside of telomeres.

Journal article

Zhang Z, He C, Gao Y, Zhang L, Song Y, Zhu T, Zhu K, Lv D, Wang J, Tian X, Ma T, Ji P, Cui W, Liu Get al., 2021, alpha-ketoglutarate delays age-related fertility decline in mammals, Aging Cell, Vol: 20, Pages: 1-15, ISSN: 1474-9718

The fecundity reduction with aging is referred as the reproductive aging which comes earlier than that of chronological aging. Since humans have postponed their childbearing age, to prolong the reproductive age becomes urgent agenda for reproductive biologists. In the current study, we examined the potential associations of α‐ketoglutarate (α‐KG) and reproductive aging in mammals including mice, swine, and humans. There is a clear tendency of reduced α‐KG level with aging in the follicle fluids of human. To explore the mechanisms, mice were selected as the convenient animal model. It is observed that a long term of α‐KG administration preserves the ovarian function, the quality and quantity of oocytes as well as the telomere maintaining system in mice. α‐KG suppresses ATP synthase and alterations of the energy metabolism trigger the nutritional sensors to down‐regulate mTOR pathway. These events not only benefit the general aging process but also maintain ovarian function and delay the reproductive decline. Considering the safety of the α‐KG as a naturally occurring molecule in energy metabolism, its utility in reproduction of large mammals including humans deserves further investigation.

Journal article

Ramlaul K, Fu W, Li H, Garrido NDM, He L, Trivedi M, Cui W, Aylett CHS, Wu Get al., 2021, Architecture of the Tuberous Sclerosis protein complex, Journal of Molecular Biology, Vol: 433, ISSN: 0022-2836

The Tuberous Sclerosis Complex (TSC) protein complex (TSCC), comprising TSC1, TSC2, and TBC1D7, is widely recognised as a key integration hub for cell growth and intracellular stress signals upstream of the mammalian target of rapamycin complex 1 (mTORC1). The TSCC negatively regulates mTORC1 by acting as a GTPase-activating protein (GAP) towards the small GTPase Rheb. Both human TSC1 and TSC2 are important tumour suppressors, and mutations in them underlie the disease tuberous sclerosis.We used single-particle cryo-EM to reveal the organisation and architecture of the complete human TSCC. We show that TSCC forms an elongated scorpion-like structure, consisting of a central “body”, with a “pincer” and a “tail” at the respective ends. The “body” is composed of a flexible TSC2 HEAT repeat dimer, along the surface of which runs the TSC1 coiled-coil backbone, breaking the symmetry of the dimer. Each end of the body is structurally distinct, representing the N- and C-termini of TSC1; a “pincer” is formed by the highly flexible N-terminal TSC1 core domains and a barbed “tail” makes up the TSC1 coiled-coil-TBC1D7 junction. The TSC2 GAP domain is found abutting the centre of the body on each side of the dimerisation interface, poised to bind a pair of Rheb molecules at a similar separation to the pair in activated mTORC1.Our architectural dissection reveals the mode of association and topology of the complex, casts light on the recruitment of Rheb to the TSCC, and also hints at functional higher order oligomerisation, which has previously been predicted to be important for Rheb-signalling suppression.

Journal article

Wang X, Yang S, Li S, Zhao L, Hao Y, Qin J, Zhang L, Zhang C, Bian W, Zuo L, Gao X, Zhu B, Lei XG, Gu Z, Cui W, Xu X, Li Z, Zhu B, Li Y, Chen S, Guo H, Zhang H, Sun J, Zhang M, Hui Y, Zhang X, Liu X, Sun B, Wang L, Qiu Q, Zhang Y, Li X, Liu W, Xue R, Wu H, Shao D, Li J, Zhou Y, Li S, Yang R, Pedersen OB, Yu Z, Ehrlich SD, Ren Fet al., 2020, Aberrant gut microbiota alters host metabolome and impacts renal failure in humans and rodents, Gut, Vol: 69, Pages: 2131-2142, ISSN: 0017-5749

OBJECTIVE: Patients with renal failure suffer from symptoms caused by uraemic toxins, possibly of gut microbial origin, as deduced from studies in animals. The aim of the study is to characterise relationships between the intestinal microbiome composition, uraemic toxins and renal failure symptoms in human end-stage renal disease (ESRD). DESIGN: Characterisation of gut microbiome, serum and faecal metabolome and human phenotypes in a cohort of 223 patients with ESRD and 69 healthy controls. Multidimensional data integration to reveal links between these datasets and the use of chronic kidney disease (CKD) rodent models to test the effects of intestinal microbiome on toxin accumulation and disease severity. RESULTS: A group of microbial species enriched in ESRD correlates tightly to patient clinical variables and encode functions involved in toxin and secondary bile acids synthesis; the relative abundance of the microbial functions correlates with the serum or faecal concentrations of these metabolites. Microbiota from patients transplanted to renal injured germ-free mice or antibiotic-treated rats induce higher production of serum uraemic toxins and aggravated renal fibrosis and oxidative stress more than microbiota from controls. Two of the species, Eggerthella lenta and Fusobacterium nucleatum, increase uraemic toxins production and promote renal disease development in a CKD rat model. A probiotic Bifidobacterium animalis decreases abundance of these species, reduces levels of toxins and the severity of the disease in rats. CONCLUSION: Aberrant gut microbiota in patients with ESRD sculpts a detrimental metabolome aggravating clinical outcomes, suggesting that the gut microbiota will be a promising target for diminishing uraemic toxicity in those patients. TRIAL REGISTRATION NUMBER: This study was registered at ClinicalTrials.gov (NCT03010696).

Journal article

Lv C, Li F, Li X, Tian Y, Zhang Y, Sheng X, Song Y, Meng Q, Yuan S, Luan L, Andl T, Feng X, Jiao B, Xu M, Plikus MV, Dai X, Lengner C, Cui W, Ren F, Shuai J, Millar SE, Yu Zet al., 2020, Author Correction: MiR-31 promotes mammary stem cell expansion and breast tumorigenesis by suppressing Wnt signaling antagonists., Nature Communications, Vol: 11, Pages: 1-4, ISSN: 2041-1723

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

Journal article

Sheng X, Lin Z, Lv C, Shao C, Bi X, Deng M, Xu J, Guerrero-Juarez CF, Li M, Wu X, Zhao R, Yang X, Li G, Liu X, Wang Q, Nie Q, Cui W, Gao S, Zhang H, Liu Z, Cong Y, Plikus MV, Lengner CJ, Andersen B, Ren F, Yu Zet al., 2020, Cycling Stem Cells Are Radioresistant and Regenerate the Intestine., Cell Reports, Vol: 32, Pages: 107952-107952, ISSN: 2211-1247

A certain number of epithelial cells in intestinal crypts are DNA damage resistant and contribute to regeneration. However, the cellular mechanism underlying intestinal regeneration remains unclear. Using lineage tracing, we show that cells marked by an Msi1 reporter (Msi1+) are right above Lgr5high cells in intestinal crypts and exhibit DNA damage resistance. Single-cell RNA sequencing reveals that the Msi1+ cells are heterogeneous with the majority being intestinal stem cells (ISCs). The DNA damage-resistant subpopulation of Msi1+ cells is characterized by low-to-negative Lgr5 expression and is more rapidly cycling than Lgr5high radiosensitive crypt base columnar stem cells (CBCs). This enables an efficient repopulation of the intestinal epithelium at early stage when Lgr5high cells are not emerging. Furthermore, relative to CBCs, Msi1+ cells preferentially produce Paneth cells during homeostasis and upon radiation repair. Together, we demonstrate that the DNA damage-resistant Msi1+ cells are cycling ISCs that maintain and regenerate the intestinal epithelium.

Journal article

Tan GC, Wong YP, Cui W, Dibb Net al., 2020, Construction of a doxycycline inducible lentivirus that expresses stem cell-specific miR-302 cluster, MALAYSIAN JOURNAL OF PATHOLOGY, Vol: 42, Pages: 91-97, ISSN: 0126-8635

Journal article

Zhao R, Du S, Liu Y, Lv C, Song Y, Chen X, Zhang B, Li D, Gao S, Cui W, Plikus M, Hou X, Wu K, Liu Z, Liu Z, Cong Y, Li Y, Yu Zet al., 2020, Mucoadhesive-to-penetrating controllable peptosomes-in-microspheres co-loaded with anti-miR-31 oligonucleotide and Curcumin for targeted colorectal cancer therapy, THERANOSTICS, Vol: 10, Pages: 3594-3611, ISSN: 1838-7640

Journal article

Diksin M, Rowlinson J, Kondrashov A, Denning C, Hughes J, Constantin-Teodosiu T, Cui W, Scurr D, Ahmad O, Gell C, Onion D, Croxall N, Smith S, Rahman Ret al., 2019, CHARACTERISATION OF THE POST-SURGICAL INVASIVE TUMOUR NICHE USING ASTROCYTE-GLIOBLASTOMA ORGANOIDS AND DECELLULARISED HUMAN BRAIN, 24th Annual Scientific Meeting and Education Day of the Society-for-Neuro-Oncology (SNO) / 3rd SNO-SCIDOT Joint Conference on Therapeutic Delivery to the CNS, Publisher: OXFORD UNIV PRESS INC, Pages: 264-265, ISSN: 1522-8517

Conference paper

Li G, Tian X, Lv D, Zhang L, Zhang Z, Wang J, Yang M, Tao J, Ma T, Wu H, Ji P, Wu Y, Lian Z, Cui W, Liu Get al., 2019, NLRP7 is expressed in ovine ovary and associated with in vitro preimplantation embryo development, Reproduction, Vol: 158, Pages: 415-427, ISSN: 0022-4251

NLRP (NACHT, LRR and PYD domains-containing proteins) family plays the pivotal roles in mammalian reproduction. Mutation of NLRP7 is often associated with human recurrent hydatidiform moles. Few studies regarding the functions of NLRP7 have been performed in another mammalian species rather than humans. In the current study, for the first time, the function of NLRP7 has been explored in ovine ovary. NLRP7 protein was mainly located in ovarian follicles and in the in vitro pre-implantation embryos. To identify its origin, 763bp partial CDS of NLRP7 deriving from sheep cumulus oocyte complexes (COCs) was cloned, it showed a great homology with Homo sapiens. The high levels of mRNA and protein of NLRP7 were steadily expressed in oocytes, parthenogenetic embryos or IVF embryos. NLRP7 knockdown by the combination of siRNA and shRNA jeopardized both the parthenogenetic and IVF embryo development. The results strongly suggest that NLRP7 plays an important role in ovine reproduction. The potential mechanisms of NLRP7 will be fully investigated in the future.

Journal article

Xu W, Hua H, Chiu Y, Li G, Zhi H, Yu Z, Ren F, Luo Y, Cui Wet al., 2019, CD146 Regulates Growth Factor-Induced mTORC2 Activity Independent of PI3K and mTORC1 Pathways, Cell Reports, Vol: 29, Pages: 1311-1322.E5, ISSN: 2211-1247

The mechanistic target of rapamycin complex 2(mTORC2) coordinates cell proliferation, survival,and metabolism with environmental inputs, yet howextracellular stimuli such as growth factors (GFs)activate mTORC2 remains enigmatic. Here wedemonstrate that in human endothelial cells,activation of mTORC2 signaling by GFs is mediatedby transmembrane cell adhesion protein CD146.Upon GF stimulation, the cytoplasmic tail of CD146is phosphorylated, which permits its positivelycharged, juxtamembrane KKGK motif to interactwith Rictor, the defining subunit of mTORC2. The formation of the CD146-Rictor/mTORC2 complexprotects Rictor from ubiquitin-proteasome-mediated degradation, thereby specifically upregulatingmTORC2 activity with no intervention of thePI3K and mTORC1 pathways. This CD146-mediatedmTORC2 activation in response to GF stimulationpromotes cell proliferation and survival. Therefore,our findings identify a molecular mechanism bywhich extracellular stimuli regulate mTORC2 activity,linking environmental cues with mTORC2 regulation.

Journal article

Diksin M, Rowlinson J, Kondrashov A, Denning C, Hughes J, Constantin-Teodo T, Cui W, Gell C, Onion D, Croxall N, Smith S, Rahman Ret al., 2019, CHARACTERISATION OF THE INVASIVE TUMOUR NICHE USING ASTROCYTE-GLIOBLASTOMA ORGANOIDS AND DECELLULARISED HUMAN BRAIN, Meeting of the British-Neuro-Oncology-Society (BNOS), Publisher: OXFORD UNIV PRESS INC, Pages: 7-7, ISSN: 1522-8517

Conference paper

Xu W, Hua H, Chiu Y-H, Li G, Zhi H, Yu Z, Ren F, Luo Y, Cui Wet al., 2019, CD146 Regulates Growth Factor-Induced mTORC2 Activity Independent of the PI3K and mTORC1 Pathways, Cell Reports, Vol: 29, Pages: 1311-1322.e5, ISSN: 2211-1247

Journal article

Tian Y, Xu J, Li Y, Zhao R, Du S, Lv C, Wu W, Liu R, Sheng X, Song Y, Bi X, Li G, Li M, Wu X, Lou P, You H, Cui W, Sun J, Shuai J, Ren F, Zhang B, Guo M, Hou X, Wu K, Xue L, Zhang H, Plikus MV, Cong Y, Lengner CJ, Liu Z, Yu Zet al., 2019, MicroRNA-31 reduces inflammatory signaling and promotes regeneration in colon epithelium, and delivery of mimics in microspheres reduces colitis in mice, Gastroenterology, Vol: 156, Pages: 2281-2296.e6, ISSN: 0016-5085

BACKGROUND & AIMS: Levels of microRNA 31 (MIR31) are increased in intestinal tissues from patients with inflammatory bowel diseases and colitis-associated neoplasias. We investigated the effects of this microRNA on intestinal inflammation by studying mice with colitis. METHODS: We obtained colon biopsy samples from 82 patients with ulcerative colitis (UC), 79 patients with Crohn's disease (CD), and 34 healthy individuals (controls) at Shanghai Tenth People's Hospital. MIR31- knockout mice and mice with conditional disruption of Mir31 specifically in the intestinal epithelium (MIR31 conditional knockouts) were given dextran sulfate sodium (DSS) or 2,4,6-trinitrobenzene sulfonic acid (TNBS) to induce colitis. We performed chromatin immunoprecipitation (ChIP) and luciferase assays to study proteins that regulate expression of MIR31, including STAT3 and p65, in LOVO colorectal cancer cells and organoids derived from mouse colon cells. Partially hydrolyzed alpha-lactalbumin (alpha-La) was used to generate peptosome nanoparticles, and MIR31 mimics were loaded onto their surface using electrostatic adsorption. Peptosome-MIR31 mimic particles were encapsulated into oxidized konjac glucomannan (OKGM) microspheres, which were administered by enema into the large intestines of mice with DSS-induced colitis. Intestinal tissues were collected and analyzed by histology and immunohistochemistry. RESULTS: Levels of MIR31 were increased in inflamed mucosa from patients with CD or UC, and from mice with colitis, compared with controls. STAT3 and NF-κB activated transcription of MIR31 in colorectal cancer cells and organoids in response to TNF and IL6. MIR31-knockout and conditional-knockout mice developed more severe colitis in response to DSS and TNBS, with increased immune responses, compared to control mice. MIR31 bound to 3' untranslated regions of Il17ra and Il7r mRNAs (which encode receptors for the inflammatory cytokine IL17) and Il16st mRNA (which encodes GP130, a c

Journal article

Cui W, Zhang S, Bell E, Zhi H, Brown S, Muhammad Imran SA, Azuara Vet al., 2019, OCT4 and PAX6 determine the dual function of SOX2 in human ESCs as a key pluripotent or neural factor, Stem Cell Research and Therapy, Vol: 10, ISSN: 1757-6512

BackgroundSox2 is a well-established pluripotent transcription factor that plays an essential role in establishing and maintaining pluripotent stem cells (PSCs). It is also thought to be a linage specifier that governs PSC neural lineage specification upon their exiting the pluripotent state. However, the exact role of SOX2 in human PSCs was still not fully understood. In this study, we studied the role of SOX2 in human embryonic stem cells (hESCs) by gain- and loss-of-function approaches and explored the possible underlying mechanisms.ResultsWe demonstrate that knockdown of SOX2 induced hESC differentiation to endoderm-like cells, whereas overexpression of SOX2 in hESCs enhanced their pluripotency under self-renewing culture conditions but promoted their neural differentiation upon replacing the culture to non-self-renewal conditions. We show that this culture-dependent dual function of SOX2 was probably attributed to its interaction with different transcription factors predisposed by the culture environments. Whilst SOX2 interacts with OCT4 under self-renewal conditions, we found that, upon neural differentiation, PAX6, a key neural transcription factor, is upregulated and shows interaction with SOX2. The SOX2-PAX6 complex has different gene regulation pattern from that of SOX2-OCT4 complex.ConclusionsOur work provides direct evidence that SOX2 is necessarily required for hESC pluripotency; however, it can also function as a neural factor, depending on the environmental input. OCT4 and PAX6 might function as key SOX2-interacting partners that determine the function of SOX2 in hESCs.

Journal article

Luo Y, Xu W, Li G, Cui Wet al., 2018, Weighing in on mTOR complex 2 signaling: The expanding role in cell metabolism, Oxidative Medicine and Cellular Longevity, Vol: 2018, ISSN: 1942-0900

In all eukaryotes, the mechanistic target of rapamycin (mTOR) signaling emerges as a master regulator of homeostasis, which integrates environmental inputs, including nutrients, energy, and growth factors, to regulate many fundamental cellular processes such as cell growth and metabolism. mTOR signaling functions through two structurally and functionally distinct complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), which correspond to two major branches of signal output. While mTORC1 is well characterized for its structure, regulation, and function in the last decade, information of mTORC2 signaling is only rapidly expanding in recent years, from structural biology, signaling network, to functional impact. Here we review the recent advances in many aspects of the mTORC2 signaling, with particular focus on its involvement in the control of cell metabolism and its physiological implications in metabolic diseases and aging.

Journal article

Ramasamy TS, Ong ALC, Cui W, 2018, Impact of Three-Dimentional Culture Systems on Hepatic Differentiation of Puripotent Stem Cells and Beyond., Adv Exp Med Biol, Vol: 1077, Pages: 41-66, ISSN: 0065-2598

Generation of functional hepatocytes from human pluripotent stem cells (hPSCs) is a vital tool to produce large amounts of human hepatocytes, which hold a great promise for biomedical and regenerative medicine applications. Despite a tremendous progress in developing the differentiation protocols recapitulating the developmental signalling and stages, these resulting hepatocytes from hPSCs yet achieve maturation and functionality comparable to those primary hepatocytes. The absence of 3D milieu in the culture and differentiation of these hepatocytes may account for this, at least partly, thus developing an optimal 3D culture could be a step forward to achieve this aim. Hence, review focuses on current development of 3D culture systems for hepatic differentiation and maturation and the future perspectives of its application.

Journal article

Shi J, Ma X, Su Y, Song Y, Tian Y, Yuan S, Zhang X, Yang D, Zhang H, Shuai J, Cui W, Ren F, Plikus MV, Chen Y, Luo J, Yu Zet al., 2018, MiR-31 mediates inflammatory signaling to promote re-epithelialization during skin wound healing, Journal of Investigative Dermatology, Vol: 138, Pages: 2253-2263, ISSN: 0022-202X

Wound healing is essential for skin repair after injury, and consists of hemostasis, inflammation, re-epithelialization and remodeling phases. Successful re-epithelialization, which relies on proliferation and migration of epidermal keratinocytes, requires reduction in tissue inflammation. Therefore, understanding the molecular mechanism underlying the transition from inflammation to re-epithelialization will help to better understand the principles of wound healing. Currently, the in vivo functions of specific microRNAs in wound healing are not fully understood. We observed that miR-31 expression is strongly induced in wound edge keratinocytes, and is directly regulated by the activity of NF-κB and STAT3 signaling pathways during inflammation phase. We utilized miR-31 loss-of-function mouse models to demonstrate that miR-31 promotes keratinocyte proliferation and migration. Mechanistically, miR-31 activates the RAS/MAPK signaling by directly targeting Rasa1, Spred1, Spred2 and Spry4, which are negative regulators of the RAS/MAPK pathway. Knockdown of these miR-31 targets at least partially rescues the delayed scratch wound re-epithelialization phenotype observed in vitro in miR-31 knockdown keratinocytes. Taken together, these findings identify miR-31 as an important cell-autonomous mediator during the transition from inflammation to re-epithelialization phases of wound healing, suggesting a therapeutic potential for miR-31 in skin injury repair.

Journal article

Li Y, Wang J, Ren F, Zhang W, Zhang H, Zhao L, Zhang M, Cui W, Wang X, Guo Het al., 2018, Lactoferrin Promotes Osteogenesis through TGF-ss Receptor II Binding in Osteoblasts and Activation of Canonical TGF-ss Signaling in MC3T3-E1 Cells and C57BL/6J Mice, JOURNAL OF NUTRITION, Vol: 148, Pages: 1285-1292, ISSN: 0022-3166

BackgroundLactoferrin (LF), as a major functional protein in dairy products, is known to modulate bone anabolic effects. However, the underlying molecular mechanisms remain unclear; the receptor of LF in osteoblast differentiation has not been identified.ObjectiveThe aims of the study were to 1) illuminate whether the receptor of LF in osteoblast differentiation is transforming growth factor-β (TGF-β) receptor (TβR) II and 2) determine whether the TGF-β signaling pathway is activated by LF in promoting osteogenesis in vitro and in vivo, in addition to P38 and extracellular signal-regulated kinase (ERK) pathways.MethodsWe utilized co-immunoprecipitation to detect any binding of LF to TβRII. Subsequently, the role of the TGF-β signaling pathway involved in LF-induced osteoblast proliferation and differentiation was determined by inhibition of TβRI activity by inhibition and knockout of TβRII expression by small guide RNA (sgRNAs) in MC3T3-E1 cells. In addition, 4-wk-old male C57BL/6J mice were orally administered 100 mg LF/kg body weight for 16 wk, after which any activation of the TGF-β signaling pathway in vivo was measured by Western blots.ResultsLF was shown to directly interact with the TβRII protein and to activate the TGF-β signaling pathway in MC3T3-E1 cells. Inhibition of TβRI activity and knockout TβRII expression both attenuated the stimulation of LF in osteoblast proliferation and differentiation by 30–50%. LF-induced activation of TGF-β canonical signaling resulted in upregulation of osteogenic factors. Moreover, the expression of phosphorylated-drosophila mothers against decapentaplegic protein 2 (SMAD2) was increased by 1-fold after LF treatment in the femoral tissue of mice.ConclusionsThis study provides evidence identifying TβRII as an LF receptor in LF-induced osteoblast differentiation. In addition, the TβRII-dependent TGF-β canonical signaling pathways were p

Journal article

Lv C, Li F, Li X, Tian Y, Zhang Y, Sheng X, Song Y, Meng Q, Yuan S, Luan L, Andl T, Feng X, Jiao B, Xu M, Plikus MV, Dai X, Lengner C, Cui W, Ren F, Shuai J, Millar SE, Yu Zet al., 2017, MiR-31 promotes mammary stem cell expansion and breast tumorigenesis by suppressing Wnt signaling antagonists, Nature Communications, Vol: 8, ISSN: 2041-1723

MicroRNA-mediated post-transcriptional regulation plays key roles in stem cell self-renewal and tumorigenesis. However, the in vivo functions of specific microRNAs in controlling mammary stem cell (MaSC) activity and breast cancer formation remain poorly understood. Here we show that miR-31 is highly expressed in MaSC-enriched mammary basal cell population and in mammary tumors, and is regulated by NF-κB signaling. We demonstrate that miR-31 promotes mammary epithelial proliferation and MaSC expansion at the expense of differentiation in vivo. Loss of miR-31 compromises mammary tumor growth, reduces the number of cancer stem cells, as well as decreases tumor-initiating ability and metastasis to the lung, supporting its pro-oncogenic function. MiR-31 modulates multiple signaling pathways, including Prlr/Stat5, TGFβ and Wnt/β-catenin. Particularly, it activates Wnt/β-catenin signaling by directly targeting Wnt antagonists, including Dkk1. Importantly, Dkk1 overexpression partially rescues miR31-induced mammary defects. Together, these findings identify miR-31 as the key regulator of MaSC activity and breast tumorigenesis.

Journal article

Tian Y, Ma X, Lv C, Sheng X, Li, Song Y, Andl T, Plikus MV, Sun J, Ren F, Shuai J, Lengner CJ, Cui W, Yu Zet al., 2017, Stress responsive miR-31 is a major modulator of mouse intestinal stem cells during regeneration and tumorigenesis, eLife, Vol: 6, Pages: 1-30, ISSN: 2050-084X

Intestinal regeneration and tumorigenesis are believed to be driven by intestinal stemcells (ISCs). Elucidating mechanisms underlying ISC activation during regeneration andtumorigenesis can help uncover the underlying principles of intestinal homeostasis and diseaseincluding colorectal cancer. Here we show that miR-31 drives ISC proliferation, and protects ISCsagainst apoptosis, both during homeostasis and regeneration in response to ionizing radiationinjury. Furthermore, miR-31 has oncogenic properties, promoting intestinal tumorigenesis.Mechanistically, miR-31 acts to balance input from Wnt, BMP, TGFb signals to coordinate control ofintestinal homeostasis, regeneration and tumorigenesis. We further find that miR-31 is regulated bythe STAT3 signaling pathway in response to radiation injury. These findings identify miR-31 as acritical modulator of ISC biology, and a potential therapeutic target for a broad range of intestinalregenerative disorders and cancers.

Journal article

Zhu J, Cui W, Dai Y-F, 2017, Production of inbred offspring by intracytoplasmic sperm injection of oocytes from juvenile female mice, Reproduction, Fertility and Development, Vol: 30, Pages: 451-458, ISSN: 1031-3613

The aim of the present study was to determine whether the use of oocytes from juvenile female mice would improve the efficiency of intracytoplasmic sperm injection (ICSI). In the present study, 15 adult and 14 juvenile C57BL6/J female mice were superovulated, with 17.8 oocytes per mouse harvested from adults, significantly lower than the 40.2 harvested from juveniles (P<0.01). Sixty and 233 oocytes were harvested from C57BL/6J adult and juvenile mice respectively, activated in 10mM SrCl2+5μgmL-1 cytochalasin B for 5-6h and cultured in potassium simplex optimisation medium (KSOM) for 3.5 days, with no differences in morula and blastocyst rates between groups (91.7% vs 96.6%; P>0.05). Twelve hours after injection of human chorionic gonadotrophin, oocytes were harvested from C57BL/6J juvenile mice into KSOM, randomly divided into groups and activated with the same method mentioned above at 0, 2, 4 or 6h and then cultured in KSOM for 3.5 days. There was no significant difference in morula and blastocyst rates among the different groups (P>0.05). Oocytes from juvenile mice activated in 10mM SrCl2 for 2h were subjected to ICSI and the rates of pronuclear formation and Day 1 cleavage were significantly improved compared with the control group (P<0.01). ICSI combined with activation of oocytes from inbred mouse strains (C57BL/6J, C57BL/6N and 129Svev) successfully produced pups. The fertility of some these mice resulting from ICSI was tested, and the animals proved fertile. In conclusion, superovulated juvenile mice can yield more useable oocytes than adult mice, but additional activation is essential for full development of ICSI oocytes harvested from juvenile inbred mice.

Journal article

Imran SAM, Cui W, 2017, The Role of TRF2 in Maintaining and Protecting Neural Cell Properties., 64th Annual Scientific Meeting of the Society-for-Reproductive-Investigation (SRI), Publisher: SAGE PUBLICATIONS INC, Pages: 235A-235A, ISSN: 1933-7191

Conference paper

Cui W, Yu J, 2016, Proliferation, survival and metabolism: the role of PI3K/AKT/mTOR signalling in pluripotency and cell fate determination, Development, Vol: 143, Pages: 3050-3060, ISSN: 0950-1991

Phosphatidylinositide 3 kinases (PI3Ks) and their downstream mediators AKT and mammaliantarget of rapamycin (mTOR) constitute the core components of the PI3K/AKT/mTORsignalling cascade, regulating cell proliferation, survival and metabolism. Although thesefunctions are well defined in the context of tumorigenesis, recent studies – in particular thoseusing pluripotent stem cells – have highlighted the importance of this pathway to developmentand cellular differentiation. Here we review the recent in vitro and in vivo evidence for the rolePI3K/Akt/mTOR signalling plays in the control of pluripotency and differentiation, with aparticular focus on the molecular mechanisms underlying these functions.

Journal article

Ovando-Roche P, Cui W, 2015, Identification of Factors in Regulating a Protein Ubiquitination by Immunoprecipitation: a Case Study of TRF2 on Human REST4 Ubiquitination., Bio-protocol, Vol: 5, ISSN: 2331-8325

Ubiquitination is the first step of the ubiquitin-proteasome pathway that regulates cells for their homeostatic functions and is an enzymatic, protein post-translational modification process in which ubiquitin is transferred to a target protein substrate by a set of three ubiquitin enzymes (Weissman et al., 2011; Bhattacharyya et al., 2014; Ristic et al., 2014). Given the importance of this process, it is plausible that ubiquitination is under strict control by many factors and that the regulatory machineries are protein-specific. An assay for the detection of a specific protein ubiquitination will enable us to examine whether a factor has a function to regulate the ubiquitination of this protein. Here we describe a protocol that detects the ubiquitination status of the human REST4 protein in cultured cells, a neural alternative splicing isoform of REST (RE-1 silencing transcription factor), that antagonizes the repressive function of REST on neural differentiation and neuron formation. Using this protocol, we show that the telomere binding protein TRF2 stabilizes the expression of the human REST4 by inhibiting its ubiquitination. This indicates that TRF2 plays a positive role in neural differentiation (Ovando-Roche et al., 2014). This protocol is also useful for the detection of ubiquitination of other proteins of interest.

Journal article

This data is extracted from the Web of Science and reproduced under a licence from Thomson Reuters. You may not copy or re-distribute this data in whole or in part without the written consent of the Science business of Thomson Reuters.

Request URL: http://wlsprd.imperial.ac.uk:80/respub/WEB-INF/jsp/search-html.jsp Request URI: /respub/WEB-INF/jsp/search-html.jsp Query String: respub-action=search.html&id=00460033&limit=30&person=true