Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
2.
Lik Sprava ; (2): 49-53, 2001.
Article in Ukrainian | MEDLINE | ID: mdl-11519431

ABSTRACT

A comprehensive evaluation has been done of parameters characterizing cell-bound immunity, cytokinic profile, stress hormones (hydrocortisone), and C-reactive protein in a comparative aspect in those patients having undergone laparoscopic or conventional colectomy. The above indices were determined before surgery, in the early postoperative period, 24 h and 7 days subsequent to surgery. Laparoscopic surgical intervention was found out to lower somewhat the level of immunosuppression early in the postoperative course, which fact is manifested by retaining of expression HLA-DR on monocytes of the peripheral blood. Further randomized investigations should allow the final judgement about the degree of surgical stress.


Subject(s)
Homeostasis/immunology , Intestinal Diseases/immunology , Intestine, Large/immunology , Adult , Aged , C-Reactive Protein/analysis , Colectomy , Cytokines/blood , Female , HLA-DR Antigens/blood , Humans , Hydrocortisone/blood , Intestinal Diseases/surgery , Intestine, Large/surgery , Laparoscopy , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Postoperative Period , Prospective Studies , Time Factors
3.
Kidney Int ; 58(6): 2291-300, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11115063

ABSTRACT

BACKGROUND: Genetic ablation of cyclooxygenase-2 (COX-2) resulted in cystic renal dysplasia and early death in adult mice. The ontologic development of the renal pathology and the biochemical and physiological abnormalities associated with the dysplasia are unknown. METHODS: Mice homozygous for a targeted deletion of COX-2 (-/-) were compared with wild-type littermates (+/+). Somatic and kidney growth and renal histology were studied at the day of birth and at a number of postnatal ages. Systolic blood pressure, urinalysis, urine osmolality, serum and urine chemistries, and inulin clearance were evaluated in adult animals. RESULTS: Beginning at postnatal day 10 (PN10), kidney growth was suppressed in -/- animals, while somatic growth and heart growth were unaffected. By PN10, -/- kidneys had thin nephrogenic cortexes and crowded, small, subcapsular glomeruli. The pathology increased with age with progressive outer cortical dysplasia, cystic subcapsular glomeruli, loss of proximal tubular mass, and tubular atrophy and cyst formation. Adult -/- kidneys had profound diffuse tubular cyst formation, outer cortical glomerular hypoplasia and periglomerular fibrosis, inner cortical nephron hypertrophy, and diffuse interstitial fibrosis. The glomerular filtration rate was reduced by more than 50% in -/- animals (6.82 +/- 0.65 mL/min/kg) compared with wild-type controls (14.7 +/- 1.01 mL/min/kg, P < 0. 001). Plasma blood urea nitrogen and creatinine were elevated in null animals compared with controls. Blood pressure, urinalysis, urine osmolality, and other plasma chemistries were unaffected by the deletion of COX-2. CONCLUSIONS: Deficiency of COX-2 results in progressive and specific renal architectural disruption and functional deterioration beginning in the final phases of nephrogenesis. Tissue-specific and time-dependent expression of COX-2 appears necessary for normal postnatal renal development and the maintenance of normal renal architecture and function.


Subject(s)
Isoenzymes/genetics , Kidney/abnormalities , Multicystic Dysplastic Kidney/enzymology , Multicystic Dysplastic Kidney/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Animals , Blood Pressure , Blood Urea Nitrogen , Creatinine/blood , Creatinine/urine , Cyclooxygenase 2 , Disease Models, Animal , Disease Progression , Drinking , Electrolytes/blood , Electrolytes/urine , Female , Genotype , Glomerular Filtration Rate , Inulin/pharmacokinetics , Kidney/enzymology , Kidney/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multicystic Dysplastic Kidney/pathology , Organ Size , Osmolar Concentration , Phenotype , Pregnancy , Urinalysis , Urine/chemistry
4.
Transplantation ; 70(2): 288-92, 2000 Jul 27.
Article in English | MEDLINE | ID: mdl-10933150

ABSTRACT

BACKGROUND: A successful kidney transplant from a living-related donor (LRD) remains the most effective renal replacement therapy for children with end-stage renal failure. The use of LRD kidneys results in decreased time on dialysis, increased graft survival, and better function compared with kidneys transplanted from cadaver donors. We retrospectively analyzed data from the United Network of Organ Sharing (UNOS) Scientific Renal Transplant Registry to determine risk factors for graft loss in children who received an LRD kidney. METHODS: Data was obtained from the UNOS Scientific Renal Transplant Registry on 2418 children ranging in age from 0 to 18 years who underwent an LRD kidney transplantation between January 1988 and December 1994. Multivariate analysis of graft survival was performed using Kaplan-Meier and Cox regression models. RESULTS: The effects of age, pretransplantation dialysis, early rejection, and race were found to significantly affect graft survival. Gender, peak panel-reactive antibody, and ABO blood type were not found to be significant risk factors. Infants <2 years of age initially had the worst graft survival; however, over time their results stabilized, and at 7 years estimated graft survival was good (71%). Adolescents ranging in age from 13-18 years had the best initial graft survival, but as time went on graft survival worsened (55%). Patients who underwent pretransplantation dialysis had a relative risk for graft loss of 1.77 (P<0.001), whereas those who had an early rejection had a relative risk for graft loss of 1.41 (P<0.002). African-Americans had a significantly higher relative risk for graft loss than either Caucasians (1.57, P<0.0005) or Hispanics (2.01, P<0.0003). CONCLUSIONS: Predictors of graft survival for children who receive LRD kidney transplants include age at transplantation, pretransplantation dialysis, early rejection, and race. Over time, adolescents and African-Americans seem to have the lowest graft survival.


Subject(s)
Graft Survival , Kidney Transplantation/immunology , Living Donors , Adolescent , Child , Child, Preschool , Female , Graft Rejection , Humans , Infant , Kidney Failure, Chronic/surgery , Kidney Transplantation/mortality , Male , Multivariate Analysis , Parents , Racial Groups , Regression Analysis , Renal Dialysis , Survival Rate , Time Factors
5.
Am J Physiol Regul Integr Comp Physiol ; 279(2): R522-30, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10938241

ABSTRACT

To evaluate the presence and regulation of the renin-angiotensin system (RAS) in metanephric organ culture, embryonic day 14 (E14) rat metanephroi were cultured for 6 days. mRNAs for renin and both ANG II receptors (AT(1) and AT(2)) are expressed at E14, and all three genes continue to be expressed in culture. Renin mRNA is localized to developing tubules and ureteral branches in the cultured explants. At E14, renin immunostaining is found in isolated cells scattered within the mesenchyme. As differentiation progresses, renin localizes to the ureteric epithelium, developing tubules and glomeruli. E14 metanephroi contain ANG II, and peptide production persists in culture. Renin activity is present at E14 (6.13 +/- 0.61 pg ANG I. kidney(-1). h(-1)) and in cultured explants (28.84 +/- 1. 13 pg ANG I. kidney(-1). h(-1)). Renin activity in explants is increased by ANG II treatment (70.1 +/- 6.36 vs. 40.97 +/- 1.94 pg ANG I. kidney(-1). h(-1) in control). This increase is prevented by AT(1) blockade, whereas AT(2) antagonism has no effect. These studies document an operational local RAS and a previously undescribed positive-feedback mechanism for renin generation in avascular, cultured developing metanephroi. This novel expression pattern and regulatory mechanism highlight the unique ability of developing renal cells to express an active RAS.


Subject(s)
Kidney/embryology , Renin-Angiotensin System/physiology , Angiotensin II/metabolism , Animals , Embryo, Mammalian/metabolism , Embryo, Mammalian/physiology , Immunohistochemistry , In Situ Hybridization , Organ Culture Techniques , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Receptors, Angiotensin/physiology , Renin/genetics , Renin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
Pediatr Nephrol ; 14(4): 328-31, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10775080

ABSTRACT

Hypocomplementemic urticarial vasculitis syndrome (HUVS) is well described in adults but is quite rare in children. We report a pediatric case of HUVS initially diagnosed as juvenile rheumatoid arthritis and then as Henoch-Schönlein purpura. Beginning at 3 years of age, our patient developed polyarthritis with hypocomplementemia. She subsequently experienced an intermittent purpuric rash beginning at age 4 years, and she continued to have episodic arthritis and rash for years. Hematuria and proteinuria were noted at 12 years of age; renal biopsy revealed membranoproliferative glomerulonephritis with membranous features. Serum complement evaluation revealed activation of the classical pathway, consistent with HUVS. Therapy with oral dapsone led to improvement in proteinuria. HUVS should be considered in the differential diagnosis of pediatric patients with glomerulonephritis, urticarial rash, arthritis/arthralgias, and obstructive pulmonary disease.


Subject(s)
Complement System Proteins/analysis , Urticaria/complications , Vasculitis/blood , Vasculitis/complications , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis/complications , Arthritis/drug therapy , Child , Dapsone/therapeutic use , Female , Glomerulonephritis, Membranoproliferative/complications , Glomerulonephritis, Membranoproliferative/pathology , Humans , Kidney/pathology , Proteinuria/complications , Proteinuria/drug therapy , Syndrome , Urticaria/drug therapy
7.
J Am Soc Nephrol ; 10(10): 2125-34, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10505689

ABSTRACT

The expression of vascular endothelial growth factor (VEGF) and its receptors Flt-1 and Flk-1 in the rat kidney was examined during ontogeny using Northern blot analysis and immunocytochemistry. In prevascular embryonic kidneys (embryonic day 14 [E14]), immunoreactive Flt-1 and Flk-1 were observed in isolated angioblasts, whereas VEGF was not detected. Angioblasts aligned forming cords before morphologically differentiating into endothelial cells. In late fetal kidneys (E19), immunoreactive VEGF was detected in glomerular epithelial and tubular cells, whereas Flt-1 and Flk-1 were expressed in contiguous endothelial cells. To determine whether VEGF induces endothelial cell differentiation and vascular development in the kidney, the effect of recombinant human VEGF (5 ng/ml) was examined on rat metanephric organ culture, a model known to recapitulate nephrogenesis in the absence of vessels. After 6 d in culture in serum-free, defined media, metanephric kidney growth and morphology were assessed. DNA content was higher in VEGF-treated explants (1.9 +/- 0.17 microg/kidney, n = 9) than in paired control explants (1.4 +/- 0.10 microg/kidney, n = 9) (P < 0.05). VEGF induced proliferation of tubular epithelial cells, as indicated by an increased number of tubules and tubular proliferating cell nuclear antigen-containing cells. VEGF induced upregulation of Flk-1 and Flt-1 expression, as assessed by Western blot analysis. Developing endothelial cells were identified and localized using immunocytochemistry and electron microscopy. Flt-1, Flk-1, and angiotensin-converting enzyme-containing cells were detected in VEGF-treated explants, whereas control explants were negative. These studies confirmed previous reports indicating that the expression of VEGF and its receptors is temporally and spatially associated with kidney vascularization and identified angioblasts expressing Flt-1 and Flk-1 in prevascular embryonic kidneys. The data indicate that VEGF expression is downregulated in standard culture conditions and that VEGF stimulates growth of embryonic kidney explants by expanding both endothelium and epithelium, resulting in vasculogenesis and enhanced tubulogenesis. These data suggest that VEGF plays a critical role in renal development by promoting endothelial cell differentiation, capillary formation, and proliferation of tubular epithelia.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelium, Vascular/embryology , Kidney/embryology , Lymphokines/metabolism , Neovascularization, Physiologic , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Cell Differentiation , Culture Techniques , Embryonic and Fetal Development/physiology , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Female , Humans , Immunohistochemistry , Lymphokines/genetics , Lymphokines/pharmacology , Molecular Sequence Data , Polymerase Chain Reaction , Pregnancy , RNA, Messenger/analysis , Rats , Reference Values , Sensitivity and Specificity , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Ren Fail ; 21(3-4): 283-91, 1999.
Article in English | MEDLINE | ID: mdl-10416205

ABSTRACT

Kidney morphogenesis is accomplished by the coordinated interaction of molecular signals that culminate in the production of an organ that is architecturally and functionally ready for extrauterine, free life. In humans, nephrogenesis is completed before birth. However the kidney continues to mature both from a functional and anatomical point of view. Throughout its development, the kidney is susceptible to a variety of injurious agents. This brief review considers the basic mechanisms of kidney organogenesis and functional maturation. To illustrate some concepts, the renal alterations caused by interference with a normal regulatory system, the renin-angiotensin system is discussed.


Subject(s)
Kidney Diseases/physiopathology , Kidney/embryology , Kidney/growth & development , Animals , Humans , Kidney/drug effects , Kidney/physiology , Kidney Diseases/chemically induced
9.
Curr Opin Pediatr ; 11(2): 135-40, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10202623

ABSTRACT

Anatomical development of the kidney is achieved by the reciprocal induction of the ureteric bud and the metanephric mesenchyma. This interaction triggers the process of nephrogenesis and culminates in the formation of the mature kidney. In vivo, nephrogenesis is coordinated with renal vascularization. In fact, vascular precursors, epithelial progenitors, and mesenchymal cells communicate with one another in a highly organized fashion. As a result of this complex interaction, a mature kidney, architecturally and functionally ready for extrauterine life, is produced. This review deals with the relevant molecules and mechanisms governing nephrovascular development.


Subject(s)
Kidney/embryology , Humans , Kidney/anatomy & histology , Kidney/blood supply , Kidney/growth & development , Renal Circulation
10.
Am J Physiol ; 273(5): F731-8, 1997 11.
Article in English | MEDLINE | ID: mdl-9374836

ABSTRACT

Renal juxtaglomerular (JG) cells are specialized myoepithelioid cells located in the afferent arteriole at the entrance to the glomerulus. Their main function and distinctive feature is the synthesis and release of renin, the key hormone-enzyme of the renin-angiotensin system that regulates arterial blood pressure. Despite their relevance to health and disease, not much is known about factors that confer and/or maintain JG cell identity. To identify genes uniquely expressed in JG cells, we used a cell culture model and RNA differential display. JG cells cultured for 2 days express renin and renin mRNA, but after 10 days in culture they no longer contain or release renin and renin mRNA is reduced 700-fold. We report one cDNA differentially expressed in the 2-day JG cell culture that detects a 2.6-kb mRNA expressed at higher levels in newborn than adult kidney. Screening a 2-day culture JG cell cDNA library yielded clones representing differentially spliced transcripts. These cDNAs encode one unique protein (Zis) containing zinc fingers and domains characteristic of splicing factors and RNA binding proteins. Northern blot analysis confirmed Zis mRNA expression in differentiated JG cells, and identified an additional unique 1.5-kb transcript. The Zis transcripts are developmentally regulated in kidney and a number of other organs. The features of the Zis protein and its organ distribution suggest a possible role in regulation of transcription and/or splicing, both important steps for controlling developmentally expressed genes.


Subject(s)
Aging/metabolism , Gene Expression Regulation, Developmental , Juxtaglomerular Apparatus/metabolism , Kidney/metabolism , RNA-Binding Proteins/biosynthesis , Transcription, Genetic , Amino Acid Sequence , Animals , Animals, Newborn , Base Sequence , Cells, Cultured , Cloning, Organism , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/growth & development , Male , Molecular Sequence Data , Organ Specificity , RNA Splicing , RNA, Messenger/biosynthesis , RNA-Binding Proteins/chemistry , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Renin/biosynthesis , Zinc Fingers
11.
Microsc Res Tech ; 39(3): 254-60, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9372498

ABSTRACT

Renal vascularization and nephrogenesis occur simultaneously following a tightly regulated developmental program influenced by growth factors, extracellular matrix components and cell membrane receptors. Both processes of angiogenesis and vasculogenesis probably participate in the formation of renal vessels. The origin and fate of the various renal vascular cells and the molecular mechanisms that initiate and guide intrarenal vascularization are fundamental questions that remain to be answered.


Subject(s)
Kidney/blood supply , Kidney/embryology , Animals , Blood Vessels/embryology , Humans , Morphogenesis , Neovascularization, Physiologic
12.
Semin Nephrol ; 17(5): 492-501, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9316216

ABSTRACT

All the components of the renin-angiotensin system (RAS) are highly expressed in the developing kidney in a unique spatial and temporal pattern that is associated with nephrogenesis, vascularization, and the proper architectural and functional development of this organ. Pharmacological inhibition of the RAS results in structural and functional developmental abnormalities of the kidneys in several animal species, including humans. Similarly, altered renal morphology and functional abnormalities have been described in mice with targeted inactivation of the angiotensinogen (Agt) and the angiotensin-converting enzyme (ACE) genes. In contrast, inactivation of angiotensin receptors have not resulted in renal morphological abnormalities, suggesting a redundancy at this level of the RAS cascade that prevents the development of renal pathology. More importantly, inactivation of the ACE or Ao genes results in a renal phenotype remarkably similar to that obtained with pharmacological inhibition of the RAS. Taken together, the available information suggests that angiotensin is necessary for normal kidney development and for the maintenance of the functional and structural integrity of the adult kidney.


Subject(s)
Angiotensin II/biosynthesis , Kidney/embryology , Receptors, Angiotensin/biosynthesis , Adult , Angiotensin II/analysis , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Embryonic and Fetal Development/physiology , Female , Humans , Kidney/pathology , Mice , Pregnancy , Rabbits , Receptors, Angiotensin/analysis , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology
13.
Am J Physiol ; 273(2 Pt 2): R503-9, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9277532

ABSTRACT

Chronic unilateral ureteral obstruction (UUO) in early development activates the intrarenal renin-angiotensin system and leads to profound renal vasoconstriction, renal growth arrest, and interstitial fibrosis. To investigate the response of the AT1 and AT2 subtypes of the angiotensin II (ANG II) receptors to UUO, Sprague-Dawley rats underwent UUO or control sham operation in the first 48 h of life and were studied 1-28 days later. Renal mRNA for renin, AT1 and AT2 receptor, and receptor binding and distribution were determined. In contrast to controls, renin mRNA increased from 14 to 28 days in the obstructed kidney. After ipsilateral UUO, AT1 mRNA was suppressed at 1 day, but had increased compared with controls at 28 days. AT2 receptor mRNA fell rapidly in all kidneys from 1 to 3 days of age, after which it remained undetectable. Compared with the intact opposite kidney, AT2 mRNA was suppressed in the obstructed kidney 1 day after UUO. Compared with controls, AT1 and AT2 receptor binding was decreased by ipsilateral UUO at 1 day, whereas AT1 binding was increased at 28 days. Renal ANG II content was increased in the obstructed compared with the intact opposite kidney 28 days after UUO. In view of the increase in renal renin and angiotensin II production resulting from UUO, increased renal AT1 mRNA and receptor binding are likely to contribute to the vasoconstriction and interstitial fibrosis of the neonatal kidney after prolonged UUO.


Subject(s)
Animals, Newborn/physiology , Gene Expression Regulation, Developmental , Receptors, Angiotensin/genetics , Ureteral Obstruction/metabolism , Angiotensin II/metabolism , Animals , Animals, Newborn/metabolism , Gene Expression , Kidney/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/metabolism , Renin/genetics
14.
Dev Biol ; 183(2): 139-49, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9126290

ABSTRACT

To determine whether low oxygen is a stimulus for endothelial cell differentiation and vascular development in the kidney, we examined the effect of low oxygen on rat metanephric organ culture, a model known to recapitulate nephrogenesis in the absence of vessels. After 6 days in culture in standard (20% O2) or low oxygen (1-3% O2) conditions, metanephric kidney growth and morphology were assessed by DNA measurement, and light and electron microscopy. DNA content was higher in 3% O2-treated explants (2.5 +/- 0.17 microgram/kidney, n = 9) than in 20% O2 explants (1.5 +/- 0.09 microgram/kidney, n = 9), P < 0.05. Low oxygen induced proliferation of tubular epithelial cells, resulting in enhanced number of tubules of similar size. Endothelial cells forming capillaries were localized in 3% O2 explants by light and electron microscopy and by immunocytochemistry using endothelial cell markers. Flt-1, Flk-1, and ACE-containing cells were detected in 3% O2-treated explants, whereas 20% O2 explants were virtually negative. VEGF mRNA levels were 10-fold higher in 3% O2-treated explants than in 20% O2-treated explants. Addition of anti-VEGF antibodies to 3% O2-treated explants prevented low oxygen-induced growth and endothelial cell differentiation and proliferation. Our data indicate that low oxygen stimulates growth by cell proliferation and induces tubulogenesis, endothelial cell differentiation, and vasculogenesis in metanephric kidneys in culture. Upregulation of VEGF expression by low oxygen and prevention of low oxygen-induced tubulogenesis and vasculogenesis by anti-VEGF antibodies indicate that these changes were mediated by VEGF. These data suggest that low oxygen is the stimulus to initiate renal vascularization.


Subject(s)
Endothelial Growth Factors/physiology , Kidney/blood supply , Lymphokines/physiology , Neovascularization, Physiologic/drug effects , Oxygen/pharmacology , Animals , Biomarkers/analysis , Cell Division , DNA/analysis , Endothelial Growth Factors/genetics , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/ultrastructure , Epithelium , Kidney/embryology , Kidney Tubules/embryology , Lymphokines/genetics , Morphogenesis , Neovascularization, Physiologic/physiology , Organ Culture Techniques , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Am J Physiol ; 272(2 Pt 2): R662-8, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9124492

ABSTRACT

Angiotensin II (ANG II) and its receptors, AT1 and AT2, may modulate kidney development. To define the temporal and spatial distribution of AT1 and AT2 receptors and their mRNAs during nephrogenesis, fetal, newborn, and adult rat kidneys were studied using reverse transcription-polymerase chain reaction and radioligand binding autoradiography. AT1 expression was minimal at embryonic day 14 (E14), highly expressed at E20, and persisted into adulthood. Conversely, AT2 expression was easily detected from E14 through postnatal day 7 but was undetectable by postnatal day 28. At E14, 76% of the receptors were AT2, 24% were AT1, and both were found in the undifferentiated mesenchyme. By E17, AT1 comprised 40% of the receptors and localized to mature nephron segments, whereas AT2 remained within both condensed mesenchyme and differentiating epithelia. The dissociation constants for AT1 and AT2 were 0.45 +/- 0.09 nM and 0.73 +/- 0.15 nM, respectively, at E17, similar to adult values. By E20, AT1 and AT2 colocalized to the outer medullary stripe, deep nephrons, medullary rays, and blood vessels, while AT2 continued to predominate in the actively differentiating cortex. The presence of both subtypes of receptors capable of binding ANG II during early nephrogenesis and the time-dependent and structure-specific regulation of receptor localization confirm a regulated developmental program for receptor expression and suggest important roles for AT1 and AT2 in renal morphogenesis.


Subject(s)
Animals, Newborn/metabolism , Embryo, Mammalian/metabolism , Kidney/embryology , Kidney/metabolism , Receptors, Angiotensin/metabolism , Aging/metabolism , Animals , Animals, Newborn/growth & development , Embryo, Mammalian/physiology , Embryonic and Fetal Development , Kidney/growth & development , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/genetics
17.
Am J Kidney Dis ; 26(3): 409-31, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7645550

ABSTRACT

Molecular, cellular, and physiological studies indicate that the renin-angiotensin system (RAS) is highly expressed during early kidney development. We propose that a major function of the RAS during early embryonic development is the modulation of growth processes that lead the primitive kidney into a properly differentiated and architecturally organized organ suited for independent extrauterine life. As development progresses, the RAS acquires new and overlapping functions such as the endocrine and paracrine regulation of blood pressure and renal hemodynamics. Disease states in adult mammals often result in expression of RAS genes and phenotypic changes resembling the embryonic pattern, emphasizing the importance of undertaking developmental studies. Because of their importance in health and disease, the immediate challenge is to identify the mechanisms that regulate the unique development of the RAS and its role(s) in normal and abnormal growth processes.


Subject(s)
Aging/physiology , Kidney/physiology , Renin-Angiotensin System/physiology , Animals , Humans , Kidney/embryology , Kidney/enzymology , Kidney/ultrastructure , Peptidyl-Dipeptidase A/metabolism , Receptors, Angiotensin/metabolism
18.
Am J Physiol ; 267(4 Pt 2): R865-78, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7943427

ABSTRACT

Living organisms are the result of precise and complex associations of regulatory systems in which active biomolecules interact with one another and respond to the challenges of growth and development, alterations in the environment, and disease. Understanding of body homeostasis may be accomplished at various levels of scientific endeavor. Physiological research has brought about an enormous understanding of the fundamental principles that sustain life in health and disease. The field of molecular biology has provided new tools and strategies with which to examine physiological processes as viewed from the level of fundamental biomolecules. The integration of both fields as "molecular physiology" has provided the opportunity for another level of scientific understanding and the opening of new avenues of research. Renin is one such molecule that participates in the control of several diverse physiological responses including changes in blood pressure, fluid and electrolyte homeostasis, renal function, and perhaps some elements of growth and differentiation. Because of the authors' bias, this review article will use renin to introduce many of the techniques of molecular biology and illustrate the areas of ongoing and potential interdependent activities resulting in the emerging field of molecular physiology.


Subject(s)
Molecular Biology/methods , Physiology/methods , Renin/physiology , Animals , Animals, Genetically Modified , Cloning, Molecular , Gene Expression , Humans , Mutagenesis , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Promoter Regions, Genetic , RNA, Messenger/analysis , Recombination, Genetic , Renin/biosynthesis , Renin/genetics , Transfection
19.
Kidney Int ; 45(5): 1333-9, 1994 May.
Article in English | MEDLINE | ID: mdl-8072245

ABSTRACT

Unilateral ureteral obstruction (UUO) in the neonate increases ipsilateral renal renin gene expression, an effect which is mediated by renal nerves. To determine whether neonatal UUO alters the number of renal cortical cells secreting renin and whether this change is modulated by renal nerve activity, newborn Sprague-Dawley rats were subjected to left UUO, right uninephrectomy, or sham operation and studied four weeks thereafter. To evaluate the importance of renal nerves in this response, an additional group of animals underwent chemical sympathectomy with guanethidine. Ureteral obstruction was associated with marked reduction in renal mass in the obstructed kidney and contralateral compensatory hypertrophy, changes which were not altered by sympathectomy. Renin messenger RNA and renal renin content were elevated in the obstructed kidney. The number of cells secreting renin, measured by the reverse hemolytic plaque assay, was markedly increased in the obstructed kidney (45 +/- 18 plaques/slide vs. 11 +/- 1 plaques/slide in sham animals), but not in the opposite kidney or following uninephrectomy. This effect was not significantly altered by sympathectomy. There was no change in the amount of renin secreted per cell or in the secretory response to Ca++. These results show that UUO results in recruitment of cells not previously secreting renin by a mechanism independent of renal nerve activity. This recruitment occurs without alteration of the quantity of renin secreted per cell or in the normal regulatory effect of Ca++ on renin secretion. An increase in the number of renin-secreting cells may contribute to the activation of the renin-angiotensin system, and thus to the vasoconstriction observed following ureteral obstruction.


Subject(s)
Kidney Cortex/metabolism , Renin/metabolism , Ureteral Obstruction/metabolism , Animals , Animals, Newborn , Cell Movement , Gene Expression , Guanethidine/pharmacology , Hemolytic Plaque Technique , Kidney Cortex/innervation , Kidney Cortex/pathology , Nephrectomy , Norepinephrine/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Renin/genetics , Sympathetic Nervous System/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...