Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
J Clin Transl Sci ; 6(1): e13, 2022.
Article in English | MEDLINE | ID: mdl-35211339

ABSTRACT

INTRODUCTION: A national survey characterized training and career development for translational researchers through Clinical and Translational Science Award (CTSA) T32/TL1 programs. This report summarizes program goals, trainee characteristics, and mentorship practices. METHODS: A web link to a voluntary survey was emailed to 51 active TL1 program directors and administrators. Descriptive analyses were performed on aggregate data. Qualitative data analysis used open coding of text followed by an axial coding strategy based on the grounded theory approach. RESULTS: Fifty out of 51 (98%) invited CTSA hubs responded. Training program goals were aligned with the CTSA mission. The trainee population consisted of predoctoral students (50%), postdoctoral fellows (30%), and health professional students in short-term (11%) or year-out (9%) research training. Forty percent of TL1 programs support both predoctoral and postdoctoral trainees. Trainees are diverse by academic affiliation, mostly from medicine, engineering, public health, non-health sciences, pharmacy, and nursing. Mentor training is offered by most programs, but mandatory at less than one-third of them. Most mentoring teams consist of two or more mentors. CONCLUSIONS: CTSA TL1 programs are distinct from other NIH-funded training programs in their focus on clinical and translational research, cross-disciplinary approaches, emphasis on team science, and integration of multiple trainee types. Trainees in nearly all TL1 programs were engaged in all phases of translational research (preclinical, clinical, implementation, public health), suggesting that the CTSA TL1 program is meeting the mandate of NCATS to provide training to develop the clinical and translational research workforce.

2.
J Clin Transl Sci ; 4(6): 556-561, 2020 Jun 29.
Article in English | MEDLINE | ID: mdl-33942017

ABSTRACT

Clinical and Translational Science Award (CTSA) TL1 trainees and KL2 scholars were surveyed to determine the immediate impact of the COVID-19 pandemic on training and career development. The most negative impact was lack of access to research facilities, clinics, and human subjects, plus for KL2 scholars lack of access to team members and need for homeschooling. TL1 trainees reported having more time to think and write. Common strategies to maintain research productivity involved time management, virtual connections with colleagues, and shifting to research activities not requiring laboratory/clinic settings. Strategies for mitigating the impact of the COVID-19 pandemic on training and career development are described.

3.
Pediatr Res ; 82(5): 727-732, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28853726

ABSTRACT

The conduct of clinical trials in small pediatric subspecialties such as pediatric nephrology is hampered by both clinical demands on the pediatric nephrologist and the small number of appropriate patients available for such studies. The American Society of Pediatric Nephrology Therapeutics Development Committee (TDC) was established to (1) identify the various stakeholders with interests and/or expertise related to clinical trials in children with kidney disease and (2) develop more effective partnerships among all parties regarding strategies for successful clinical trial development and execution. This article discusses the rationale, structure, and function of the TDC, the status of progress toward its goals, and the insights gained to date that may be useful for other subspecialties that face similar challenges.


Subject(s)
Clinical Trials as Topic/methods , Kidney Diseases/therapy , Nephrology/methods , Pediatrics/methods , Research Design , Age Factors , Child , Clinical Trials as Topic/standards , Consensus , Humans , Kidney Diseases/diagnosis , Kidney Diseases/epidemiology , Nephrology/standards , Pediatrics/standards , Practice Guidelines as Topic , Research Design/standards , Stakeholder Participation , Treatment Outcome , Workflow
4.
Kidney Int ; 91(4): 781-783, 2017 04.
Article in English | MEDLINE | ID: mdl-28314579

ABSTRACT

In this issue, Bierzynska et al. report the results of analyzing a cohort of children with steroid-resistant nephrotic syndrome (SRNS) of childhood, correlating genetic studies with clinical outcomes. This approach has the potential to advance both our research into the causes and treatments of childhood SRNS and our ability to stratify patients into groups that are likely to be responsive to specific treatments. Here, we discuss some of the strengths and limitations of this report.


Subject(s)
Nephrotic Syndrome , Cohort Studies , Humans
5.
Adv Chronic Kidney Dis ; 24(2): 107-116, 2017 03.
Article in English | MEDLINE | ID: mdl-28284376

ABSTRACT

Accumulating evidence suggests that the central locus for the progression of CKD is the renal proximal tubule. As injured tubular epithelial cells dedifferentiate in attempted repair, they stimulate inflammation and recruit myofibroblasts. At the same time, tissue loss stimulates remnant nephron hypertrophy. Increased tubular transport workload eventually exceeds the energy-generating capacity of the hypertrophied nephrons, leading to anerobic metabolism, acidosis, hypoxia, endoplasmic reticulum stress, and the induction of additional inflammatory and fibrogenic responses. The result is a vicious cycle of injury, misdirected repair, maladaptive responses, and more nephron loss. Therapy that might be advantageous at one phase of this progression pathway could be deleterious during other phases. Thus, interrupting this downward spiral requires narrowly targeted approaches that promote healing and adequate function without generating further entry into the progression cycle.


Subject(s)
Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Nephritis, Interstitial/complications , Nephritis, Interstitial/physiopathology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/physiopathology , Acute Kidney Injury/complications , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Cell Dedifferentiation , Disease Progression , Extracellular Matrix/metabolism , Fibrosis , Homeostasis , Humans , Hyperplasia/pathology
6.
Kidney Int ; 90(4): 797-808, 2016 10.
Article in English | MEDLINE | ID: mdl-27503806

ABSTRACT

The function of hypoxia-inducible factor-1α (HIF-1α) in chronic kidney disease is disputed. Here we report that interactions of HIF-1α with transforming growth factor-ß (TGF-ß) signaling may promote its fibrotic effects. Knockout of HIF-1α is protective against glomerulosclerosis and glomerular type-I collagen accumulation in a mouse podocyte ablation model. Transcriptional analysis of cultured renal cells showed that α2(I) collagen expression is directly regulated by HIF-1α binding to a functional hypoxia-responsive element in its promoter at -335 relative to the transcription start site. Activation of COL1A2 transcription by HIF-1α occurred in the absence of hypoxia and is strongly enhanced by TGF-ß signaling. TGF-ß, in addition to increasing HIF-1α levels, increased both HIF-1α binding to the COL1A2 promoter and HIF-1α N-terminal transactivation domain activity. These effects of TGF-ß on HIF-1α were inhibited in Smad3-null mouse embryonic fibroblasts, suggesting a requirement for Smad3. Phosphorylated Smad3 also associated with the -335 hypoxia-responsive element of the COL1A2 promoter independent of a Smad DNA binding sequence. Smad3 binding to the -335 hypoxia-responsive element required HIF-1α both in vitro and in kidney lysate from the disease model, suggesting formation of an HIF-1α-Smad3 transcriptional complex. Thus, HIF-1α-Smad3 has a novel interaction in glomerulosclerosis.


Subject(s)
Collagen Type I/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Glomerulus/pathology , Renal Insufficiency, Chronic/pathology , Smad3 Protein/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Podocytes , Promoter Regions, Genetic , Renal Insufficiency, Chronic/metabolism , Sclerosis , Signal Transduction , Smad3 Protein/genetics , Transforming Growth Factor beta1/metabolism
7.
PLoS One ; 11(5): e0155534, 2016.
Article in English | MEDLINE | ID: mdl-27187580

ABSTRACT

Fibrosis is a final common pathway leading to loss of kidney function, in which the fibrogenic cytokine, transforming growth factor ß (TGF-ß), plays a central role. While previous studies showed that TGF-ß antagonism by various means prevents fibrosis in mouse models, clinical approaches based on these findings remain elusive. 1D11 is a neutralizing antibody to all three isoforms of TGF-ß. In both adriamycin (ADR)-induced nephropathy and NEP25 podocyte ablation nephropathy, thrice-weekly intraperitoneal administration of 1D11 from the day of disease induction until the mice were sacrificed (day 14 for ADR and day 28 for NEP25), significantly reduced glomerular COL1A2 mRNA accumulation and histological changes. Consistent with our previous findings, proteinuria remained overt in the mice treated with 1D11, suggesting distinct mechanisms for proteinuria and fibrogenesis. Podocyte numbers determined by WT1 staining were significantly reduced in NEP25-model glomeruli as expected, while WT1-positive cells were preserved in mice receiving 1D11. Even when 1D11 was administered after the onset of proteinuria on day 3, 1D11 preserved WT1-positive cell numbers in glomeruli and significantly reduced glomerular scar score (2.5 ± 0.2 [control IgG] vs. 1.8 ± 0.2 [1D11], P < 0.05) and glomerular COL1A2 mRNA expression (19.3 ± 4.4 [control IgG] vs. 8.4 ± 2.4 [1D11] fold increase over the healthy control, P < 0.05). Transmission electron microscopy revealed loss of podocytes and denuded glomerular basement membrane in NEP25 mice with disease, whereas podocytes remained attached to the basement membrane, though effaced and swollen, in those receiving 1D11 from day 3. Together, these data suggest that TGF-ß neutralization by 1D11 prevents glomerular fibrosis even when started after the onset of proteinuria. While overt proteinuria and podocyte effacement persist, 1D11 prevents total podocytes detachment, which might be a key event activating fibrogenic events in glomeruli.


Subject(s)
Antibodies, Monoclonal/pharmacology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Proteinuria/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Biomarkers , Disease Models, Animal , Doxorubicin/adverse effects , Fibrosis , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/ultrastructure , Male , Mice , Podocytes/metabolism , Podocytes/ultrastructure , Proteinuria/drug therapy , Proteinuria/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
9.
Pediatr Nephrol ; 30(2): 193-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25370779

ABSTRACT

At a recent meeting of international experts on clinical aspects of progressive chronic kidney disease (CKD), an extensive analysis of extant clinical trials was used to develop more effective and economical surrogate markers for CKD outcomes in adults. This article describes the reasons for this undertaking, the methods and conclusions of the meeting, and the relevance of these findings to pediatric nephrology.


Subject(s)
Biomarkers/analysis , Renal Insufficiency, Chronic/pathology , Adult , Child , Disease Progression , Female , Glomerular Filtration Rate , Humans , Male , Renal Insufficiency, Chronic/drug therapy , Treatment Outcome
10.
Cell Signal ; 26(10): 2276-83, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24905473

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling plays an important and complex role in renal fibrogenesis. The seemingly simple TGF-ß/Smad cascade is intensively regulated at several levels, including crosstalk with other signaling pathways. Epidermal growth factor (EGF) is a potent mitogen for epithelial cells and is elevated in diseased kidneys. In this study, we examined its effect on TGF-ß-induced fibrotic changes in human proximal tubular epithelial cells. Simultaneous treatment with EGF specifically inhibited basal and TGF-ß-induced type-I collagen and α-smooth muscle actin (αSMA) expression at both mRNA and protein levels. These effects were prevented by inhibition of either the EGF receptor kinase or its downstream MEK kinase but not by blockade of either the JNK or PI3K pathway. Overexpression of a constitutively active MEK1 construct mimicked the inhibitory effect of EGF. Further, EGF suppressed Smad transcriptional activities, as shown by reduced activation of ARE-luc and SBE-luc. Both reductions were prevented by MEK inhibition. However, EGF did not block Smad2 or Smad3 phosphorylation by TGF-ß, or Smad2/3 nuclear import. Finally EGF induced the phosphorylation and expression of TGIF, a known TGF-ß/Smad repressor. Both the phosphorylation and the induction were blocked by a MEK inhibitor. Overexpression of TGIF abolished TGF-ß-induced αSMA promoter activity. Together these results suggest that EGF inhibits two TGF-ß-stimulated markers of EMT through EGF receptor tyrosine kinase and downstream ERK activation, but not through PI3K or JNK. The inhibition results from effector mechanisms downstream of Smads, and most likely involves the transcriptional repressor, TGIF.


Subject(s)
Epidermal Growth Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Transforming Growth Factor beta1/pharmacology , Actins/genetics , Actins/metabolism , Cell Line , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Enzyme Activation/drug effects , Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Homeodomain Proteins/metabolism , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Phosphorylation , Promoter Regions, Genetic , Repressor Proteins/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism
11.
Pediatr Nephrol ; 29(2): 193-202, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23715783

ABSTRACT

In chronic kidney disease, ongoing failure of individual nephrons leads to the progressive loss of renal function. This process results in part from a cellular and molecular response to injury that represents an attempt to maintain homeostasis but instead initiates a program that damages the nephron. As nephrons are lost, compensation by the remaining nephrons exacerbates glomerular pathophysiology. The delivery of excessive amounts of biologically active molecules to the distal nephron and tubulointerstitium generates inflammation and cellular dedifferentiation. Energy requirements of hyperfunctioning nephrons exceed the metabolic substrate available to the renal tubule, and inadequacy of the local vascular supply promotes hypoxia/ischemia and consequent acidosis and reactive oxygen species generation. In this way, mechanisms activated to maintain biological balance ultimately lead to demise of the nephron.


Subject(s)
Nephrons/pathology , Nephrons/physiopathology , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Disease Progression , Humans
12.
Exp Cell Res ; 319(19): 2928-37, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24080014

ABSTRACT

Regulation of TGF-ß1/Smad3 signaling in fibrogenesis is complex. Previous work by our lab suggests that ERK MAP kinase phosphorylates the linker region (LR) of Smad3 to enhance TGF-ß-induced collagen-I accumulation. However the roles of the individual Smad3LR phosphorylation sites (T179, S204, S208 and S213) in the collagen-I response to TGF-ß are not clear. To address this issue, we tested the ability of Smad3 constructs expressing wild-type Smad3 or Smad3 with mutated LR phosphorylation sites to reconstitute TGF-ß-stimulated COL1A2 promoter activity in Smad3-null or -knockdown cells. Blocking ERK in fibroblasts and renal mesangial cells inhibited both S204 phosphorylation and Smad3-mediated COL1A2 promoter activity. Mutations replacing serine at S204 or S208 in the linker region decreased Smad3-mediated COL1A2 promoter activity, whereas mutating T179 enhanced basal COL1A2 promoter activity and did not prevent TGF-ß stimulation. Interestingly, mutation of all four Smad3LR sites (T179, S204, S208 and S213) was not inhibitory, suggesting primacy of the two inhibitory sites. These results suggest that in these mesenchymal cells, phosphorylation of the T179 and possibly S213 sites may act as a brake on the signal, whereas S204 phosphorylation by ERK in some manner releases that brake. Renal epithelial cells (HKC) respond differently from MEF or mesangial cells; blocking ERK neither changed TGF-ß-stimulated S204 phosphorylation nor prevented Smad3-mediated COL1A2 promoter activity in HKC. Furthermore, re-expression of wild type-Smad3 or the S204A-Smad3 mutant in Smad3-knockdown HKC reconstituted Smad3-mediated COL1A2 promoter activity. Collectively, these data suggest that Serine-204 phosphorylation in the Smad3LR is a critical event by which ERK enhances Smad3-mediated COL1A2 promoter activity in mesenchymal cells.


Subject(s)
Collagen Type I/metabolism , Serine/metabolism , Smad3 Protein/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Collagen Type I/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mutation/genetics , Phenotype , Signal Transduction/physiology , Trans-Activators/genetics , Transcriptional Activation/physiology , Transforming Growth Factor beta/genetics
13.
Am J Physiol Renal Physiol ; 305(9): F1323-31, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23946285

ABSTRACT

Hypoxia-inducible factors (HIFs) are transcription factors consisting of an oxygen-sensitive α-subunit binding to a stable ß-subunit. HIFs regulate multiple signaling pathways that could contribute to fibrogenesis, supporting their potential role in hypoxia-mediated renal fibrosis. We previously reported that HIF-1 is upregulated and required for transforming growth factor (TGF)-ß induction of collagen in renal tubular cells. Here, we performed in vitro and in vivo studies of potential glomerular crosstalk between TGF-ß and normoxic HIF signaling. HIF-α has two major isoforms, HIF-1α and HIF-2α with different target gene sets. In cultured human mesangial cells, TGF-ß1 treatment increased both HIF-1α and HIF-2α expression in normoxia. TGF-ß1 did not increase HIF-1α/2α mRNA levels nor decrease the rate of protein degradation, suggesting that it enhances HIF-1α/2α expression through translation. TGF-ß receptor (ALK5) kinase activity was required for increased, TGF-ß-stimulated HIF-α expression in response to TGF-ß, and inhibiting PI3-kinase markedly decreased HIF-α expression. Blocking HIF-1α/2α expression using siRNA decreased basal and TGF-ß1-stimulated type I collagen expression, while overexpressing nondegradable HIF-α increased the collagen response, with HIF-2α being significantly more effective than HIF-1α. In adriamycin-induced mouse glomerulosclerosis, HIF-2α target genes were upregulated in sclerosing glomeruli. Taken together, our data demonstrate potential signaling interaction between TGF-ß and HIFs to promote renal fibrogenesis in normoxia and suggest that the HIF-2α isoform is more important during glomerulosclerosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Collagen/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mesangial Cells/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , Cells, Cultured , Fibrosis , Glomerulosclerosis, Focal Segmental/metabolism , Humans , Male , Mice , Mice, 129 Strain , Oxygen , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism
14.
J Cell Sci ; 126(Pt 16): 3697-712, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23781022

ABSTRACT

Increased intraglomerular pressure is an important pathogenic determinant of kidney fibrosis in the progression of chronic kidney disease, and can be modeled by exposing glomerular mesangial cells (MC) to mechanical stretch. MC produce extracellular matrix and profibrotic cytokines, including connective tissue growth factor (CTGF) when stretched. We show that p21-activated kinase 1 (Pak1) is activated by stretch in MC in culture and in vivo in a process marked by elevated intraglomerular pressures. Its activation is essential for CTGF upregulation. Rac1 is an upstream regulator of Pak1 activation. Stretch induces transactivation of the type I transforming growth factor ß1 receptor (TßRI) independently of ligand binding. TßRI transactivation is required not only for Rac1/Pak1 activation, but also for activation of the canonical TGFß signaling intermediate Smad3. We show that Smad3 activation is an essential requirement for CTGF upregulation in MC under mechanical stress. Pak1 regulates Smad3 C-terminal phosphorylation and transcriptional activation. However, a second signaling pathway, that of RhoA/Rho-kinase and downstream Erk activation, is also required for stretch-induced CTGF upregulation in MC. Importantly, this is also regulated by Pak1. Thus, Pak1 serves as a novel central mediator in the stretch-induced upregulation of CTGF in MC.


Subject(s)
Connective Tissue Growth Factor/metabolism , Mesangial Cells/metabolism , Receptors, Transforming Growth Factor beta/metabolism , p21-Activated Kinases/metabolism , Animals , Connective Tissue Growth Factor/genetics , Humans , Mesangial Cells/physiology , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, Transforming Growth Factor beta/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Signal Transduction , Transcriptional Activation , Up-Regulation , p21-Activated Kinases/genetics
15.
Am J Physiol Renal Physiol ; 305(4): F485-94, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23761672

ABSTRACT

Transforming growth factor (TGF)-ß is a major mediator of kidney fibrosis. In the past decade it was recognized that, besides canonical Smad signaling, many other signaling pathways participate in the process of TGF-ß-induced fibrogenesis. One such pathway involves mammalian target of rapamycin complex (mTORC)1. We recently reported that the hypoxia-inducible factor (HIF)-1 is essential for TGF-ß-induced collagen expression regardless of ambient oxygen tension. A modulator of HIF expression other than oxygen tension is mTORC1. We therefore sought to evaluate a possible role for mTORC1 activity in TGF-ß-induced fibrogenesis. mTORC1 activity was increased in human mesangial cells treated with TGF-ß in a TGF-ß receptor-dependent manner. Short hairpin (sh)RNA to Smad3 decreased, while overexpression of Smad3 increased, the mTORC1 activity, suggesting that TGF-ß stimulation of mTORC1 also requires Smad3. Pretreatment with rapamycin or shRNA for a regulatory molecule of mTORC1, Raptor, reduced TGF-ß-induced COL1A2-luc activity and collagen I protein expression. mTORC1 inhibition also prevented the TGF-ß-stimulated increase in both hypoxia-responsive element (HRE) activity and HIF-1α protein expression, while activation of mTORC1 by active Rheb increased basal but not TGF-ß-induced HRE activity. shRNA to Smad3 reduced HRE activity, while overexpression of Smad3 increased HIF-1α protein expression and activity in an mTORC1-dependent manner. Lastly, overexpression of HIF-1α bypassed the inhibitory effect of mTORC1 blockade on collagen expression. These results suggest that Smad3/mTORC1 interaction to promote HIF-1 expression is a key step in normoxic kidney fibrogenesis.


Subject(s)
Collagen/metabolism , Fibrosis/metabolism , Glomerular Mesangium/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Multiprotein Complexes/metabolism , Smad3 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism , Blotting, Western , Cell Culture Techniques , Collagen/drug effects , Glomerular Mesangium/cytology , Humans , Hypoxia/metabolism , Immunoprecipitation , Mechanistic Target of Rapamycin Complex 1 , RNA, Small Interfering/genetics , Signal Transduction , Sirolimus/pharmacology , Transfection , Transforming Growth Factor beta/pharmacology
16.
J Biol Chem ; 287(43): 35815-24, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22942286

ABSTRACT

SARA has been shown to be a regulator of epithelial cell phenotype, with reduced expression during TGF-ß1-mediated epithelial-to-mesenchymal transition. Examination of the pathways that might play a role in regulating SARA expression identified phosphatidylinositol 3-kinase (PI3K) pathway inhibition as sufficient to reduce SARA expression. The mechanism of PI3K inhibition-mediated SARA down-regulation differs from that induced by TGF-ß1 in that, unlike TGF-ß1, PI3K-dependent depletion of SARA was apparent within 6 h and did not occur at the mRNA or promoter level but was blocked by inhibition of proteasome-mediated degradation. This effect was independent of Akt activity because neither reducing nor enhancing Akt activity modulated the expression of SARA. Therefore, this is likely a direct effect of p85α action, and co-immunoprecipitation of SARA and p85α confirmed that these proteins interact. Both SARA and PI3K have been shown to be associated with endosomes, and either LY294002 or p85α knockdown enlarged SARA-containing endocytic vesicles. Inhibition of clathrin-mediated endocytosis blocked SARA down-regulation, and a localization-deficient mutant SARA was protected against down-regulation. As inhibiting PI3K can activate the endosomal fusion-regulatory small GTPase Rab5, we expressed GTPase-deficient Rab5 and observed endosomal enlargement and reduced SARA protein expression, similar to that seen with PI3K inhibition. Importantly, either interference with PI3K via LY294002 or p85α knockdown, or constitutive activity of the Rab5 pathway, enhanced the expression of smooth muscle α-actin. Together, these data suggest that although TGF-ß1 can induce epithelial-to-mesenchymal transition through reduction in SARA expression, SARA is also basally regulated by its interaction with PI3K.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Epithelial-Mesenchymal Transition/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Serine Endopeptidases/metabolism , Transforming Growth Factor beta1/metabolism , rab5 GTP-Binding Proteins/metabolism , Actins/biosynthesis , Actins/genetics , Cell Line , Chromones/pharmacology , Class Ia Phosphatidylinositol 3-Kinase/genetics , Endocytosis/drug effects , Endocytosis/physiology , Enzyme Inhibitors/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Serine Endopeptidases/genetics , Time Factors , Transforming Growth Factor beta1/genetics , rab5 GTP-Binding Proteins/genetics
17.
Kidney Int ; 82(5): 525-36, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22534961

ABSTRACT

Multiple transforming growth factor (TGF)-ß-induced fibrogenic signals have been described in vitro. To evaluate mechanisms in vivo, we used an adriamycin nephropathy model in 129x1/Svj mice that display massive proteinuria by days 5 to 7 and pathological findings similar to human focal segmental glomerulosclerosis by day 14. TGF-ß mRNA expression increased after day 7 along with nuclear translocation of the TGF-ß receptor-specific transcription factor Smad3. Inhibiting TGF-ß prevented both pathological changes and type-I collagen and fibronectin mRNA expression, but proteinuria persisted. Renal Akt was phosphorylated in adriamycin-treated mice, suggesting PI3-kinase activation. Expression of mRNA for the p110γ isozyme of PI3-kinase was specifically increased and p110γ colocalized with nephrin by immunohistochemistry early in disease. Nephrin levels subsequently decreased. Inhibition of p110γ by AS605240 preserved nephrin expression and prevented proteinuria. In cultured podocytes, adriamycin stimulated p110γ expression. AS605240, but not a TGF-ß receptor kinase inhibitor, prevented adriamycin-induced cytoskeletal disorganization and apoptosis, supporting a role for p110γ in podocyte injury. AS605240, at a dose that decreased proteinuria, prevented renal collagen mRNA expression in vivo but did not affect TGF-ß-stimulated collagen induction in vitro. Thus, PI3-kinase p110γ mediates initial podocyte injury and proteinuria, both of which precede TGF-ß-mediated glomerular scarring.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Doxorubicin , Glomerulosclerosis, Focal Segmental/enzymology , Kidney/enzymology , Proteinuria/enzymology , Signal Transduction , Smad3 Protein/metabolism , Animals , Apoptosis , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Collagen Type I/genetics , Collagen Type I/metabolism , Disease Models, Animal , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/prevention & control , Immunohistochemistry , Kidney/drug effects , Kidney/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Phosphorylation , Podocytes/enzymology , Podocytes/pathology , Protein Kinase Inhibitors/pharmacology , Proteinuria/chemically induced , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Time Factors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation
18.
Pediatr Nephrol ; 27(6): 901-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21947270

ABSTRACT

In chronic kidney disease (CKD), once injury from any number of disease processes reaches a threshold, there follows an apparently irreversible course toward decline in kidney function. The tubulointerstitium may play a key role in this common progression pathway. Direct injury, high metabolic demands, or stimuli from various other forms of renal dysfunction activate tubular cells. These, in turn, interact with interstitial tissue elements and inflammatory cells, causing further pathologic changes in the renal parenchyma. The tissue response to these changes thus generates a feed-forward loop of kidney injury and progressive loss of function. This article reviews the mechanisms of this negative cycle mediating CKD.


Subject(s)
Kidney Diseases/pathology , Kidney Tubules/pathology , Nephritis, Interstitial/pathology , Cell Communication , Chronic Disease , Disease Progression , Feedback, Physiological , Fibrosis , Humans , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Kidney Tubules/metabolism , Kidney Tubules/physiopathology , Nephritis, Interstitial/metabolism , Signal Transduction
19.
Am J Physiol Renal Physiol ; 300(4): F898-905, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21209004

ABSTRACT

Increasing evidence suggests that chronic kidney disease may develop following acute kidney injury and that this may be due, in part, to hypoxia-related phenomena. Hypoxia-inducible factor (HIF) is stabilized in hypoxic conditions and regulates multiple signaling pathways that could contribute to renal fibrosis. As transforming growth factor (TGF)-ß is known to mediate renal fibrosis, we proposed a profibrotic role for cross talk between the TGF-ß1 and HIF-1α signaling pathways in kidney cells. Hypoxic incubation increased HIF-1α protein expression in cultured human renal tubular epithelial cells and mouse embryonic fibroblasts. TGF-ß1 treatment further increased HIF-1α expression in cells treated with hypoxia and also increased HIF-1α in normoxic conditions. TGF-ß1 did not increase HIF-1α mRNA levels nor decrease the rate of protein degradation, suggesting that it enhances normoxic HIF-1α translation. TGF-ß receptor (ALK5) kinase activity was required for increased HIF-1α expression in response to TGF-ß1, but not to hypoxia. A dominant negative Smad3 decreased the TGF-ß-stimulated reporter activity of a HIF-1α-sensitive hypoxia response element. Conversely, a dominant negative HIF-1α construct decreased Smad-binding element promoter activity in response to TGF-ß. Finally, blocking HIF-1α transcription with a biochemical inhibitor, a dominant negative construct, or gene-specific knockdown decreased basal and TGF-ß1-stimulated type I collagen expression, while HIF-1α overexpression increased both. Taken together, our data demonstrate cooperation in signaling between Smad3 and HIF-1α and suggest a new paradigm in which HIF-1α is necessary for normoxic, TGF-ß1-stimulated renal cell fibrogenesis.


Subject(s)
Collagen/metabolism , Epithelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Kidney/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Blotting, Western , Cell Line , Cells, Cultured , Epithelial Cells/drug effects , Humans , Kidney/cytology , Kidney/drug effects , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection , Transforming Growth Factor beta/pharmacology
20.
J Biol Chem ; 285(40): 30741-51, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20650890

ABSTRACT

Transforming growth factor ß (TGF-ß) promotes tissue fibrosis via the receptor-specific Smad pathway and non-canonical pathways. We recently reported that TGF-ß1-stimulated collagen expression by cultured kidney cells requires integrin-dependent activation of focal adhesion kinase (FAK) and consequent ERK MAP kinase activity leading to Smad3 linker region phosphorylation. Here, we defined a role for αvß3-integrin in this non-canonical pathway. A human kidney tubular cell line in which ß1-integrin was knocked down (ß1-k/d) demonstrated enhanced type I collagen mRNA expression and promoter activity. A second shRNA to either αv-integrin or ß3-integrin, but not to another αv-binding partner, ß6-integrin, abrogated the enhanced COL1A2 promoter activity in ß1-k/d cells. Although αvß3-integrin surface expression levels were not different, αvß3-integrins colocalized with sites of focal adhesion significantly more in ß1-k/d cells, and activated αvß3-integrin was detected only in ß1-k/d cells. Further, the collagen response was decreased by a function-blocking antibody or a peptide inhibitor of αvß3-integrin. In cells lacking αvß3-integrin, the responses were attenuated, whereas the response was enhanced in αvß3-overexpressing cells. Rac1 and ERK, previously defined mediators for this non-canonical pathway, showed increased activities in ß1-k/d cells. Finally, inhibition of αvß3-integrin decreased Rac1 activity and COL1A2 promoter activity in ß1-k/d cells. Together, our results indicate that decreasing ß1 chain causes αvß3-integrin to become functionally dominant and promotes renal cell fibrogenesis via Rac1-mediated ERK activity.


Subject(s)
Collagen/biosynthesis , Gene Expression Regulation , Integrin alphaVbeta3/metabolism , Integrin beta Chains/metabolism , Kidney Tubules/metabolism , Transforming Growth Factor beta1/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Line , Collagen/genetics , Collagen Type I , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/genetics , Focal Adhesions/metabolism , Gene Knockdown Techniques , Humans , Integrin alphaVbeta3/genetics , Integrin beta Chains/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...