Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
2.
Philos Trans R Soc Lond B Biol Sci ; 378(1867): 20210070, 2023 01 02.
Article En | MEDLINE | ID: mdl-36374130

Effective restoration planning tools are needed to mitigate global carbon and biodiversity crises. Published spatial assessments of restoration potential are often at large scales or coarse resolutions inappropriate for local action. Using a Tanzanian case study, we introduce a systematic approach to inform landscape restoration planning, estimating spatial variation in cost-effectiveness, based on restoration method, logistics, biomass modelling and uncertainty mapping. We found potential for biomass recovery across 77.7% of a 53 000 km2 region, but with some natural spatial discontinuity in moist forest biomass, that was previously assigned to human causes. Most areas with biomass deficit (80.5%) were restorable through passive or assisted natural regeneration. However, cumulative biomass gains from planting outweighed initially high implementation costs meaning that, where applicable, this method yielded greater long-term returns on investment. Accounting for ecological, funding and other uncertainty, the top 25% consistently cost-effective sites were within protected areas and/or moderately degraded moist forest and savanna. Agro-ecological mosaics had high biomass deficit but little cost-effective restoration potential. Socio-economic research will be needed to inform action towards environmental and human development goals in these areas. Our results highlight value in long-term landscape restoration investments and separate treatment of savannas and forests. Furthermore, they contradict previously asserted low restoration potential in East Africa, emphasizing the importance of our regional approach for identifying restoration opportunities across the tropics. This article is part of the theme issue 'Understanding forest landscape restoration: reinforcing scientific foundations for the UN Decade on Ecosystem Restoration'.


Ecosystem , Trees , Humans , Biodiversity , Forests , Biomass , Conservation of Natural Resources/methods
3.
J Immunol Methods ; 474: 112666, 2019 11.
Article En | MEDLINE | ID: mdl-31525363

Identity testing of biopharmaceutical products is conducted at multiple steps in the manufacturing process, for drug product lot release, and often for product importation. Because of the chemical and structural similarities of antibody-based products, they present a unique challenge for the development of a QC friendly identity assay where specificity is the critical attribute. Here we report on the development of a novel, rapid and highly specific assay designed to simplify identity testing of antibody-based biopharmaceutical products. A lateral flow immunoassay platform (LFIA) was optimized and used to develop seven identity-specific tests against therapeutic monoclonal antibodies. The specificity of each assay was verified against 10-40 antibody products. An average linear range of antibody detection from 50 to 10,000 ng/ml was observed, allowing minimal sample dilution to be performed. The optimized LFIA platform consistently produced a strong visual signal and showed no false positive results. Three of the seven LFIA-based identity assays have been successfully validated for product release, in accordance with ICH validation guidelines. Additional tests will be validated as products reach the commercial phase. We demonstrate that a lateral flow-based identity assay is an ideal analytical tool for identity testing of antibody therapeutics. The assay platform can easily be adapted for new antibody products and it can be quickly transferred and validated for product testing.


Antibodies, Monoclonal/analysis , Biological Products/analysis , Immunoassay , Predictive Value of Tests , Reproducibility of Results , Time Factors , Workflow
4.
Hum Mol Genet ; 28(5): 751-763, 2019 03 01.
Article En | MEDLINE | ID: mdl-30388220

Autosomal dominant polycystic kidney disease (ADPKD) is among the most common monogenic disorders mainly associated with PKD1/PC1 mutations. We show herein that renal regulation in Pc1 dosage-reduced and -increased mouse models converge toward stimulation of c-Myc expression along with ß-catenin, delineating c-Myc as a key Pkd1 node in cystogenesis. Enhanced renal c-Myc-induced ADPKD in SBM transgenic mice lead conversely to striking upregulation of Pkd1/Pc1 expression and ß-catenin activation, lending credence for reciprocal crosstalk between c-Myc and Pc1. In adult SBM kidneys, c-Myc is strongly enriched on Pkd1 promoter with RNA pol II, consistent with Pkd1 upregulation during cystogenesis. Similar c-Myc direct binding at birth uncovers an equivalent role on Pkd1 regulation during renal developmental program. Concurrent with enriched c-Myc binding, recruitment of active chromatin modifying co-factors by c-Myc at the Pkd1 regulatory region probably opens chromatin to stimulate transcription. A similar transcriptional activation by c-Myc is also likely operant on endogenous human PKD1 gene from our transactivation analysis in response to human c-MYC upregulation. Genetic ablation of c-Myc in Pc1-reduced and -increased mouse models significantly attenuates cyst growth, proliferation and PKD progression. Our study determined a dual role for c-Myc, as a major contributor in Pc1-induced cystogenesis and in a feed-forward regulatory Pkd1-c-Myc loop mechanism that may also prevail in human ADPKD.


Gene Expression Regulation , Genetic Predisposition to Disease , Proto-Oncogene Proteins c-myc/metabolism , TRPP Cation Channels/genetics , Animals , Base Sequence , Binding Sites , Biomarkers , Cell Line , Disease Models, Animal , Disease Progression , Gene Dosage , Genetic Association Studies , Humans , Immunohistochemistry , Mice , Mice, Knockout , Models, Biological , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Protein Binding , Signal Transduction , TRPP Cation Channels/metabolism , Transcription, Genetic
5.
J Feline Med Surg ; 21(12): 1069-1079, 2019 12.
Article En | MEDLINE | ID: mdl-30461328

OBJECTIVES: The aim of this study was to design and carry out a preliminary evaluation of a urine point-of-care test for kidney injury molecule 1 (KIM-1) in healthy and diseased cats. METHODS: Part of the feline KIM-1 gene was amplified, ligated into a plasmid with a signal peptide and monomeric human IgGFc, and transfected into a mammalian cell line. Supernatant was purified and tested for the fusion protein by gel electrophoresis and Western blot. Mice were immunized three times with purified proteins, and hybridomas were generated from splenocytes. Antibodies were tested by ELISA for detection of recombinant KIM-1 and naturally occurring KIM-1 in disease-state urine. Next, a lateral flow assay (LFA) with capture and detection antibodies was constructed, and tested with 34 urine samples from healthy and diseased cats. Antibodies were also tested for reactivity with formalin-fixed paraffin-embedded kidney tissue. RESULTS: Three antibodies were assessed. Antibodies detected between 0.4 and 60 ng/ml feline KIM-1 fusion protein in the LFA. Urine samples from healthy cats yielded faint bands in the LFA corresponding to optical density (OD) values of 4.8-8.8. Samples from cats with suspected or confirmed acute kidney injury (AKI) had OD values ranging from 1.6-20.5. Urine KIM-1 varied over multiple days in cats with sepsis or urethral obstruction despite normalizing serum creatinine concentration. In tissue sections, KIM-1 antibodies labeled tubular cells with morphological features of injury. CONCLUSIONS AND RELEVANCE: A practical patient-side assay for detection of KIM-1 in feline urine has been developed. Preliminary results show marked though transient increases in cats with sepsis and urethral obstruction-associated AKI, and expression in injured tubules. Although initial data indicating that the LFA is sensitive and specific for KIM-1 in cats with AKI are promising, values associated with different types of injury, urine collection, urine storage and specific gravity need to be investigated.


Acute Kidney Injury/veterinary , Cat Diseases/diagnosis , Hepatitis A Virus Cellular Receptor 1/isolation & purification , Immunoassay/veterinary , Kidney/injuries , Point-of-Care Testing , Acute Kidney Injury/diagnosis , Acute Kidney Injury/urine , Animals , Biomarkers/urine , Cat Diseases/urine , Cats , Immunoassay/methods , Male
6.
Sci Rep ; 8(1): 16408, 2018 11 06.
Article En | MEDLINE | ID: mdl-30401798

Severe equine asthma is a chronic inflammatory condition of the lower airways similar to adult-onset asthma in humans. Exacerbations are characterized by bronchial and bronchiolar neutrophilic inflammation, mucus hypersecretion and airway constriction. In this study we analyzed the gene expression response of the bronchial epithelium within groups of asthmatic and non-asthmatic animals following exposure to a dusty hay challenge. After challenge we identified 2341 and 120 differentially expressed genes in asthmatic and non-asthmatic horses, respectively. Gene set enrichment analysis of changes in gene expression after challenge identified 587 and 171 significantly enriched gene sets in asthmatic and non-asthmatic horses, respectively. Gene sets in asthmatic animals pertained, but were not limited, to cell cycle, neutrophil migration and chemotaxis, wound healing, hemostasis, coagulation, regulation of body fluid levels, and the hedgehog pathway. Furthermore, transcription factor target enrichment analysis in the asthmatic group showed that transcription factor motifs with the highest enrichment scores for up-regulated genes belonged to the E2F transcription factor family. It is postulated that engagement of hedgehog and E2F pathways in asthmatic horses promotes dysregulated cell proliferation and abnormal epithelial repair. These fundamental lesions may prevent re-establishment of homeostasis and perpetuate inflammation.


Asthma/genetics , Asthma/pathology , Bronchi/pathology , Cell Cycle/genetics , Respiratory Mucosa/metabolism , Animals , Cell Movement/genetics , Gene Expression Profiling , Hedgehog Proteins/metabolism , Homeostasis/genetics , Horses , Neutrophils/cytology , Sequence Analysis, RNA
7.
PeerJ ; 6: e5759, 2018.
Article En | MEDLINE | ID: mdl-30324028

BACKGROUND: Severe equine asthma is a chronic inflammatory disease of the lung in horses similar to low-Th2 late-onset asthma in humans. This study aimed to determine the utility of RNA-Seq to call gene sequence variants, and to identify sequence variants of potential relevance to the pathogenesis of asthma. METHODS: RNA-Seq data were generated from endobronchial biopsies collected from six asthmatic and seven non-asthmatic horses before and after challenge (26 samples total). Sequences were aligned to the equine genome with Spliced Transcripts Alignment to Reference software. Read preparation for sequence variant calling was performed with Picard tools and Genome Analysis Toolkit (GATK). Sequence variants were called and filtered using GATK and Ensembl Variant Effect Predictor (VEP) tools, and two RNA-Seq predicted sequence variants were investigated with both PCR and Sanger sequencing. Supplementary analysis of novel sequence variant selection with VEP was based on a score of <0.01 predicted with Sorting Intolerant from Tolerant software, missense nature, location within the protein coding sequence and presence in all asthmatic individuals. For select variants, effect on protein function was assessed with Polymorphism Phenotyping 2 and screening for non-acceptable polymorphism 2 software. Sequences were aligned and 3D protein structures predicted with Geneious software. Difference in allele frequency between the groups was assessed using a Pearson's Chi-squared test with Yates' continuity correction, and difference in genotype frequency was calculated using the Fisher's exact test for count data. RESULTS: RNA-Seq variant calling and filtering correctly identified substitution variants in PACRG and RTTN. Sanger sequencing confirmed that the PACRG substitution was appropriately identified in all 26 samples while the RTTN substitution was identified correctly in 24 of 26 samples. These variants of uncertain significance had substitutions that were predicted to result in loss of function and to be non-neutral. Amino acid substitutions projected no change of hydrophobicity and isoelectric point in PACRG, and a change in both for RTTN. For PACRG, no difference in allele frequency between the two groups was detected but a higher proportion of asthmatic horses had the altered RTTN allele compared to non-asthmatic animals. DISCUSSION: RNA-Seq was sensitive and specific for calling gene sequence variants in this disease model. Even moderate coverage (<10-20 counts per million) yielded correct identification in 92% of samples, suggesting RNA-Seq may be suitable to detect sequence variants in low coverage samples. The impact of amino acid alterations in PACRG and RTTN proteins, and possible association of the sequence variants with asthma, is of uncertain significance, but their role in ciliary function may be of future interest.

8.
BMC Genomics ; 18(1): 708, 2017 Sep 08.
Article En | MEDLINE | ID: mdl-28886691

BACKGROUND: Severe equine asthma is a naturally occurring lung inflammatory disease of mature animals characterized by neutrophilic inflammation, bronchoconstriction, mucus hypersecretion and airway remodeling. Exacerbations are triggered by inhalation of dust and microbial components. Affected animals eventually are unable of aerobic performance. In this study transcriptomic differences between asthmatic and non-asthmatic animals in the response of the bronchial epithelium to an inhaled challenge were determined. RESULTS: Paired endobronchial biopsies were obtained pre- and post-challenge from asthmatic and non-asthmatic animals. The transcriptome, determined by RNA-seq and analyzed with edgeR, contained 111 genes differentially expressed (DE) after challenge between horses with and without asthma, and 81 of these were upregulated. Genes involved in neutrophil migration and activation were in central location in interaction networks, and related gene ontology terms were significantly overrepresented. Relative abundance of specific gene products as determined by immunohistochemistry was correlated with differential gene expression. Gene sets involved in neutrophil chemotaxis, immune and inflammatory response, secretion, blood coagulation and apoptosis were overrepresented among up-regulated genes, while the rhythmic process gene set was overrepresented among down-regulated genes. MMP1, IL8, TLR4 and MMP9 appeared to be the most important proteins in connecting the STRING protein network of DE genes. CONCLUSIONS: Several differentially expressed genes and networks in horses with asthma also contribute to human asthma, highlighting similarities between severe human adult and equine asthma. Neutrophil activation by the bronchial epithelium is suggested as the trigger of the inflammatory cascade in equine asthma, followed by epithelial injury and impaired repair and differentiation. Circadian rhythm dysregulation and the sonic Hedgehog pathway were identified as potential novel contributory factors in equine asthma.


Asthma/genetics , Bronchi/metabolism , Gene Expression Profiling , Respiratory Mucosa/metabolism , Animals , Gene Ontology , Horses , Inflammation/genetics
9.
Am J Vet Res ; 78(2): 186-194, 2017 Feb.
Article En | MEDLINE | ID: mdl-28140640

OBJECTIVE To determine whether canine protein C (CnPC) had antichemotactic effects on canine neutrophils, whether endothelial protein C receptor (EPCR) was expressed on canine neutrophils, and the role of EPCR in neutrophil chemotaxis. SAMPLE Neutrophils isolated from blood samples from healthy dogs (n = 6) and sick dogs with (2) or without (3) an inflammatory leukogram. PROCEDURES Neutrophils were analyzed by reverse transcriptase PCR assay and flow cytometry for detection of EPCR mRNA and protein expression, respectively. Neutrophils were incubated with CnPC zymogen or canine activated protein C (CnAPC), with or without RCR-379 (an anti-human EPCR antibody). Neutrophils were then allowed to migrate through a filter membrane toward a chemokine. Untreated neutrophils served as positive control samples. Migration was quantified by fluorescence measurement, and chemotaxis index (Chx) values (fluorescence of test sample/fluorescence of positive control sample) were computed. RESULTS The cDNA for EPCR was amplified, and EPCR expression was detected on neutrophil surfaces. Obtained Chx values were significantly higher in cells treated with RCR-379 than in cells treated with CnPC or CnAPC alone. The Chx values for neutrophils treated with RCR-379 were not significantly different from 1, whereas those for neutrophils treated without RCR-379 were significantly less than 1. The effects of RCR-379 on neutrophil migration were independent of concentration or activation status of protein C. CONCLUSIONS AND CLINICAL RELEVANCE Canine neutrophils expressed EPCR, and inhibition of neutrophil chemotaxis by CnPC and CnAPC depended on EPCR. Interventions with EPCR signaling may have therapeutic application in dogs.


Blood Coagulation Factors/drug effects , Dog Diseases/blood , Neutrophils/drug effects , Protein C/pharmacology , Receptors, Cell Surface/drug effects , Animals , Blood Coagulation Tests , Chemotaxis, Leukocyte/drug effects , Dogs/blood , Flow Cytometry/veterinary , Polymerase Chain Reaction/veterinary
10.
PLoS One ; 10(1): e0115042, 2015.
Article En | MEDLINE | ID: mdl-25606855

The potato cyst nematode, Globodera rostochiensis, is an important pest of potato. Like other pathogens, plant parasitic nematodes are presumed to employ effector proteins, secreted into the apoplast as well as the host cytoplasm, to alter plant cellular functions and successfully infect their hosts. We have generated a library of ORFs encoding putative G. rostochiensis putative apoplastic effectors in vectors for expression in planta. These clones were assessed for morphological and developmental effects on plants as well as their ability to induce or suppress plant defenses. Several CLAVATA3/ESR-like proteins induced developmental phenotypes, whereas predicted cell wall-modifying proteins induced necrosis and chlorosis, consistent with roles in cell fate alteration and tissue invasion, respectively. When directed to the apoplast with a signal peptide, two effectors, an ubiquitin extension protein (GrUBCEP12) and an expansin-like protein (GrEXPB2), suppressed defense responses including NB-LRR signaling induced in the cytoplasm. GrEXPB2 also elicited defense response in species- and sequence-specific manner. Our results are consistent with the scenario whereby potato cyst nematodes secrete effectors that modulate host cell fate and metabolism as well as modifying host cell walls. Furthermore, we show a novel role for an apoplastic expansin-like protein in suppressing intra-cellular defense responses.


Gene Expression Regulation/physiology , Helminth Proteins/biosynthesis , Host-Pathogen Interactions/physiology , Nematoda/physiology , Plant Diseases/parasitology , Solanum tuberosum/parasitology , Animals , Helminth Proteins/genetics
11.
PLoS One ; 9(4): e96217, 2014.
Article En | MEDLINE | ID: mdl-24777050

Secretoglobin family 1A member 1 (SCGB 1A1) is a small protein mainly secreted by mucosal epithelial cells of the lungs and uterus. SCGB 1A1, also known as club (Clara) cell secretory protein, represents a major constituent of airway surface fluid. The protein has anti-inflammatory properties, and its concentration is reduced in equine recurrent airway obstruction (RAO) and human asthma. RAO is characterized by reversible airway obstruction, bronchoconstriction and neutrophilic inflammation. Direct effects of SCGB 1A1 on neutrophil functions are unknown. We have recently identified that the SCGB1A1 gene is triplicated in equids and gives rise to two distinct proteins. In this study we produced the endogenously expressed forms of SCGBs (SCGB 1A1 and 1A1A) as recombinant proteins, and analyzed their effects on reactive oxygen species production, phagocytosis, chemotaxis and neutrophil extracellular trap (NET) formation ex vivo. We further evaluated whether NETs are present in vivo in control and inflamed lungs. Our data show that SCGB 1A1A but not SCGB 1A1 increase neutrophil oxidative burst and phagocytosis; and that both proteins markedly reduce neutrophil chemotaxis. SCGB 1A1A reduced chemotaxis significantly more than SCGB 1A1. NET formation was significantly reduced in a time- and concentration-dependent manner by SCGB 1A1 and 1A1A. SCGB mRNA in bronchial biopsies, and protein concentration in bronchoalveolar lavage fluid, was lower in horses with RAO. NETs were present in bronchoalveolar lavage fluid from horses with exacerbated RAO, but not in fluid from horses with RAO in remission or in challenged healthy horses. These findings indicate that SCGB 1A1 and 1A1A have overlapping and diverging functions. Considering disparities in the relative abundance of SCGB 1A1 and 1A1A in airway secretions of animals with RAO suggests that these functional differences may contribute to the pathogenesis of RAO and other neutrophilic inflammatory lung diseases.


Extracellular Traps/drug effects , Extracellular Traps/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Phagocytosis/drug effects , Reactive Oxygen Species/metabolism , Secretoglobins/pharmacology , Airway Obstruction/genetics , Airway Obstruction/metabolism , Animals , Base Sequence , Bronchoalveolar Lavage Fluid/chemistry , Chemotaxis/drug effects , Dose-Response Relationship, Drug , Horses , Humans , Interleukin-8/pharmacology , Molecular Sequence Data , Neutrophils/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recurrence , Respiratory Burst/drug effects , Secretoglobins/genetics , Secretoglobins/metabolism , Time Factors
12.
Mol Phylogenet Evol ; 69(3): 430-6, 2013 Dec.
Article En | MEDLINE | ID: mdl-23988306

Secretoglobin family 1A member 1 (SCGB 1A1) is a small anti-inflammatory and immunomodulatory protein that is abundantly secreted in airway surface fluids. We recently reported the existence of three distinct SCGB1A1 genes in the domestic horse genome as opposed to the single gene copy consensus present in other mammals. The origin of SCGB1A1 gene triplication and the evolutionary relationship of the three genes amongst Equidae family members are unknown. For this study, SCGB1A1 genomic data were collected from various Equus individuals including E. caballus, E. przewalskii, E. asinus, E. grevyi, and E. quagga. Three SCGB1A1 genes in E. przewalskii, two SCGB1A1 genes in E. asinus, and a single SCGB1A1 gene in E. grevyi and E. quagga were identified. Sequence analysis revealed that the non-synonymous nucleotide substitutions between the different equid genes coded for 17 amino acid changes. Most of these changes localized to the SCGB 1A1 central cavity that binds hydrophobic ligands, suggesting that this area of SCGB 1A1 evolved to accommodate diverse molecular interactions. Three-dimensional modeling of the proteins revealed that the size of the SCGB 1A1 central cavity is larger than that of SCGB 1A1A. Altogether, these findings suggest that evolution of the SCGB1A1 gene may parallel the separation of caballine and non-caballine species amongst Equidae, and may indicate an expansion of function for SCGB1A1 gene products.


Equidae/genetics , Gene Duplication , Phylogeny , Uteroglobin/genetics , Amino Acid Sequence , Animals , Base Sequence , Evolution, Molecular , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Sequence Alignment , Sequence Analysis, DNA
13.
BMC Genomics ; 13: 712, 2012 Dec 19.
Article En | MEDLINE | ID: mdl-23253434

BACKGROUND: Secretoglobin 1A1 (SCGB 1A1), also called Clara cell secretory protein, is the most abundantly secreted protein of the airway. The SCGB1A1 gene has been characterized in mammals as a single copy in the genome. However, analysis of the equine genome suggested that horses might have multiple SCGB1A1 gene copies. Non-ciliated lung epithelial cells produce SCGB 1A1 during inhalation of noxious substances to counter airway inflammation. Airway fluid and lung tissue of horses with recurrent airway obstruction (RAO), a chronic inflammatory lung disease affecting mature horses similar to environmentally induced asthma of humans, have reduced total SCGB 1A1 concentration. Herein, we investigated whether horses have distinct expressed SCGB1A1 genes; whether the transcripts are differentially expressed in tissues and in inflammatory lung disease; and whether there is cell specific protein expression in tissues. RESULTS: We identified three SCGB1A1 gene copies on equine chromosome 12, contained within a 512-kilobase region. Bioinformatic analysis showed that SCGB1A1 genes differ from each other by 8 to 10 nucleotides, and that they code for different proteins. Transcripts were detected for SCGB1A1 and SCGB1A1A, but not for SCGB1A1P. The SCGB1A1P gene had most inter-individual variability and contained a non-sense mutation in many animals, suggesting that SCGB1A1P has evolved into a pseudogene. Analysis of SCGB1A1 and SCGB1A1A sequences by endpoint-limiting dilution PCR identified a consistent difference affecting 3 bp within exon 2, which served as a gene-specific "signature". Assessment of gene- and organ-specific expression by semiquantitative RT-PCR of 33 tissues showed strong expression of SCGB1A1 and SCGB1A1A in lung, uterus, Fallopian tube and mammary gland, which correlated with detection of SCGB 1A1 protein by immunohistochemistry. Significantly altered expression of the ratio of SCGB1A1A to SCGB1A1 was detected in RAO-affected animals compared to controls, suggesting different roles for SCGB 1A1 and SCGB 1A1A in this inflammatory condition. CONCLUSIONS: This is the first report of three SCGB1A1 genes in a mammal. The two expressed genes code for proteins predicted to differ in function. Alterations in the gene expression ratio in RAO suggest cell and tissue specific regulation and functions. These findings may be important for understanding of lung and reproductive conditions.


Gene Expression Profiling , Horses/genetics , Uteroglobin/genetics , Airway Obstruction/genetics , Animals , Base Sequence , Gene Dosage/genetics , Genomics , Horse Diseases/genetics , Molecular Sequence Data , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Hum Mol Genet ; 19(7): 1174-89, 2010 Apr 01.
Article En | MEDLINE | ID: mdl-20053665

While high levels of Pkd1 expression are detected in tissues of patients with autosomal dominant polycystic kidney disease (ADPKD), it is unclear whether enhanced expression could be a pathogenetic mechanism for this systemic disorder. Three transgenic mouse lines were generated from a Pkd1-BAC modified by introducing a silent tag via homologous recombination to target a sustained wild-type genomic Pkd1 expression within the native tissue and temporal regulation. These mice specifically overexpressed the Pkd1 transgene in extrarenal and renal tissues from approximately 2- to 15-fold over Pkd1 endogenous levels in a copy-dependent manner. All transgenic mice reproducibly developed tubular and glomerular cysts leading to renal insufficiency. Interestingly, Pkd1(TAG) mice also exhibited renal fibrosis and calcium deposits in papilla reminiscent of nephrolithiasis as frequently observed in ADPKD. Similar to human ADPKD, these mice consistently displayed hepatic fibrosis and approximately 15% intrahepatic cysts of the bile ducts affecting females preferentially. Moreover, a significant proportion of mice developed cardiac anomalies with severe left-ventricular hypertrophy, marked aortic arch distention and/or valvular stenosis and calcification that had profound functional impact. Of significance, Pkd1(TAG) mice displayed occasional cerebral lesions with evidence of ruptured and unruptured cerebral aneurysms. This Pkd1(TAG) mouse model demonstrates that overexpression of wild-type Pkd1 can trigger the typical adult renal and extrarenal phenotypes resembling human ADPKD.


Disease Models, Animal , Polycystic Kidney Diseases/genetics , TRPP Cation Channels/genetics , Animals , Blood Pressure , Gene Expression , Heart/physiopathology , Kidney/pathology , Kidney/physiopathology , Life Expectancy , Liver/pathology , Mice , Mice, Transgenic , Phenotype , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/physiopathology , TRPP Cation Channels/metabolism , Up-Regulation
15.
J Biol Chem ; 283(20): 13817-24, 2008 May 16.
Article En | MEDLINE | ID: mdl-18356167

The transcriptional regulator Cux1 (CDP, Cutl1) is aberrantly expressed in mouse models for polycystic kidney disease. Here we show that p75-Cux1, the shortest isoform of Cux1, transcribed from an alternative promoter within intron 20, is also deregulated in polycystic kidneys derived from Pkd1 mutant embryos. To determine the role of the p75-Cux1 isoform in cystogenesis, we generated transgenic mice expressing p75-CUX1 in the kidneys and other tissues. Strikingly, these animals developed polycystic kidneys at variable penetrance and severity, correlating with transgene expression levels. Histological and marker analysis of p75-CUX1-derived polycystic kidneys revealed renal cysts derived from the tubular nephron, supporting a model of autosomal dominant polycystic kidney disease. Transgenic p75-CUX1 kidneys additionally showed an up-regulation of the protooncogene c-myc and a down-regulation of the cyclin-dependent kinase inhibitor p27. Chromatin affinity purification experiments confirmed the direct interaction of Cux1 with the c-myc and p27 promoters. These molecular alterations were accompanied by an increase in cilia length and in the proliferative index of epithelial cells lining the cysts. Together, these results identify an important role for the short isoform of CUX1 in polycystic kidney disease development.


Gene Expression Regulation , Homeodomain Proteins/chemistry , Homeodomain Proteins/physiology , Kidney/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Polycystic Kidney Diseases/genetics , Repressor Proteins/chemistry , Repressor Proteins/physiology , Animals , Cilia/metabolism , Heterozygote , Homozygote , In Situ Hybridization , Kidney/embryology , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Models, Genetic , Phenotype , Polycystic Kidney Diseases/metabolism , Protein Isoforms
16.
Mol Cell Biol ; 26(4): 1538-48, 2006 Feb.
Article En | MEDLINE | ID: mdl-16449663

The pathogenetic mechanisms underlying autosomal dominant polycystic kidney disease (ADPKD) remain to be elucidated. While there is evidence that Pkd1 gene haploinsufficiency and loss of heterozygosity can cause cyst formation in mice, paradoxically high levels of Pkd1 expression have been detected in the kidneys of ADPKD patients. To determine whether Pkd1 gain of function can be a pathogenetic process, a Pkd1 bacterial artificial chromosome (Pkd1-BAC) was modified by homologous recombination to solely target a sustained Pkd1 expression preferentially to the adult kidney. Several transgenic lines were generated that specifically overexpressed the Pkd1 transgene in the kidneys 2- to 15-fold over Pkd1 endogenous levels. All transgenic mice reproducibly developed tubular and glomerular cysts and renal insufficiency and died of renal failure. This model demonstrates that overexpression of wild-type Pkd1 alone is sufficient to trigger cystogenesis resembling human ADPKD. Our results also uncovered a striking increased renal c-myc expression in mice from all transgenic lines, indicating that c-myc is a critical in vivo downstream effector of Pkd1 molecular pathways. This study not only produced an invaluable and first PKD model to evaluate molecular pathogenesis and therapies but also provides evidence that gain of function could be a pathogenetic mechanism in ADPKD.


Polycystic Kidney, Autosomal Dominant/genetics , Proteins/genetics , Animals , Base Sequence , Chromosomes, Artificial, Bacterial/genetics , DNA/genetics , Disease Models, Animal , Gene Expression , Genes, myc , Humans , Kidney/abnormalities , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Organ Specificity , Polycystic Kidney, Autosomal Dominant/etiology , Polycystic Kidney, Autosomal Dominant/metabolism , Recombination, Genetic , Signal Transduction , TRPP Cation Channels
...