Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Rev. Assoc. Med. Bras. (1992, Impr.) ; 69(11): e20230334, 2023. tab
Article in English | LILACS-Express | LILACS | ID: biblio-1521483

ABSTRACT

SUMMARY OBJECTIVE: Autosomal dominant polycystic kidney disease is an inherited kidney disorder with mutations in polycystin-1 or polycystin-2. Autosomal recessive polycystic kidney disease is a severe form of polycystic kidney disease that is characterized by enlarged kidneys and congenital hepatic fibrosis. Mutations at PKHD1 are responsible for all typical forms of autosomal recessive polycystic kidney disease. METHODS: We evaluated the children diagnosed with polycystic kidney disease between October 2020 and May 2022. The diagnosis was established by family history, ultrasound findings, and/or genetic analysis. The demographic, clinical, and laboratory findings were evaluated retrospectively. RESULTS: There were 28 children (male/female: 11:17) evaluated in this study. Genetic analysis was performed in all patients (polycystin-1 variants in 13, polycystin-2 variants in 7, and no variants in 8 patients). A total of 18 variants in polycystin-1 and polycystin-2 were identified and 9 (50%) of them were not reported before. A total of eight novel variants were identified as definite pathogenic or likely pathogenic mutations. There was no variant detected in the PKDH1 gene. CONCLUSION: Our results highlighted molecular features of Turkish children with polycystic kidney disease and demonstrated novel variations that can be utilized in clinical diagnosis and prognosis.

2.
Chinese Journal of Neurology ; (12): 1135-1142, 2022.
Article in Chinese | WPRIM | ID: wpr-958009

ABSTRACT

Objective:To explore the clinical phenotype and gene characteristics of a case of TSC2/PKD1 adjacency gene syndrome, so as to improve the clinical understanding of the disease.Methods:A case of TSC2/PKD1 adjacency gene syndrome diagnosed in the Department of Neurology of the Children′s Hospital Affiliated to Zhengzhou University was analyzed retrospectively. The clinical data, laboratory examination, imaging characteristics and gene variation characteristics of the child were summarized.Results:The patient was a 17 months old girl, with the main complaint of "intermittent convulsion with 17 months of underdevelopment". The clinical manifestations were epileptic seizures, which were in the form of a series of spastic seizures, absence seizures, focal seizures, and depigmentation spots can be seen in the trunk and neck. Cranial magnetic resonance imaging showed multiple patchy signals in the cortex and subcortical areas of the bilateral cerebral hemispheres, multiple small nodular shadows under the ependyma of the bilateral lateral ventricles, the heart color Doppler ultrasound showed patent foramen ovale and pericardial effusion, and the abdomen color Doppler ultrasound showed polycystic kidney. Ophthalmic color Doppler ultrasound showed that there were localized small swelling lesions around the optic disc of the left eye. The whole exon gene sequencing of the pedigree showed the proband had partial deletion of TSC2 gene (NM_000548) at chromosome position chr16: 2125799-2185690. The real-time quantitative detection system verified that exons 23-42 were deleted, and all exons of PKD1 gene were deleted (NM_001009944), and multiple ligation dependent probe amplification verified that exons 1-46 were deleted, and no downstream gene deletion was found. The overall deletion size was about 60 kb. Both of the girl's father and mother had normal phenotypes and were wild-type.Conclusions:TSC2/PKD1 adjacency gene syndrome is relatively rare. It can have clinical manifestations of tuberous sclerosis/autosomal dominant polycystic kidney disease. Most of the nervous system and kidney are seriously affected, and the prognosis is poor. TSC2/PKD1 gene deletion and variation is the genetic cause of the TSC2/PKD1 adjacency gene syndrome.

3.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 66-72, 2020.
Article in Chinese | WPRIM | ID: wpr-873186

ABSTRACT

Objective:To observe the effect of Danggui Buxuetang on lung histopathology and protein kinase D1 (PKD1), nuclear transcription factor-κB (NF-κB) and manganese superoxide dismutase (MnSOD)-mediated oxidative stress pathway in rats with pulmonary fibrosis induced by bleomycin, so as to explore the mechanism of intervention of pulmonary fibrosis.Method:Thirty-two male SPF SD rats were randomly divided into sham operation group, model group, Danggui Buxuetang group and prednisone group, with 8 rats in each group. Except the sham operation group, the other groups were prepared through the intratracheal instillation with bleomycin. After modeling for 24 h, the rats of Danggui Buxuetang group were administered with Danggui Buxuetang (0.81 g·kg-1). The rats of prednisone group were given aqueous solution of prednisone (0.005 g·kg-1). The rats of sham operation group and model group were given the same volume of saline. After 14 days of administration, blood was collected from the femoral artery, serum was separated, and the lungs were taken by thoracotomy. The pathological changes of rat lung tissues were observed by hematoxylin-eosin staining (HE) and Masson trichrome staining, and graded by Szapiel score and Ashcroft score at the same time. The content of serum malondialdehyde (MDA), and the activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px) were determined. Real-time fluorescent quantitative polymerase chain reaction (Real-time PCR) and Western blot were used to measure mRNA and protein expressions of PKD1, NF-κB, MnSOD.Result:Compared with the rats in sham operation group, the rats in model group had higher Szapiel scores and Ashcroft scores (P<0.05), higher serum MDA content , but lower SOD, CAT and GSH-Px activities(P<0.01), moreover, the rat lung tissues in model group had higher mRNA and protein expressions of PKD1, NF-κB and MnSOD (P<0.01) than those in sham operation group. Compared with the rats in model group, the Szapiel scores and Ashcroft scores of the rats in Danggui Buxuetang group were decreased significantly(P<0.05). The serum MDA content was decreased significantly, and SOD, CAT, GSH-Px activities were increased, whereas mRNA and protein expressions of PKD1, NF-κB, MnSOD in the rat lung tissues were decreased(P<0.05,P<0.01).Conclusion:Danggui Buxuetang can reduce the degree of pulmonary fibrosis by regulating the anti-oxidation pathway of PKD1/NF-κB/MnSOD mitochondrial nucleus and improving the body's antioxidant capacity.

4.
Chinese Journal of Clinical Thoracic and Cardiovascular Surgery ; (12): 569-573, 2020.
Article in Chinese | WPRIM | ID: wpr-822553

ABSTRACT

@#Objective    To explore the effects of PKD1 gene on mouse aortic smooth muscle (MOVAS) cells autophagy. Methods    The shRNA and over-expression lentiviral vectors for the target gene of PKD1 were constructed. MOVAS cells were infected by a number of successful packaging shRNA (PKD1 knockdown) or ETS-1 (PKD1 over-expressing) lentiviral vectors, and qPCR was used to test interference and over-expressing effects. Then qPCR and Western blotting were used to detect the expression levels of autophagy markers including Atg5, Beclin1 and LC3 in control group, shPKD1 group and ETS-1 group. Results    Compared with the control group, PKD1 mRNA level was decreased in the shPKD1 group (P<0.05); ETS-1 and PKD1 mRNA levels were increased in the ETS-1 group (P<0.05). In contrast with the control group, the mRNA levels of autophagy markers including Atg5 (P<0.05) and Beclin1 (P<0.01) were obviously decreased in the shPKD1 group, but they were obviously increased in the ETS-1 group (P<0.001). Protein levels of Atg5, Beclin1 and LC3 were significantly decreased in the shPKD1 group (P<0.05), but they were increased obviously in the ETS-1 group (P<0.05) in contrast with the control group. Conclusion    PKD1 gene is involved in MOVAS cells autophagy, low expression of PKD1 gene can inhibit autophagy and high expression of PKD1 promotes autophagy in vascular smooth muscle cells.

5.
Journal of Pharmaceutical Practice ; (6): 241-244, 2020.
Article in Chinese | WPRIM | ID: wpr-821478

ABSTRACT

Objective Breast cancer is one of the deadliest malignancies in the world. ebracteolatain A (EA) is a kind of acetylphloroglucinol extracted from ebracteolatain. To explore the specific mechanism of EA inhibiting the proliferation of breast cancer cell MCF-7, so as to provide a new approach for the clinical treatment of breast cancer. Methods EA with different concentrations were added to breast cancer cell MCF-7 to detect changes in PKD1 protein expression. The plasmid with overexpressed PKD1 was constructed and transfected into cells, and the mRNA and protein expression levels of PKD1 were detected by real-time fluorescence quantitative PCR and Western Blot assay. CCK-8 assay was used to detect changes in cell proliferation capacity. Western Blot assay was used to detect the expression level of PKD1 and its related signaling pathways. Results EA inhibited the expression of PKD1 protein in breast cancer cells with a dose-dependent manner (P< 0.05). When transfected with the overexpressed plasmid, PKD1 was significantly increased in mRNA and protein levels (P<0.001). At the same time, PKD1 overexpression significantly reversed inhibition of EA on MCF-7 proliferation (P<0.001). It was confirmed by signaling pathway analysis that EA might affect the proliferation ability of breast cancer cells by inhibiting PKD1-mediated MEK/ERK and PI3K/AKT signaling activity (P<0.05). Conclusion EA could inhibit the proliferation of breast cancer cells by regulating PKD1-mediated MEK/ERK and PI3K/AKT signaling pathways.

6.
Journal of Shanghai Jiaotong University(Medical Science) ; (12): 744-749, 2019.
Article in Chinese | WPRIM | ID: wpr-843398

ABSTRACT

Objective: To explore the potential mechanism of male reproductive failure in autosomal dominant polycystic kidney disease (ADPKD) patients and analyze the outcomes of assisted reproductive technology treatment. Methods: Next-generation sequencing was performed for genetic diagnosis of 8 ADPKD patients, who came to International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, for genetic counseling. The semen of ADPKD patients and normal males who came for pre-pregnancy consultation was collected by masturbation for sperm analysis. The ultrastructure of sperm was observed by transmission electron microscopy. Outcomes of 7 patients with ADPKD who chose preimplantation genetic testing (PGT) were compared with those of 7 patients who were dystrophin (DMD) gene mutation carriers, undergoing the PGT in the same period. Results: Eight patients with ADPKD were heterozygous for polycystin 1 (PKD1) gene. Key parameters of sperm motion including progressive motility sperm percentage, curvilinear velocity, straight-line velocity, average path velocity, amplitude of lateral head displacement were much lower than those of normal semen, showing mild to severe oligozoospermia. One ADPKD patient with severe oligoathenospermia manifested bilateral seminal vesicle cysts. Transmission electron microscopy showed that the central microtubules of the sperm flagella of ADPKD patients were absent and the surrounding double microtubules were disorganized. There was no significant difference in the number of eggs, fertilization rate, cleavage rate, effective embryo rate and excellent embryo rate between the ADPKD patients and the DMD gene mutation carriers, but the ADPKD patients were prone to early abortion. Conclusion: Male reproductive failure caused by ADPKD may be related to many factors such as abnormal structure of sperm flagella and genital cysts. Further, PKD1 mutation may play a role in embryo implantation and early development.

7.
Childhood Kidney Diseases ; : 64-66, 2018.
Article in English | WPRIM | ID: wpr-739197

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent hereditary renal disease and causes terminal chronic renal failure. ADPKD is characterized by bilateral multiple renal cysts, which are produced by mutations of the PKD1 and PKD2 genes. PKD1 is located on chromosome 16 and encodes a protein that is involved in cell cycle regulation and intracellular calcium transport in epithelial cells and is responsible for 85% of ADPKD cases. Although nine cases of unilateral ADPKD with contralateral kidney agenesis have been reported, there have been no reports of early childhood ADPKD. Here, we report the only case of unilateral ADPKD with contralateral kidney dysplasia in the world in a four year-old girl who was intrauterinely diagnosed since she was 20 weeks old and followed for four years until present.


Subject(s)
Female , Humans , Calcium , Cell Cycle , Chromosomes, Human, Pair 16 , Epithelial Cells , Kidney , Kidney Failure, Chronic , Polycystic Kidney, Autosomal Dominant
8.
Childhood Kidney Diseases ; : 28-30, 2018.
Article in English | WPRIM | ID: wpr-739185

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent hereditary renal disease and causes terminal chronic renal failure. ADPKD is characterized by bilateral multiple renal cysts, which are produced by mutations of the PKD1 and PKD2 genes. PKD1 is located on chromosome 16 and encodes a protein that is involved in cell cycle regulation and intracellular calcium transport in epithelial cells and is responsible for 85% of ADPKD cases. Although nine cases of unilateral ADPKD with contralateral kidney agenesis have been reported, there have been no reports of early childhood ADPKD. Here, we report the only case of unilateral ADPKD with contralateral kidney dysplasia in the world in a four year-old girl who was intrauterinely diagnosed since she was 20 weeks old and followed for four years until present.


Subject(s)
Female , Humans , Calcium , Cell Cycle , Chromosomes, Human, Pair 16 , Epithelial Cells , Kidney , Kidney Failure, Chronic , Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant
9.
Chinese Journal of Pathophysiology ; (12): 643-649, 2018.
Article in Chinese | WPRIM | ID: wpr-701174

ABSTRACT

AIM:To investigate the angiogenic effect and mechanisms of astragaloside IV(AS-IV)in rats with myocardial infarction via protein kinase D 1(PKD1)-histone deacetylase 5(HDAC5)-vascular endothelial growth factor (VEGF)signaling pathway.METHODS:The classic model of myocardial infarction by ligation of the left anterior de-scending coronary artery was replicated,and the rats were randomly divided into model group,AS-IV group,and AS-IV+CID755673(PKD1 inhibitor)group.The sham operation control group and DMSO control group were also set up.All the rats were given intravenous injection via caudal vein.The rats were sacrificed 4 weeks later,and segmental heart samples were used for HE staining and Masson staining.The expression of PKD1,HDAC5 and VEGF was analyzed by immunohis-tochemistry,RT-PCR and and Western blot.RESULTS:Compared with sham operation group and DMSO group,the myo-cardium in model group showed disordered arrangement, accompanied with necrotic myocardial cells and obvious fibrosis tissue.After treatment with AS-IV,the morphological changes of myocardium were obviously improved,and the number of new blood vessels increased significantly.However,after treatment with AS-IV+CID755673,the myocardial tissues of the rats became disordered again,with increased necrotic cells and some closed vessels.The mRNA and protein expression of PKD1,HDAC5 and VEGF in myocardial tissue in model group was significantly lower than that in sham operation and DMSO groups(P<0.05).The expression in AS-IV group was significantly higher than that in model group(P<0.01), while that in AS-IV+CID755673 group was significantly lower than that in AS-IV group(P<0.05).CONCLUSION:AS-IV promotes the angiogenesis of myocardial tissues in the rats after myocardial infarction partly by regulating the PKD 1-HDAC5-VEGF signaling pathway.

10.
Tissue Engineering and Regenerative Medicine ; (6): 793-801, 2018.
Article in English | WPRIM | ID: wpr-718786

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the combined effect of low-level laser treatment (LLLT) and recombinant human bone morphological protein-2 (rhBMP-2) applied to hypoxic-cultured MC3T3-E1 osteoblastic cells and to determine possible signaling pathways underlying differentiation and mineralization of osteoblasts under hypoxia. METHODS: MC3T3-E1 cells were cultured under 1% oxygen tension for 72 h. Cell cultures were divided into four groups: normoxia control, low-level laser (LLL) alone, rhBMP-2 combined with LLLT, and rhBMP-2 under hypoxia. Laser irradiation was applied at 0, 24, and 48 h. Cells were treated with rhBMP-2 at 50 ng/mL. Alkaline phosphatase activity was measured at 3, 7, and 14 days to evaluate osteoblastic differentiation. Cell mineralization was determined with Alizarin red S staining at 7 and 14 days. Western blot assays were performed to evaluate whether p38/protein kinase D (PKD) signaling was involved. RESULTS: The results indicate that LLLT and rhBMP-2 synergistically increased alkaline phosphatase (ALP) activity and mineralization. Western blot analyses showed that expression of type I collagen, runt-related transcription factor 2 (RUNX2), and Osterix (Osx), increased and expression of hypoxia-inducible factor 1-alpha (HIF-1α), decreased more in the LLLT and rhBMP-2 combined group than in the rhBMP-2 or LLL alone groups. Moreover, LLLT and rhBMP-2 stimulated p38 phosphorylation and rhBMP-2 and LLLT increased Prkd1 phosphorylation. CONCLUSION: Combined treatment with rhBMP-2 and LLL induced differentiation and mineralization of hypoxiccultured MC3T3-E1 osteoblasts by activating p38/PKD signaling in vitro.


Subject(s)
Humans , Alkaline Phosphatase , Hypoxia , Blotting, Western , Cell Culture Techniques , Collagen Type I , In Vitro Techniques , Low-Level Light Therapy , Miners , Osteoblasts , Oxygen , Phosphorylation , Phosphotransferases , Transcription Factors
11.
Journal of Clinical Pediatrics ; (12): 420-423, 2018.
Article in Chinese | WPRIM | ID: wpr-694696

ABSTRACT

Objective To explore the clinical phenotype and genotype characteristics of infantile polycystic kidney. Method The clinical data of polycystic kidney disease in one infant were retrospectively analyzed, and the correlation between clinical phenotype and genotype was analyzed. Results In this infant the polycystic kidney was discoved in the fetal period, and shortness of breath, foaming at the mouth were present after birth. Abdominal magnetic resonance imaging indicated that there was medullary sponge kidney in both kidneys accompanied by mild effusion and cysts were found in the right kidney. Gene detection showed a missense mutation c.1123 C>T (Arg375Trp) of exon15 in PKHD1 gene. The exon31 of PKHD1 gene had a missense mutation c.3617G>T (Gly1206Val), which was a new missense mutation. The exon18 of PKD1 gene had a missense mutation c.7211G>A (Arg2404Gln), which is a complex heterozygous mutant of homozygote. All of the mutations are missense mutations. The infant was improved and discharged after treatment, and renal function was normal during 4 months of follow-up. Conclusion Gene detection can be used for early diagnosis of infantile polycystic kidney disease. Newborns with two missense mutations can survive, and exon31 mutation c.3617G>T (Gly1206Val) is a new finding.

12.
Tissue Engineering and Regenerative Medicine ; (6): 433-441, 2017.
Article in English | WPRIM | ID: wpr-655772

ABSTRACT

Hypoxia suppresses osteoblastic differentiation and the bone-forming capacity. As the leading osteoinductive growth factor used clinically in bone-related regenerative medicine, recombinant human bone morphogenic protein-2 (rhBMP- 2) has yielded promising results in unfavorable hypoxic clinical situations. Although many studies have examined the effects of rhBMP-2 on osteoblastic differentiation, mineralization and the related signaling pathways, those of rhBMP-2 on osteoblastic cells remain unknown, particularly under hypoxic conditions. Therefore, this study was conducted under a 1% oxygen tension to examine the differentiating effects of rhBMP-2 on osteoblastic cells under hypoxia. rhBMP-2 could also induce the differentiation and mineralization of Osteoblastic (MC3T3-E1) cells under1%hypoxic conditions. rhBMP-2 could also induce the differentiation and mineralization of MC3T3-E1 cells under 1% hypoxic conditions. rhBMP-2 increased the alkaline phosphatase {ALP} activity in a time dependent manner, and expression of ALP, collagen type-1 (Col-1) and osteocalcin (OC) mRNAwere up-regulated significantly in a time- and concentration-dependent manner. In addition, the area of the mineralized nodules increased gradually in a concentration-dependent manner. Western blot analysis, which was performed to identify the signaling pathways underlying rhBMP-2-induced osteoblastic differentiation under hypoxic conditions, showed that rhBMP-2 significantly promoted the phosphorylation of the p38 mitogen-activated protein kinase (MAPK) in a time-dependent manner. A pretreatment with SB203580, a p38 MAPK inhibitor, inhibited the rhBMP-2-mediated differentiation and mineralization. Moreover, the phosphorylation of p38 induced by rhBMP-2 was inhibited in response to a pretreatment of the cells with Go6976, a protein kinase D {PKD) inhibitor. These findings suggest that rhBMP-2 induces the differentiation and mineralization of MC3T3-E1 cells under hypoxic conditions via activation of the PKD and p38 MAPK signaling pathways.


Subject(s)
Humans , Alkaline Phosphatase , Hypoxia , Blotting, Western , Collagen , Miners , Osteoblasts , Osteocalcin , Oxygen , p38 Mitogen-Activated Protein Kinases , Phosphorylation , Protein Kinases , Regenerative Medicine
13.
Article in English | IMSEAR | ID: sea-176040

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a common hereditary kidney disease caused due to a mutation in PKD1 gene and the PKD2 gene located at chromosome level 16 and chromosome 4. ADPKD often leads to progressive kidney (renal) failure, primarily due to continued enlargement of the cysts and replacement of normal kidney tissue. The present case is of a 70-year-old male diabetic patient with ADPKD along with acute pyelonephritis caused by multi-drug resistant Staphylococcus D group and Candida albicans, treated with a new antibiotic adjuvant entity ceftriaxone/sulbactam/disodium edetate, and fluconazole recovered completely.

14.
Genomics & Informatics ; : 16-22, 2012.
Article in English | WPRIM | ID: wpr-155519

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by formation of multiple fluid-filled cysts that expand over time and destroy renal architecture. The proteins encoded by the PKD1 and PKD2 genes, mutations in which account for nearly all cases of ADPKD, may help guard against cystogenesis. Previously developed mouse models of PKD1 and PKD2 demonstrated an embryonic lethal phenotype and massive cyst formation in the kidney, indicating that PKD1 and PKD2 probably play important roles during normal renal tubular development. However, their precise role in development and the cellular mechanisms of cyst formation induced by PKD1 and PKD2 mutations are not fully understood. To address this question, we presently created Pkd2 knockout and PKD2 transgenic mouse embryo fibroblasts. We used a mouse oligonucleotide microarray to identify messenger RNAs whose expression was altered by the overexpression of the PKD2 or knockout of the Pkd2. The majority of identified mutations was involved in critical biological processes, such as metabolism, transcription, cell adhesion, cell cycle, and signal transduction. Herein, we confirmed differential expressions of several genes including aquaporin-1, according to different PKD2 expression levels in ADPKD mouse models, through microarray analysis. These data may be helpful in PKD2-related mechanisms of ADPKD pathogenesis.


Subject(s)
Animals , Mice , Biological Phenomena , Cell Adhesion , Cell Cycle , Embryonic Structures , Fibroblasts , Kidney , Mice, Transgenic , Microarray Analysis , Oligonucleotide Array Sequence Analysis , Phenotype , Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Proteins , RNA, Messenger , Signal Transduction
15.
Braz. j. med. biol. res ; 44(7): 606-617, July 2011. ilus
Article in English | LILACS | ID: lil-595695

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common human life-threatening monogenic disorders. The disease is characterized by bilateral, progressive renal cystogenesis and cyst and kidney enlargement, often leading to end-stage renal disease, and may include extrarenal manifestations. ADPKD is caused by mutation in one of two genes, PKD1 and PKD2, which encode polycystin-1 (PC1) and polycystin-2 (PC2), respectively. PC2 is a non-selective cation channel permeable to Ca2+, while PC1 is thought to function as a membrane receptor. The cyst cell phenotype includes increased proliferation and apoptosis, dedifferentiation, defective planar polarity, and a secretory pattern associated with extracellular matrix remodeling. The two-hit model for cyst formation has been recently extended by the demonstration that early gene inactivation leads to rapid and diffuse development of renal cysts, while inactivation in adult life is followed by focal and late cyst formation. Renal ischemia/reperfusion, however, can function as a third hit, triggering rapid cyst development in kidneys with Pkd1 inactivation induced in adult life. The PC1-PC2 complex behaves as a sensor in the primary cilium, mediating signal transduction via Ca2+ signaling. The intracellular Ca2+ homeostasis is impaired in ADPKD, being apparently responsible for the cAMP accumulation and abnormal cell proliferative response to cAMP. Activated mammalian target for rapamycin (mTOR) and cell cycle dysregulation are also significant features of PKD. Based on the identification of pathways altered in PKD, a large number of preclinical studies have been performed and are underway, providing a basis for clinical trials in ADPKD and helping the design of future trials.


Subject(s)
Humans , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Apoptosis/genetics , Calcium/metabolism , Disease Progression , Gene Silencing , Mutation , Membrane Proteins/genetics , Polycystic Kidney, Autosomal Dominant/metabolism
16.
Article in Portuguese | LILACS | ID: lil-685675

ABSTRACT

A doença renal policística do adulto é uma desordem genética de caráter autossômico dominante, caracterizada por progressivo desenvolvimento e crescimento de cistos renais, que culminam com a falência renal terminal na meia-idade. Outras manifestações clínicas associadas incluem cistos hepáticos e pancreáticos, hipertensão, aneurismas cerebrais e defeitos cardiovasculares. Aspectos celulares e moleculares dos mecanismos de cistogênese envolvem proliferação e apoptose celular, remodelamento da matriz extracelular, secreção e acúmulo de fluidos. Geneticamente heterogênea, na maioria dos casos (aproximadamente 85%) são mutações no gene PKD1, localizado no cromossomo 16p13.3, com o segundo gene, PKD2, localizado nos intervalos do cromossomo 4q13-q23, respondendo por 15% das mutações, ambos já seqüenciados e caracterizados. Existem evidências da interação comum das proteínas policistinas 1 e 2 em rotas de eventos de adesão extracelular e transporte iônico, possibilitando a regulação do fluxo de Ca++ transmembrana. Inúmeros trabalhos vêm tentando correlacionar o genótipo mutado ao fenótipo expresso em termos da progressão e severidade daautosomal dominant polycystic kidney disease. A análise das mutações autosomal dominant polycystic kidney disease é fundamental para a compreensão dos mecanismos de atuação envolvidos na doença. Métodos de detecção baseados na reação em cadeia da polimerase têm sido amplamente empregados, como a reação em cadeia da polimerase via transcriptase reversa, protein truncation test, single-strand conformational polymorphism e denaturing high performance liquid chromatography, importantes ferramentas que auxiliam no seqüenciamento e caracterização dessas mutações


Adult polycystic kidney disease is an autosomal dominant genetic disorder, characterized by progressive development and growth of renal cysts, which result in terminal renal failure in middle age. Other associated clinical manifestations include hepatic and pancreatic cysts, hypertension, cerebral aneurysms and cardiovascular disorders. Cellular and mononuclear aspects of the mechanisms of cytogenesis comprehend cellular proliferation and apoptosis, remodeling of the extracellular matrix, secretion and accumulation of fluids. This disease is genetically heterogeneous; in most cases (approximately 85%), the gene involved is PKD1, which is located on chromosome 16p13.3. In the remaining cases (15%), the disease is caused by mutational changes in another gene (PKD2), which is located at chromosome intervals 4q13-q23. Both genes have been sequenced and characterized. There is evidence of the common interaction of polycystins 1 and 2 in pathways of extracellular adhesion and ionic transportation events, which promotes the regulation of transmembrane Ca++ flow. Several studies have tried to correlate the mutant genotype with the phenotype expressed in terms of progression and severity of the autosomal dominant polycystic kidney disease. Analyzing mutations in the autosomal dominant polycystic kidney disease is crucial for understanding the action mechanisms involved in this disease. Detection methods based on the polymerase chain reaction have been widely used, such as the reverse transcriptase polymerase chain reaction, protein truncation test, single-strand conformational polymorphism, and denaturing high performance liquid chromatography. They are important tools that help sequencing and characterizing those mutations


Subject(s)
Humans , Adult , Polycystic Kidney, Autosomal Recessive/diagnosis , Polycystic Kidney, Autosomal Recessive/genetics , Adult/psychology
17.
Article in Portuguese | LILACS | ID: lil-691674

ABSTRACT

A doença renal policística do adulto é uma desordem genética de caráter autossômicodominante, caracterizada pelo progressivo desenvolvimento e crescimento de cistos renais,que podem levar à doença renal terminal durante a fase adulta do indivíduo. Outrasmanifestações clínicas associadas incluem cistos hepáticos e pancreáticos, hipertensão,aneurismas cerebrais e alterações cardiovasculares. Aspectos celulares e moleculares dosmecanismos de cistogênese envolvem proliferação e apoptose celular, remodelamento damatriz extracelular, secreção e acúmulo de líquidos. Geneticamente heterogênea, na maioriados casos (~ 85%) são mutações no gene PKD1, localizado no cromossomo 16p13.3, como segundo gene, PKD2, localizado nos intervalos do cromossomo 4q13-q23, respondendopor 15% de mutações, ambos já seqüenciados e caracterizados, ocorrendo ainda um terceirogene, PKD3, porém ainda pouco estudado. Existem evidências da interação comum dasproteínas policistinas 1 e 2, associadas com proteínas ciliares em rotas de eventos deadesões extracelulares e transportes iônicos, possibilitando a regulação do fluxo de Cl- eCa2+ transmembrana.


Adult polycystic kidney disease is an autosomal dominant genetic disorder, characterizedby progressive development and growth of renal cysts, which may lead to terminal renalfailure during adulthood. Other associated clinical manifestations include hepatic andpancreatic cysts, hypertension, cerebral aneurysms and cardiovascular disorders. Cellularand mononuclear aspects of the mechanisms of cytogenesis comprehend cellular proliferationand apoptosis, remodeling of the extracellular matrix, secretion and accumulation of fluids.This disease is genetically heterogeneous; in most cases (approximately 85%), the geneinvolved is PKD1, which is located on chromosome 16p13.3. In the remaining cases (15%),the disease is caused by mutational changes in another gene (PKD2), which is located atchromosome intervals 4q13-q23. Both genes have been sequenced and characterized. Thereis also a third gene, PKD3, which has been little studied. There is evidence of the commoninteraction of polycystins 1 and 2, associated with ciliary proteins in pathways of extracellularadhesion and ionic transportation events, which promotes the regulation of Cl- andtransmembrane Ca2+ flow.


Subject(s)
Adult , Kidney Diseases , Molecular Biology , Polycystic Kidney, Autosomal Dominant
18.
Korean Journal of Nephrology ; : 867-877, 2004.
Article in English | WPRIM | ID: wpr-43796

ABSTRACT

BACKGROUND: PKD2 gene, encoding polycystin-2 protein is a cause of autosomal dominant polycystic kidney disease (ADPKD) forms cyst in the kidney, liver, and pancreas and result cardiac defects. Polycystin-2 protein interacts with polycystin-1 to produce calcium-permeable non-selective cation current but this signaling pathway was unknown well. To identify expression profiles of mutant PKD2 gene overexpressed HEK 293 cells and EF-hand motif function, we execute microarray experiment using mutant (m1, m1/m2) PKD2 overexpressed cells. METHODS: cDNA probes were prepared by labeling mRNA from mutant (m1, m1/m2) PKD2 overexpressed cells with Cy5-dUTP and Cy3-dUTP through reverse transcription. The mixed probes of each sample were then hybridized with 14, 080 cDNA microarray, and the fluorescent signals were scanned by Arraywox scanner (Applied Precision LLC. Northeastern, USA). The values of Cy5-dUTP and Cy3- dUTP on each spot were analyzed and calculated by ImaGene 5.1 software (BioDiscovery Inc., El Segundo, CA, USA). Normalization was performed by using subtract mean and all genes. RESULTS: It PKD2 over-expressed cells, up-regulated genes were 11 containing PTK2 and down-regulated genes were 27 including MAP2, ABS. In mutant (m1) PKD2 genes, up-regulated genes were 20 and down-regulated genes were 121. In mutant (m1/m2) PKD2 genes, up-regulated genes were 363 and down-regulated genes were 360, respectively. Based on the clustering data, up-regulated genes were 5 containing IL4R and SIL1, and down-regulated genes were 6 including SAE1 and CPSF4. CONCLUSION: Microarray analysis may provide invaluable information on the role of mutant PKD2 gene product. In this study, we have used a high-density cDNA microarray technique to assess the gene expression profile of mutant (m1, m1/m2) PKD2 overexpression cells. Value of up- and down-regulated genes normalized by Genesight 3.2 version software compared with M/A plot data to identify confidence of data. Selected genes numbers were different, but the level of fold value was similar. Finally, we identified that several genes maybe involve in PKD2 function.


Subject(s)
DNA, Complementary , HEK293 Cells , Kidney , Liver , Microarray Analysis , Oligonucleotide Array Sequence Analysis , Pancreas , Polycystic Kidney, Autosomal Dominant , Reverse Transcription , RNA, Messenger , Transcriptome
19.
Korean Journal of Nephrology ; : 714-726, 1998.
Article in Korean | WPRIM | ID: wpr-159050

ABSTRACT

We analysed the 49 probands of autosomal dominant polycystic kidney disease (ADPKD) in Korea to elucidate clinical and genetic characteristics. 1) Family history of renal disease or ADPKD was taken from 44% of probands; hypertension 88%, cerebrovascular attack 64%, end stage renal disease 16%. 2) From the family screening with renal ultrasonography, we have confirmed dominant trait in 24 families. We found 2 families which have suspicious new mutations. 3) We performed linkage analysis of 15 families. The PKD-1 to non PKD-1 ratio was 13 : 2. 4) The male to femal ratio was 17 : 32 and age at diagnosis was 41 (24-65)years (mean (range)) in male, 45 (26-68) years in female. 5) The factors leading to the diagnosis of ADPKD were flank pain (23%), incidental finding (17%), palpable abdominal mass (11%), headache (9%) and gross hematuria (9%) 3) Hypertension (80%), azotemia (43%), flank pain (42%), renal calcification (42%), gross hematuria (33%) and cyst hemorrhage (25%) were renal complications. There were 2 renal cell carcinoma cases. 4)Liver cyst (82%) was the most common extrarenal manifestations. There were colonic diverticulosis (13%), cerebral artery aneurysm (7%), adrenal cyst (4%) and pancreatic cyst (2%). Results of our study revealed the clinical and genetic characteristics of ADPKD in Korea. We found only 44% of family history of renal disease, variable initial manifestations, variable renal and extrarenal complications. And we also found the similar percentage of PKD-2 (13%) to that (5-15%) of western countries, but it is necessary to study with more patients and families.


Subject(s)
Female , Humans , Male , Aneurysm , Azotemia , Carcinoma, Renal Cell , Cerebral Arteries , Diagnosis , Diverticulosis, Colonic , Flank Pain , Headache , Hematuria , Hemorrhage , Hypertension , Incidental Findings , Kidney Failure, Chronic , Korea , Mass Screening , Pancreatic Cyst , Polycystic Kidney, Autosomal Dominant , Ultrasonography
20.
Chinese Pharmacological Bulletin ; (12)1987.
Article in Chinese | WPRIM | ID: wpr-565071

ABSTRACT

Autosomal dominant polycystic kidney disease(ADPKD),a common inherited disease,is characterized by massive enlargement of fluid-filled renal cysts.Progressively enlarging cysts compromise normal renal parenchyma,reduce renal function and lead to renal failure.Up to now,the treatment options for ADPKD have been limited to renal replacement therapy by dialysis or by transplantation for patients with end-stage renal failure.Inhibition of cyst fluid secretion,suppression of cyst epithelial cell growth and prevention of renal failure are new approaches to treat PKD.

SELECTION OF CITATIONS
SEARCH DETAIL