Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(11): e0259732, 2021.
Article in English | MEDLINE | ID: mdl-34780505

ABSTRACT

Mesenchymal stem cell derived extracellular vesicles (MSC-EVs) are bioactive particles that evoke beneficial responses in recipient cells. We identified a role for MSC-EV in immune modulation and cellular salvage in a model of SARS-CoV-2 induced acute lung injury (ALI) using pulmonary epithelial cells and exposure to cytokines or the SARS-CoV-2 receptor binding domain (RBD). Whereas RBD or cytokine exposure caused a pro-inflammatory cellular environment and injurious signaling, impairing alveolar-capillary barrier function, and inducing cell death, MSC-EVs reduced inflammation and reestablished target cell health. Importantly, MSC-EV treatment increased active ACE2 surface protein compared to RBD injury, identifying a previously unknown role for MSC-EV treatment in COVID-19 signaling and pathogenesis. The beneficial effect of MSC-EV treatment was confirmed in an LPS-induced rat model of ALI wherein MSC-EVs reduced pro-inflammatory cytokine secretion and respiratory dysfunction associated with disease. MSC-EV administration was dose-responsive, demonstrating a large effective dose range for clinical translation. These data provide direct evidence of an MSC-EV-mediated improvement in ALI and contribute new insights into the therapeutic potential of MSC-EVs in COVID-19 or similar pathologies of respiratory distress.


Subject(s)
Acute Lung Injury/complications , Acute Lung Injury/virology , COVID-19/pathology , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Pneumonia/complications , Pneumonia/virology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Disease Models, Animal , Extracellular Vesicles/ultrastructure , Humans , Immunomodulation , Male , Models, Biological , Pneumonia/pathology , Rats, Sprague-Dawley , SARS-CoV-2/physiology , Signal Transduction , THP-1 Cells
2.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L723-L737, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30652491

ABSTRACT

Secreted exosomes are bioactive particles that elicit profound responses in target cells. Using targeted metabolomics and global microarray analysis, we identified a role of exosomes in promoting mitochondrial function in the context of pulmonary arterial hypertension (PAH). Whereas chronic hypoxia results in a glycolytic shift in pulmonary artery smooth muscle cells (PASMCs), exosomes restore energy balance and improve O2 consumption. These results were confirmed in a hypoxia-induced mouse model and a semaxanib/hypoxia rat model of PAH wherein exosomes improved the mitochondrial dysfunction associated with disease. Importantly, exosome exposure increased PASMC expression of pyruvate dehydrogenase (PDH) and glutamate dehydrogenase 1 (GLUD1), linking exosome treatment to the TCA cycle. Furthermore, we show that although prolonged hypoxia induced sirtuin 4 expression, an upstream inhibitor of both GLUD1 and PDH, exosomes reduced its expression. These data provide direct evidence of an exosome-mediated improvement in mitochondrial function and contribute new insights into the therapeutic potential of exosomes in PAH.


Subject(s)
Exosomes/metabolism , Exosomes/transplantation , Mesenchymal Stem Cells/metabolism , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/therapy , Animals , Cells, Cultured , Citric Acid Cycle , Disease Models, Animal , Glutamate Dehydrogenase/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/metabolism , Models, Biological , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Rats , Rats, Sprague-Dawley , Sirtuins/metabolism
3.
Regen Med ; 10(7): 815-39, 2015.
Article in English | MEDLINE | ID: mdl-26568079

ABSTRACT

AIM: Identification of mechanistic pathways for selected renal cell (SRC) therapeutic bioactivity in rodent models of chronic kidney disease. MATERIALS & METHODS: In vivo and in vitro functional bioassays applied to investigate regenerative outcomes associated with delivery of SRC to diseased rodent kidney. RESULTS: In vivo, SRC reduces chronic infiltration by monocytes/macrophages. SRC attenuates NF-κB and PAI-1 responses while simultaneously promoting host tubular cell expansion through trophic cues. In vitro, SRC-derived conditioned media attenuates TNF-α-induced NF-κB response, TGF-ß-mediated PAI-1 response and increases expression of transcripts associated with cell cycle regulation. Observed bioactive responses were from vesicle and nonvesicle-associated factors, including specific miRNAs. CONCLUSION: We identify a paracrine mechanism for SRC immunomodulatory and trophic cues on host renal tissues, catalyzing long-term functional benefits in vivo.


Subject(s)
Gene Expression Regulation , Kidney Tubules/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Renal Insufficiency, Chronic/metabolism , Transforming Growth Factor beta1/biosynthesis , Animals , Disease Models, Animal , Kidney Tubules/pathology , Macrophages/pathology , NF-kappa B/genetics , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Rats , Rats, Transgenic , Rats, Zucker , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Transforming Growth Factor beta1/genetics
4.
Cell Transplant ; 22(6): 1023-39, 2013.
Article in English | MEDLINE | ID: mdl-22889490

ABSTRACT

New treatment paradigms that slow or reverse progression of chronic kidney disease (CKD) are needed to relieve significant patient and healthcare burdens. We have shown that a population of selected renal cells (SRCs) stabilized disease progression in a mass reduction model of CKD. Here, we further define the cellular composition of SRCs and apply this novel therapeutic approach to the ZSF1 rat, a model of severe progressive nephropathy secondary to diabetes, obesity, dyslipidemia, and hypertension. Injection of syngeneic SRCs into the ZSF1 renal cortex elicited a regenerative response that significantly improved survival and stabilized disease progression to renal structure and function beyond 1 year posttreatment. Functional improvements included normalization of multiple nephron structures and functions including glomerular filtration, tubular protein handling, electrolyte balance, and the ability to concentrate urine. Improvements to blood pressure, including reduced levels of circulating renin, were also observed. These functional improvements following SRC treatment were accompanied by significant reductions in glomerular sclerosis, tubular degeneration, and interstitial inflammation and fibrosis. Collectively, these data support the utility of a novel renal cell-based approach for slowing renal disease progression associated with diabetic nephropathy in the setting of metabolic syndrome, one of the most common causes of end-stage renal disease.


Subject(s)
Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Disease Progression , Kidney Function Tests , Kidney/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Blood Pressure/drug effects , Cell Tracking , Diabetic Nephropathies/drug therapy , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Glomerular Filtration Rate/drug effects , Kidney/drug effects , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Mice , Rats , Rats, Inbred Lew , Survival Analysis
5.
Cells Tissues Organs ; 196(4): 374-84, 2012.
Article in English | MEDLINE | ID: mdl-22584161

ABSTRACT

Dedifferentiation and proliferation of resident tubular epithelial cells is a mechanism of action potentially contributing to repair and regeneration in kidneys presenting with ischemic or chronic disease. To more efficiently develop cell and tissue engineering technologies for the kidney, we have developed molecular assays to evaluate the acquisition of a pluripotent state associated with stem/progenitor cell phenotype during induction of a regenerative response within the kidneys of rats with chronic kidney disease (CKD) following therapeutic intervention. Intrarenal delivery of selected bioactive renal cells leads to significant upregulation of pluripotency-associated SOX2 mRNA within the diseased kidney tissue from 1 to 24 weeks after treatment. The overall regenerative response index was assessed by quantitative composite expression of CD24, NODAL and LEFTY1 proteins, which were induced within 1 week of cell treatment and peaked at 12 weeks after treatment, reaching statistical significance (p < 0.05) compared to untreated CKD controls. Molecular assays that incorporate the assessment of SOX2 and the regenerative response index may prove to be valuable tools for the detection and monitoring of the tissue response after the delivery of regenerative treatments for CKD, thereby significantly shortening the developmental timelines associated with such therapies.


Subject(s)
Cell Transplantation/methods , Kidney Diseases/therapy , Kidney/physiology , Regenerative Medicine/methods , Animals , Chronic Disease , Disease Models, Animal , Kidney/cytology , Kidney/metabolism , Kidney Diseases/metabolism , Male , Rats , Rats, Inbred Lew , Regeneration/physiology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tissue Engineering
6.
Tissue Eng Part A ; 18(9-10): 1025-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22136657

ABSTRACT

Urinary pathology requiring urinary diversion, partial or full bladder replacement, is a significant clinical problem affecting ~14,000 individuals annually in the United States alone. The use of gastrointestinal tissue for urinary diversion or bladder reconstruction/replacement surgeries is frequently associated with complications. To try and alleviate or reduce the frequency of these complications, tissue engineering and regenerative medicine strategies have been developed using bio-absorbable materials seeded with cells derived from the bladder. However, bladder-sourced cells may not always be suitable for such applications, especially in patients with bladder cancer. In this study, we describe the isolation and characterization of smooth muscle cells (SMCs) from porcine adipose and peripheral blood that are phenotypically and functionally indistinguishable from bladder-derived SMCs. In a preclinical Good Laboratory Practice study, we demonstrate that autologous adipose- and peripheral blood-derived SMCs may be used to seed synthetic, biodegradable tubular scaffold structures and that implantation of these seeded scaffolds into a porcine cystectomy model leads to successful de novo regeneration of a tubular neo-organ composed of urinary-like neo-tissue that is histologically identical to native bladder. The ability to create urologic structures de novo from scaffolds seeded by autologous adipose- or peripheral blood-derived SMCs will greatly facilitate the translation of urologic tissue engineering technologies into clinical practice.


Subject(s)
Adipose Tissue/cytology , Guided Tissue Regeneration/methods , Tissue Engineering/methods , Urinary Bladder/surgery , Animals , Female , Fluorescent Antibody Technique , Male , Myocytes, Smooth Muscle/cytology , Reverse Transcriptase Polymerase Chain Reaction , Swine , Tissue Scaffolds/chemistry
7.
Lipids Health Dis ; 10: 171, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21957910

ABSTRACT

BACKGROUND: Therapeutically bioactive cell populations are currently understood to promote regenerative outcomes in vivo by leveraging mechanisms of action including secretion of growth factors, site specific engraftment and directed differentiation. Constitutive cellular populations undoubtedly participate in the regenerative process. Adipose tissue represents a source of therapeutically bioactive cell populations. The potential of these cells to participate in various aspects of the regenerative process has been demonstrated broadly. However, organ association of secretory and developmental markers to specific peri-organ adipose depots has not been investigated. To characterize this topographical association, we explored the potential of cells isolated from the stromal vascular fraction (SVF) of kidney sourced adipose to express key renal associated factors. RESULTS: We report that renal adipose tissue is a novel reservoir for EPO expressing cells. Kidney sourced adipose stromal cells demonstrate hypoxia regulated expression of EPO and VEGF transcripts. Using iso-electric focusing, we demonstrate that kidney and non-kidney sourced adipose stromal cells present unique patterns of EPO post-translational modification, consistent with the idea that renal and non-renal sources are functionally distinct adipose depots. In addition, kidney sourced adipose stromal cells specifically express the key renal developmental transcription factor WT1. CONCLUSIONS: Taken together, these data are consistent with the notion that kidney sourced adipose stromal (KiSAS) cells may be primed to recreate a regenerative micro-environment within the kidney. These findings open the possibility of isolating solid-organ associated adipose derived cell populations for therapeutic applications in organ-specific regenerative medicine products.


Subject(s)
Adipocytes, White/metabolism , Gene Expression Regulation , Intra-Abdominal Fat/cytology , Kidney/cytology , Regenerative Medicine/methods , Adipocytes, White/cytology , Animals , Biomarkers , Cell Hypoxia , Cell Separation , Cells, Cultured , Erythropoietin/genetics , Erythropoietin/metabolism , Humans , Intra-Abdominal Fat/metabolism , Kidney/metabolism , Male , Organ Specificity , Protein Processing, Post-Translational , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
8.
J Cell Physiol ; 226(10): 2702-11, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21792927

ABSTRACT

Myocardin (MYOCD) is a smooth and cardiac muscle-specific transcriptional coactivator that is required for the proper expression of contraction-related genes. Through its function to transactivate effector genes, MYOCD plays an essential role in mediating the switch between contractile and non-contractile phenotypes, particularly in smooth muscle cells (SMC). There are at least two known transcript variants of MYOCD that are expressed in SMC, differing only by the presence (+) or absence (Δ) of Exon 11. To date, no functional role has been assigned to the domain encoded by Exon 11, nor have any notable differences between the ability of each isoform to activate contraction-related genes been observed. In this study we compared sequences for Exon 11 among several mammalian species and identified a highly conserved, putative target sequence for glycogen synthase kinase 3 (GSK3) phosphorylation, suggesting a regulatory role for Exon 11 that can be modulated by alternative splicing. The function of Exon 11 was investigated by altering MYOCD splice selection in cultured porcine SMC with small interfering RNAs (siRNA) and specific chemical inhibitors, resulting in a relative increase in expression of ΔExon 11 variants in the endogenous pool of MYOCD mRNA. The relative increase in ΔExon 11 mRNAs correlated with a reduction of contractile phenotype in the porcine SMC as evidenced by morphological assessment and molecular analysis of effector genes. Together, these data suggest that MYOCD ΔExon 11 may participate in modulating SMC phenotype, potentially acting as a dominant-negative repressor of contraction-related genes.


Subject(s)
Alternative Splicing/physiology , Myocytes, Smooth Muscle/physiology , Nuclear Proteins/genetics , Trans-Activators/genetics , Amino Acid Sequence , Animals , Aorta/cytology , Carotid Arteries/cytology , Conserved Sequence , Genetic Variation , In Vitro Techniques , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phenotype , Swine , Trans-Activators/chemistry , Trans-Activators/metabolism , Urinary Bladder/cytology
9.
Tissue Eng Part C Methods ; 17(8): 843-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21595545

ABSTRACT

Adipose tissue contains a heterogeneous cell population composed of endothelial cells, adipocytes, smooth muscle cells (SMC), and mesenchymal progenitors and stromal cells that meet the criteria put forth by the International Society for Cellular Therapy as defining mesenchymal stem cells (MSC). In this study, we expanded the stromal vascular fraction (SVF) of human adipose tissue and characterized the resulting adherent primary cell cultures by quantitative reverse transcription-polymerase chain reaction, antigen expression, protein fingerprinting, growth kinetics, in vitro tri-lineage differentiation bioactivity, and functional responses to small molecules modulating SMC-related developmental pathways and compared the results to those obtained with functionally validated MSC cultures. SVF-derived initial cultures (P0) were expanded in a defined medium that was not optimized for MSC growth conditions, neither were recombinant cytokines or growth factors added to the media to direct differentiation. The adherent cell cultures derived from SVF expansion under these conditions had markedly distinct phenotypic and biological properties relative to functionally validated MSC cultures. SVF-derived adherent cell cultures retained characteristics consistent with the SMC subpopulation within adipose tissue--phenotype, gene, and protein expression--that were independent of passage number and source of SVF (n=4 independent donors). SVF-derived cells presented significantly less robust in vitro tri-lineage differentiation bioactivity relative to validated MSC. Expanded SVF cells and MSC had opposite responses to the thromboxane A2 mimetic U46619, demonstrating an unambiguous functional distinction between the two cell types. Taken together, these data support the conclusions that SVF cells expanded under the conditions described in these studies are accurately described as adipose-derived SMC and represent a cellular subpopulation of adipose SVF that is separate and distinct from other classes of adipose-derived cells.


Subject(s)
Adipose Tissue/cytology , Mesenchymal Stem Cells/cytology , Myocytes, Smooth Muscle/cytology , Stromal Cells/cytology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adipocytes/cytology , Biopsy , Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Differentiation , Cell Proliferation , Humans , Phenotype , Thromboxane A2/metabolism
10.
Cell Transplant ; 20(11-12): 1771-90, 2011.
Article in English | MEDLINE | ID: mdl-21439130

ABSTRACT

Development of a tissue-engineered neo-kidney augment (NKA) requires evaluation of defined, therapeutically relevant cell and cell/biomaterial composites (NKA constructs) for regenerative potential in mammalian kidney. Previous work identified primary renal cell populations that extended survival and improved renal function in a rodent model of chronic kidney disease (CKD). This study extends that work toward the goal of developing NKA by (i) screening in vivo inflammatory and fibrotic responses to acellular biomaterials delivered to healthy rodent renal parenchyma, (ii) evaluating the functionality of renal cell/biomaterial combinations in vitro, (iii) generating NKA constructs by combining therapeutically relevant cell populations with biocompatible biomaterial, and (iv) evaluating in vivo neokidney tissue development in response to NKA constructs delivered to healthy rodent renal parenchyma. Gelatin and hyaluronic acid (HA)-based hydrogels elicited the least inflammatory and fibrotic responses in renal parenchyma relative to polycaprolactone (PCL) and poly(lactic-co-glycolic acid) (PLGA) beads or particles and were associated with neovascularization and cellular infiltration by 4 weeks postimplantation. Renal cell populations seeded onto gelatin or HA-based hydrogels were viable and maintained a tubular epithelial functional phenotype during an in vitro maturation of 3 days as measured by transcriptomic, proteomic, secretomic, and confocal immunofluorescence assays. In vivo delivery of cell-seeded NKA constructs (bioactive renal cells + gelatin hydrogels) to healthy rodent renal parenchyma elicited neokidney tissue formation at 1 week postimplantation. To investigate a potential mechanism by which NKA constructs could impact a disease state, the effect of conditioned media on TGF-ß signaling pathways related to tubulo-interstitial fibrosis associated with CKD progression was evaluated. Conditioned medium was observed to attenuate TGF-ß-induced epithelial-mesenchymal transition (EMT) in vitro in a human proximal tubular cell line (HK2).


Subject(s)
Kidney/cytology , Tissue Engineering , Animals , Cell Adhesion , Cell Survival , Cells, Cultured , Dogs , Epithelial-Mesenchymal Transition/drug effects , Gelatin/chemistry , Gene Expression Profiling , Humans , Hydrogels/chemistry , Kidney/metabolism , Kidney/pathology , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Proteome/analysis , Rats , Rats, Inbred Lew , Transforming Growth Factor beta/pharmacology
11.
Appl Immunohistochem Mol Morphol ; 19(2): 184-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20930617

ABSTRACT

Bladder tissue has been regenerated in humans with neurogenic bladder using an implant produced from autologous urothelial (UC) and smooth muscle cells (SMC) expanded from bladder biopsies seeded onto a biodegradable synthetic scaffold. As the majority of bladder cancers are urothelial carcinomas (aka, transitional cell carcinoma), this 2-cell type autologous sourcing strategy presents significant challenges to product development. Entire bladders have been regenerated in cystectomized animals using a single-cell-type sourcing strategy: implants were seeded with bladder-derived SMC-only. Applying the bladder SMC-only sourcing strategy to produce clinical implants for bladder replacement or urinary diversion in bladder cancer patients requires methods for screening SMC cultures for the presence of potentially cancerous UC cells to provide evidence of SMC culture purity before seeding the scaffold. In this report, we show a 10-fold to 100-fold improvement in the sensitivity of qualitative and quantitative reverse-transcription PCR (qRT-PCR)-based assays for detecting UC positive for Cytokeratin 5 (CK5) in mixed SMC/UC cultures when the cell population was first subjected to magnetic activated cell sorting to enrich for cells expressing the epithelial cell adhesion molecule (known as EPCAM or CD326), a marker known to be present in normal UC and upregulated in the cancerous UC.


Subject(s)
Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Cell Adhesion Molecules/analysis , Keratin-5/analysis , Myocytes, Smooth Muscle/pathology , Urothelium/pathology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Culture Techniques , Epithelial Cell Adhesion Molecule , Flow Cytometry , Humans , Keratin-5/genetics , Keratin-5/metabolism , Magnetics , Myocytes, Smooth Muscle/metabolism , Organ Culture Techniques , Regeneration , Reverse Transcriptase Polymerase Chain Reaction/methods , Tissue Engineering/methods , Tissue Scaffolds , Transplantation, Autologous , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urinary Bladder, Neurogenic/genetics , Urinary Bladder, Neurogenic/metabolism , Urinary Bladder, Neurogenic/pathology , Urothelium/metabolism
12.
Tissue Eng Part C Methods ; 17(3): 261-73, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20846053

ABSTRACT

Chronic kidney disease (CKD) is a global health problem; the growing gap between the number of patients awaiting transplant and organs actually transplanted highlights the need for new treatments to restore renal function. Regenerative medicine is a promising approach from which treatments for organ-level disorders (e.g., neurogenic bladder) have emerged and translated to clinics. Regenerative templates, composed of biodegradable material and autologous cells, isolated and expanded ex vivo, stimulate native-like organ tissue regeneration after implantation. A critical step for extending this strategy from bladder to kidney is the ability to isolate, characterize, and expand functional renal cells with therapeutic potential from diseased tissue. In this study, we developed methods that yield distinct subpopulations of primary kidney cells that are compatible with process development and scale-up. These methods were translated to rodent, large mammal, and human kidneys, and then to rodent and human tissues with advanced CKD. Comparative in vitro studies demonstrated that phenotype and key functional attributes were retained consistently in ex vivo cultures regardless of species or disease state, suggesting that autologous sourcing of cells that contribute to in situ kidney regeneration after injury is feasible, even with biopsies from patients with advanced CKD.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Kidney Failure, Chronic/pathology , Kidney/cytology , Kidney/pathology , Adolescent , Adult , Animals , Biopsy , Cell Proliferation , Cells, Cultured , Dogs , Erythropoietin/metabolism , Female , Humans , Infant , Kidney/metabolism , Male , Middle Aged , Rats , Reproducibility of Results
13.
Am J Physiol Renal Physiol ; 299(5): F1026-39, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20826573

ABSTRACT

Established chronic kidney disease (CKD) may be identified by severely impaired renal filtration that ultimately leads to the need for dialysis or kidney transplant. Dialysis addresses only some of the sequelae of CKD, and a significant gap persists between patients needing transplant and available organs, providing impetus for development of new CKD treatment modalities. Some postulate that CKD develops from a progressive imbalance between tissue damage and the kidney's intrinsic repair and regeneration processes. In this study we evaluated the effect of kidney cells, delivered orthotopically by intraparenchymal injection to rodents 4-7 wk after CKD was established by two-step 5/6 renal mass reduction (NX), on the regeneration of kidney function and architecture as assessed by physiological, tissue, and molecular markers. A proof of concept for the model, cell delivery, and systemic effect was demonstrated with a heterogeneous population of renal cells (UNFX) that contained cells from all major compartments of the kidney. Tubular cells are known contributors to kidney regeneration in situ following acute injury. Initially tested as a control, a tubular cell-enriched subpopulation of UNFX (B2) surprisingly outperformed UNFX. Two independent studies (3 and 6 mo in duration) with B2 confirmed that B2 significantly extended survival and improved renal filtration (serum creatinine and blood urea nitrogen). The specificity of B2 effects was verified by direct comparison to cell-free vehicle controls and an equivalent dose of non-B2 cells. Quantitative histological evaluation of kidneys at 6 mo after treatment confirmed that B2 treatment reduced severity of kidney tissue pathology. Treatment-associated reduction of transforming growth factor (TGF)-ß1, plasminogen activator inhibitor (PAI)-1, and fibronectin (FN) provided evidence that B2 cells attenuated canonical pathways of profibrotic extracellular matrix production.


Subject(s)
Kidney Failure, Chronic/therapy , Kidney Tubules/cytology , Kidney/cytology , Animals , Blotting, Western , Cell Separation , Cell Transplantation , DNA/biosynthesis , DNA/genetics , Erythroid Cells , Flow Cytometry , Fluorescent Antibody Technique , Glomerular Filtration Rate/physiology , Homeostasis , Kidney/physiopathology , Kidney Failure, Chronic/physiopathology , Male , Nephrectomy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Recovery of Function , Survival , Y Chromosome/genetics , gamma-Glutamyltransferase/metabolism
14.
Biotechniques ; 48(2): 153-5, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20359300

ABSTRACT

Three-dimensional collagen gel contraction is the standard assay utilized for functionally quantifying a variety of cell types, in particular smooth muscle cells (SMCs) and myofibroblasts. Here, we have developed a method to effectively reduce the three-dimensional parameters of the standard collagen gel into a single, linear measurement. Cell/collagen suspensions that are cast into glass capillary tubes provide several advantages over the well plate format, such as eliminating the need for digital imaging equipment and software to quantify the amount of cellular contraction. In addition, capillary tube gels require significantly fewer cells and far less reagents than standard methods.


Subject(s)
Collagen/metabolism , Cytological Techniques/methods , Muscle Contraction/physiology , Myocytes, Smooth Muscle/physiology , Calcium/metabolism , Humans , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Urinary Bladder/cytology , Urinary Bladder/metabolism , Urinary Bladder/physiology
15.
Development ; 133(12): 2435-45, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16720880

ABSTRACT

In the mouse embryo, the splanchnic mesodermal cells of the anterior heart field (AHF) migrate from the pharynx to contribute to the early myocardium of the outflow tract (OT) and right ventricle (RV). Recent studies have attempted to distinguish the AHF from other precardiac populations, and to determine the genetic and molecular mechanisms that regulate its development. Here, we have used an Fgf8lacZ allele to demonstrate that Fgf8 is expressed within the developing AHF. In addition, we use both a hypomorphic Fgf8 allele (Fgf8neo) and Cre-mediated gene ablation to show that Fgf8 is essential for the survival and proliferation of the AHF. Nkx2.5Cre is expressed in the AHF, primary heart tube and pharyngeal endoderm, while TnT-Cre is expressed only within the specified heart tube myocardium. Deletion of Fgf8 by Nkx2.5Cre results in a significant loss of the Nkx2.5Cre lineage and severe OT and RV truncations by E9.5, while the remaining heart chambers (left ventricle and atria) are grossly normal. These defects result from significant decreases in cell proliferation and aberrant cell death in both the pharyngeal endoderm and splanchnic mesoderm. By contrast, ablation of Fgf8 in the TnT-Cre domain does not result in OT or RV defects, providing strong evidence that Fgf8 expression is crucial in the pharyngeal endoderm and/or overlying splanchnic mesoderm of the AHF at a stage prior to heart tube elongation. Analysis of downstream signaling components, such as phosphorylated-Erk and Pea3, identifies the AHF splanchnic mesoderm itself as a target for Fgf8 signaling.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Heart/anatomy & histology , Heart/embryology , Signal Transduction/physiology , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Branchial Region/anatomy & histology , Branchial Region/metabolism , Cardiovascular Abnormalities , Craniofacial Abnormalities , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/pathology , Embryo, Mammalian/physiology , Endoderm/cytology , Endoderm/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 8/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Heart/growth & development , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Knockout , Neural Crest/cytology , Neural Crest/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Development ; 132(17): 3847-57, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16049112

ABSTRACT

During kidney morphogenesis, the formation of nephrons begins when mesenchymal nephron progenitor cells aggregate and transform into epithelial vesicles that elongate and assume an S-shape. Cells in different regions of the S-shaped body subsequently differentiate into the morphologically and functionally distinct segments of the mature nephron. Here, we have used an allelic series of mutations to determine the role of the secreted signaling molecule FGF8 in nephrogenesis. In the absence of FGF8 signaling, nephron formation is initiated, but the nascent nephrons do not express Wnt4 or Lim1, and nephrogenesis does not progress to the S-shaped body stage. Furthermore, the nephron progenitor cells that reside in the peripheral zone, the outermost region of the developing kidney, are progressively lost. When FGF8 signaling is severely reduced rather than eliminated, mesenchymal cells differentiate into S-shaped bodies. However, the cells within these structures that normally differentiate into the tubular segments of the mature nephron undergo apoptosis, resulting in the formation of kidneys with severely truncated nephrons consisting of renal corpuscles connected to collecting ducts by an abnormally short tubular segment. Thus, unlike other FGF family members, which regulate growth and branching morphogenesis of the collecting duct system, Fgf8 encodes a factor essential for gene regulation and cell survival at distinct steps in nephrogenesis.


Subject(s)
Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/genetics , Nephrons/cytology , Nephrons/metabolism , Alleles , Animals , Cell Survival , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Mesoderm/metabolism , Mice , Mice, Transgenic , Nephrons/embryology , Phenotype , Signal Transduction , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Cord/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...