Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Aging (Albany NY) ; 16(4): 3068-3087, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38385965

ABSTRACT

Advanced age is the largest risk factor for many diseases and several types of cancer, including colorectal cancer (CRC). Senescent cells are known to accumulate with age in various tissues, where they can modulate the surrounding tissue microenvironment through their senescence associated secretory phenotype (SASP). Recently, we showed that there is an increased number of senescent cells in the colons of CRC patients and demonstrated that senescent fibroblasts and their SASP create microniches in the colon that are conducive to CRC onset and progression. However, the composition of the SASP is heterogenous and cell-specific, and the precise senescence profile of colon fibroblasts has not been well-defined. To generate a SASP atlas of human colon fibroblasts, we induced senescence in primary human colon fibroblasts using various in vitro methods and assessed the resulting transcriptome. Using RNASequencing and further validation by quantitative RT-PCR and Luminex assays, we define and validate a 'core senescent profile' that might play a significant role in shaping the colon microenvironment. We also performed KEGG analysis and GO analyses to identify key pathways and biological processes that are differentially regulated in colon fibroblast senescence. These studies provide insights into potential driver proteins involved in senescence-associated diseases, like CRC, which may lead to therapies to improve overall health in the elderly and to prevent CRC.


Subject(s)
Cellular Senescence , Neoplasms , Humans , Aged , Phenotype , Neoplasms/metabolism , Fibroblasts/metabolism , Colon , Tumor Microenvironment
2.
Immunity ; 56(8): 1876-1893.e8, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37480848

ABSTRACT

Acute graft-versus-host disease (aGVHD) remains a major limitation of allogeneic stem cell transplantation (SCT), and severe intestinal manifestation is the major cause of early mortality. Intestinal microbiota control MHC class II (MHC-II) expression by ileal intestinal epithelial cells (IECs) that promote GVHD. Here, we demonstrated that genetically identical mice of differing vendor origins had markedly different intestinal microbiota and ileal MHC-II expression, resulting in discordant GVHD severity. We utilized cohousing and antibiotic treatment to characterize the bacterial taxa positively and negatively associated with MHC-II expression. A large proportion of bacterial MHC-II inducers were vancomycin sensitive, and peri-transplant oral vancomycin administration attenuated CD4+ T cell-mediated GVHD. We identified a similar relationship between pre-transplant microbes, HLA class II expression, and both GVHD and mortality in a large clinical SCT cohort. These data highlight therapeutically tractable mechanisms by which pre-transplant microbial taxa contribute to GVHD independently of genetic disparity.


Subject(s)
Gastrointestinal Microbiome , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mice , Animals , Vancomycin , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Transplantation, Homologous/adverse effects
3.
Horm Cancer ; 11(1): 52-62, 2020 02.
Article in English | MEDLINE | ID: mdl-32002807

ABSTRACT

Primary aldosteronism (PA) is the most common cause of secondary hypertension with a high prevalence among patients with resistant hypertension. Despite the recent discovery of somatic variants in aldosterone-producing adenoma (APA)-associated PA, causes for PA due to bilateral aldosterone production (bilateral hyperaldosteronism; BHA) remain unknown. Herein, we identified rare gene variants in ATP2B4, in a cohort of patients with BHA. ATP2B4 belongs to the same family of Ca-ATPases as ATP2B3, which is involved in the pathogenesis of APA. Endogenous ATP2B4 expression was characterized in adrenal tissue, and the gene variants were functionally analyzed for effects on aldosterone synthase (CYP11B2) expression, steroid production in basal and agonist-stimulated conditions, and for changes in biophysical properties of channel properties. Knockdown of ATP2B4 in HAC15 exhibited reduced angiotensin II stimulation in one of four shRNA clones. Stable HAC15 cell lines with doxycycline (dox) - inducible wild-type and variant forms of ATP2B4 - were generated, and dox-induced upregulation of ATP2B4 mRNA and protein was confirmed. However, ATP2B4 variants did not alter basal or agonist-stimulated CYP11B2 expression. Whole-cell recordings in HAC15 cells indicated robust endogenous ATP2B4 conductance in native cells but reduced conductance with overexpressed WT and variant ATP2B4. The previously defined PA-causing ATP2B3 variant served as a positive control and exhibited elevated CYP11B2 mRNA. In conclusion, while this study did not confirm a pathogenic role for ATP2B4 variants in BHA, we describe the sequencing analysis for familial and sporadic BHA and outline a template for the thorough in vitro characterization of gene variants.


Subject(s)
Electrophysiology/methods , Genetic Variation/genetics , High-Throughput Nucleotide Sequencing/methods , Hyperaldosteronism/genetics , Plasma Membrane Calcium-Transporting ATPases/genetics , Cell Line, Tumor , Female , Humans , Male
4.
Hypertension ; 75(3): 645-649, 2020 03.
Article in English | MEDLINE | ID: mdl-31983310

ABSTRACT

Driver somatic mutations for aldosterone excess have been found in ≈90% of aldosterone-producing adenomas (APAs) using an aldosterone synthase (CYP11B2)-guided sequencing approach. In the present study, we identified a novel somatic CACNA1H mutation (c.T4289C, p.I1430T) in an APA without any currently known aldosterone-driver mutations using CYP11B2 immunohistochemistry-guided whole exome sequencing. The CACNA1H gene encodes a voltage-dependent T-type calcium channel alpha-1H subunit. Germline variants in this gene are known as a cause of familial hyperaldosteronism IV. Targeted next-generation sequencing detected identical CACNA1H variants in 2 additional APAs in a cohort of the University of Michigan, resulting in a prevalence of 4% (3/75) in APAs. We tested the functional effect of the variant on adrenal cell aldosterone production and CYP11B2 mRNA expression using the human adrenocortical HAC15 cell line with a doxycycline-inducible CACNA1HI1430T mutation. Doxycycline treatment increased CYP11B2 mRNA levels as well as aldosterone production, supporting a pathological role of the CACNA1H p.I1430T mutation on the development of primary aldosteronism. In conclusion, somatic CACNA1H mutation is a genetic cause of APAs. Although the prevalence of this mutation is low, this study will provide better understanding of molecular mechanism of inappropriate aldosterone production in APAs.


Subject(s)
Adenoma/genetics , Adrenal Cortex Neoplasms/genetics , Aldosterone/biosynthesis , Calcium Channels, T-Type/genetics , Hyperaldosteronism/etiology , Adenoma/complications , Adenoma/metabolism , Adrenal Cortex Neoplasms/complications , Adrenal Cortex Neoplasms/metabolism , Angiotensin II/pharmacology , Calcium Signaling , Cell Line, Tumor , Cytochrome P-450 CYP11B2/biosynthesis , Cytochrome P-450 CYP11B2/genetics , Doxycycline/pharmacology , Enzyme Induction/drug effects , Genetic Vectors/drug effects , High-Throughput Nucleotide Sequencing , Humans , Lentivirus/genetics , Mutation, Missense , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Exome Sequencing
5.
Endocr Relat Cancer ; 26(2): 217-225, 2019 02.
Article in English | MEDLINE | ID: mdl-30475217

ABSTRACT

Several somatic mutations specific to aldosterone-producing adenomas (APAs) have been described. A small proportion of adrenocortical carcinomas (ACCs) are associated with hyperaldosteronism, either primary aldosteronism or hyperreninemic hyperaldosteronism. However, it is unknown whether they harbor mutations of the same spectrum as APAs. The objective of this study is to describe the clinical phenotype and molecular genotype of ACCs with hyperaldosteronism, particularly the analysis for common APA-associated genetic changes. Patients were identified by retrospective chart review at a specialized referral center and by positive staining for CYP11B2 of tissue microarrays. Twenty-five patients with ACC and hyperaldosteronism were initially identified by retrospective chart review, and tissue for further analysis was available on 13 tumors. Seven patients were identified by positive staining for CYP11B2 in a tissue microarray, of which two were already identified in the initial chart review. Therefore, a total number of 18 patients with a diagnosis of ACC and features of either primary aldosteronism or hyperreninemic hyperaldosteronism were therefore included in the final study. Mutational status for a select list of oncogenes, tumor suppressor genes and genes known to carry mutations in APAs were analyzed by next-generation sequencing. Review of clinical data suggested autonomous aldosterone production in the majority of cases, while for some cases, hyperreninemic hyperaldosteronism was the more likely mechanism. The mutational landscape of ACCs associated with hyperaldosteronism was not different from ACCs with a different hormonal phenotype. None of the ACCs harbored mutations of known APA-associated genes, suggesting an alternative mechanism conferring aldosterone production.


Subject(s)
Adrenocortical Carcinoma/blood , High-Throughput Nucleotide Sequencing/methods , Hyperaldosteronism/etiology , Adult , Aged , Female , Humans , Male , Middle Aged , Mutation , Retrospective Studies , Young Adult
6.
Fam Cancer ; 16(4): 561-566, 2017 10.
Article in English | MEDLINE | ID: mdl-28389767

ABSTRACT

Melanomas are associated with several hereditary conditions. We present a large family with several family members affected with primary melanomas and dysplastic nevi as well as thyroid cancer and other malignant tumors. Clinical work-up did not reveal a mutation in any of the genes usually considered with evaluation for predisposition to melanoma (BRCA1/2, CDKN2A, CDK4, PTEN, TP53). Whole exome sequencing of five affected family members showed a new variant in POT1. POT1 is associated with the telomere shelterin complex that regulates telomere protection and telomerase access. Germline mutations in POT1 were recently shown to be associated with hereditary predisposition to melanoma. Our findings support a role of POT1 germline mutations in cancer predisposition beyond melanoma development, suggesting a broader phenotype of the POT1-associated tumor predisposition syndrome that might also include thyroid cancer as well as possibly other malignant tumors.


Subject(s)
Germ-Line Mutation , Melanoma/genetics , Skin Neoplasms/genetics , Telomere-Binding Proteins/genetics , Adult , Aged , Exome , Female , Genetic Predisposition to Disease , Humans , Male , Melanoma/pathology , Neoplastic Syndromes, Hereditary/genetics , Pedigree , Shelterin Complex , Skin Neoplasms/pathology , Exome Sequencing
7.
J Bone Miner Res ; 32(8): 1640-1643, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28394026

ABSTRACT

The recognition of hereditary causes of primary hyperparathyroidism (pHPT) is important because clinical care and surveillance differ significantly between sporadic and hereditary pHPT. In addition, the increasing number of genetic tests poses a challenge to classify mutations as benign or pathogenic. Functional work-up of variants remains a mainstay to provide evidence for pathogenicity. We describe a 52-year-old male patient with recurrent pHPT since age 35 years. Despite several neck surgeries with complete parathyroidectomy, he experienced persistent pHPT, necessitating repeated surgery for a forearm autotransplant, which finally resulted in unmeasurable parathyroid hormone (PTH) levels. Genetic testing revealed a new CDC73 variant (c.238-8G>A [IVS2-8G>A]), initially classified as a variant of uncertain significance. Parathyroid tissue from the initial surgeries showed loss of heterozygosity. Using an RT-PCR approach, we show that the mutation leads to the use of a cryptic splice site in peripheral mononuclear cells. In addition, a minigene approach confirms the use of the cryptic splice site in a heterologous cell system. The novel c.238-8G>A CDC73 variant activates a cryptic splice site, and the functional data provided justify the classification as a likely pathogenic variant. Our results underscore the importance of functional work-up for variant classification in the absence of other available data, such as presence in disease-specific databases, other syndromic clinical findings, or family history. In addition, the presented case exemplifies the importance to consider a hereditary condition in young patients with pHPT, particularly those with multi-gland involvement. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Hyperparathyroidism/genetics , Point Mutation , RNA Splice Sites , Tumor Suppressor Proteins/genetics , Humans , Hyperparathyroidism/metabolism , Hyperparathyroidism/pathology , Male , Middle Aged , Tumor Suppressor Proteins/metabolism
8.
J Mol Endocrinol ; 57(1): 1-11, 2016 07.
Article in English | MEDLINE | ID: mdl-27099398

ABSTRACT

Somatic and germline mutations in the inward-rectifying K(+) channel (KCNJ5) are a common cause of primary aldosteronism (PA) in aldosterone-producing adenoma and familial hyperaldosteronism type III, respectively. Dysregulation of adrenal cell calcium signaling represents one mechanism for mutated KCNJ5 stimulation of aldosterone synthase (CYP11B2) expression and aldosterone production. However, the mechanisms stimulating acute and chronic production of aldosterone by mutant KCNJ5 have not been fully characterized. Herein, we defined the effects of the T158A KCNJ5 mutation (KCNJ5(T158A)) on acute and chronic regulation of aldosterone production using an adrenal cell line with a doxycycline-inducible KCNJ5(T158A) gene (HAC15-TRE-KCNJ5(T158A)). Doxycycline incubation caused a time-dependent increase in KCNJ5(T158A) and CYP11B2 mRNA and protein levels. Electrophysiological analyses confirm the loss of inward rectification and increased Na(+) permeability in KCNJ5(T158A)-expressing cells. KCNJ5(T158A) expression also led to the activation of CYP11B2 transcriptional regulators, NURR1 and ATF2. Acutely, KCNJ5(T158A) stimulated the expression of total and phosphorylated steroidogenic acute regulatory protein (StAR). KCNJ5(T158A) expression increased the synthesis of aldosterone and the hybrid steroids 18-hydroxycortisol and 18-oxocortisol, measured with liquid chromatography-tandem mass spectrometry (LC-MS/MS). All of these stimulatory effects of KCNJ5(T158A) were inhibited by the L-type Ca(2+) channel blocker, verapamil. Overall, KCNJ5(T158A)increases CYP11B2 expression and production of aldosterone, corticosterone and hybrid steroids by upregulating both acute and chronic regulatory events in aldosterone production, and verapamil blocks KCNJ5(T158A)-mediated pathways leading to aldosterone production.


Subject(s)
Aldosterone/biosynthesis , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Mutation , Adrenal Glands/metabolism , Calcium Channel Blockers/pharmacology , Cell Line, Tumor , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gene Expression , Gene Expression Regulation/drug effects , Genetic Vectors/genetics , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Steroids/biosynthesis , Transcription, Genetic , Transduction, Genetic
9.
Mol Cell Endocrinol ; 384(1-2): 102-8, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24472523

ABSTRACT

Aberrant expression of gonadotropin-releasing hormone receptor (GnRHR) has been reported in human adrenal tissues including aldosterone-producing adenoma (APA). However, the details of its expression and functional role in adrenals are still not clear. In this study, quantitative RT-PCR analysis revealed the mean level of GnRHR mRNA was significantly higher in APAs than in human normal adrenal (NA) (P=0.004). GnRHR protein expression was detected in human NA and neoplastic adrenal tissues. In H295R cells transfected with GnRHR, treatment with GnRH resulted in a concentration-dependent increase in CYP11B2 reporter activity. Chronic activation of GnRHR with GnRH (100nM), in a cell line with doxycycline-inducible GnRHR (H295R-TR/GnRHR), increased CYP11B2 expression and aldosterone production. These agonistic effects were inhibited by blockers for the calcium signaling pathway, KN93 and calmidazolium. These results suggest GnRH, through heterotopic expression of its receptor, may be a potential regulator of CYP11B2 expression levels in some cases of APA.


Subject(s)
Adrenal Gland Neoplasms/genetics , Adrenocortical Adenoma/genetics , Cytochrome P-450 CYP11B2/genetics , Gene Expression Regulation, Neoplastic , Gonadotropin-Releasing Hormone/genetics , Receptors, LHRH/genetics , Adrenal Cortex/metabolism , Adrenal Cortex/pathology , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Adrenocortical Adenoma/metabolism , Adrenocortical Adenoma/pathology , Aldosterone/biosynthesis , Benzylamines/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Cell Line, Tumor , Cytochrome P-450 CYP11B2/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Humans , Imidazoles/pharmacology , Receptors, LHRH/metabolism , Sulfonamides/pharmacology
10.
J Mol Endocrinol ; 52(2): 159-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24403568

ABSTRACT

Purkinje cell protein 4 (PCP4) is a calmodulin (CaM)-binding protein that accelerates calcium association and dissociation with CaM. It has been previously detected in aldosterone-producing adenomas (APA), but details on its expression and function in adrenocortical tissues have remained unknown. Therefore, we performed the immunohistochemical analysis of PCP4 in the following tissues: normal adrenal (NA; n=15), APA (n=15), cortisol-producing adenomas (n=15), and idiopathic hyperaldosteronism cases (IHA; n=5). APA samples (n=45) were also submitted to quantitative RT-PCR of PCP4, CYP11B1, and CYP11B2, as well as DNA sequencing for KCNJ5 mutations. Transient transfection analysis using PCP4 siRNA was also performed in H295R adrenocortical carcinoma cells, following ELISA analysis, and CYP11B2 luciferase assays were also performed after PCP4 vector transfection in order to study the regulation of PCP4 protein expression. In our findings, PCP4 immunoreactivity was predominantly detected in APA and in the zona glomerulosa of NA and IHA. In APA, the mRNA levels of PCP4 were significantly correlated with those of CYP11B2 (P<0.0001) and were significantly higher in cases with KCNJ5 mutation than WT (P=0.005). Following PCP4 vector transfection, CYP11B2 luciferase reporter activity was significantly higher than controls in the presence of angiotensin-II. Knockdown of PCP4 resulted in a significant decrease in CYP11B2 mRNA levels (P=0.012) and aldosterone production (P=0.011). Our results indicate that PCP4 is a regulator of aldosterone production in normal, hyperplastic, and neoplastic human adrenocortical cells.


Subject(s)
Adrenal Cortex/metabolism , Aldosterone/biosynthesis , Nerve Tissue Proteins/metabolism , Adenoma/genetics , Adenoma/pathology , Adrenal Cortex/drug effects , Adrenal Cortex/pathology , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Angiotensin II/pharmacology , Blotting, Western , Cell Line , Colforsin/pharmacology , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , DNA/metabolism , Enzyme Assays , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Luciferases/metabolism , Nerve Tissue Proteins/genetics , Plasmids/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Transfection
11.
J Clin Endocrinol Metab ; 98(11): E1861-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24037882

ABSTRACT

CONTEXT: Primary aldosteronism is a heterogeneous group of disorders comprising both sporadic and familial forms. Mutations in the KCNJ5 gene, which encodes the inward rectifier K(+) channel 4 (G protein-activated inward rectifier K(+) channel 4, Kir3.4), cause familial hyperaldosteronism type III (FH-III) and are involved in the pathogenesis of sporadic aldosterone-producing adenomas. OBJECTIVE: The objective of the study was to characterize the effects of a newly described KCNJ5 mutation in vitro. PATIENTS AND METHODS: The index case is a 62-year-old woman affected by primary aldosteronism, who underwent left adrenalectomy after workup for adrenal adenoma. Exon 1 of KCNJ5 was PCR amplified from adrenal tissue and peripheral blood and sequenced. Electrophysiological and gene expression studies were performed to establish the functional effects of the new mutation on the membrane potential and adrenal cell CYP11B2 expression. RESULTS: KCNJ5 sequencing in the index case revealed a new p.Y152C germline mutation; interestingly, the phenotype of the patient was milder than most of the previously described FH-III families. The tyrosine-to-cysteine substitution resulted in pathological Na(+) permeability, cell membrane depolarization, and disturbed intracellular Ca(2+) homeostasis, effects similar, albeit smaller, to the ones demonstrated for other KCNJ5 mutations. Gene expression studies revealed an increased expression of CYP11B2 and its transcriptional regulator NR4A2 in HAC15 adrenal cells overexpressing KCNJ5(Y152C) compared to the wild-type channel. The effect was clearly Ca(2+)-dependent, because it was abolished by the calcium channel blocker nifedipine. CONCLUSIONS: Herein we describe a new germline mutation in KCNJ5 responsible for FH-III.


Subject(s)
Adenoma/genetics , Adrenal Gland Neoplasms/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Adenoma/pathology , Adenoma/surgery , Adrenal Gland Neoplasms/pathology , Adrenal Gland Neoplasms/surgery , Adrenalectomy , Calcium/metabolism , Cytochrome P-450 CYP11B2/genetics , Family Health , Female , HEK293 Cells , Humans , Hyperaldosteronism/genetics , Hyperaldosteronism/pathology , Hyperaldosteronism/surgery , Middle Aged , Point Mutation
12.
J Clin Endocrinol Metab ; 97(8): E1567-72, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22628608

ABSTRACT

CONTEXT: Primary aldosteronism is a heterogeneous disease that includes both sporadic and familial forms. A point mutation in the KCNJ5 gene is responsible for familial hyperaldosteronism type III. Somatic mutations in KCNJ5 also occur in sporadic aldosterone producing adenomas (APA). OBJECTIVE: The objective of the study was to define the effect of the KCNJ5 mutations on gene expression and aldosterone production using APA tissue and human adrenocortical cells. METHODS: A microarray analysis was used to compare the transcriptome profiles of female-derived APA samples with and without KCNJ5 mutations and HAC15 adrenal cells overexpressing either mutated or wild-type KCNJ5. Real-time PCR validated a set of differentially expressed genes. Immunohistochemical staining localized the KCNJ5 expression in normal adrenals and APA. RESULTS: We report a 38% (18 of 47) prevalence of KCNJ5 mutations in APA. KCNJ5 immunostaining was highest in the zona glomerulosa of NA and heterogeneous in APA tissue, and KCNJ5 mRNA was 4-fold higher in APA compared with normal adrenals (P < 0.05). APA with and without KCNJ5 mutations displayed slightly different gene expression patterns, notably the aldosterone synthase gene (CYP11B2) was more highly expressed in APA with KCNJ5 mutations. Overexpression of KCNJ5 mutations in HAC15 increased aldosterone production and altered expression of 36 genes by greater than 2.5-fold (P < 0.05). Real-time PCR confirmed increases in CYP11B2 and its transcriptional regulator, NR4A2. CONCLUSIONS: KCNJ5 mutations are prevalent in APA, and our data suggest that these mutations increase expression of CYP11B2 and NR4A2, thus increasing aldosterone production.


Subject(s)
Adenoma/genetics , Adrenal Cortex/metabolism , Adrenal Gland Neoplasms/genetics , Aldosterone/biosynthesis , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Mutation , Adenoma/metabolism , Adrenal Cortex/cytology , Adrenal Gland Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Humans
13.
Mol Cell Endocrinol ; 350(2): 151-62, 2012 Mar 24.
Article in English | MEDLINE | ID: mdl-21839803

ABSTRACT

Aldosterone is the major mineralocorticoid synthesized by the adrenal and plays an important role in the regulation of systemic blood pressure through the absorption of sodium and water. Aldosterone production is regulated tightly by selective expression of aldosterone synthase (CYP11B2) in the adrenal outermost zone, the zona glomerulosa. Angiotensin II (Ang II), potassium (K(+)) and adrenocorticotropin (ACTH) are the main physiological agonists which regulate aldosterone secretion. Aldosterone production is regulated within minutes of stimulation (acutely) through increased expression and phosphorylation of the steroidogenic acute regulatory (StAR) protein and over hours to days (chronically) by increased expression of the enzymes involved in the synthesis of aldosterone, particularly CYP11B2. Imbalance in any of these processes may lead to several disorders of aldosterone excess. In this review we attempt to summarize the key molecular events involved in the acute and chronic phases of aldosterone secretion.


Subject(s)
Aldosterone/biosynthesis , Metabolic Networks and Pathways/physiology , Adrenocorticotropic Hormone/pharmacology , Aldosterone/chemistry , Angiotensin II/pharmacology , Animals , Humans , Hyperaldosteronism/enzymology , Hyperaldosteronism/etiology , Hyperaldosteronism/genetics , Hyperaldosteronism/metabolism , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Models, Biological , Potassium/pharmacology , Steroids/biosynthesis , Time Factors
14.
Int J Diabetes Dev Ctries ; 29(4): 143-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20336195

ABSTRACT

The neuropathies of the peripheral, central and autonomic nervous systems are known to be caused by hyperglycemia, a consequence of the deregulation of glucose in diabetes. Several in vivo models such as streptozotocin-induced diabetic rats, mice and Chinese hamsters have been used to study the pathogenesis of diabetic neuropathy because of their resemblance to human pathology. However, these in vivo models have met with strong ethical oppositions. Further, the system complexity has inherent limitations of inconvenience of analyzing ephemeral molecular events and crosstalk of signal transduction pathways. Alternative in vitro models have been selected and put to effective use in diabetic studies. We critically review the use of these in vitro models such as primary cultures of dorsal root ganglia, Schwann cells and neural tissue as well as neural cell lines which have proved to be excellent systems for detailed study. We also assess the use of embryo cultures for the study of hyperglycemic effects on development, especially of the nervous system. These systems function as useful models to scrutinize the molecular events underlying hyperglycemia-induced stress in neuronal systems and have been very effectively used for the same. This comprehensive overview of advantages and disadvantages of in vitro systems that are currently in use will be of interest especially for comparative assessment of results and for appropriate choice of models for experiments in diabetic neuropathy.

SELECTION OF CITATIONS
SEARCH DETAIL
...