Network


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

Hotspot


Dive into the research topics where Lele Jiang is active.

Publication


Featured researches published by Lele Jiang.


Nature Medicine | 2007

Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-1.

Heiko Johnen; Shu Lin; Tamara Kuffner; David A. Brown; Vicky Wang-Wei Tsai; Asne R. Bauskin; Liyun Wu; Greg J. Pankhurst; Lele Jiang; Simon Junankar; Mark Hunter; W. Douglas Fairlie; Nicola J. Lee; Ronaldo F. Enriquez; Paul A. Baldock; Eva Corey; Fred S. Apple; MaryAnn M. Murakami; En Ju Lin; Chuansong Wang; Matthew J. During; Amanda Sainsbury; Herbert Herzog; Samuel N. Breit

Anorexia and weight loss are part of the wasting syndrome of late-stage cancer, are a major cause of morbidity and mortality in cancer, and are thought to be cytokine mediated. Macrophage inhibitory cytokine-1 (MIC-1) is produced by many cancers. Examination of sera from individuals with advanced prostate cancer showed a direct relationship between MIC-1 abundance and cancer-associated weight loss. In mice with xenografted prostate tumors, elevated MIC-1 levels were also associated with marked weight, fat and lean tissue loss that was mediated by decreased food intake and was reversed by administration of antibody to MIC-1. Additionally, normal mice given systemic MIC-1 and transgenic mice overexpressing MIC-1 showed hypophagia and reduced body weight. MIC-1 mediates its effects by central mechanisms that implicate the hypothalamic transforming growth factor-β receptor II, extracellular signal–regulated kinases 1 and 2, signal transducer and activator of transcription-3, neuropeptide Y and pro-opiomelanocortin. Thus, MIC-1 is a newly defined central regulator of appetite and a potential target for the treatment of both cancer anorexia and weight loss, as well as of obesity.


Growth Factors Journal | 2011

The TGF-β superfamily cytokine, MIC-1/GDF15: A pleotrophic cytokine with roles in inflammation, cancer and metabolism

Samuel N. Breit; Heiko Johnen; Andrew D. Cook; Vicky Wang-Wei Tsai; Mohammad G. Mohammad; Tamara Kuffner; Hong Ping Zhang; Christopher P. Marquis; Lele Jiang; Glen P. Lockwood; Michelle Lee-Ng; Yasmin Husaini; Liyun Wu; John A. Hamilton; David A. Brown

Macrophage inhibitory cytokine-1 (MIC-1/GDF15) is associated with cardiovascular disease, inflammation, body weight regulation and cancer. Its serum levels facilitate the diagnosis and prognosis of cancer and vascular disease. Furthermore, its serum levels are a powerful predictor of all-cause mortality, suggesting a fundamental role in biological processes associated with ageing. In cancer, the data available suggest that MIC-1/GDF15 is antitumorigenic, but this may not always be the case as disease progresses. Cancer promoting effects of MIC-1/GDF15 may be due, in part, to effects on antitumour immunity. This is suggested by the anti-inflammatory and immunosuppressive properties of MIC-1/GDF15 in animal models of atherosclerosis and rheumatoid arthritis. Furthermore, in late-stage cancer, large amounts of MIC-1/GDF15 in the circulation suppress appetite and mediate cancer anorexia/cachexia, which can be reversed by monoclonal antibodies in animals. Available data suggest MIC-1/GDF15 may be an important molecule mediating the interplay between cancer, obesity and chronic inflammation.


FEBS Letters | 2010

The enigma of the CLIC proteins: Ion channels, redox proteins, enzymes, scaffolding proteins?

Dene R. Littler; Stephen J. Harrop; Sophia C. Goodchild; Juanita M. Phang; Andrew V. Mynott; Lele Jiang; Stella M. Valenzuela; Michele Mazzanti; Louise J. Brown; Samuel N. Breit; Paul M. G. Curmi

Chloride intracellular channel proteins (CLICs) are distinct from most ion channels in that they have both soluble and integral membrane forms. CLICs are highly conserved in chordates, with six vertebrate paralogues. CLIC‐like proteins are found in other metazoans. CLICs form channels in artificial bilayers in a process favoured by oxidising conditions and low pH. They are structurally plastic, with CLIC1 adopting two distinct soluble conformations. Phylogenetic and structural data indicate that CLICs are likely to have enzymatic function. The physiological role of CLICs appears to be maintenance of intracellular membranes, which is associated with tubulogenesis but may involve other substructures.


Journal of Immunology | 2009

High Levels of Human Antigen-Specific CD4+ T Cells in Peripheral Blood Revealed by Stimulated Coexpression of CD25 and CD134 (OX40)

John Zaunders; Mee Ling Munier; Nabila Seddiki; Sarah Pett; Susanna Ip; Michelle Bailey; Yin Xu; Kai Brown; Wayne B. Dyer; Min Kim; Robert De Rose; Stephen J. Kent; Lele Jiang; Samuel N. Breit; Sean Emery; Anthony L. Cunningham; David A. Cooper; Anthony D. Kelleher

Ag-specific human CD4+ memory T lymphocytes have mostly been studied using assays of proliferation in vitro. Intracellular cytokine and ELISPOT assays quantify effector cell populations but barely detect responses to certain recall Ags that elicit strong proliferative responses, e.g., tetanus toxoid, that comprise non-Th1 CD4+ cells. We have found that culturing whole blood with Ag for 40–48 h induces specific CD4+ T cells to simultaneously express CD25 and CD134. This new technique readily detects responses to well-described CD4+ T cell recall Ags, including preparations of mycobacteria, CMV, HSV-1, influenza, tetanus toxoid, Candida albicans, and streptokinase, as well as HIV-1 peptides, with high specificity. The assay detects much higher levels of Ag-specific cells than intracellular cytokine assays, plus the cells retain viability and can be sorted for in vitro expansion. Furthermore, current in vitro assays for human CD4+ memory T lymphocytes are too labor-intensive and difficult to standardize for routine diagnostic laboratories, whereas the whole-blood CD25+CD134+ assay combines simplicity of setup with a straightforward cell surface flow cytometry readout. In addition to revealing the true extent of Ag-specific human CD4+ memory T lymphocytes, its greatest use will be as a simple in vitro monitor of CD4+ T cell responses to Ags such as tuberculosis infection or vaccines.


The Journal of Neuroscience | 2008

CLIC1 function is required for beta-amyloid-induced generation of reactive oxygen species by microglia.

Rosemary Milton; Rosella Abeti; Stefania Averaimo; Silvia DeBiasi; Laura Vitellaro; Lele Jiang; Paul M. G. Curmi; Samuel N. Breit; Michael R. Duchen; Michele Mazzanti

The Alzheimers disease (AD) brain is characterized by plaques containing β-amyloid (Aβ) protein surrounded by astrocytes and reactive microglia. Activation of microglia by Aβ initiates production of reactive oxygen species (ROS) by the plasmalemmal NADPH oxidase; the resultant oxidative stress is thought to contribute to neurodegeneration in AD. We have previously shown that Aβ upregulates a chloride current mediated by the chloride intracellular channel 1 (CLIC1) protein in microglia. We now demonstrate that Aβ promotes the acute translocation of CLIC1 from the cytosol to the plasma membrane of microglia, where it mediates a chloride conductance. Both the Aβ induced Cl− conductance and ROS generation were prevented by pharmacological inhibition of CLIC1, by replacement of chloride with impermeant anions, by an anti-CLIC1 antibody and by suppression of CLIC1 expression using siRNA. Thus, the CLIC1-mediated Cl− conductance is required for Aβ-induced generation of neurotoxic ROS by microglia. Remarkably, CLIC1 activation is itself dependent on oxidation by ROS derived from the activated NADPH oxidase. We therefore propose that CLIC1 translocation from the cytosol to the plasma membrane, in response to redox modulation by NADPH oxidase-derived ROS, provides a feedforward mechanism that facilitates sustained microglial ROS generation by the NAPDH oxidase.


Journal of Cell Science | 2012

Intracellular chloride channel protein CLIC1 regulates macrophage function through modulation of phagosomal acidification.

Lele Jiang; Kanin Salao; Hui Li; Joanna M. Rybicka; Robin M. Yates; Xu Wei Luo; Xin Xin Shi; Tamara Kuffner; Vicky Wang-Wei Tsai; Yasmin Husaini; Liyun Wu; David A. Brown; Thomas Grewal; Louise J. Brown; Paul M. G. Curmi; Samuel N. Breit

Summary Intracellular chloride channel protein 1 (CLIC1) is a 241 amino acid protein of the glutathione S transferase fold family with redox- and pH-dependent membrane association and chloride ion channel activity. Whilst CLIC proteins are evolutionarily conserved in Metazoa, indicating an important role, little is known about their biology. CLIC1 was first cloned on the basis of increased expression in activated macrophages. We therefore examined its subcellular localisation in murine peritoneal macrophages by immunofluorescence confocal microscopy. In resting cells, CLIC1 is observed in punctate cytoplasmic structures that do not colocalise with markers for endosomes or secretory vesicles. However, when these macrophages phagocytose serum-opsonised zymosan, CLIC1 translocates onto the phagosomal membrane. Macrophages from CLIC1−/− mice display a defect in phagosome acidification as determined by imaging live cells phagocytosing zymosan tagged with the pH-sensitive fluorophore Oregon Green. This altered phagosomal acidification was not accompanied by a detectable impairment in phagosomal-lysosomal fusion. However, consistent with a defect in acidification, CLIC1−/− macrophages also displayed impaired phagosomal proteolytic capacity and reduced reactive oxygen species production. Further, CLIC1−/− mice were protected from development of serum transfer induced K/BxN arthritis. These data all point to an important role for CLIC1 in regulating macrophage function through its ion channel activity and suggest it is a suitable target for the development of anti-inflammatory drugs.


Nephrology Dialysis Transplantation | 2012

Macrophage inhibitory cytokine-1 (MIC-1/GDF15) and mortality in end-stage renal disease

Samuel N. Breit; Juan Jesus Carrero; Vicky Wang-Wei Tsai; Nasreen Yagoutifam; Wei Luo; Tamara Kuffner; Asne R. Bauskin; Liyun Wu; Lele Jiang; Peter Bárány; Olof Heimbürger; Mary-Ann Murikami; Fred S. Apple; Christopher P. Marquis; Laurence Macia; Shu Lin; Amanda Sainsbury; Herbert Herzog; Matthew Law; Peter Stenvinkel; David A. Brown

BACKGROUND Elevated macrophage inhibitory cytokine-1 (MIC-1/GDF15) levels in serum mediate anorexia and weight loss in some cancer patients and similarly elevated levels occur in chronic kidney disease (CKD). Serum MIC-1/GDF15 is also elevated in chronic inflammatory diseases and predicts atherosclerotic events independently of traditional risk factors. The relationship between chronic inflammation, decreasing body mass index (BMI) and increased mortality in CKD is not well understood and is being actively investigated. MIC-1/GDF15 may link these features of CKD. METHODS Cohorts of incident dialysis patients from Sweden (n = 98) and prevalent hemodialysis patients from the USA (n = 381) had serum MIC-1/GDF15, C-reactive protein (CRP) levels and BMI measured at study entry. Additional surrogate markers of nutritional adequacy, body composition and inflammation were assessed in Swedish patients. Patients were followed for all-cause mortality. RESULTS In the Swedish cohort, serum MIC-1/GDF15 was associated with decreasing BMI, measures of nutrition and markers of oxidative stress and inflammation. Additionally, high serum MIC-1/GDF15 levels identified patients with evidence of protein-energy wasting who died in the first 3 years of dialysis. The ability of serum MIC-1/GDF15 to predict mortality in the first 3 years of dialysis was confirmed in the USA cohort. In both cohorts, serum MIC-1/GDF15 level was an independent marker of mortality when adjusted for age, CRP, BMI, history of diabetes mellitus and/or cardiovascular disease and glomerular filtration rate or length of time on dialysis at study entry. CONCLUSIONS MIC-1/GDF15 is a novel independent serum marker of mortality in CKD capable of significantly improving the mortality prediction of other established markers. MIC-1/GDF15 may mediate protein-energy wasting in CKD and represent a novel therapeutic target for this fatal complication.


PLOS ONE | 2013

TGF-b Superfamily Cytokine MIC-1/GDF15 Is a Physiological Appetite and Body Weight Regulator

Vicky Wang-Wei Tsai; Laurence Macia; Heiko Johnen; Tamara Kuffner; Rakesh Manadhar; Sebastian B. Jørgensen; Ka Ki Michelle Lee-Ng; Hong Ping Zhang; Liyun Wu; Christopher P. Marquis; Lele Jiang; Yasmin Husaini; Shu Lin; Herbert Herzog; David A. Brown; Amanda Sainsbury; Samuel N. Breit

The TGF-b superfamily cytokine MIC-1/GDF15 circulates in all humans and when overproduced in cancer leads to anorexia/cachexia, by direct action on brain feeding centres. In these studies we have examined the role of physiologically relevant levels of MIC-1/GDF15 in the regulation of appetite, body weight and basal metabolic rate. MIC-1/GDF15 gene knockout mice (MIC-1−/−) weighed more and had increased adiposity, which was associated with increased spontaneous food intake. Female MIC-1−/− mice exhibited some additional alterations in reduced basal energy expenditure and physical activity, possibly owing to the associated decrease in total lean mass. Further, infusion of human recombinant MIC-1/GDF15 sufficient to raise serum levels in MIC-1−/− mice to within the normal human range reduced body weight and food intake. Taken together, our findings suggest that MIC-1/GDF15 is involved in the physiological regulation of appetite and energy storage.


Biochimica et Biophysica Acta | 2014

CLIC proteins, ezrin, radixin, moesin and the coupling of membranes to the actin cytoskeleton: a smoking gun?

Lele Jiang; Juanita M. Phang; Jiang Yu; Stephen J. Harrop; Anna Sokolova; Anthony P. Duff; Krystyna E. Wilk; Heba Alkhamici; Samuel N. Breit; Stella M. Valenzuela; Louise J. Brown; Paul M. G. Curmi

The CLIC proteins are a highly conserved family of metazoan proteins with the unusual ability to adopt both soluble and integral membrane forms. The physiological functions of CLIC proteins may include enzymatic activity in the soluble form and anion channel activity in the integral membrane form. CLIC proteins are associated with the ERM proteins: ezrin, radixin and moesin. ERM proteins act as cross-linkers between membranes and the cortical actin cytoskeleton. Both CLIC and ERM proteins are controlled by Rho family small GTPases. CLIC proteins, ERM and Rho GTPases act in a concerted manner to control active membrane processes including the maintenance of microvillar structures, phagocytosis and vesicle trafficking. All of these processes involve the interaction of membranes with the underlying cortical actin cytoskeleton. The relationships between Rho GTPases, CLIC proteins, ERM proteins and the membrane:actin cytoskeleton interface are reviewed. Speculative models are proposed involving the formation of localised multi-protein complexes on the membrane surface that assemble via multiple weak interactions. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.


Genesis | 2010

Generation and characterization of mice with null mutation of the chloride intracellular channel 1 gene.

Min Ru Qiu; Lele Jiang; Klaus I. Matthaei; Simone M. Schoenwaelder; Tamara Kuffner; Pierre Mangin; Joanne E. Joseph; Joyce Low; David E. Connor; Stella M. Valenzuela; Paul M. G. Curmi; Louise J. Brown; Martyn P. Mahaut-Smith; Shaun P. Jackson; Samuel N. Breit

CLIC1 belongs to a family of highly conserved and widely expressed intracellular chloride ion channel proteins existing in both soluble and membrane integrated forms. To study the physiological and biological role of CLIC1 in vivo, we undertook conditional gene targeting to engineer Clic1 gene knock‐out mice. This represents creation of the first gene knock‐out of a vertebrate CLIC protein family member. We first generated a Clic1 Knock‐in (Clic1FN) allele, followed by Clic1 knock‐out (Clic1−/−) mice by crossing Clic1FN allele with TNAP‐cre mice, resulting in germline gene deletion through Cre‐mediated recombination. Mice heterozygous or homozygous for these alleles are viable and fertile and appear normal. However, Clic1−/− mice show a mild platelet dysfunction characterized by prolonged bleeding times and decreased platelet activation in response to adenosine diphosphate stimulation linked to P2Y12 receptor signaling. genesis 48:127–136, 2010.

Collaboration


Dive into the Lele Jiang's collaboration.

Top Co-Authors

Avatar

Samuel N. Breit

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

David A. Brown

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Tamara Kuffner

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yasmin Husaini

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Liyun Wu

St. Vincent's Health System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul M. G. Curmi

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Ping Zhang

University of New South Wales

View shared research outputs
Researchain Logo
Decentralizing Knowledge