Network


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

Hotspot


Dive into the research topics where Hui Y. Lan is active.

Publication


Featured researches published by Hui Y. Lan.


Hypertension | 2003

Uric Acid Stimulates Monocyte Chemoattractant Protein-1 Production in Vascular Smooth Muscle Cells Via Mitogen-Activated Protein Kinase and Cyclooxygenase-2

John Kanellis; Susumu Watanabe; Jin H. Li; Duk Hee Kang; Ping Li; Takahiko Nakagawa; Ann Wamsley; David Sheikh-Hamad; Hui Y. Lan; Lili Feng; Richard J. Johnson

Abstract— Previous studies have reported that uric acid stimulates vascular smooth muscle cell (VSMC) proliferation in vitro. We hypothesized that uric acid may also have direct proinflammatory effects on VSMCs. Crystal‐ and endotoxin‐free uric acid was found to increase VSMC monocyte chemoattractant protein‐1 (MCP‐1) expression in a time‐ and dose‐dependent manner, peaking at 24 hours. Increased mRNA and protein expression occurred as early as 3 hours after uric acid incubation and was partially dependent on posttranscriptional modification of MCP‐1 mRNA. In addition, uric acid activated the transcription factors nuclear factor‐[kappa]B and activator protein‐1, as well as the MAPK signaling molecules ERK p44/42 and p38, and increased cyclooxygenase‐2 (COX‐2) mRNA expression. Inhibition of p38 (with SB 203580), ERK 44/42 (with UO126 or PD 98059), or COX‐2 (with NS398) each significantly suppressed uric acid–induced MCP‐1 expression at 24 hours, implicating these pathways in the response to uric acid. The ability of both n‐acetyl‐cysteine and diphenyleneionium (antioxidants) to inhibit uric acid–induced MCP‐1 production suggested involvement of intracellular redox pathways. Uric acid regulates critical proinflammatory pathways in VSMCs, suggesting it may have a role in the vascular changes associated with hypertension and vascular disease.


Cell Stem Cell | 2010

A Subpopulation of CD26+ Cancer Stem Cells with Metastatic Capacity in Human Colorectal Cancer

Roberta Pang; Wl Law; Andrew Cy Chu; Jensen T. Poon; Colin S.C. Lam; Ariel K.M. Chow; Lui Ng; Leonard W.H. Cheung; Xiao R. Lan; Hui Y. Lan; Victoria P.Y. Tan; Thomas Cheung Yau; Ronnie Tung-Ping Poon; Benjamin C.Y. Wong

Recent evidence suggests that a subpopulation of cancer cells, cancer stem cells (CSCs), is responsible for tumor growth in colorectal cancer. However, the role of CSCs in colorectal cancer metastasis is unclear. Here, we identified a subpopulation of CD26(+) cells uniformly present in both the primary and metastatic tumors in colorectal cancer patients with liver metastasis. Furthermore, in patients without distant metastasis at the time of presentation, the presence of CD26(+) cells in their primary tumors predicted distant metastasis on follow-up. Isolated CD26(+) cells, but not CD26(-) cells, led to development of distant metastasis when injected into the mouse cecal wall. CD26(+) cells were also associated with enhanced invasiveness and chemoresistance. Our findings have uncovered a critical role of CSCs in metastatic progression of cancer. Furthermore, the ability to predict metastasis based on analysis of CSC subsets in the primary tumor may have important clinical implication as a selection criterion for adjuvant therapy.


Journal of The American Society of Nephrology | 2011

TGF-β/Smad3 Signaling Promotes Renal Fibrosis by Inhibiting miR-29

Wei Qin; Arthur C.K. Chung; Xiao R. Huang; Xiao-Ming Meng; David Hui; Cheuk-Man Yu; Joseph J.Y. Sung; Hui Y. Lan

TGF-β/Smad3 signaling promotes fibrosis, but the development of therapeutic interventions involving this pathway will require the identification and ultimate targeting of downstream fibrosis-specific genes. In this study, using a microRNA microarray and real-time PCR, wild-type mice had reduced expression of miR-29 along with the development of progressive renal fibrosis in obstructive nephropathy. In contrast, Smad3 knockout mice had increased expression of miR-29 along with the absence of renal fibrosis in the same model of obstruction. In cultured fibroblasts and tubular epithelial cells, Smad3 mediated TGF-β(1)-induced downregulation of miR-29 by binding to the promoter of miR-29. Furthermore, miR-29 acted as a downstream inhibitor and therapeutic microRNA for TGF-β/Smad3-mediated fibrosis. In vitro, overexpression of miR-29b inhibited, but knockdown of miR-29 enhanced, TGF-β(1)-induced expression of collagens I and III by renal tubular cells. Ultrasound-mediated gene delivery of miR-29b either before or after established obstructive nephropathy blocked progressive renal fibrosis. In conclusion, miR-29 is a downstream inhibitor of TGF-β/Smad3-mediated fibrosis and may have therapeutic potential for diseases involving fibrosis.


Journal of The American Society of Nephrology | 2003

Inhibition of Renal Fibrosis by Gene Transfer of Inducible Smad7 Using Ultrasound-Microbubble System in Rat UUO Model

Hui Y. Lan; Wei Mu; Naruya Tomita; Xiao R. Huang; Jin H. Li; Hong-Jian Zhu; Ryuichi Morishita; Richard J. Johnson

TGF-beta is a key mediator in renal fibrosis. Kidney-targeted gene therapy with anti-TGF-beta strategies is expected to have therapeutic potential, but this has been hampered by concerns over the safety and practicability of viral vectors and the inefficiency of nonviral transfection techniques. The present study explored the potential role of TGF-beta/Smad signaling in renal fibrosis in vivo and developed a safe and effective gene therapy to specifically block TGF-beta signaling and renal fibrosis in a rat unilateral ureteral obstruction (UUO) model by transferring a doxycycline-regulated Smad7 gene or control empty vectors using an ultrasound-microbubble (Optison)-mediated system. The Smad7 transgene expression was tightly controlled by addition of doxycycline in the daily drinking water. Groups of six rats were sacrificed at day 7, and the transfection rate, Smad7 transgene expression, and tubulointerstitial fibrosis including alpha-smooth muscle actin and collagen matrix mRNA and protein expression were determined. Compared with the non-ultrasound treatment, the combination of ultrasound with Optison largely increased the transfection rate of FITC-ODN and Smad7 transgene expression up to a 1000-fold, and this was found in all kidney tissues. Compared with normal rats, Smad7 expression within the UUO kidney was significantly reduced, and this was associated with up to a sixfold increase in Smad2 and Smad3 activation and severe tubulointerstitial fibrosis. In contrast, treatment with inducible Smad7 resulted in a fivefold increase in Smad7 expression with complete inhibition of Smad2 and Smad3 activation and tubulointerstitial fibrosis in terms of tubulointerstitial myofibroblast accumulation (85% downward arrow ) and collagen I and III mRNA and protein expression (60 to 70% downward arrow ). In conclusion, the ultrasound-mediated inducible Smad7 gene transfer is a safe, effective, and controllable gene therapy. TGF-beta-mediated renal fibrosis is regulated positively by Smad2/3, but negatively by Smad7. Target blockade of TGF-beta/Smad signaling by expression of Smad7 may provide a new therapeutic potential for renal fibrosis.


Nephrology | 2005

Transforming growth factor-beta and Smad signalling in kidney diseases.

Wansheng Wang; Vijay Koka; Hui Y. Lan

SUMMARY:  Extensive studies have demonstrated that transforming growth factor‐beta (TGF‐β) plays an important role in the progression of renal diseases. TGF‐β exerts its biological functions mainly through its downstream signalling molecules, Smad2 and Smad3. It is now clear that Smad3 is critical for TGF‐βs pro‐fibrotic effect, whereas the functions of Smad2 in fibrosis in response to TGF‐β still need to be determined. Our recent studies have demonstrated that Smad signalling is also a critical pathway for renal fibrosis induced by other pro‐fibrotic factors, such as angiotensin II and advanced glycation end products (AGE). These pro‐fibrotic factors can activate Smads directly and independently of TGF‐β. They can also cause renal fibrosis via the ERK/p38 MAP kinase–Smad signalling cross‐talk pathway. In contrast, blockade of Smad2/3 activation by overexpression of an inhibitory Smad7 prevents collagen matrix production induced by TGF‐β, angiotensin II, high glucose and AGE and attenuates renal fibrosis in various animal models including rat obstructive kidney, remnant kidney and diabetic kidney diseases. Results from these studies indicate that Smad signalling is a key and final common pathway of renal fibrosis. In addition, TGF‐β has anti‐inflammatory and immune‐regulatory properties. Our most recent studies demonstrated that TGF‐β transgenic mice are protected against renal inflammation in mouse obstructive and diabetic models. Upregulation of renal Smad7, thereby blocking NF.κB activation via induction of IκBα, is a central mechanism by which TGF‐β inhibits renal inflammation. In conclusion, TGF‐β signals through Smad2/3 to mediate renal fibrosis, whereas induction of Smad7 inhibits renal fibrosis and inflammation. Thus, targeting Smad signalling by overexpression of Smad7 may have great therapeutic potential for kidney diseases.


Nature Reviews Nephrology | 2016

TGF-[beta]: the master regulator of fibrosis

Xiao-Ming Meng; David J. Nikolic-Paterson; Hui Y. Lan

Transforming growth factor-β (TGF-β) is the primary factor that drives fibrosis in most, if not all, forms of chronic kidney disease (CKD). Inhibition of the TGF-β isoform, TGF-β1, or its downstream signalling pathways substantially limits renal fibrosis in a wide range of disease models whereas overexpression of TGF-β1 induces renal fibrosis. TGF-β1 can induce renal fibrosis via activation of both canonical (Smad-based) and non-canonical (non-Smad-based) signalling pathways, which result in activation of myofibroblasts, excessive production of extracellular matrix (ECM) and inhibition of ECM degradation. The role of Smad proteins in the regulation of fibrosis is complex, with competing profibrotic and antifibrotic actions (including in the regulation of mesenchymal transitioning), and with complex interplay between TGF-β/Smads and other signalling pathways. Studies over the past 5 years have identified additional mechanisms that regulate the action of TGF-β1/Smad signalling in fibrosis, including short and long noncoding RNA molecules and epigenetic modifications of DNA and histone proteins. Although direct targeting of TGF-β1 is unlikely to yield a viable antifibrotic therapy due to the involvement of TGF-β1 in other processes, greater understanding of the various pathways by which TGF-β1 controls fibrosis has identified alternative targets for the development of novel therapeutics to halt this most damaging process in CKD.


Journal of Clinical Investigation | 1999

Monocyte chemoattractant protein-1 promotes macrophage-mediated tubular injury, but not glomerular injury, in nephrotoxic serum nephritis

Gregory H. Tesch; Andreas Schwarting; Koji Kinoshita; Hui Y. Lan; Barrett J. Rollins; Vicki Rubin Kelley

Monocyte chemoattractant protein-1 (MCP-1) is upregulated in renal parenchymal cells during kidney disease. To investigate whether MCP-1 promotes tubular and/or glomerular injury, we induced nephrotoxic serum nephritis (NSN) in MCP-1 genetically deficient mice. Mice were analyzed when tubules and glomeruli were severely damaged in the MCP-1-intact strain (day 7). MCP-1 transcripts increased fivefold in MCP-1-intact mice. MCP-1 was predominantly localized within cortical tubules (90%), and most cortical tubules were damaged, whereas few glomerular cells expressed MCP-1 (10%). By comparison, there was a marked reduction (>40%) in tubular injury in MCP-1-deficient mice (histopathology, apoptosis). MCP-1-deficient mice were not protected from glomerular injury (histopathology, proteinuria, macrophage influx). Macrophage accumulation increased adjacent to tubules in MCP-1-intact mice compared with MCP-1-deficient mice (70%, P < 0.005), indicating that macrophages recruited by MCP-1 induce tubular epithelial cell (TEC) damage. Lipopolysaccharide-activated bone marrow macrophages released molecules that induced TEC death that was not dependent on MCP-1 expression by macrophages or TEC. In conclusion, MCP-1 is predominantly expressed by TEC and not glomeruli, promotes TEC and not glomerular damage, and increases activated macrophages adjacent to TEC that damage TEC during NSN. Therefore, we suggest that blockage of TEC MCP-1 expression is a therapeutic strategy for some forms of kidney disease.


Journal of The American Society of Nephrology | 2010

miR-192 Mediates TGF-β/Smad3-Driven Renal Fibrosis

Arthur C.K. Chung; Xiao R. Huang; Xiao-Ming Meng; Hui Y. Lan

TGF-beta/Smad3 promotes renal fibrosis, but the mechanisms that regulate profibrotic genes remain unclear. We hypothesized that miR-192, a microRNA expressed in the kidney may mediate renal fibrosis in a Smad3-dependent manner. Microarray and real-time PCR demonstrated a tight association between upregulation of miR-192 in the fibrotic kidney and activation of TGF-beta/Smad signaling. Deletion of Smad7 promoted miR-192 expression and enhanced Smad signaling and fibrosis in obstructive kidney disease. In contrast, overexpression of Smad7 to block TGF-beta/Smad signaling inhibited miR-192 expression and renal fibrosis in the rat 5/6 nephrectomy model; in vitro, overexpression of Smad7 in tubular epithelial cells abolished TGF-beta1-induced miR-192 expression. Furthermore, Smad3 but not Smad2 mediated TGF-beta1-induced miR-192 expression by binding to the miR-192 promoter. Last, overexpression of a miR-192 mimic promoted and addition of a miR-192 inhibitor blocked TGF-beta1-induced collagen matrix expression. Taken together, miR-192 may be a critical downstream mediator of TGF-beta/Smad3 signaling in the development of renal fibrosis.


Journal of The American Society of Nephrology | 2003

Chymase Is Upregulated in Diabetic Nephropathy: Implications for an Alternative Pathway of Angiotensin II–Mediated Diabetic Renal and Vascular Disease

Xiao R. Huang; Wei Y. Chen; Luan D. Truong; Hui Y. Lan

Angiotensin II (AngII) has been shown to play a critical role in diabetic nephropathy and vasculopathy. Although it is well recognized that an angiotensin-converting enzyme (ACE)-dependent AngII-generating system is a major source of intrarenal AngII production, it is here reported that the chymase-dependent AngII-generating system is upregulated in the human diabetic kidney. This becomes particularly strong in those with hypertension. In the normal kidney, while ACE was constitutively expressed by most kidney cells, chymase was weakly expressed by mesangial cells (MC) and vascular smooth muscle cells (VSMC) only. In the diabetic kidney, while ACE expression was significantly upregulated (1 to 3-fold) by tubular epithelial cells (TEC) and infiltrating mononuclear cells, there was also markedly increased chymase expression (10 to 15-fold) by both MC and VSMC, with strong deposition in the collagen-rich extracellular matrix including both diffuse and nodular glomerulosclerosis, tubulointerstitial fibrosis, and vascular sclerosis. Interestingly, while ACE expression showed no difference in patients with or without hypertension, upregulation of chymase in hypertensive patients was much stronger than that seen in those without hypertension (4 to 7-fold, P < 0.001). Correlation analysis showed that, in contrast to the ACE expression, upregulation of chymase correlated significantly with the increase in BP and the severity of collagen matrix deposition within the glomerulus, tubulointerstitium, and arterial walls (all with P < 0.001). In conclusion, the present study demonstrates that chymase, as an alternative AngII-generating enzyme, is markedly upregulated in the diabetic kidney and may be associated with the development of diabetic/hypertensive nephropathy. In addition, differential expression of ACE and chymase in the diabetic kidney indicates that both ACE and chymase may be of equal importance for AngII-mediated diabetic nephropathy and vascular disease. Results from this study suggest that blockade of both AngII-generating pathways may provide additional beneficial effect on diabetic nephropathy.


The FASEB Journal | 2003

Advanced glycation end products activate Smad signaling via TGF-β-dependent and -independent mechanisms: implications for diabetic renal and vascular disease

Jin H. Li; Xiao R. Huang; Hong-Jian Zhu; Matthew D. Oldfield; Mark E. Cooper; Luan D. Truong; Richard J. Johnson; Hui Y. Lan

While it is thought that advanced glycation end products (AGEs) act by stimulating transforming growth factor (TGF)‐β to mediate diabetic injury, we report that AGEs can activate TGF‐β signaling, Smads, and mediate diabetic scarring directly and independently of TGF‐β. AGEs activate Smad2/3 in renal and vascular cells at 5 min, peaking over 15–30 min before TGF‐β synthesis at 24 h and occurs in TGF‐β receptor I and II mutant cells. This is mediated by RAGE and ERK/p38 mitogen‐activated protein kinases (MAPKs). In addition, AGEs also activate Smads at 24 h via the classic TGF‐β‐dependent pathway. A substantial inhibition of AGE‐ induced Smad activation and collagen synthesis by ERK/p38 MAPK inhibitors, but not by TGF‐β blockade, suggests that the MAPK‐Smad signaling crosstalk pathway is a key mechanism in diabetic scarring. Prevention of AGE‐induced Smad activation and collagen synthesis by overexpression of Smad7 indicates that Smad signaling may play a critical role in diabetic complications. This is further supported by the findings that activation of Smad2/3 in human diabetic nephropathy and vasculopathy is associated with local deposition of AGEs and up‐ regulation of RAGE. Thus, AGEs act by activating Smad signaling to mediate diabetic complications via both TGF‐β‐dependent and ‐independent pathways, shedding new light on the pathogenesis of diabetic organ injury.

Collaboration


Dive into the Hui Y. Lan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiao R. Huang

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Wei Mu

University of Florida

View shared research outputs
Top Co-Authors

Avatar

Arthur C.K. Chung

Hong Kong Baptist University

View shared research outputs
Top Co-Authors

Avatar

Xiao-Ru Huang

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Xiao Ru Huang

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Richard J. Johnson

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiao-Ming Meng

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge