Network


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

Hotspot


Dive into the research topics where Neal Paragas is active.

Publication


Featured researches published by Neal Paragas.


Nature Medicine | 2011

The Ngal reporter mouse detects the response of the kidney to injury in real time

Neal Paragas; Andong Qiu; Qing-Yin Zhang; Benjamin Samstein; Shixian Deng; Kai M. Schmidt-Ott; Melanie Viltard; Wenqiang Yu; Catherine S. Forster; Gangli Gong; Yidong Liu; Ritwij Kulkarni; Kiyoshi Mori; Avtandil Kalandadze; Adam J. Ratner; Prasad Devarajan; Donald W. Landry; Chyuan-Sheng Lin; Jonathan Barasch

Many proteins have been proposed to act as surrogate markers of organ damage, yet for many candidates the essential biomarker characteristics that link the protein to the injured organ have not yet been described. We generated an Ngal reporter mouse by inserting a double-fusion reporter gene encoding luciferase-2 and mCherry (Luc2-mC) into the Ngal (Lcn2) locus. The Ngal-Luc2-mC reporter accurately recapitulated the endogenous message and illuminated injuries in vivo in real time. In the kidney, Ngal-Luc2-mC imaging showed a sensitive, rapid, dose-dependent, reversible, and organ- and cell-specific relationship with tubular stress, which correlated with the level of urinary Ngal (uNgal). Unexpectedly, specific cells of the distal nephron were the source of uNgal. Cells isolated from Ngal-Luc2-mC mice also revealed both the onset and the resolution of the injury, and the actions of NF-κB inhibitors and antibiotics during infection. Thus, imaging of Ngal-Luc2-mC mice and cells identified injurious and reparative agents that affect kidney damage.


Nature Chemical Biology | 2010

Iron Traffics in Circulation Bound to a Siderocalin (Ngal)-Catechol Complex

Guanhu Bao; Matthew S Clifton; Trisha M. Hoette; Kiyoshi Mori; Shixian Deng; Andong Qiu; Melanie Viltard; David Y. Williams; Neal Paragas; Thomas Leete; Ritwij Kulkarni; Xiangpo Li; Belinda T. Lee; Avtandil Kalandadze; Adam J. Ratner; Juan C. Pizarro; Kai M. Schmidt-Ott; Donald W. Landry; Kenneth N. Raymond; Roland K. Strong; Jonathan Barasch

The lipocalins are secreted proteins that bind small organic molecules. Scn-Ngal [known as Neutrophil Gelatinase Associated Lipocalin, Siderocalin, Lipocalin 2] sequesters bacterial iron chelators, called siderophores, and consequently blocks bacterial growth. However, Scn-Ngal is also prominently expressed in aseptic diseases, implying that it binds additional ligands and serves additional functions. Using chemical screens, crystallography, and fluorescence methods, we report that Scn-Ngal binds iron together with a small metabolic product called catechol. The formation of the complex blocked the reactivity of iron and permitted its transport once introduced into circulation in vivo. Scn-Ngal then recycled its iron in endosomes by a pH sensitive mechanism. Since catechols derive from bacterial and mammalian metabolism of dietary compounds, the Scn-Ngal:catechol:iron complex represents an unforeseen microbial-host interaction, which mimics Scn-Ngal:siderophore interactions, but instead traffics iron in aseptic tissues. These results identify an endogenous siderophore, which may link the disparate roles of Scn-Ngal in different diseases.


Current Opinion in Nephrology and Hypertension | 2006

Neutrophil gelatinase-associated lipocalin-mediated iron traffic in kidney epithelia

Kai M. Schmidt-Ott; Kiyoshi Mori; Avtandil Kalandadze; Jau-Yi Li; Neal Paragas; Thomas Nicholas; Prasad Devarajan; Jonathan Barasch

Purpose of reviewNeutrophil gelatinase-associated lipocalin (NGAL) is a member of the lipocalin superfamily of carrier proteins. NGAL is the first known mammalian protein which specifically binds organic molecules called siderophores, which are high-affinity iron chelators. Here, we review the expression, siderophore-dependent biological activities and clinical significance of NGAL in epithelial development and in kidney disease. Recent findingsNGAL expression is rapidly induced in the nephron in response to renal epithelial injury. This has led to the establishment of NGAL assays that detect renal damage in the human. Additionally, only when complexed with siderophore and iron as a trimer, NGAL induces mesenchymal–epithelial transition (or nephron formation) in embryonic kidney in vitro and protects adult kidney from ischemia–reperfusion injury in vivo. While the structure of the NGAL: siderophore: iron complex has thus far only been solved for bacterially synthesized siderophores, new evidence suggests the presence of mammalian siderophore-like molecules. SummaryNGAL is rapidly and massively induced in renal epithelial injury and NGAL: siderophore: iron complexes may comprise a physiological renoprotective mechanism. The data have implications for the diagnosis and treatment of acute renal injury.


Developmental Cell | 2009

Scara5 is a ferritin receptor mediating non-transferrin iron delivery.

Jau Yi Li; Neal Paragas; Renée M. Ned; Andong Qiu; Melanie Viltard; Thomas Leete; Ian R. Drexler; Xia Chen; Simone Sanna-Cherchi; Farah Mohammed; David Y. Williams; Chyuan Sheng Lin; Kai M. Schmidt-Ott; Nancy C. Andrews; Jonathan Barasch

Developing organs require iron for a myriad of functions, but embryos deleted of the major adult transport proteins, transferrin or its receptor transferrin receptor1 (TfR1(-/-)), still initiate organogenesis, suggesting that non-transferrin pathways are important. To examine these pathways, we developed chimeras composed of fluorescence-tagged TfR1(-/-) cells and untagged wild-type cells. In the kidney, TfR1(-/-) cells populated capsule and stroma, mesenchyme and nephron, but were underrepresented in ureteric bud tips. Consistently, TfR1 provided transferrin to the ureteric bud, but not to the capsule or the stroma. Instead of transferrin, we found that the capsule internalized ferritin. Since the capsule expressed a novel receptor called Scara5, we tested its role in ferritin uptake and found that Scara5 bound serum ferritin and then stimulated its endocytosis from the cell surface with consequent iron delivery. These data implicate cell type-specific mechanisms of iron traffic in organogenesis, which alternatively utilize transferrin or non-transferrin iron delivery pathways.


Journal of The American Society of Nephrology | 2005

Novel Regulators of Kidney Development from the Tips of the Ureteric Bud

Kai M. Schmidt-Ott; Jun Yang; Xia Chen; Howard Wang; Neal Paragas; Kiyoshi Mori; Jau-Yi Li; Benson Lu; Frank Costantini; Mario Schiffer; Erwin P. Bottinger; Jonathan Barasch

Mammalian nephrogenesis depends on the interaction between the ureteric bud and the metanephric mesenchyme. As the ureteric bud undergoes branching and segmentation, the stalks differentiate into the collecting system of the mature kidney, while the tip cells interact with the adjacent cells of the metanephric mesenchyme, inducing their conversion into nephrons. This induction is mediated by secreted factors. For identifying novel mediators, the tips of the ureteric tree were isolated and microarray analyses were performed using manually refined, multistep gene ontology annotations. For identifying conserved factors, two databases were developed, one from mouse E12.5 and one from rat E13.5 ureteric buds. The overlap of mouse and rat data sets yielded 20 different transcripts that were enriched in the ureteric bud compared with metanephric mesenchyme and predicted to code for secreted proteins. Real-time reverse transcriptase-PCR and in situ hybridization confirmed these identifications. One of the genes that was highly specific to the ureteric bud tip was cytokine-like factor 1 (CLF-1). Recombinant CLF-1 in complex with its physiologic ligand, cardiotrophin-like cytokine (CLC), triggered phosphorylation of signal transducer and activator of transcription 3 in mesenchyme, a pathway characteristic of mesenchymal-to-epithelial conversion. Indeed, when applied to isolated rat metanephric mesenchyme, CLF-1/CLC (3 nM) induced mature nephron structures expressing glomerular and tubular markers. These results underline the power of this first comprehensive gene expression analysis of the ureteric bud tip to identify bioactive molecules.


Acta Physiologica | 2013

Neutrophil gelatinase‐associated lipocalin: pathophysiology and clinical applications

Eugenia Singer; Lajos Markó; Neal Paragas; Jonathan Barasch; Duska Dragun; Dominik N. Müller; Klemens Budde; Kai M. Schmidt-Ott

Neutrophil gelatinase‐associated lipocalin (NGAL), a 25 kDa protein produced by injured nephron epithelia, is one of the most promising new markers of renal epithelial injury. In contrast to serum creatinine and urinary output, which are the measures of kidney function, NGAL is specifically induced in the damaged nephron and then released into blood and urine, where it can be readily measured. Careful proof‐of‐concept studies using defined animal models have uncovered the sources and trafficking of NGAL in acute kidney injury (AKI) and have addressed the contributions of renal and non‐renal sources. Clinical studies indicate that NGAL, unlike creatinine, is a marker responsive to tissue stress and nephron injury, but less so to adaptive hemodynamic responses. In certain clinical settings, NGAL is an earlier marker compared with serum creatinine. In addition, clinical studies have shown that NGAL is a powerful predictor of poor clinical outcomes, which can be used to risk stratify patients when combined with serum creatinine. NGAL has important limitations, including its responsiveness in systemic inflammation, which is partially uncoupled from its response to kidney injury and which needs to be considered when interpreting NGAL results clinically. This review covers the biology and pathophysiology of NGAL and summarizes the results of the growing body of clinical studies that have addressed the utility of NGAL in the early diagnosis of AKI, in the distinction of intrinsic AKI and in the prognostic assessment of broad patient populations.


Development | 2007

β-catenin/TCF/Lef controls a differentiation-associated transcriptional program in renal epithelial progenitors

Kai M. Schmidt-Ott; T. Néstor H Masckauchán; Xia Chen; Benjamin J. Hirsh; Abby Sarkar; Jun Yang; Neal Paragas; Valerie A. Wallace; Daniel Dufort; Paul Pavlidis; Bernd Jagla; Jan Kitajewski; Jonathan Barasch

In the embryonic kidney, progenitors in the metanephric mesenchyme differentiate into specialized renal epithelia in a defined sequence characterized by the formation of cellular aggregates, conversion into polarized epithelia and segmentation along a proximal-distal axis. This sequence is reiterated throughout renal development to generate nephrons. Here, we identify global transcriptional programs associated with epithelial differentiation utilizing an organ culture model of rat metanephric mesenchymal differentiation, which recapitulates the hallmarks of epithelialization in vivo in a synchronized rather than reiterative fashion. We observe activation of multiple putative targets ofβ -catenin/TCF/Lef-dependent transcription coinciding with epithelial differentiation. We show in cultured explants that isolated activation ofβ -catenin signaling in epithelial progenitors induces, in a TCF/Lef-dependent manner, a subset of the transcripts associated with epithelialization, including Pax8, cyclin D1 (Ccnd1) and Emx2. This is associated with anti-apoptotic and proliferative effects in epithelial progenitors, whereas cells with impaired TCF/Lef-dependent transcription are progressively depleted from the epithelial lineage. In vivo, TCF/Lef-responsive genes comprise a conserved transcriptional program in differentiating renal epithelial progenitors and β-catenin-containing transcriptional complexes directly bind to their promoter regions. Thus,β -catenin/TCF/Lef-mediated transcriptional events control a subset of the differentiation-associated transcriptional program and thereby participate in maintenance, expansion and stage progression of the epithelial lineage.


The New England Journal of Medicine | 2013

Mutations in DSTYK and Dominant Urinary Tract Malformations

Simone Sanna-Cherchi; R.V. Sampogna; Natalia Papeta; Katelyn E. Burgess; Shannon N. Nees; Brittany J. Perry; Murim Choi; Monica Bodria; Yuanli Liu; Patricia L. Weng; Vladimir J. Lozanovski; Miguel Verbitsky; F. Lugani; Roel Sterken; Neal Paragas; Gianluca Caridi; Alba Carrea; M. Dagnino; Anna Materna-Kiryluk; G. Santamaria; C. Murtas; Nadica Ristoska-Bojkovska; Claudia Izzi; Nilgun Kacak; Beatrice Bianco; S. Giberti; Maddalena Gigante; G. Piaggio; Loreto Gesualdo; D. Kosuljandic Vukic

BACKGROUND Congenital abnormalities of the kidney and the urinary tract are the most common cause of pediatric kidney failure. These disorders are highly heterogeneous, and the etiologic factors are poorly understood. METHODS We performed genomewide linkage analysis and whole-exome sequencing in a family with an autosomal dominant form of congenital abnormalities of the kidney or urinary tract (seven affected family members). We also performed a sequence analysis in 311 unrelated patients, as well as histologic and functional studies. RESULTS Linkage analysis identified five regions of the genome that were shared among all affected family members. Exome sequencing identified a single, rare, deleterious variant within these linkage intervals, a heterozygous splice-site mutation in the dual serine-threonine and tyrosine protein kinase gene (DSTYK). This variant, which resulted in aberrant splicing of messenger RNA, was present in all affected family members. Additional, independent DSTYK mutations, including nonsense and splice-site mutations, were detected in 7 of 311 unrelated patients. DSTYK is highly expressed in the maturing epithelia of all major organs, localizing to cell membranes. Knockdown in zebrafish resulted in developmental defects in multiple organs, which suggested loss of fibroblast growth factor (FGF) signaling. Consistent with this finding is the observation that DSTYK colocalizes with FGF receptors in the ureteric bud and metanephric mesenchyme. DSTYK knockdown in human embryonic kidney cells inhibited FGF-stimulated phosphorylation of extracellular-signal-regulated kinase (ERK), the principal signal downstream of receptor tyrosine kinases. CONCLUSIONS We detected independent DSTYK mutations in 2.3% of patients with congenital abnormalities of the kidney or urinary tract, a finding that suggests that DSTYK is a major determinant of human urinary tract development, downstream of FGF signaling. (Funded by the National Institutes of Health and others.).


Journal of Clinical Investigation | 2014

α–Intercalated cells defend the urinary system from bacterial infection

Neal Paragas; Ritwij Kulkarni; Max Werth; Kai M. Schmidt-Ott; Catherine S. Forster; Rong Deng; Qing-Yin Zhang; Eugenia Singer; Alexander D. Klose; Tian Huai Shen; Kevin P. Francis; Sunetra Ray; Soundarapandian Vijayakumar; Samuel Seward; Mary E. Bovino; Katherine Xu; Yared Takabe; Fábio E. Amaral; Sumit Mohan; Rebecca Wax; Kaitlyn Corbin; Simone Sanna-Cherchi; Kiyoshi Mori; Lynne L. Johnson; Thomas L. Nickolas; Vivette D. D’Agati; Chyuan-Sheng Lin; Andong Qiu; Qais Al-Awqati; Adam J. Ratner

α-Intercalated cells (A-ICs) within the collecting duct of the kidney are critical for acid-base homeostasis. Here, we have shown that A-ICs also serve as both sentinels and effectors in the defense against urinary infections. In a murine urinary tract infection model, A-ICs bound uropathogenic E. coli and responded by acidifying the urine and secreting the bacteriostatic protein lipocalin 2 (LCN2; also known as NGAL). A-IC-dependent LCN2 secretion required TLR4, as mice expressing an LPS-insensitive form of TLR4 expressed reduced levels of LCN2. The presence of LCN2 in urine was both necessary and sufficient to control the urinary tract infection through iron sequestration, even in the harsh condition of urine acidification. In mice lacking A-ICs, both urinary LCN2 and urinary acidification were reduced, and consequently bacterial clearance was limited. Together these results indicate that A-ICs, which are known to regulate acid-base metabolism, are also critical for urinary defense against pathogenic bacteria. They respond to both cystitis and pyelonephritis by delivering bacteriostatic chemical agents to the lower urinary system.


Journal of The American Society of Nephrology | 2011

The Sweet Pee Model for Sglt2 Mutation

Joseph P. Ly; Tuncer Onay; Karen Sison; Gavasker A. Sivaskandarajah; Venkata Sabbisetti; Lingli Li; Joseph V. Bonventre; Ann M. Flenniken; Neal Paragas; Jon M. Barasch; S. Lee Adamson; Lucy R. Osborne; Janet Rossant; Jurgen Schnermann; Susan E. Quaggin

Inhibiting renal glucose transport is a potential pharmacologic approach to treat diabetes. The renal tubular sodium-glucose transporter 2 (SGLT2) reabsorbs approximately 90% of the filtered glucose load. An animal model with sglt2 dysfunction could provide information regarding the potential long-term safety and efficacy of SGLT2 inhibitors, which are currently under clinical investigation. Here, we describe Sweet Pee, a mouse model that carries a nonsense mutation in the Slc5a2 gene, which results in the loss of sglt2 protein function. The phenotype of Sweet Pee mutants was remarkably similar to patients with mutations in the Scl5a2 gene. The Sweet Pee mutants had improved glucose tolerance, higher urinary excretion of calcium and magnesium, and growth retardation. Renal physiologic studies demonstrated a prominent distal osmotic diuresis without enhanced natriuresis. Sweet Pee mutants did not exhibit increased KIM-1 or NGAL, markers of acute tubular injury. After induction of diabetes, Sweet Pee mice had better overall glycemic control than wild-type control mice, but had a higher risk for infection and an increased mortality rate (70% in homozygous mutants versus 10% in controls at 20 weeks). In summary, the Sweet Pee model allows study of the long-term benefits and risks associated with inhibition of SGLT2 for the management of diabetes. Our model suggests that inhibiting SGLT2 may improve glucose control but may confer increased risks for infection, malnutrition, volume contraction, and mortality.

Collaboration


Dive into the Neal Paragas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kai M. Schmidt-Ott

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catherine S. Forster

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Prasad Devarajan

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas L. Nickolas

Columbia University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge