Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Nat Prod ; 87(4): 764-773, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38423998

ABSTRACT

The brevicidines represent a novel class of nonribosomal antimicrobial peptides that possess remarkable potency and selectivity toward highly problematic and resistant Gram-negative pathogenic bacteria. A recently discovered member of the brevicidine family, coined brevicidine B (2), comprises a single amino acid substitution (from d-Tyr2 to d-Phe2) in the amino acid sequence of the linear moiety of brevicidine (1) and was reported to exhibit broader antimicrobial activity against both Gram-negative (MIC = 2-4 µgmL-1) and Gram-positive (MIC = 2-8 µgmL-1) pathogens. Encouraged by this, we herein report the first total synthesis of the proposed structure of brevicidine B (2), building on our previously reported synthetic strategy to access brevicidine (1). In agreement with the original isolation paper, pleasingly, synthetic 2 demonstrated antimicrobial activity toward Escherichia coli, Pseudomonas aeruginosa, and Klebsiella pneumoniae (MIC = 4-8 µgmL-1). Interestingly, however, synthetic 2 was inactive toward all of the tested Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus strains. Substitution of d-Phe2 with its enantiomer, and other hydrophobic residues, yields analogues that were either inactive or only exhibited activity toward Gram-negative strains. The striking difference in the biological activity of our synthetic 2 compared to the reported natural compound warrants the re-evaluation of the original natural product for purity or possible differences in relative configuration. Finally, the evaluation of synthetic 1 and 2 in a human kidney organoid model of nephrotoxicity revealed substantial toxicity of both compounds, although 1 was less toxic than 2 and polymyxin B. These results indicate that modification to position 2 may afford a strategy to mitigate the nephrotoxicity of brevicidine.


Subject(s)
Anti-Bacterial Agents , Microbial Sensitivity Tests , Structure-Activity Relationship , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Molecular Structure , Pseudomonas aeruginosa/drug effects , Humans , Depsipeptides/pharmacology , Depsipeptides/chemistry , Depsipeptides/chemical synthesis , Klebsiella pneumoniae/drug effects , Escherichia coli/drug effects , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Peptides, Cyclic/pharmacology , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry
2.
Methods Mol Biol ; 2664: 69-83, 2023.
Article in English | MEDLINE | ID: mdl-37423983

ABSTRACT

Kidney organoids differentiated from human pluripotent stem cells (hPSC) have advanced the study of kidney diseases by providing an in vitro system that outperforms traditional monolayer cell culture and complements animal models. This chapter describes a simple two-stage protocol that generates kidney organoids in suspension culture in less than 2 weeks. In the first stage, hPSC colonies are differentiated into nephrogenic mesoderm. In the second stage of the protocol, renal cell lineages develop and self-organize into kidney organoids that contain fetal-like nephrons with proximal and distal tubule segmentation. A single assay generates up to 1000 organoids, thereby providing a rapid and cost-efficient method for the bulk production of human kidney tissue. Applications include the study of fetal kidney development, genetic disease modelling, nephrotoxicity screening, and drug development.


Subject(s)
Kidney , Pluripotent Stem Cells , Animals , Humans , Nephrons , Cell Culture Techniques/methods , Cell Differentiation , Organoids
3.
ACS Infect Dis ; 8(12): 2413-2429, 2022 12 09.
Article in English | MEDLINE | ID: mdl-36413173

ABSTRACT

With the post-antibiotic era rapidly approaching, many have turned their attention to developing new treatments, often by structural modification of existing antibiotics. Polymyxins, a family of lipopeptide antibiotics that are used as a last line of defense in the clinic, have recently developed resistance and exhibit significant nephrotoxicity issues. Using thiol-ene chemistry, the facile preparation of six unique S-lipidated building blocks was demonstrated and used to generate lipopeptide mimetics upon incorporation into solid-phase peptide synthesis (SPPS). We then designed and synthesized 38 polymyxin analogues, incorporating these unique building blocks at the N-terminus, or to replace hydrophobic residues at positions 6 and 7 of the native lipopeptides. Several polymyxin analogues bearing one or more S-linked lipids were found to be equipotent to polymyxin, showed minimal kidney nephrotoxicity, and demonstrated activity against several World Health Organisation (WHO) priority pathogens. The S-lipidation strategy has demonstrated potential as a novel approach to prepare innovative new lipopeptide antibiotics.


Subject(s)
Anti-Bacterial Agents , Polymyxin B , Anti-Bacterial Agents/pharmacology
4.
ACS Pharmacol Transl Sci ; 5(4): 207-215, 2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35434532

ABSTRACT

Acute kidney injury (AKI), a sudden loss of kidney function, is a common and serious condition for which there are no approved specific therapies. While there are multiple approaches to treat the underlying causes of AKI, no targets have been clinically validated. Here, we assessed a series of potent, selective competitive inhibitors of histone deacetylase 8 (HDAC8), a promising therapeutic target in an AKI setting. Using biochemical assays, zebrafish AKI phenotypic assays, and human kidney organoid assays, we show that selective HDAC8 inhibitors can lead to efficacy in increasingly stringent models. One of these, PCI-34051, was efficacious in a rodent model of AKI, further supporting the potential for HDAC8 inhibitors and, in particular, this scaffold as a therapeutic approach to AKI.

5.
Nat Rev Nephrol ; 18(5): 277-293, 2022 05.
Article in English | MEDLINE | ID: mdl-35173348

ABSTRACT

Preclinical models of human disease provide powerful tools for therapeutic discovery but have limitations. This problem is especially apparent in the field of acute kidney injury (AKI), in which clinical trial failures have been attributed to inaccurate modelling performed largely in rodents. Multidisciplinary efforts such as the Kidney Precision Medicine Project are now starting to identify molecular subtypes of human AKI. In addition, over the past decade, there have been developments in human pluripotent stem cell-derived kidney organoids as well as zebrafish, rodent and large animal models of AKI. These organoid and AKI models are being deployed at different stages of preclinical therapeutic development. However, the traditionally siloed, preclinical investigator-driven approaches that have been used to evaluate AKI therapeutics to date rarely account for the limitations of the model systems used and have given rise to false expectations of clinical efficacy in patients with different AKI pathophysiologies. To address this problem, there is a need to develop more flexible and integrated approaches, involving teams of investigators with expertise in a range of different model systems, working closely with clinical investigators, to develop robust preclinical evidence to support more focused interventions in patients with AKI.


Subject(s)
Acute Kidney Injury , Translational Research, Biomedical , Acute Kidney Injury/therapy , Animals , Female , Humans , Kidney , Male , Models, Theoretical , Zebrafish
6.
Kidney Int ; 100(6): 1214-1226, 2021 12.
Article in English | MEDLINE | ID: mdl-34534550

ABSTRACT

A multitude of disease and therapy related factors drive the frequent development of kidney disorders in cancer patients. Along with chemotherapy, the newer targeted therapeutics can also cause kidney dysfunction through on and off-target mechanisms. Interestingly, among the small molecule inhibitors approved for the treatment of cancers that harbor BRAF-kinase activating mutations, vemurafenib can trigger tubular damage and acute kidney injury. BRAF is a proto-oncogene involved in cell growth. To investigate the underlying mechanisms, we developed cell culture and mouse models of vemurafenib kidney toxicity. At clinically relevant concentrations vemurafenib induces cell-death in transformed and primary mouse and human kidney tubular epithelial cells. In mice, two weeks of daily vemurafenib treatment causes moderate acute kidney injury with histopathological characteristics of kidney tubular epithelial cells injury. Importantly, kidney tubular epithelial cell-specific BRAF gene deletion did not influence kidney function under normal conditions or alter the severity of vemurafenib-associated kidney impairment. Instead, we found that inhibition of ferrochelatase, an enzyme involved in heme biosynthesis contributes to vemurafenib kidney toxicity. Ferrochelatase overexpression protected kidney tubular epithelial cells and conversely ferrochelatase knockdown increased the sensitivity to vemurafenib-induced kidney toxicity. Thus, our studies suggest that vemurafenib-associated kidney tubular epithelial cell dysfunction and kidney toxicity is BRAF-independent and caused, in part, by off-target ferrochelatase inhibition.


Subject(s)
Ferrochelatase , Proto-Oncogene Proteins B-raf , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Indoles/toxicity , Kidney/metabolism , Mice , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Sulfonamides/toxicity , Vemurafenib
7.
J Vis Exp ; (170)2021 04 13.
Article in English | MEDLINE | ID: mdl-33938892

ABSTRACT

Kidney organoids generated from hPSCs have provided an unlimited source of renal tissue. Human kidney organoids are an invaluable tool for studying kidney disease and injury, developing cell-based therapies, and testing new therapeutics. For such applications, large numbers of uniform organoids and highly reproducible assays are needed. We have built upon our previously published kidney organoid protocol to improve the overall health of the organoids. This simple, robust 3D protocol involves the formation of uniform embryoid bodies in minimum component medium containing lipids, insulin-transferrin-selenium-ethanolamine supplement and polyvinyl alcohol with GSK3 inhibitor (CHIR99021) for 3 days, followed by culture in knock-out serum replacement (KOSR)-containing medium. In addition, agitating assays allows for reduction in clumping of the embryoid bodies and maintaining a uniform size, which is important for reducing variability between organoids. Overall, the protocol provides a fast, efficient, and cost-effective method for generating large quantities of kidney organoids.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/metabolism , Kidney/physiopathology , Organoids/metabolism , Cell Differentiation , Humans
8.
STAR Protoc ; 1(3): 100150, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377044

ABSTRACT

Kidney organoids represent a physiologically advanced model for studying the mechanisms of kidney development and disease. Here, we describe a simple two-step protocol for the differentiation of human pluripotent stem cells into kidney organoids. Our approach involves suspension culture that allows for rapid and cost-effective bulk production of organoids, which is well suited for large-scale assays such as drug screening. The organoids correspond to fetal human kidney tissue and may be of limited use for modeling adult kidney function. For complete details on the use and execution of this protocol, please refer to Przepiorski et al. (2018).


Subject(s)
Cell Culture Techniques/methods , Kidney/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Cells, Cultured , Embryoid Bodies/cytology , Humans , Paraffin Embedding
9.
J Biol Chem ; 295(48): 16328-16341, 2020 11 27.
Article in English | MEDLINE | ID: mdl-32887795

ABSTRACT

Acute kidney injury (AKI) is a common clinical condition associated with diverse etiologies and abrupt loss of renal function. In patients with sepsis, rhabdomyolysis, cancer, and cardiovascular disorders, the underlying disease or associated therapeutic interventions can cause hypoxia, cytotoxicity, and inflammatory insults to renal tubular epithelial cells (RTECs), resulting in the onset of AKI. To uncover stress-responsive disease-modifying genes, here we have carried out renal transcriptome profiling in three distinct murine models of AKI. We find that Vgf nerve growth factor inducible gene up-regulation is a common transcriptional stress response in RTECs to ischemia-, cisplatin-, and rhabdomyolysis-associated renal injury. The Vgf gene encodes a secretory peptide precursor protein that has critical neuroendocrine functions; however, its role in the kidneys remains unknown. Our functional studies show that RTEC-specific Vgf gene ablation exacerbates ischemia-, cisplatin-, and rhabdomyolysis-associated AKI in vivo and cisplatin-induced RTEC cell death in vitro Importantly, aggravation of cisplatin-induced renal injury caused by Vgf gene ablation is partly reversed by TLQP-21, a Vgf-derived peptide. Finally, in vitro and in vivo mechanistic studies showed that injury-induced Vgf up-regulation in RTECs is driven by the transcriptional regulator Sox9. These findings reveal a crucial downstream target of the Sox9-directed transcriptional program and identify Vgf as a stress-responsive protective gene in kidney tubular epithelial cells.


Subject(s)
Acute Kidney Injury/metabolism , Epithelial Cells/metabolism , Kidney Tubules/metabolism , Nerve Growth Factors/biosynthesis , SOX9 Transcription Factor/metabolism , Up-Regulation , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Epithelial Cells/pathology , Kidney Tubules/pathology , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , SOX9 Transcription Factor/genetics
10.
Am J Physiol Renal Physiol ; 318(4): F971-F978, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32150447

ABSTRACT

Acute kidney injury (AKI) remains a major global healthcare problem, and there is a need to develop human-based models to study AKI in vitro. Toward this goal, we have characterized induced pluripotent stem cell-derived human kidney organoids and their response to cisplatin, a chemotherapeutic drug that induces AKI and preferentially damages the proximal tubule. We found that a single treatment with 50 µM cisplatin induces hepatitis A virus cellular receptor 1 (HAVCR1) and C-X-C motif chemokine ligand 8 (CXCL8) expression, DNA damage (γH2AX), and cell death in the organoids but greatly impairs organoid viability. DNA damage was not specific to the proximal tubule but also affected the distal tubule and interstitial cell populations. This lack of specificity correlated with low expression of proximal tubule-specific SLC22A2/organic cation transporter 2 (OCT2) for cisplatin. To improve viability, we developed a repeated low-dose regimen of 4 × 5 µM cisplatin over 7 days and found this caused less toxicity while still inducing a robust injury response that included secretion of known AKI biomarkers and inflammatory cytokines. This work validates the use of human kidney organoids to model aspects of cisplatin-induced injury, with the potential to identify new AKI biomarkers and develop better therapies.


Subject(s)
Acute Kidney Injury/chemically induced , Antineoplastic Agents/toxicity , Cisplatin/toxicity , DNA Damage , Kidney Tubules, Proximal/drug effects , Organoids/drug effects , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Antineoplastic Agents/metabolism , Cells, Cultured , Cisplatin/metabolism , Dose-Response Relationship, Drug , Hepatitis A Virus Cellular Receptor 1/genetics , Hepatitis A Virus Cellular Receptor 1/metabolism , Histones/metabolism , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Organic Cation Transporter 2/metabolism , Organoids/metabolism , Organoids/pathology , Time Factors
11.
Am J Physiol Renal Physiol ; 317(2): F478-F488, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31188030

ABSTRACT

The hepatocyte nuclear factor-1ß (Hnf1b) transcription factor is a key regulator of kidney tubule formation and is associated with a syndrome of renal cysts and early onset diabetes. To further our understanding of Hnf1b in the developing zebrafish kidney, we performed RNA sequencing analysis of proximal tubules from hnf1b-deficient larvae. This analysis revealed an enrichment of gene transcripts encoding transporters of the solute carrier (SLC) superfamily, including multiple members of slc2 and slc5 glucose transporters. An investigation of expression of slc2a1a, slc2a2, and slc5a2 as well as a poorly studied glucose/mannose transporter encoded by slc5a9 revealed that these genes undergo dynamic spatiotemporal changes during tubule formation and maturation. A comparative analysis of zebrafish SLC genes with those expressed in mouse proximal tubules showed a substantial overlap at the level of gene families, indicating a high degree of functional conservation between zebrafish and mammalian proximal tubules. Taken together, our findings are consistent with a role for Hnf1b as a critical determinant of proximal tubule transport function by acting upstream of a large number of SLC genes and validate the zebrafish as a physiologically relevant model of the mammalian proximal tubule.


Subject(s)
Gene Expression Profiling , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/physiology , Kidney Tubules, Proximal/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Regulation, Developmental , Larva , Mice , RNA/biosynthesis , RNA/genetics , Species Specificity
13.
Semin Cell Dev Biol ; 91: 86-93, 2019 07.
Article in English | MEDLINE | ID: mdl-30172050

ABSTRACT

The intermediate mesoderm is located between the somites and the lateral plate mesoderm and gives rise to renal progenitors that contribute to the three mammalian kidney types (pronephros, mesonephros and metanephros). In this review, focusing largely on murine kidney development, we examine how the intermediate mesoderm forms during gastrulation/axis elongation and how it progressively gives rise to distinct renal progenitors along the rostro-caudal axis. We highlight some of the potential signalling cues and core transcription factor circuits that direct these processes, up to the point of early metanephric kidney formation.


Subject(s)
Kidney/embryology , Mesoderm/embryology , Mesonephros/embryology , Somites/embryology , Animals , Body Patterning/genetics , Gene Expression Regulation, Developmental , Kidney/metabolism , Mesoderm/metabolism , Mesonephros/metabolism , Mice , Organogenesis/genetics , Somites/metabolism , Transcription Factors/genetics
14.
Stem Cell Reports ; 11(2): 470-484, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30033089

ABSTRACT

Kidney organoids made from pluripotent stem cells have the potential to revolutionize how kidney development, disease, and injury are studied. Current protocols are technically complex, suffer from poor reproducibility, and have high reagent costs that restrict scalability. To overcome some of these issues, we have established a simple, inexpensive, and robust method to grow kidney organoids in bulk from human induced pluripotent stem cells. Our organoids develop tubular structures by day 8 and show optimal tissue morphology at day 14. A comparison with fetal human kidneys suggests that day-14 organoid tissue most closely resembles late capillary loop stage nephrons. We show that deletion of HNF1B, a transcription factor linked to congenital kidney defects, interferes with tubulogenesis, validating our experimental system for studying renal developmental biology. Taken together, our protocol provides a fast, efficient, and cost-effective method for generating large quantities of human fetal kidney tissue, enabling the study of normal and aberrant kidney development.


Subject(s)
Bioreactors , Cell Culture Techniques , Kidney/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Biomarkers , Cell Differentiation , Fibrosis , Gene Knockout Techniques , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Kidney/embryology , Nephrons/cytology
15.
Molecules ; 22(3)2017 Mar 13.
Article in English | MEDLINE | ID: mdl-28335401

ABSTRACT

We describe the use of the fluorescent reporter compound CDr10b to label mid-intestinal structures in zebrafish larvae after simple immersion. CDr10b is deposited into the gut where it initially fills the lumen and is excreted. Using laser-mediated injury of the intestine, we show that CDr10b provides a useful readout of the integrity and repair of the epithelial cell barrier. In addition, CDr10b specifically labels the absorptive mid-intestine segment that is analogous to the mammalian small intestine. By perturbing retinoic acid signaling, which regulates the size of the mid-intestine segment, we show that CDr10b is a valuable tool to rapidly assess developmental malformations of the intestine in live animals.


Subject(s)
Boron Compounds/administration & dosage , Intestines/ultrastructure , Zebrafish/anatomy & histology , Animals , Larva/anatomy & histology , Microscopy, Fluorescence , Signal Transduction , Tretinoin/metabolism , Zebrafish/growth & development
16.
Chem Commun (Camb) ; 51(2): 395-8, 2015.
Article in English | MEDLINE | ID: mdl-25407666

ABSTRACT

We report the development of a small fluorescent molecule, BDNCA3-D2, herein referred to as PT-Yellow. Soaking zebrafish embryos in PT-Yellow or intraperitoneal injection into adults results in non-toxic in vivo fluorescent labeling of the renal proximal tubules, the major site of blood filtrate reabsorption and a common target of injury in acute kidney injury. We demonstrate the applicability of this new compound as a rapid and simple readout for zebrafish kidney filtration and proximal tubule reabsorption function.


Subject(s)
Fluorescent Dyes/analysis , Kidney Tubules, Proximal/ultrastructure , Kidney/ultrastructure , Zebrafish/anatomy & histology , Animals , Fluorescent Dyes/administration & dosage , Kidney/anatomy & histology , Kidney Tubules, Proximal/anatomy & histology , Larva/anatomy & histology , Larva/ultrastructure , Optical Imaging , Zebrafish/growth & development
17.
Semin Nephrol ; 34(4): 437-44, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25217272

ABSTRACT

Renal tubule epithelial cells can regenerate in response to acute injury. Although this process remains poorly understood, it appears to involve the reactivation of pathways that are operative during embryonic kidney formation. A better understanding of renal regeneration may lead to the development of new therapies that can attenuate acute kidney injury or expedite recovery. The zebrafish is being used as a model to understand renal regeneration. In this review, we summarize the current knowledge on zebrafish kidney formation, describe methods for inducing acute injury, and focus on the unique capacity of the zebrafish adult kidney to undergo de novo nephron formation in response to damage.


Subject(s)
Acute Kidney Injury , Kidney/physiology , Mesonephros/physiology , Regeneration/physiology , Animals , Kidney/cytology , Kidney Tubules/cytology , Kidney Tubules/physiology , Mesonephros/cytology , Nephrons/physiology , Podocytes/physiology , Zebrafish
18.
Int J Dev Biol ; 57(5): 357-64, 2013.
Article in English | MEDLINE | ID: mdl-23873367

ABSTRACT

Histone modifying enzymes play critical roles in cell differentiation and development. In this study, we report that SMYD2 (SET and MYND domain containing protein 2), a histone lysine methyltransferase, is induced during human embryonic stem (ES) cell differentiation and it is preferentially expressed in somatic cells versus pluripotent cells. Knockdown of SMYD2 in human ES cells promotes the induction of endodermal markers during differentiation, while overexpression has opposite effects. In vivo experiments in zebrafish revealed that knockdown of smyd2a (a homologue gene of human SMYD2) causes developmental delay and aberrant tail formation, which is coincident with low expression of ntl and over induction Nodal-related genes during gastrulation. Taken together, these findings suggest that SMYD2 plays a critical role at early stages of development and in human ES cell differentiation.


Subject(s)
Cell Differentiation/genetics , Embryo, Nonmammalian/metabolism , Embryonic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/genetics , Zebrafish Proteins/genetics , Animals , Cells, Cultured , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryonic Stem Cells/cytology , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/metabolism , Humans , In Situ Hybridization , Mice , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tail/embryology , Tail/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/metabolism
19.
Nat Protoc ; 8(4): 800-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23538883

ABSTRACT

This protocol describes how to isolate primary cardiomyocytes from adult zebrafish hearts and culture them for up to 4 weeks, thereby using them as an alternative to in vivo experiments. After collagenase digestion of the ventricle, cells are exposed to increasing calcium concentrations in order to obtain high-purity cardiomyocytes. The whole isolation process can be accomplished in 4-5 h. The culture conditions we established allow the cells to preserve their mature sarcomeric integrity and contractile properties. Furthermore, adult zebrafish cardiomyocytes in culture, similarly to zebrafish in vivo heart regeneration, undergo partial dedifferentiation and, in contrast to their mammalian counterparts, are able to proliferate. Our protocol enables the study of structural and functional properties in close-to-native cardiomyocytes and allows the application of in vitro techniques and assays that are not feasible to perform in living animals.


Subject(s)
Cell Culture Techniques , Cell Separation/methods , Myocytes, Cardiac/cytology , Zebrafish , Animals , Calcium/pharmacology , Cell Dedifferentiation , Cell Proliferation , Culture Media , Fibrin/chemistry
20.
PLoS One ; 5(4): e10383, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20442782

ABSTRACT

In the vertebrates, the BMP/Smad1 and TGF-beta/Smad2 signaling pathways execute antagonistic functions in different contexts of development. The differentiation of specific structures results from the balance between these two pathways. For example, the gastrula organizer/node of the vertebrates requires a region of low Smad1 and high Smad2 signaling. In Drosophila, Mad regulates tissue determination and growth in the wing, but the function of dSmad2 in wing patterning is largely unknown. In this study, we used an RNAi loss-of-function approach to investigate dSmad2 signaling during wing development. RNAi-mediated knockdown of dSmad2 caused formation of extra vein tissue, with phenotypes similar to those seen in Dpp/Mad gain-of-function. Clonal analyses revealed that the normal function of dSmad2 is to inhibit the response of wing intervein cells to the extracellular Dpp morphogen gradient that specifies vein formation, as measured by expression of the activated phospho-Mad protein. The effect of dSmad2 depletion in promoting vein differentiation was dependent on Medea, the co-factor shared by Mad and dSmad2. Furthermore, double RNAi experiments showed that Mad is epistatic to dSmad2. In other words, depletion of Smad2 had no effect in Mad-deficient wings. Our results demonstrate a novel role for dSmad2 in opposing Mad-mediated vein formation in the wing. We propose that the main function of dActivin/dSmad2 in Drosophila wing development is to antagonize Dpp/Mad signaling. Possible molecular mechanisms for the opposition between dSmad2 and Mad signaling are discussed.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/physiology , Signal Transduction/physiology , Smad2 Protein/physiology , Transcription Factors/antagonists & inhibitors , Veins/growth & development , Wings, Animal/growth & development , Activins/metabolism , Animals , Drosophila/growth & development , Drosophila Proteins/metabolism , Female , Smad Proteins, Receptor-Regulated , Wings, Animal/blood supply
SELECTION OF CITATIONS
SEARCH DETAIL
...