Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 320(3): F359-F374, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33427061

ABSTRACT

Lysophosphatidic acid (LPA) increases platelet-derived growth factor-B (PDGFB) and connective tissue growth factor (CTGF) production and secretion by proximal tubule (PT) cells through LPA2 receptor-Gqα-αvß6-integrin-mediated activation of transforming growth factor-ß1 (TGFB1). LPA2, ß6-integrin, PDGFB, and CTGF increase in kidneys after ischemia-reperfusion injury (IRI), coinciding with fibrosis. The TGFB1 receptor antagonist SD-208 prevents increases of ß6-integrin, TGFB1-SMAD signaling, and PDGFB/CTGF expression after IRI and ameliorates fibrosis (Geng H, Lan R, Singha PK, Gilchrist A, Weinreb PH, Violette SM, Weinberg JM, Saikumar P, Venkatachalam MA. Am J Pathol 181: 1236-1249, 2012; Geng H, Lan R, Wang G, Siddiqi AR, Naski MC, Brooks AI, Barnes JL, Saikumar P, Weinberg JM, Venkatachalam MA. Am J Pathol 174: 1291-1308, 2009). We report now that LPA1 receptor signaling through epidermal growth factor receptor (EGFR)-ERK1/2-activator protein-1 cooperates with LPA2-dependent TGFB1 signaling to additively increase PDGFB/CTGF production and secretion by PT cells. Conversely, inhibition of both pathways results in greater suppression of PDGFB/CTGF production and secretion and promotes greater PT cellular differentiation than inhibiting one pathway alone. Antagonism of the LPA-generating enzyme autotaxin suppressed signaling through both pathways. After IRI, kidneys showed not only more LPA2, nuclear SMAD2/3, and PDGFB/CTGF but also increased LPA1 and autotaxin proteins, together with enhanced EGFR/ERK1/2 activation. Remarkably, the TGFB1 receptor antagonist SD-208 prevented all of these abnormalities excepting increased LPA2. SD-208 inhibits only one arm of LPA signaling: LPA2-Gqα-αvß6-integrin-dependent production of active TGFB1 and its receptor-bound downstream effects. Consequently, far-reaching protection by SD-208 against IRI-induced signaling alterations and tubule-interstitial pathology is not fully explained by our data. TGFB1-dependent feedforward modulation of LPA1 signaling is one possibility. SD-208 effects may also involve mitigation of injury caused by IRI-induced TGFB1 signaling in endothelial cells and monocytes. Our results have translational implications for using TGFB1 receptor antagonists, LPA1 and LPA2 inhibitors concurrently, and autotaxin inhibitors in acute kidney injury to prevent the development of chronic kidney disease.


Subject(s)
Acute Kidney Injury/metabolism , Cytokines/metabolism , Kidney Tubules, Proximal/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Reperfusion Injury/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Cell Line , Connective Tissue Growth Factor/metabolism , Disease Models, Animal , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney Tubules, Proximal/pathology , Lymphokines/metabolism , Male , Mice , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Rats, Sprague-Dawley , Receptors, Lysophosphatidic Acid/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/metabolism
2.
Am J Physiol Renal Physiol ; 318(5): F1086-F1099, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32174143

ABSTRACT

Unilateral ischemia-reperfusion (UIR) injury leads to progressive renal atrophy and tubulointerstitial fibrosis (TIF) and is commonly used to investigate the pathogenesis of the acute kidney injury-chronic kidney disease transition. Although it is well known that contralateral nephrectomy (CNX), even 2 wk post-UIR injury, can improve recovery, the physiological mechanisms and tubular signaling pathways mediating such improved recovery remain poorly defined. Here, we examined the renal hemodynamic and tubular signaling pathways associated with UIR injury and its reversal by CNX. Male Sprague-Dawley rats underwent left UIR or sham UIR and 2 wk later CNX or sham CNX. Blood pressure, left renal blood flow (RBF), and total glomerular filtration rate were assessed in conscious rats for 3 days before and over 2 wk after CNX or sham CNX. In the presence of a contralateral uninjured kidney, left RBF was lower (P < 0.05) from 2 to 4 wk following UIR (3.6 ± 0.3 mL/min) versus sham UIR (9.6 ± 0.3 mL/min). Without CNX, extensive renal atrophy, TIF, and tubule dedifferentiation, but minimal pimonidazole and hypoxia-inducible factor-1α positivity in tubules, were present at 4 wk post-UIR injury. Conversely, CNX led (P < 0.05) to sustained increases in left RBF (6.2 ± 0.6 mL/min) that preceded the increases in glomerular filtration rate. The CNX-induced improvement in renal function was associated with renal hypertrophy, more redifferentiated tubules, less TIF, and robust pimonidazole and hypoxia-inducible factor-1α staining in UIR injured kidneys. Thus, contrary to expectations, indexes of hypoxia are not observed with the extensive TIF at 4 wk post-UIR injury in the absence of CNX but are rather associated with the improved recovery of renal function and structure following CNX.


Subject(s)
Acute Kidney Injury/physiopathology , Kidney/blood supply , Renal Circulation , Renal Insufficiency, Chronic/etiology , Reperfusion Injury/physiopathology , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Animals , Atrophy , Cell Hypoxia , Disease Models, Animal , Disease Progression , Fibrosis , Hemodynamics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/metabolism , Kidney/pathology , Male , Nephrectomy , Rats, Sprague-Dawley , Recovery of Function , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Time Factors , Vascular Endothelial Growth Factor A/metabolism
3.
Genes Cancer ; 10(5-6): 134-149, 2019.
Article in English | MEDLINE | ID: mdl-31798766

ABSTRACT

Screening of several TNBC cell lines showed altered Smad2 and Smad3 protein levels compared to normal mammary epithelial cells, suggesting the possibility that it could play an important role in the escape of cancer cells from TGF-ß mediated growth inhibition. To assess the functional relevance of these endogenous molecules, Smad2 or Smad3 expression was knocked down individually and assessed their effects on pro-oncogenic properties of TGF-ß. Smad3 deficiency reduced growth and invasion capacity of breast cancer cells in comparison to Smad2 which had no effect. Smad3 deficiency was also found to be associated with a reduction in the expressions of TMEPAI/PMEPA1 and EMT inducing transcription factors, E-Cadherin and increased expression of cell cycle inhibitors and Vimentin. On the other hand, Smad2 deficiency had opposite effect on these regulators. Interestingly, the decreased growth, invasion and associated gene expressions were largely reversed by overexpressing TMEPAI in Smad3 knockdown cells, suggesting that Smad3-TMEPAI axis may be involved in subverting growth suppressive effects of TGF-ß into growth promotion. Similarly, altered levels of Smad proteins and TMEPAI were also noted in primary TNBC tumor tissues. Analysis of the existing databases provided additional support in terms of TMEPAI and Smad2 expression impacting the survival of TNBC patients. Taken together, our data demonstrate a novel role for Smad3 in cancer transformation and cancer progression through TMEPAI and further suggest that selective targeting of TGF-ß-Smad3-TMEPAI axis may be beneficial in triple negative breast cancer therapy and prevention.

4.
J Am Soc Nephrol ; 27(11): 3356-3367, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27000065

ABSTRACT

During recovery by regeneration after AKI, proximal tubule cells can fail to redifferentiate, undergo premature growth arrest, and become atrophic. The atrophic tubules display pathologically persistent signaling increases that trigger production of profibrotic peptides, proliferation of interstitial fibroblasts, and fibrosis. We studied proximal tubules after ischemia-reperfusion injury (IRI) to characterize possible mitochondrial pathologies and alterations of critical enzymes that govern energy metabolism. In rat kidneys, tubules undergoing atrophy late after IRI but not normally recovering tubules showed greatly reduced mitochondrial number, with rounded profiles, and large autophagolysosomes. Studies after IRI of kidneys in mice, done in parallel, showed large scale loss of the oxidant-sensitive mitochondrial protein Mpv17L. Renal expression of hypoxia markers also increased after IRI. During early and late reperfusion after IRI, kidneys exhibited increased lactate and pyruvate content and hexokinase activity, which are indicators of glycolysis. Furthermore, normally regenerating tubules as well as tubules undergoing atrophy exhibited increased glycolytic enzyme expression and inhibitory phosphorylation of pyruvate dehydrogenase. TGF-ß antagonism prevented these effects. Our data show that the metabolic switch occurred early during regeneration after injury and was reversed during normal tubule recovery but persisted and became progressively more severe in tubule cells that failed to redifferentiate. In conclusion, irreversibility of the metabolic switch, taking place in the context of hypoxia, high TGF-ß signaling and depletion of mitochondria characterizes the development of atrophy in proximal tubule cells and may contribute to the renal pathology after AKI.


Subject(s)
Acute Kidney Injury/complications , Glycolysis , Ischemia/complications , Kidney Tubules, Proximal/pathology , Kidney/blood supply , Mitochondria/metabolism , Mitochondrial Diseases/etiology , Animals , Atrophy/etiology , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
5.
Genes Cancer ; 5(9-10): 320-36, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25352949

ABSTRACT

TMEPAI (transmembrane prostate androgen-induced) is amplified at genomic, transcript and protein levels in triple-negative breast cancers and promotes TGF-ß dependent growth, motility and invasion. Tumor promotion by TMEPAI depends on two different but related actions on TGF-ß signaling. Firstly, TMEPAI binds and sequesters regulatory Smads2/3 and thereby decreases growth suppressive signaling by TGF-ß. Secondly, increased expression of TMEPAI decreases PTEN (phosphatase and tensin homolog) abundance, and thereby increases TGF-ß dependent tumor promotive PI3K/Akt signaling. These actions of TMEPAI give rise to increased cell proliferation and motility. Moreover, signaling alterations produced by high TMEPAI were associated with oncogenic Snail expression and lung metastases. Finally, an inverse correlation between TMEPAI and PTEN levels was confirmed in triple negative breast cancer tumor samples. Together, our findings suggest that TMEPAI has dually critical roles to promote TGF-ß dependent cancer cell growth and metastasis. Thus, redirected TGF-ß signaling through TMEPAI may play a pivotal role in TGF-ß mediated tumor promotion.

6.
J Am Soc Nephrol ; 25(7): 1496-507, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24511135

ABSTRACT

Preexisting CKD may affect the severity of and/or recovery from AKI. We assessed the impact of prior graded normotensive renal mass reduction on ischemia-reperfusion-induced AKI. Rats underwent 40 minutes of ischemia 2 weeks after right uninephrectomy and surgical excision of both poles of the left kidney (75% reduction of renal mass), right uninephrectomy (50% reduction of renal mass), or sham reduction of renal mass. The severity of AKI was comparable among groups, which was reflected by similarly increased serum creatinine (SCr; approximately 4.5 mg/dl) at 2 days, tubule necrosis at 3 days, and vimentin-expressing regenerating tubules at 7 days postischemia-reperfusion. However, SCr remained elevated compared with preischemia-reperfusion values, and more tubules failed to differentiate during late recovery 4 weeks after ischemia-reperfusion in rats with 75% renal mass reduction relative to other groups. Tubules that failed to differentiate continued to produce vimentin, exhibited vicarious proliferative signaling, and expressed less vascular endothelial growth factor but more profibrotic peptides. The disproportionate failure of regenerating tubules to redifferentiate in rats with 75% renal mass reduction associated with more severe capillary rarefaction and greater tubulointerstitial fibrosis. Furthermore, initially normotensive rats with 75% renal mass reduction developed hypertension and proteinuria, 2-4 weeks postischemia-reperfusion. In summary, severe (>50%) renal mass reduction disproportionately compromised tubule repair, diminished capillary density, and promoted fibrosis with hypertension after ischemia-reperfusion-induced AKI in rats, suggesting that accelerated declines of renal function may occur after AKI in patients with preexisting CKD.


Subject(s)
Acute Kidney Injury/etiology , Kidney Tubules/pathology , Kidney/anatomy & histology , Animals , Fibrosis , Kidney/surgery , Male , Organ Size , Rats , Rats, Sprague-Dawley , Severity of Illness Index
7.
Am J Pathol ; 181(4): 1236-49, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22885106

ABSTRACT

After ischemia-reperfusion injury (IRI), kidney tubules show activated transforming growth factor ß (TGF-ß) signaling and increased expression of profibrotic peptides, platelet-derived growth factor-B (PDGF-B) and connective tissue growth factor (CTGF). If tubule repair after IRI is incomplete, sustained paracrine activity of these peptides can activate interstitial fibroblast progenitors and cause fibrosis. We show that lysophosphatidic acid (LPA), a ubiquitous phospholipid that is increased at sites of injury and inflammation, signals through LPA2 receptors and Gαq proteins of cultured proximal tubule cells to transactivate latent TGF-ß in a Rho/Rho-kinase and αvß6 integrin-dependent manner. Active TGF-ß peptide then initiates signaling to increase the production and secretion of PDGF-B and CTGF. In a rat model of IRI, increased TGF-ß signaling that was initiated early during reperfusion did not subside during recovery, but progressively increased, causing tubulointerstitial fibrosis. This was accompanied by correspondingly increased LPA2 and ß6 integrin proteins and elevated tubule expression of TGF-ß1, together with PDGF-B and CTGF. Treatment with a pharmacological TGF-ß type I receptor antagonist suppressed TGF-ß signaling, decreased the expression of ß6 integrin, PDGF-B, and CTGF, and ameliorated fibrosis. We suggest that LPA-initiated autocrine signaling is a potentially important mechanism that gives rise to paracrine profibrotic signaling in injured kidney tubule cells.


Subject(s)
Antigens, Neoplasm/metabolism , Cytokines/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Integrins/metabolism , Kidney Tubules, Proximal/metabolism , Lysophospholipids/pharmacology , Receptors, Lysophosphatidic Acid/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Cytokines/genetics , Fibrosis , Gene Expression Regulation/drug effects , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Lipids/blood , Male , Mice , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Regeneration/drug effects , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Smad2 Protein/metabolism , Time Factors , Transforming Growth Factor beta/metabolism
8.
Am J Physiol Renal Physiol ; 302(9): F1210-23, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22301622

ABSTRACT

We investigated the signaling basis for tubule pathology during fibrosis after renal injury. Numerous signaling pathways are activated physiologically to direct tubule regeneration after acute kidney injury (AKI) but several persist pathologically after repair. Among these, transforming growth factor (TGF)-ß is particularly important because it controls epithelial differentiation and profibrotic cytokine production. We found that increased TGF-ß signaling after AKI is accompanied by PTEN loss from proximal tubules (PT). With time, subpopulations of regenerating PT with persistent loss of PTEN (phosphate and tension homolog) failed to differentiate, became growth arrested, expressed vimentin, displayed profibrotic JNK activation, and produced PDGF-B. These tubules were surrounded by fibrosis. In contrast, PTEN recovery was associated with epithelial differentiation, normal tubule repair, and less fibrosis. This beneficial outcome was promoted by TGF-ß antagonism. Tubule-specific induction of TGF-ß led to PTEN loss, JNK activation, and fibrosis even without prior AKI. In PT culture, high TGF-ß depleted PTEN, inhibited differentiation, and activated JNK. Conversely, TGF-ß antagonism increased PTEN, promoted differentiation, and decreased JNK activity. Cre-Lox PTEN deletion suppressed differentiation, induced growth arrest, and activated JNK. The low-PTEN state with JNK signaling and fibrosis was ameliorated by contralateral nephrectomy done 2 wk after unilateral ischemia, suggesting reversibility of the low-PTEN dysfunctional tubule phenotype. Vimentin-expressing tubules with low-PTEN and JNK activation were associated with fibrosis also after tubule-selective AKI, and with human chronic kidney diseases of diverse etiology. By preventing tubule differentiation, the low-PTEN state may provide a platform for signals initiated physiologically to persist pathologically and cause fibrosis after injury.


Subject(s)
Cell Differentiation , Kidney Tubules, Proximal/pathology , MAP Kinase Kinase 4/physiology , PTEN Phosphohydrolase/deficiency , Phenotype , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Cells, Cultured , Chronic Disease , Fibrosis , Humans , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney Tubules, Proximal/physiopathology , Male , Mice , Mice, Transgenic , Models, Animal , Rats , Rats, Sprague-Dawley , Regeneration/physiology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology
9.
Am J Physiol Renal Physiol ; 298(5): F1078-94, 2010 May.
Article in English | MEDLINE | ID: mdl-20200097

ABSTRACT

Recently published epidemiological and outcome analysis studies have brought to our attention the important role played by acute kidney injury (AKI) in the progression of chronic kidney disease (CKD) to end-stage renal disease (ESRD). AKI accelerates progression in patients with CKD; conversely, CKD predisposes patients to AKI. This research gives credence to older, well-thought-out wisdom that recovery from AKI is often not complete and is marked by residual structural damage. It also mirrors older experimental observations showing that unilateral nephrectomy, a surrogate for loss of nephrons by disease, compromises structural recovery and worsens tubulointerstitial fibrosis after ischemic AKI. Moreover, review of a substantial body of work on the relationships among reduced renal mass, hypertension, and pathology associated with these conditions suggests that impaired myogenic autoregulation of blood flow in the setting of hypertension, the arteriolosclerosis that results, and associated recurrent ischemic AKI in microscopic foci play important roles in the development of progressively increasing tubulointerstitial fibrosis. How nutrition, an additional factor that profoundly affects renal disease progression, influences these events needs reevaluation in light of information on the effects of calories vs. protein and animal vs. vegetable protein on injury and progression. Considerations based on published and emerging data suggest that a pathology that develops in regenerating tubules after AKI characterized by failure of differentiation and persistently high signaling activity is the proximate cause that drives downstream events in the interstitium: inflammation, capillary rarefaction, and fibroblast proliferation. In light of this information, we advance a comprehensive hypothesis regarding the pathophysiology of AKI as it relates to the progression of kidney disease. We discuss the implications of this pathophysiology for developing efficient therapeutic strategies to delay progression and avert ESRD.


Subject(s)
Acute Kidney Injury/physiopathology , Disease Progression , Kidney Diseases/physiopathology , Kidney Failure, Chronic/physiopathology , Acute Kidney Injury/complications , Animals , Chronic Disease , Disease Models, Animal , Fibrosis , Humans , Kidney/pathology , Kidney Diseases/etiology , Kidney Failure, Chronic/etiology
10.
Wound Repair Regen ; 18(2): 159-67, 2010.
Article in English | MEDLINE | ID: mdl-20230600

ABSTRACT

We describe the fabrication and use of an in vitro wounding device that denudes cultured epithelium in patterns designed to leave behind strips or islands of cells sufficiently narrow or small to ensure that all the remaining cells become rapidly activated and then migrate, dedifferentiate, and proliferate in near synchrony. The design ensures that signals specific to regenerating cells do not become diluted by quiescent differentiated cells that are not affected by wound-induced activation. The device consists of a flat circular disk of rubber, engraved to produce alternating ridges and grooves in patterns of concentric circles or parallel lines. The disk is mounted at the end of a pneumatically controlled piston assembly. Application of controlled pressure and circular or linear movement of the disk on cultures produced highly reproducible wounding patterns. The near-synchronous regenerative activity of cell bands or islands allowed the collection of samples large enough for biochemical studies to sensitively detect alterations involving mRNA for several early response genes and protein phosphorylation in major signaling pathways. The method is versatile, easy to use and reproducible, and should facilitate biochemical, proteomic, and genomic studies of wound-induced regeneration of cultured epithelium.


Subject(s)
Epithelium/physiology , Regeneration , Wound Healing/physiology , Animals , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , Genes, fos , Genes, jun , Microscopy , Polymerase Chain Reaction , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/metabolism
11.
Am J Pathol ; 174(4): 1291-308, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19342372

ABSTRACT

We studied autocrine transforming growth factor (TGF)beta signaling in kidney epithelium. Cultured proximal tubule cells showed regulated signaling that was high during log-phase growth, low during contact-inhibited differentiation, and rapidly increased during regeneration of wounded epithelium. Autoregulation of signaling correlated with TGFbeta receptor and Smad7 levels, but not with active TGFbeta, which was barely measurable in the growth medium. Confluent differentiated cells with low receptor and high Smad7 levels exhibited blunted responses to saturating concentrations of exogenously provided active TGFbeta, suggesting that TGFbeta signaling homeostasis was achieved by cell density-dependent modulation of signaling intermediates. Antagonism of Alk5 kinase, the TGFbeta type I receptor, dramatically accelerated the induction of differentiation in sparse, proliferating cultures and permitted better retention of differentiated features in regenerating cells of wounded, confluent cultures. Alk5 antagonism accelerated the differentiation of cells in proximal tubule primary cultures while simultaneously increasing their proliferation. Consequently, Alk5-inhibited primary cultures formed confluent, differentiated monolayers faster than untreated cultures. Furthermore, treatment with an Alk5 antagonist promoted kidney repair reflected by increased tubule differentiation and decreased tubulo-interstitial pathology during the recovery phase following ischemic injury in vivo. Our results show that autocrine TGFbeta signaling in proliferating proximal tubule cells exceeds the levels that are necessary for physiological regeneration. To that end, TGFbeta signaling is redundant and maladaptive during tubule repair by epithelial regeneration.


Subject(s)
Cell Differentiation/physiology , Kidney Tubules, Proximal/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Wound Healing/physiology , Activin Receptors/antagonists & inhibitors , Animals , Cell Proliferation , Epithelium/metabolism , Epithelium/pathology , Homeostasis/physiology , Ischemia/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Protein Serine-Threonine Kinases , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL
...