Network


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

Hotspot


Dive into the research topics where Changli Wei is active.

Publication


Featured researches published by Changli Wei.


Nature Genetics | 2005

TRPC6 is a glomerular slit diaphragm-associated channel required for normal renal function

Jochen Reiser; Krishna R. Polu; Clemens C. Möller; Peter Kenlan; Mehmet M. Altintas; Changli Wei; Christian Faul; Stephanie Herbert; Ivan Villegas; Carmen Avila-Casado; Mary McGee; Hikaru Sugimoto; Dennis Brown; Raghu Kalluri; Peter Mundel; Paula L. Smith; David E. Clapham; Martin R. Pollak

Progressive kidney failure is a genetically and clinically heterogeneous group of disorders. Podocyte foot processes and the interposed glomerular slit diaphragm are essential components of the permeability barrier in the kidney. Mutations in genes encoding structural proteins of the podocyte lead to the development of proteinuria, resulting in progressive kidney failure and focal segmental glomerulosclerosis. Here, we show that the canonical transient receptor potential 6 (TRPC6) ion channel is expressed in podocytes and is a component of the glomerular slit diaphragm. We identified five families with autosomal dominant focal segmental glomerulosclerosis in which disease segregated with mutations in the gene TRPC6 on chromosome 11q. Two of the TRPC6 mutants had increased current amplitudes. These data show that TRPC6 channel activity at the slit diaphragm is essential for proper regulation of podocyte structure and function.


Nature Medicine | 2011

Circulating urokinase receptor as a cause of focal segmental glomerulosclerosis

Changli Wei; Shafic El Hindi; Jing Li; Alessia Fornoni; Nelson Goes; Junichiro Sageshima; Dony Maiguel; S. Ananth Karumanchi; Hui Kim Yap; Moin A. Saleem; Qing-Yin Zhang; Boris Nikolic; Abanti Chaudhuri; Pirouz Daftarian; Eduardo Salido; Armando Torres; Moro O. Salifu; Minnie M. Sarwal; Franz Schaefer; Christian Morath; Vedat Schwenger; Martin Zeier; Vineet Gupta; David Roth; Maria Pia Rastaldi; George W. Burke; Phillip Ruiz; Jochen Reiser

Focal segmental glomerulosclerosis (FSGS) is a cause of proteinuric kidney disease, compromising both native and transplanted kidneys. Treatment is limited because of a complex pathogenesis, including unknown serum factors. Here we report that serum soluble urokinase receptor (suPAR) is elevated in two-thirds of subjects with primary FSGS, but not in people with other glomerular diseases. We further find that a higher concentration of suPAR before transplantation underlies an increased risk for recurrence of FSGS after transplantation. Using three mouse models, we explore the effects of suPAR on kidney function and morphology. We show that circulating suPAR activates podocyte β3 integrin in both native and grafted kidneys, causing foot process effacement, proteinuria and FSGS-like glomerulopathy. Our findings suggest that the renal disease only develops when suPAR sufficiently activates podocyte β3 integrin. Thus, the disease can be abrogated by lowering serum suPAR concentrations through plasmapheresis, or by interfering with the suPAR–β3 integrin interaction through antibodies and small molecules targeting either uPAR or β3 integrin. Our study identifies serum suPAR as a circulating factor that may cause FSGS.


Nature Medicine | 2008

Modification of kidney barrier function by the urokinase receptor

Changli Wei; Clemens C. Möller; Mehmet M. Altintas; Jing Li; Karin Schwarz; Serena Zacchigna; Liang Xie; Anna Henger; Holger Schmid; Maria Pia Rastaldi; Peter J. Cowan; Matthias Kretzler; Roberto Parrilla; Moise Bendayan; Vineet Gupta; Boris Nikolic; Raghu Kalluri; Peter Carmeliet; Peter Mundel; Jonche Reiser

Podocyte dysfunction, represented by foot process effacement and proteinuria, is often the starting point for progressive kidney disease. Therapies aimed at the cellular level of the disease are currently not available. Here we show that induction of urokinase receptor (uPAR) signaling in podocytes leads to foot process effacement and urinary protein loss via a mechanism that includes lipid-dependent activation of αvβ3 integrin. Mice lacking uPAR (Plaur−/−) are protected from lipopolysaccharide (LPS)-mediated proteinuria but develop disease after expression of a constitutively active β3 integrin. Gene transfer studies reveal a prerequisite for uPAR expression in podocytes, but not in endothelial cells, for the development of LPS-mediated proteinuria. Mechanistically, uPAR is required to activate αvβ3 integrin in podocytes, promoting cell motility and activation of the small GTPases Cdc42 and Rac1. Blockade of αvβ3 integrin reduces podocyte motility in vitro and lowers proteinuria in mice. Our findings show a physiological role for uPAR signaling in the regulation of kidney permeability.


Science Translational Medicine | 2011

Rituximab Targets Podocytes in Recurrent Focal Segmental Glomerulosclerosis

Alessia Fornoni; Junichiro Sageshima; Changli Wei; Sandra Merscher-Gomez; Robier Aguillon-Prada; Alexandra Jauregui; Jing Li; Adela Mattiazzi; Gaetano Ciancio; Linda Chen; Gaston Zilleruelo; Carolyn Abitbol; Jayanthi Chandar; Wacheree Seeherunvong; Camillo Ricordi; Masami Ikehata; Maria Pia Rastaldi; Jochen Reiser; George W. Burke

Rituximab treatment in high-risk patients with focal segmental glomerulosclerosis directly affects podocyte function and is linked to reduced incidence of recurrent proteinuria after kidney transplantation. Rituximab Prods Podocytes to Action Rituximab is a monoclonal antibody against CD20, a protein located on the surface of B cells. It is typically used to treat certain cancers and autoimmune disorders, but has also treated kidney conditions, including focal segmental glomerulosclerosis (FSGS)—a disorder that can affect both pediatric and adult patients. Recurrent FSGS is a problem for 30 to 40% of patients who have undergone kidney transplantation, and can be characterized by progression to end-stage renal disease and recurrence of proteinuria after transplant. Despite the ability of rituximab to treat FSGS, it has been unclear exactly how this drug achieves success in some patients, but not others. Fornoni and colleagues hypothesized that rituximab operates in a B cell–independent manner, targeting instead specific kidney cells called podocytes. To test this hypothesis, Fornoni et al. studied 41 patients at high risk for FSGS: 14 historical control patients who were not treated with rituximab and 27 patients who received rituximab at the time of kidney transplant. They found fewer podocytes with sphingomyelin phosphodiesterase acid-like 3b (SMPDL-3b) protein in biopsies from patients who later developed recurrent FSGS. The authors had also collected serum from all patients before transplant and then later treated normal human podocytes in culture with the sera. Serum from patients who would later develop recurrent FSGS caused a decrease in both SMPDL-3b protein and acid sphingomyelinase activity. This phenomenon was prevented by rituximab. The FSGS serum from patients also disrupted the actin cytoskeleton of cultured podocytes, but pretreatment with rituximab, or even overexpression of SMPDL-3b protein, partially prevented this phenotype. Together, these data suggest that modulation of sphingolipid-related proteins plays a role in the pathogenesis of recurrent FSGS and, moreover, that these proteins and enzymes might be targets of rituximab treatment. With the mechanism solved, rituximab may represent a new therapeutic strategy to prevent recurrent proteinuria after kidney transplantation. Here’s to happy and healthy kidneys! Focal segmental glomerulosclerosis (FSGS) is a glomerular disease characterized by proteinuria, progression to end-stage renal disease, and recurrence of proteinuria after kidney transplantation in about one-third of patients. It has been suggested that rituximab might treat recurrent FSGS through an unknown mechanism. Rituximab not only recognizes CD20 on B lymphocytes, but might also bind sphingomyelin phosphodiesterase acid-like 3b (SMPDL-3b) protein and regulate acid sphingomyelinase (ASMase) activity. We hypothesized that rituximab prevents recurrent FSGS and preserves podocyte SMPDL-3b expression. We studied 41 patients at high risk for recurrent FSGS, 27 of whom were treated with rituximab at time of kidney transplant. SMPDL-3b protein, ASMase activity, and cytoskeleton remodeling were studied in cultured normal human podocytes that had been exposed to patient sera with or without rituximab. Rituximab treatment was associated with lower incidence of posttransplant proteinuria and stabilization of glomerular filtration rate. The number of SMPDL-3b+ podocytes in postreperfusion biopsies was reduced in patients who developed recurrent FSGS. Rituximab partially prevented SMPDL-3b and ASMase down-regulation that was observed in podocytes treated with the sera of patients with recurrent FSGS. Overexpression of SMPDL-3b or treatment with rituximab was able to prevent disruption of the actin cytoskeleton and podocyte apoptosis induced by patient sera. This effect was diminished in cultured podocytes where SMPDL-3b was silenced. Our study suggests that treatment of high-risk patients with rituximab at time of kidney transplant might prevent recurrent FSGS by modulating podocyte function in an SMPDL-3b–dependent manner.


Journal of The American Society of Nephrology | 2007

Induction of TRPC6 Channel in Acquired Forms of Proteinuric Kidney Disease

Clemens C. Möller; Changli Wei; Mehmet M. Altintas; Jing Li; Anna Greka; Takamoto Ohse; Jeffrey W. Pippin; Maria Pia Rastaldi; Stefan Wawersik; Susan C. Schiavi; Anna Henger; Matthias Kretzler; Stuart J. Shankland; Jochen Reiser

Injury to podocytes and their slit diaphragms typically leads to marked proteinuria. Mutations in the TRPC6 gene that codes for a slit diaphragm-associated, cation-permeable ion channel have been shown recently to co-segregate with hereditary forms of progressive kidney failure. Herein is shown that induced expression of wild-type TRPC6 is a common feature of human proteinuric kidney diseases, with highest induction observed in membranous nephropathy. Cultured podocytes that are exposed to complement upregulate TRPC6 protein. Stimulation of receptor-operated channels in puromycin aminonucleoside-treated podocytes leads to increased calcium influx in a time- and dosage-dependent manner. Mechanistically, it is shown that TRPC6 is functionally connected to the podocyte actin cytoskeleton, which is rearranged upon overexpression of TRPC6. Transient in vivo gene delivery of TRPC6 into mice leads to expression of TRPC6 protein at the slit diaphragm and causes proteinuria. These studies suggest the involvement of TRPC6 in the pathology of nongenetic forms of proteinuric disease.


Journal of Clinical Investigation | 2007

Proteolytic processing of dynamin by cytoplasmic cathepsin L is a mechanism for proteinuric kidney disease

Sanja Sever; Mehmet M. Altintas; Sharif R. Nankoe; Clemens C. Möller; David Ko; Changli Wei; Joel Henderson; Elizabetta C. del Re; Lianne Hsing; Ann H. Erickson; Clemens D. Cohen; Matthias Kretzler; Dontscho Kerjaschki; Alexander Y. Rudensky; Boris Nikolic; Jochen Reiser

Kidney podocytes and their foot processes maintain the ultrafiltration barrier and prevent urinary protein loss (proteinuria). Here we show that the GTPase dynamin is essential for podocyte function. During proteinuric kidney disease, induction of cytoplasmic cathepsin L leads to cleavage of dynamin at an evolutionary conserved site, resulting in reorganization of the podocyte actin cytoskeleton and proteinuria. Dynamin mutants that lack the cathepsin L site, or render the cathepsin L site inaccessible through dynamin self-assembly, are resistant to cathepsin L cleavage. When delivered into mice, these mutants restored podocyte function and resolve proteinuria. Our study identifies dynamin as a critical regulator of renal permselectivity that is specifically targeted by proteolysis under pathological conditions.


Journal of The American Society of Nephrology | 2012

Circulating suPAR in Two Cohorts of Primary FSGS

Changli Wei; Howard Trachtman; Jing Li; Chuanhui Dong; Aaron L. Friedman; Jennifer Gassman; June L. McMahan; Milena Radeva; Karsten M. Heil; Agnes Trautmann; Ali Anarat; Sevinç Emre; Gian Marco Ghiggeri; Fatih Ozaltin; Dieter Haffner; Debbie S. Gipson; Frederick J. Kaskel; Dagmar-Christiane Fischer; Franz Schaefer; Jochen Reiser

Overexpression of soluble urokinase receptor (suPAR) causes pathology in animal models similar to primary FSGS, and one recent study demonstrated elevated levels of serum suPAR in patients with the disease. Here, we analyzed circulating suPAR levels in two cohorts of children and adults with biopsy-proven primary FSGS: 70 patients from the North America-based FSGS clinical trial (CT) and 94 patients from PodoNet, the Europe-based consortium studying steroid-resistant nephrotic syndrome. Circulating suPAR levels were elevated in 84.3% and 55.3% of patients with FSGS patients in the CT and PodoNet cohorts, respectively, compared with 6% of controls (P<0.0001); inflammation did not account for this difference. Multiple regression analysis suggested that lower suPAR levels associated with higher estimated GFR, male sex, and treatment with mycophenolate mofetil. In the CT cohort, there was a positive association between the relative reduction of suPAR after 26 weeks of treatment and reduction of proteinuria, with higher odds for complete remission (P=0.04). In the PodoNet cohort, patients with an NPHS2 mutation had higher suPAR levels than those without a mutation. In conclusion, suPAR levels are elevated in geographically and ethnically diverse patients with FSGS and do not reflect a nonspecific proinflammatory milieu. The associations between a change in circulating suPAR with different therapeutic regimens and with remission support the role of suPAR in the pathogenesis of FSGS.


Journal of The American Society of Nephrology | 2007

Overexpression of Interleukin-13 Induces Minimal-Change–Like Nephropathy in Rats

Kin-Wai Lai; Changli Wei; Li-Kiang Tan; Puay Hoon Tan; Chiang Gs; Caroline G. Lee; Stanley C. Jordan; Hui-Kim Yap

IL-13 has been implicated in the pathogenesis of minimal-change nephrotic syndrome. This study aimed to investigate the role of IL-13 on the development of proteinuria and expression of podocyte-related genes that are associated with nephrotic syndrome. IL-13 was overexpressed in Wistar rats through transfection of a mammalian expression vector cloned with the rat IL-13 gene, into the quadriceps by in vivo electroporation. Serum IL-13, albumin, cholesterol, and creatinine and urine albumin were measured serially. Kidneys were harvested after day 70 for histology and electron microscopy. Glomerular gene expression of nephrin, podocin, dystroglycan, B7-1, and IL-13 receptor subunits were examined using real-time PCR with hybridization probes and expressed as an index against beta-actin. Protein expression of these molecules was determined by immunofluorescence staining. The IL-13-transfected rats (n = 41) showed significant albuminuria, hypoalbuminemia, and hypercholesterolemia when compared with control rats (n = 17). No significant histologic changes were seen in glomeruli of IL-13-transfected rats. However, electron microscopy showed up to 80% of podocyte foot process fusion. Glomerular gene expression was significantly upregulated for B7-1, IL-4Ralpha, and IL-13Ralpha2 but downregulated for nephrin, podocin, and dystroglycan. Immunofluorescence staining intensity was reduced for nephrin, podocin, and dystroglycan but increased for B7-1 and IL-4Ralpha in IL-13-transfected rats compared with controls. In conclusion, these results suggest that IL-13 overexpression in the rat could lead to podocyte injury with downregulation of nephrin, podocin, and dystroglycan and a concurrent upregulation of B7-1 in the glomeruli, inducing a minimal change-like nephropathy that is characterized by increased proteinuria, hypoalbuminemia, hypercholesterolemia, and fusion of podocyte foot processes.


The New England Journal of Medicine | 2015

Soluble Urokinase Receptor and Chronic Kidney Disease

Salim Hayek; Sanja Sever; Yi An Ko; Howard Trachtman; Mosaab Awad; Shikha Wadhwani; Mehmet M. Altintas; Changli Wei; Anna L. Hotton; Audrey L. French; Laurence Sperling; Stamatios Lerakis; Arshed A. Quyyumi; Jochen Reiser

BACKGROUND Relatively high plasma levels of soluble urokinase-type plasminogen activator receptor (suPAR) have been associated with focal segmental glomerulosclerosis and poor clinical outcomes in patients with various conditions. It is unknown whether elevated suPAR levels in patients with normal kidney function are associated with future decline in the estimated glomerular filtration rate (eGFR) and with incident chronic kidney disease. METHODS We measured plasma suPAR levels in 3683 persons enrolled in the Emory Cardiovascular Biobank (mean age, 63 years; 65% men; median suPAR level, 3040 pg per milliliter) and determined renal function at enrollment and at subsequent visits in 2292 persons. The relationship between suPAR levels and the eGFR at baseline, the change in the eGFR over time, and the development of chronic kidney disease (eGFR <60 ml per minute per 1.73 m(2) of body-surface area) were analyzed with the use of linear mixed models and Cox regression after adjustment for demographic and clinical variables. RESULTS A higher suPAR level at baseline was associated with a greater decline in the eGFR during follow-up; the annual change in the eGFR was -0.9 ml per minute per 1.73 m(2) among participants in the lowest quartile of suPAR levels as compared with -4.2 ml per minute per 1.73 m(2) among participants in the highest quartile (P<0.001). The 921 participants with a normal eGFR (≥ 90 ml per minute per 1.73 m(2)) at baseline had the largest suPAR-related decline in the eGFR. In 1335 participants with a baseline eGFR of at least 60 ml per minute per 1.73 m(2), the risk of progression to chronic kidney disease in the highest quartile of suPAR levels was 3.13 times as high (95% confidence interval, 2.11 to 4.65) as that in the lowest quartile. CONCLUSIONS An elevated level of suPAR was independently associated with incident chronic kidney disease and an accelerated decline in the eGFR in the groups studied. (Funded by the Abraham J. and Phyllis Katz Foundation and others.).


Journal of Clinical Investigation | 2011

CD2AP in mouse and human podocytes controls a proteolytic program that regulates cytoskeletal structure and cellular survival

Suma Yaddanapudi; Mehmet M. Altintas; Andreas D. Kistler; Isabel Fernandez; Clemens C. Möller; Changli Wei; Vasil Peev; Jan Flesche; Anna Lena Forst; Jing Li; Jaakko Patrakka; Zhijie Xiao; Florian Grahammer; Mario Schiffer; Tobias Lohmüller; Thomas Reinheckel; Changkyu Gu; Tobias B. Huber; Wenjun Ju; Markus Bitzer; Maria Pia Rastaldi; Phillip Ruiz; Karl Tryggvason; Andrey S. Shaw; Christian Faul; Sanja Sever; Jochen Reiser

Kidney podocytes are highly differentiated epithelial cells that form interdigitating foot processes with bridging slit diaphragms (SDs) that regulate renal ultrafiltration. Podocyte injury results in proteinuric kidney disease, and genetic deletion of SD-associated CD2-associated protein (CD2AP) leads to progressive renal failure in mice and humans. Here, we have shown that CD2AP regulates the TGF-β1-dependent translocation of dendrin from the SD to the nucleus. Nuclear dendrin acted as a transcription factor to promote expression of cytosolic cathepsin L (CatL). CatL proteolyzed the regulatory GTPase dynamin and the actin-associated adapter synaptopodin, leading to a reorganization of the podocyte microfilament system and consequent proteinuria. CD2AP itself was proteolyzed by CatL, promoting sustained expression of the protease during podocyte injury, and in turn increasing the apoptotic susceptibility of podocytes to TGF-β1. Our study identifies CD2AP as the gatekeeper of the podocyte TGF-β response through its regulation of CatL expression and defines a molecular mechanism underlying proteinuric kidney disease.

Collaboration


Dive into the Changli Wei's collaboration.

Top Co-Authors

Avatar

Jochen Reiser

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mehmet M. Altintas

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Pia Rastaldi

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge