Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Publication year range
1.
PLoS One ; 14(10): e0223496, 2019.
Article in English | MEDLINE | ID: mdl-31596895

ABSTRACT

The N-type Ca2+ channel (Cav2.2) is distributed in sympathetic nerves that innervate the tubules, the vessels, and the juxtaglomerular granular cells of the kidney. However, the role of N-type Ca2+ channels in renal disease remains unknown. To address this issue, Cav2.2 knockout mice were utilized. Immunoreactive Cav2.2 was undetectable in normal kidneys of C57BL/6N mice, but it became positive in the interstitial S100-positive nerve fibers after unilateral ureteral obstruction (UUO). There were no significant differences in mean blood pressure, heart rate, and renal function between wild-type littermates and Cav2.2-knockout mice at baseline, as well as after UUO. Cav2.2 deficiency significantly reduced the EVG-positive fibrotic area, alpha-SMA expression, the production of type I collagen, and the hypoxic area in the obstructed kidneys. The expression of tyrosine hydroxylase, a marker for sympathetic neurons, was significantly increased in the obstructed kidneys of wild-type mice, but not in Cav2.2-knockout mice. These data suggest that increased Cav2.2 is implicated in renal nerve activation leading to the progression of renal fibrosis. Blockade of Cav2.2 might be a novel therapeutic approach for preventing renal fibrosis.


Subject(s)
Calcium Channels, N-Type/deficiency , Kidney Diseases/metabolism , Actins/genetics , Actins/metabolism , Animals , Calcium Channels, N-Type/genetics , Cell Hypoxia , Collagen Type I/genetics , Collagen Type I/metabolism , Fibrosis , Kidney/metabolism , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Ureteral Obstruction/complications
2.
Article in English | MEDLINE | ID: mdl-26161140

ABSTRACT

BACKGROUND: The process of epithelial-mesenchymal transition (EMT), which is generally defined by phenotypic changes of injured tubules such as loss of epithelial markers or acquisition of mesenchymal markers, implies various activating steps, including proliferation, migration, and ability to produce extracellular matrix proteins. We established here a novel approach for the detection of tubular cell migration into the interstitium during renal fibrosis in vivo. RESULTS: Using an osmotic pump, bromodeoxyuridine (BrdU) was continuously given to 7-week-old Wistar rats for 4 weeks, and BrdU-positive cells were detected by immunostaining. BrdU-positive cells were present in aquaporin-1-positive proximal tubules, but not in the interstitium of BrdU-treated rat kidneys. After unilateral ureteral obstruction (UUO), some BrdU-positive tubular cells protruded from the basement membrane and migrated into the interstitium. Interstitial BrdU-positive cells were co-localized with alpha-smooth muscle actin, fibroblast specific protein-1, vimentin, and type I collagen, but not with CD68 or CD3. No BrdU-positive cells were observed in the interstitium of sham-operated kidneys. The number of BrdU-positive cells migrating into the interstitium significantly increased and peaked at 8 days after UUO. CONCLUSIONS: Long-term BrdU labeling marked some of the proximal tubular cells and enabled us to detect tubular cell migration into the interstitium after UUO. This simple method might be useful to detect EMT in vivo.

3.
Biomed Res Int ; 2014: 376191, 2014.
Article in English | MEDLINE | ID: mdl-24883308

ABSTRACT

Activin, a member of the TGF-ß superfamily, regulates cell growth and differentiation in various cell types. Activin A acts as a negative regulator of renal development as well as tubular regeneration after renal injury. However, it remains unknown whether activin A is involved in renal fibrosis. To clarify this issue, we utilized a rat model of unilateral ureteral obstruction (UUO). The expression of activin A was significantly increased in the UUO kidneys compared to that in contralateral kidneys. Activin A was detected in glomerular mesangial cells and interstitial fibroblasts in normal kidneys. In UUO kidneys, activin A was abundantly expressed by interstitial α-SMA-positive myofibroblasts. Administration of recombinant follistatin, an activin antagonist, reduced the fibrotic area in the UUO kidneys. The number of proliferating cells in the interstitium, but not in the tubules, was significantly lower in the follistatin-treated kidneys. Expression of α-SMA, deposition of type I collagen and fibronectin, and CD68-positive macrophage infiltration were significantly suppressed in the follistatin-treated kidneys. These data suggest that activin A produced by interstitial fibroblasts acts as a potent profibrotic factor during renal fibrosis. Blockade of activin A action may be a novel approach for the prevention of renal fibrosis progression.


Subject(s)
Fibrosis/drug therapy , Follistatin/administration & dosage , Inhibin-beta Subunits/biosynthesis , Ureteral Obstruction/drug therapy , Animals , Cell Proliferation/drug effects , Fibrosis/genetics , Fibrosis/pathology , Humans , Inhibin-beta Subunits/antagonists & inhibitors , Inhibin-beta Subunits/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Rats , Transforming Growth Factor beta/genetics , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology
4.
Mod Rheumatol ; 24(4): 618-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24252014

ABSTRACT

OBJECTIVES: To examine the efficacy and safety of multi-target therapy using tacrolimus (TAC), mycophenolate mofetil (MMF) and a steroid as initial treatment for active lupus nephritis (LN). METHODS: We conducted a retrospective analysis of the data of 16 consecutive patients who received the multi-target therapy for active Classes III-V LN at our department. We also compared the outcomes of the multi-target therapy with those of TAC therapy (TAC + steroid), a study of which we had conducted previously in 13 patients with active LN (TAC group). RESULTS: All the patients treated with multi-target therapy achieved complete remission (CR) (mean, 4.6 ± 3.8 months; range, 1-15 months). The clinical profiles of the patients of the multi-target group were similar to those of the TAC group at baseline, except for a significantly higher level of proteinuria (4.6 ± 2.8 vs. 2.5 ± 2.1 g/gCr, p = 0.033) in the former. The CR rate at 6 months was significantly higher in the multi-target group as compared with that in the TAC group (81% vs. 38%, p = 0.018). Two cases of serious adverse events were associated with cytomegalovirus infection in the multi-target group, namely gastric ulcer and pancytopenia, both of which were successfully treated by antiviral therapy. CONCLUSIONS: Multi-target therapy was effective as initial treatment for active LN, with CR achieved early and in a high percentage of patients. Although this therapy was generally well tolerated, it is important to bear in mind the associated risk of cytomegalovirus infection.


Subject(s)
Immunosuppressive Agents/therapeutic use , Lupus Nephritis/drug therapy , Mycophenolic Acid/analogs & derivatives , Prednisolone/therapeutic use , Tacrolimus/therapeutic use , Adult , Drug Therapy, Combination , Female , Humans , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Mycophenolic Acid/adverse effects , Mycophenolic Acid/therapeutic use , Prednisolone/adverse effects , Remission Induction , Retrospective Studies , Tacrolimus/adverse effects , Treatment Outcome , Young Adult
5.
Am J Physiol Renal Physiol ; 304(6): F665-73, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23324177

ABSTRACT

N-type Ca(2+) channels are densely distributed in sympathetic nerves that innervate renal tubules. However, the role of N-type Ca(2+) channels in renal fibrosis remains unknown. To address this issue, we examined the difference between the effects of amlodipine (an L-type Ca(2+) channel blocker) and cilnidipine (a dual L/N-type Ca(2+) channel blocker) on fibrotic changes using a rat unilateral ureteral obstruction (UUO) model. The expression of both L-type and N-type Ca(2+) channels was significantly upregulated in UUO kidneys compared with that in contralateral kidneys. There were no significant differences in mean blood pressure among the rats tested. Both amlodipine and cilnidipine significantly attenuated fibrotic changes in UUO kidneys. The antifibrotic effect of cilnidipine was more potent than that of amlodipine. Amlodipine as well as cilnidipine reduced type III collagen deposition, α-smooth muscle actin (α-SMA) expression, and interstitial cell proliferation. In addition, cilnidipine significantly reduced deposition of type I collagen and macrophage infiltration in UUO kidneys. With the use of in vivo bromodeoxyuridine labeling, label-retaining cells (LRCs) were identified as a population of tubular cells that participate in epithelial-mesenchymal transition after UUO. Some LRCs migrated into the interstitium, expressed α-SMA and vimentin, and produced several extracellular matrixes in UUO kidneys. The number of interstitial LRCs was significantly decreased by cilnidipine but not amlodipine. These data suggest that N-type Ca(2+) channels contribute to multiple steps of renal fibrosis, and its blockade may thus be a useful therapeutic approach for prevention of renal fibrosis.


Subject(s)
Amlodipine/therapeutic use , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/metabolism , Dihydropyridines/therapeutic use , Ureteral Obstruction/metabolism , Actins/metabolism , Amlodipine/pharmacology , Animals , Cadherins/metabolism , Calcium Channels, L-Type/metabolism , Cell Proliferation/drug effects , Dihydropyridines/pharmacology , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Kidney/drug effects , Macrophages/drug effects , Male , Rats , Rats, Wistar , Ureteral Obstruction/drug therapy
7.
Am J Physiol Renal Physiol ; 302(6): F694-702, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22169012

ABSTRACT

Recovery after acute kidney injury is impaired in the elderly, but the precise mechanism for such age-related incompetence remains unclear. By in vivo bromodeoxyuridine (BrdU) labeling, renal progenitor cells (label-retaining cells; LRCs) were identified in tubules of normal rat kidney and were shown to be the origin of proliferating cells after injury. In the present study, the involvement of LRCs in the age-related decline of tubular recovery after injury was examined. After 1 wk of BrdU labeling followed by a 2-wk chase period, ischemia-reperfusion injury was induced in 7-wk-, 7-mo-, and 12-mo-old rats. Age-related decreases in DNA synthesis and cell proliferation in renal tubules after injury were found. The number of LRCs also significantly declined with age. At 24 h after reperfusion, the number of LRCs significantly increased in all ages of rats tested. There was no significant difference in the ratio of LRC division among rats of different ages. The area of the rat endothelial cell antigen (RECA)-1-positive capillary network declined with age. When renal tubules isolated from rats treated with BrdU label were cocultured with human umbilical vein endothelial cells (HUVEC), the number of LRCs significantly increased compared with tubules cultured without HUVEC. These data suggest that the reduced capacity of tubular regeneration in the aging kidney is partly explained by the shortage of LRC reserves. The size of the LRC pool might be regulated by the surrounding peritubular capillary network.


Subject(s)
Aging/physiology , Kidney/cytology , Kidney/physiology , Regeneration/physiology , Stem Cells/physiology , Animals , Bromodeoxyuridine , Cells, Cultured , Coculture Techniques , Endothelial Cells , Gene Expression Regulation , Humans , Kidney/injuries , Male , Rats , Rats, Wistar , Reperfusion Injury , Staining and Labeling
8.
Am J Physiol Renal Physiol ; 301(2): F387-95, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21561997

ABSTRACT

Renal proximal tubular epithelium can regenerate after various insults. To examine whether the tubular repair process is regulated by surrounding peritubular capillaries, we established an in vitro human tubulogenesis model that mimics in vivo tubular regeneration after injury. In this model, HGF, a potent renotropic factor, dose dependently induced tubular structures in human renal proximal tubular epithelial cells cultured in gels. Consistent with regenerating tubular cells after injury, HGF-induced tubular structures expressed a developmental gene, Pax-2, and a mesenchymal marker, vimentin, and formed a lumen with aquaporin-1 expression. Electron microscopic analysis showed the presence of microvilli on the apical site of the lumen, suggesting that these structures are morphologically equivalent to renal tubules in vivo. When cocultured with human umbilical vein endothelial cells (HUVEC), HGF-induced tubular formation was significantly enhanced. This could not be reproduced by the addition of VEGF, basic FGF, or PDGF. Protein array revealed that HUVEC produced various matrix metalloproteinases (MMPs). The stimulatory effects of coculture with HUVEC or HUVEC-derived conditional medium were almost completely abolished by addition of the tissue inhibitor of metalloproteinase (TIMP)-1 or TIMP-2. These data suggest that endothelial cell-derived factors including MMPs play a critical role in tubulogenesis and imply a potential role of peritubular capillary endothelium as a source of factor(s) required for tubular recovery after injury.


Subject(s)
Hepatocyte Growth Factor/physiology , Kidney Tubules, Proximal/physiology , Regeneration , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Ischemia/pathology , Kidney Tubules, Proximal/blood supply , Kidney Tubules, Proximal/pathology , Male , Matrix Metalloproteinases/metabolism , Rats , Rats, Wistar
9.
Endocr J ; 55(1): 1-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17827789

ABSTRACT

The research described in this review suggests a novel and important role for activin A in the developmental and repair processes of the kidney (Table 1). The results obtained in these studies indicate that activin A is a negative regulator of kidney development and plays an essential part in kidney diseases, such as acute renal failure or renal fibrosis. It is also possible that activin A is a key player in the pathophysiological processes of other kidney diseases, such as congenital urogenital abnormalities, renal cystic disease and renal cell carcinoma. Activin A is thus a potential target for therapeutic interventions in kidney diseases. To address this issue, more detailed analysis on the regulation of activin production, modulation of activin activity and activin target genes is required.


Subject(s)
Activins/physiology , Kidney/embryology , Kidney/physiology , Regeneration/physiology , Activin Receptors/physiology , Animals , Autocrine Communication/physiology , Follistatin/physiology , Humans , Kidney Diseases/etiology , Models, Biological , Organogenesis/genetics , Organogenesis/physiology , Ureter/embryology , Wolffian Ducts/embryology
SELECTION OF CITATIONS
SEARCH DETAIL