Network


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

Hotspot


Dive into the research topics where Shuei-Liong Lin is active.

Publication


Featured researches published by Shuei-Liong Lin.


American Journal of Pathology | 2010

Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis.

Benjamin D. Humphreys; Shuei-Liong Lin; Akio Kobayashi; Thomas E. Hudson; Brian T. Nowlin; Joseph V. Bonventre; M. Todd Valerius; Andrew P. McMahon; Jeremy S. Duffield

Understanding the origin of myofibroblasts in kidney is of great interest because these cells are responsible for scar formation in fibrotic kidney disease. Recent studies suggest epithelial cells are an important source of myofibroblasts through a process described as the epithelial-to-mesenchymal transition; however, confirmatory studies in vivo are lacking. To quantitatively assess the contribution of renal epithelial cells to myofibroblasts, we used Cre/Lox techniques to genetically label and fate map renal epithelia in models of kidney fibrosis. Genetically labeled primary proximal epithelial cells cultured in vitro from these mice readily induce markers of myofibroblasts after transforming growth factor beta(1) treatment. However, using either red fluorescent protein or beta-galactosidase as fate markers, we found no evidence that epithelial cells migrate outside of the tubular basement membrane and differentiate into interstitial myofibroblasts in vivo. Thus, although renal epithelial cells can acquire mesenchymal markers in vitro, they do not directly contribute to interstitial myofibroblast cells in vivo. Lineage analysis shows that during nephrogenesis, FoxD1-positive((+)) mesenchymal cells give rise to adult CD73(+), platelet derived growth factor receptor beta(+), smooth muscle actin-negative interstitial pericytes, and these FoxD1-derivative interstitial cells expand and differentiate into smooth muscle actin(+) myofibroblasts during fibrosis, accounting for a large majority of myofibroblasts. These data indicate that therapeutic strategies directly targeting pericyte differentiation in vivo may productively impact fibrotic kidney disease.


American Journal of Pathology | 2008

Pericytes and Perivascular Fibroblasts Are the Primary Source of Collagen-Producing Cells in Obstructive Fibrosis of the Kidney

Shuei-Liong Lin; Tatiana Kisseleva; David A. Brenner; Jeremy S. Duffield

Understanding the origin of scar-producing myofibroblasts is vital in discerning the mechanisms by which fibrosis develops in response to inflammatory injury. Using a transgenic reporter mouse model expressing enhanced green fluorescent protein (GFP) under the regulation of the collagen type I, alpha 1 (coll1a1) promoter and enhancers, we examined the origins of coll1a1-producing cells in the kidney. Here we show that in normal kidney, both podocytes and pericytes generate coll1a1 transcripts as detected by enhanced GFP, and that in fibrotic kidney, coll1a1-GFP expression accurately identifies myofibroblasts. To determine the contribution of circulating immune cells directly to scar production, wild-type mice, chimeric with bone marrow from coll-GFP mice, underwent ureteral obstruction to induce fibrosis. Histological examination of kidneys from these mice showed recruitment of small numbers of fibrocytes to the fibrotic kidney, but these fibrocytes made no significant contribution to interstitial fibrosis. Instead, using kinetic modeling and time course microscopy, we identified coll1a1-GFP-expressing pericytes as the major source of interstitial myofibroblasts in the fibrotic kidney. Our studies suggest that either vascular injury or vascular factors are the most likely triggers for pericyte migration and differentiation into myofibroblasts. Therefore, our results serve to refocus fibrosis research to injury of the vasculature rather than injury to the epithelium.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Macrophage Wnt7b is critical for kidney repair and regeneration

Shuei-Liong Lin; Bing Li; Sujata Rao; Eun Jin Yeo; Thomas E. Hudson; Brian T. Nowlin; Huaying Pei; Lijun Chen; Jie Zheng; Thomas J. Carroll; Jeffrey W. Pollard; Andrew P. McMahon; Richard A. Lang; Jeremy S. Duffield

Macrophages are required for tissue homeostasis through their role in regulation of the immune response and the resolution of injury. Here we show, using the kidney as a model, that the Wnt pathway ligand Wnt7b is produced by macrophages to stimulate repair and regeneration. When macrophages are inducibly ablated from the injured kidney, the canonical Wnt pathway response in kidney epithelial cells is reduced. Furthermore, when Wnt7b is somatically deleted in macrophages, repair of injury is greatly diminished. Finally, injection of the Wnt pathway regulator Dkk2 enhances the repair process and suggests a therapeutic option. Because Wnt7b is known to stimulate epithelial responses during kidney development, these findings suggest that macrophages are able to rapidly invade an injured tissue and reestablish a developmental program that is beneficial for repair and regeneration.


Journal of Immunology | 2009

Bone Marrow Ly6Chigh Monocytes Are Selectively Recruited to Injured Kidney and Differentiate into Functionally Distinct Populations

Shuei-Liong Lin; Ana P. Castano; Brian T. Nowlin; Mark L. Lupher; Jeremy S. Duffield

Roles for monocyte/macrophages (Mφ) in directing the development of tissue fibrosis are increasingly recognized. Macrophages form a heterogeneous group of inflammatory leukocytes, and the mechanisms by which they acquire heterogeneity and its functional significance are unclear. We used the unilateral ureteral obstruction model of progressive kidney fibrosis to explore macrophage heterogeneity and function further. Unilateral ureteral obstruction kidney Mφs form three distinct subpopulations defined by the marker Ly6C, all of which are derived from a single Ly6Chigh bone marrow monocyte population selectively recruited to the kidney. Conditional ablation of these Mφs in vivo in CD11b-DTR mice is potently antifibrotic. The mRNA transcription profile of these populations is consistent with differential functional roles for each subpopulation, with Ly6Clow macrophages transcribing genes consistent with selective profibrotic or M2-type function. Furthermore, bone marrow chimerism studies indicate that although resident kidney macrophages proliferate markedly to comprise up to 40% of the inflammatory macrophage population, they do not contribute to fibrosis. Our data identify Ly6C as a marker of functionally discrete tissue macrophage subsets and support a model of selective recruitment of Ly6Chigh bone marrow monocytes to the kidney that differentiate into three populations of kidney macrophages, including a profibrotic Ly6Clow population.


American Journal of Pathology | 2011

Targeting Endothelium-Pericyte Cross Talk by Inhibiting VEGF Receptor Signaling Attenuates Kidney Microvascular Rarefaction and Fibrosis

Shuei-Liong Lin; Fan Chi Chang; Claudia Schrimpf; Yi Ting Chen; Ching Fang Wu; Vin-Cent Wu; Wen-Chih Chiang; Frank Kuhnert; Calvin J. Kuo; Yung-Ming Chen; Kwan Dun Wu; Tun-Jun Tsai; Jeremy S. Duffield

Microvascular pericytes and perivascular fibroblasts have recently been identified as the source of scar-producing myofibroblasts that appear after injury of the kidney. We show that cross talk between pericytes and endothelial cells concomitantly dictates development of fibrosis and loss of microvasculature after injury. When either platelet-derived growth factor receptor (R)-β signaling in pericytes or vascular endothelial growth factor (VEGF)R2 signaling in endothelial cells was blocked by circulating soluble receptor ectodomains, both fibrosis and capillary rarefaction were markedly attenuated during progressive kidney injury. Blockade of either receptor-mediated signaling pathway prevented pericyte differentiation and proliferation, but VEGFR2 blockade also attenuated recruitment of inflammatory macrophages throughout disease progression. Whereas injury down-regulated angiogenic VEGF164, the dys-angiogenic isomers VEGF120 and VEGF188 were up-regulated, suggesting that pericyte-myofibroblast differentiation triggers endothelial loss by a switch in secretion of VEGF isomers. These findings link fibrogenesis inextricably with microvascular rarefaction for the first time, add new significance to fibrogenesis, and identify novel therapeutic targets.


Kidney International | 2011

Platelet-derived growth factor receptor signaling activates pericyte-myofibroblast transition in obstructive and post-ischemic kidney fibrosis

Yi Ting Chen; Fan Chi Chang; Ching Fang Wu; Yu Hsiang Chou; Huan Lun Hsu; Wen-Chih Chiang; Juqun Shen; Yung-Ming Chen; Kwan Dun Wu; Tun-Jun Tsai; Jeremy S. Duffield; Shuei-Liong Lin

Pericytes are the major source of scar-producing myofibroblasts following kidney injury; however, the mechanisms of this transition are unclear. To clarify this, we examined Collagen 1 (α1)-green fluorescent protein (GFP) reporter mice (pericytes and myofibroblasts express GFP) following ureteral obstruction or ischemia-reperfusion injury and focused on the role of platelet-derived growth factor (PDGF)-receptor (PDGFR) signaling in these two different injury models. Pericyte proliferation was noted after injury with reactivation of α-smooth muscle actin expression, a marker of the myofibroblast phenotype. PDGF expression increased in injured tubules, endothelium, and macrophages after injury, whereas PDGFR subunits α and β were expressed exclusively in interstitial GFP-labeled pericytes and myofibroblasts. When PDGFRα or PDGFRβ activation was inhibited by receptor-specific antibody following injury, proliferation and differentiation of pericytes decreased. The antibodies also blunted the injury-induced transcription of PDGF, transforming growth factor β1, and chemokine CCL2. They also reduced macrophage infiltration and fibrosis. Imatinib, a PDGFR tyrosine kinase inhibitor, attenuated pericyte proliferation and kidney fibrosis in both fibrogenic models. Thus, PDGFR signaling is involved in pericyte activation, proliferation, and differentiation into myofibroblasts during progressive kidney injury. Hence, pericytes may be a novel target to prevent kidney fibrosis by means of PDGFR signaling blockade.


Science Translational Medicine | 2009

Serum Amyloid P Inhibits Fibrosis Through FcγR-Dependent Monocyte-Macrophage Regulation in Vivo

Ana P. Castano; Shuei-Liong Lin; Teresa Surowy; Brian T. Nowlin; Swathi A. Turlapati; Tejas V. Patel; Ajay K. Singh; Shawn Li; Mark L. Lupher; Jeremy S. Duffield

The fibrosis that occus in kidney after tissue injury can be inhibited by serum amyloid protein, which stimulates the cytokine interleukin 10. Suppressing Fibrosis When it comes to repairing damaged tissues or organs, there can be too much of a good thing. Although sometimes the body’s natural repair processes lead to complete restoration of normal tissue after an injury, at other times these processes continue unrestrained, leading to the replacement of normal tissue with fibrous scars—potentially causing organ failure and death. Such fibrosis is a common underlying feature of a wide variety of diseases, including cirrhosis of the liver, cardiovascular disease, rheumatoid arthritis, and progressive kidney disease. Even though fibroproliferative diseases are a leading cause of death in Western societies, there are currently no therapies that target fibrosis directly. Now, Duffield and colleagues describe a potential antifibrotic therapy for fibrosis of the kidney. The body’s repair processes are triggered by a range of harmful events, such as physical trauma, infections, toxin-induced damage, or an inadequate supply of blood. Tissue repair progresses through several stages, including an inflammatory response with an influx of white blood cells like monocytes, apoptosis and necrosis, the activation of wound-healing myofibroblasts, and the deposition and remodeling of extracellular matrix. When this matrix expands in an apparently uncontrolled manner, fibrous scars—consisting primarily of collagen—are formed. Compounds that inhibit the formation of these scars would be welcome tools for treating fibroproliferative diseases. Duffield’s group sought to determine whether serum amyloid P (SAP)—a protein previously shown to inhibit fibrosis in the lung and heart—might also suppress fibrosis in the kidney. SAP can bind ligands on the surface of apoptotic cells as well as Fcγ receptors on certain immune cells, and binding of SAP to the apoptotic cell ligands induces Fcγ receptor–dependent phagocytosis. These observations suggest that SAP might localize to sites of injury, aiding in the removal of wounded tissue. Duffield’s team showed that administration of human SAP to mice with experimentally induced kidney injury markedly decreased fibrosis. The SAP was recruited to the injured kidney where it was associated with dead cells. In the lung and heart, SAP appears to act by inhibiting fibrocytes, which directly produce collagen matrix. In the kidney, however, fibrocytes do not participate in fibrosis. Instead, the researchers determined, kidney fibrosis in mice depends on inflammatory monocytes and macrophages. SAP binds to and suppresses these cells, an effect that depends on expression of the anti-inflammatory cytokine IL-10 and regulated binding to Fcγ receptors, which are expressed on monocytes and macrophages. Given that a form of human SAP is now being tested in a phase 1 clinical trial in healthy volunteers, we may soon know whether it suppresses fibrotic disease in humans. New therapies that target chronic inflammation with fibrosis are urgently required. Increasing evidence points to innate activation of inflammatory cells in driving chronic organ fibrosis. Serum amyloid P is a naturally circulating soluble pattern recognition receptor, a member of the family of pentraxin proteins. It links danger-associated molecular pattern recognition to Fcγ receptor–mediated phagocytosis. Here we show that fibrosis progression in the mouse kidney is significantly inhibited by therapeutic administration of human serum amyloid P, regulated by activating Fcγ receptors, and dependent on inflammatory monocytes and macrophages, but not fibrocytes. Human serum amyloid P–mediated inhibition of mouse kidney fibrosis correlated with specific binding of human serum amyloid P to cell debris and with subsequent suppression of inflammatory monocytes and kidney macrophages in vitro and in vivo, and was dependent on regulated binding to activating Fcγ receptors and interleukin-10 expression. These studies uncover previously unidentified roles for Fcγ receptors in sterile inflammation and highlight serum amyloid P as a potential antifibrotic therapy through local generation of interleukin-10.


Journal of The American Society of Nephrology | 2014

Long-Term Risk of Coronary Events after AKI

Vin-Cent Wu; Che-Hsiung Wu; Tao-Min Huang; Cheng-Yi Wang; Chun-Fu Lai; Chih-Chung Shiao; Chia-Hsui Chang; Shuei-Liong Lin; Yen-Yuan Chen; Yung-Ming Chen; Tzong-Shinn Chu; Wen-Chih Chiang; Kwan-Dun Wu; Pi-Ru Tsai; Likwang Chen; Wen-Je Ko

The incidence rate of AKI in hospitalized patients is increasing. However, relatively little attention has been paid to the association of AKI with long-term risk of adverse coronary events. Our study investigated hospitalized patients who recovered from de novo dialysis-requiring AKI between 1999 and 2008 using patient data collected from inpatient claims from Taiwan National Health Insurance. We used Cox regression with time-varying covariates to adjust for subsequent CKD and ESRD after discharge. Results were further validated by analysis of a prospectively constructed database. Among 17,106 acute dialysis patients who were discharged, 4869 patients recovered from dialysis-requiring AKI (AKI recovery group) and were matched with 4869 patients without AKI (non-AKI group). The incidence rates of coronary events were 19.8 and 10.3 per 1000 person-years in the AKI recovery and non-AKI groups, respectively. AKI recovery associated with higher risk of coronary events (hazard ratio [HR], 1.67; 95% confidence interval [95% CI], 1.36 to 2.04) and all-cause mortality (HR, 1.67; 95% CI, 1.57 to 1.79) independent of the effects of subsequent progression to CKD and ESRD. The risk levels of de novo coronary events after hospital discharge were similar in patients with diabetes alone and patients with AKI alone (P=0.23). Our results reveal that AKI with recovery associated with higher long-term risks of coronary events and death in this cohort, suggesting that AKI may identify patients with high risk of future coronary events. Enhanced postdischarge follow-up of renal function of patients who have recovered from temporary dialysis may be warranted.


Journal of The American Society of Nephrology | 2012

Pericyte TIMP3 and ADAMTS1 Modulate Vascular Stability after Kidney Injury

Claudia Schrimpf; Cuiyan Xin; Gabriella Campanholle; Sean E. Gill; William B. Stallcup; Shuei-Liong Lin; George E. Davis; Sina A. Gharib; Benjamin D. Humphreys; Jeremy S. Duffield

Kidney pericytes are progenitors of scar-forming interstitial myofibroblasts that appear after injury. The function of kidney pericytes as microvascular cells and how these cells detach from peritubular capillaries and migrate to the interstitial space, however, are poorly understood. Here, we used an unbiased approach to identify genes in kidney pericytes relevant to detachment and differentiation in response to injury in vivo, with a particular focus on genes regulating proteolytic activity and angiogenesis. Kidney pericytes rapidly activated expression of a disintegrin and metalloprotease with thrombospondin motifs-1 (ADAMTS1) and downregulated its inhibitor, tissue inhibitor of metalloproteinase 3 (TIMP3) in response to injury. Similarly to brain pericytes, kidney pericytes bound to and stabilized capillary tube networks in three-dimensional gels and inhibited metalloproteolytic activity and angiogenic signaling in endothelial cells. In contrast, myofibroblasts did not have these vascular stabilizing functions despite their derivation from kidney pericytes. Pericyte-derived TIMP3 stabilized and ADAMTS1 destabilized the capillary tubular networks. Furthermore, mice deficient in Timp3 had a spontaneous microvascular phenotype in the kidney resulting from overactivated pericytes and were more susceptible to injury-stimulated microvascular rarefaction with an exuberant fibrotic response. Taken together, these data support functions for kidney pericytes in microvascular stability, highlight central roles for regulators of extracellular proteolytic activity in capillary homoeostasis, and identify ADAMTS1 as a marker of activation of kidney pericytes.


Proceedings of the National Academy of Sciences of the United States of America | 2013

LRP-6 is a coreceptor for multiple fibrogenic signaling pathways in pericytes and myofibroblasts that are inhibited by DKK-1

Shuyu Ren; Bryce G. Johnson; Yujiro Kida; Colin Ip; Kathryn C. Davidson; Shuei-Liong Lin; Akio Kobayashi; Richard A. Lang; Anna-Katerina Hadjantonakis; Randall T. Moon; Jeremy S. Duffield

Fibrosis of vital organs is a major public health problem with limited therapeutic options. Mesenchymal cells including microvascular mural cells (pericytes) are major progenitors of scar-forming myofibroblasts in kidney and other organs. Here we show pericytes in healthy kidneys have active WNT/β-catenin signaling responses that are markedly up-regulated following kidney injury. Dickkopf-related protein 1 (DKK-1), a ligand for the WNT coreceptors low-density lipoprotein receptor-related proteins 5 and 6 (LRP-5 and LRP-6) and an inhibitor of WNT/β-catenin signaling, effectively inhibits pericyte activation, detachment, and transition to myofibroblasts in vivo in response to kidney injury, resulting in attenuated fibrogenesis, capillary rarefaction, and inflammation. DKK-1 blocks activation and proliferation of established myofibroblasts in vitro and blocks pericyte proliferation to PDGF, pericyte migration, gene activation, and cytoskeletal reorganization to TGF-β or connective tissue growth factor. These effects are largely independent of inhibition of downstream β-catenin signaling. DKK-1 acts predominantly by inhibiting PDGF-, TGF-β–, and connective tissue growth factor-activated MAPK and JNK signaling cascades, acting via LRP-6 with associated WNT ligand. Biochemically, LRP-6 interacts closely with PDGF receptor β and TGF-β receptor 1 at the cell membrane, suggesting that it may have roles in pathways other than WNT/β-catenin. In summary, DKK-1 blocks many of the changes in pericytes required for myofibroblast transition and attenuates established myofibroblast proliferation/activation by mechanisms dependent on LRP-6 and WNT ligands but not the downstream β-catenin pathway.

Collaboration


Dive into the Shuei-Liong Lin's collaboration.

Top Co-Authors

Avatar

Yung-Ming Chen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Kwan-Dun Wu

Fu Jen Catholic University

View shared research outputs
Top Co-Authors

Avatar

Wen-Chih Chiang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Tun-Jun Tsai

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Vin-Cent Wu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Tzong-Shinn Chu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chun-Fu Lai

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yu-Feng Lin

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Fan Chi Chang

National Taiwan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge