Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 416
Filter
1.
Int J Mol Sci ; 23(2)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35054901

ABSTRACT

Mesenchymal stem cells (MSCs) can differentiate into endoderm lineages, especially parathyroid-hormone (PTH)-releasing cells. We have previously reported that tonsil-derived MSC (T-MSC) can differentiate into PTH-releasing cells (T-MSC-PTHCs), which restored the parathyroid functions in parathyroidectomy (PTX) rats. In this study, we demonstrate quality optimization by standardizing the differentiation rate for a better clinical application of T-MSC-PTHCs to overcome donor-dependent variation of T-MSCs. Quantitation results of PTH mRNA copy number in the differentiated cells and the PTH concentration in the conditioned medium confirmed that the differentiation efficiency largely varied depending on the cells from each donor. In addition, the differentiation rate of the cells from all the donors greatly improved when differentiation was started at a high cell density (100% confluence). The large-scale expression profiling of T-MSC-PTHCs by RNA sequencing indicated that those genes involved in exiting the differentiation and the cell cycle were the major pathways for the differentiation of T-MSC-PTHCs. Furthermore, the implantation of the T-MSC-PTHCs, which were differentiated at a high cell density embedded in hyaluronic acid, resulted in a higher serum PTH in the PTX model. This standardized efficiency of differentiation into PTHC was achieved by initiating differentiation at a high cell density. Our findings provide a potential solution to overcome the limitations due to donor-dependent variation by establishing a standardized differentiation protocol for the clinical application of T-MSC therapy in treating hypoparathyroidism.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/metabolism , Palatine Tonsil/cytology , Parathyroid Hormone/biosynthesis , Biomarkers , Calcium/metabolism , Cell Culture Techniques , Cells, Cultured , Contact Inhibition , Extracellular Space/metabolism , Gene Expression Profiling , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Humans , Mesenchymal Stem Cells/cytology
2.
Bioconjug Chem ; 32(5): 909-915, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33890782

ABSTRACT

Polymeric small interfering RNA (siRNA) conjugate was elaborated to sequentially circumvent the predefined biological barriers encountered in the journey of transcellular delivery of siRNA into cytosol. Herein, classic ring-opening polymerization was employed for synthesis of well-defined poly(amino acid) derivatives possessing an array of carboxyl groups in an attempt to resemble the structural characteristics of hyaluronan. Furthermore, the hyaluronan-like synthetic was conjugated with a multiple of siRNA through a glutathione (GSH)-responsive disulfide linkage. The siRNA conjugate appeared to utilize the hyaluronan-specific receptors of CD44 for cell internalization, indicating similar functionalities to our hyaluronan-mimicking synthetic. Furthermore, the carboxyl groups of hyaluronan-like synthetics were designed to be selectively detached in subcellular acidic endosomes/lysosomes and transform into the cytomembrane-disruptive flanking ethylenediamine moieties, which appeared to be crucial in facilitating translocation of siRNA payloads from entrapment and degradation in lysosomes toward the cytosol. Eventually, active siRNA could be smoothly released from the synthetic due to the GSH cleavage disulfide linkage (disulfide), consequently accounting for potent RNA knockdown activities (>90%) toward cancerous cells. In addition, appreciable knockdown of parathyroid hormone was also achieved from our proposed siRNA conjugates in parathyroid cells. Hence, the elaborated siRNA conjugate showed tremendous potential in treatment of hyperparathyroidism, and could be developed further for systemic RNA interference (RNAi) therapeutics. Moreover, this study could also be the first example of a synthetic mimic to hyaluronan acquiring its functionalities, which could have important implications for further development of biomimic materials in pursuit of biomedical applications.


Subject(s)
Drug Carriers/chemistry , Parathyroid Hormone/biosynthesis , Polymers/chemistry , RNA Interference , Biological Transport , Cell Line , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics
3.
Proc Natl Acad Sci U S A ; 115(16): E3749-E3758, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29618612

ABSTRACT

The pathogenesis of parathyroid gland hyperplasia is poorly understood, and a better understanding is essential if there is to be improvement over the current strategies for prevention and treatment of secondary hyperparathyroidism. Here we investigate the specific role of Klotho expressed in the parathyroid glands (PTGs) in mediating parathyroid hormone (PTH) and serum calcium homeostasis, as well as the potential interaction between calcium-sensing receptor (CaSR) and Klotho. We generated mouse strains with PTG-specific deletion of Klotho and CaSR and dual deletion of both genes. We show that ablating CaSR in the PTGs increases PTH synthesis, that Klotho has a pivotal role in suppressing PTH in the absence of CaSR, and that CaSR together with Klotho regulates PTH biosynthesis and PTG growth. We utilized the tdTomato gene in our mice to visualize and collect PTGs to reveal an inhibitory function of Klotho on PTG cell proliferation. Chronic hypocalcemia and ex vivo PTG culture demonstrated an independent role for Klotho in mediating PTH secretion. Moreover, we identify an interaction between PTG-expressed CaSR and Klotho. These findings reveal essential and interrelated functions for CaSR and Klotho during parathyroid hyperplasia.


Subject(s)
Glucuronidase/physiology , Parathyroid Glands/metabolism , Parathyroid Hormone/biosynthesis , Receptors, G-Protein-Coupled/physiology , Animals , Bone and Bones/pathology , Calcium/metabolism , Calcium, Dietary/administration & dosage , Female , Fibroblast Growth Factor-23 , Glucuronidase/deficiency , Glucuronidase/genetics , Homeostasis , Hypercalcemia/genetics , Hypercalcemia/pathology , Hyperparathyroidism/genetics , Hyperparathyroidism/pathology , Hyperplasia , Hypocalcemia/metabolism , Hypophosphatemia/genetics , Hypophosphatemia/pathology , Immunoprecipitation , Kidney/pathology , Klotho Proteins , Male , Mice , Parathyroid Glands/pathology , Parathyroid Hormone/genetics , Protein Interaction Mapping , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Calcium-Sensing , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics
4.
Int J Mol Sci ; 21(8)2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32331456

ABSTRACT

Primary hyperparathyroidism is a common endocrinopathy that is mainly caused by benign parathyroid adenomas. The frequency, clinical presentation and complications of the disease show significant differences between genders, with the majority of cases being reported in postmenopausal women. Due to this gender predilection, several studies have investigated the role of sex hormones in the pathogenesis of the disease and their potential use as targets for optimal and gender-specific management. Epigenetic mechanisms that regulate gene transcription may also contribute to these differences between genders. In this review, we outline what is currently known regarding the role of sex hormones and the recent data on the role of non-coding RNAs in the differences between genders in primary hyperparathyroidism due to sporadic parathyroid adenomas.


Subject(s)
Parathyroid Neoplasms/epidemiology , Parathyroid Neoplasms/etiology , Disease Susceptibility , Epigenesis, Genetic , Female , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Gonadal Steroid Hormones/genetics , Gonadal Steroid Hormones/metabolism , Humans , Male , Parathyroid Hormone/biosynthesis , Parathyroid Neoplasms/metabolism , Prevalence , Sex Factors
5.
Pediatr Blood Cancer ; 65(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-28843052

ABSTRACT

We report the case of a 14-year-old male with metastatic alveolar rhabdomyosarcoma, presenting with hypercalcaemia (3.89 mmol/l) and elevated parathyroid hormone (PTH) level (10.2 pmol/l). Imaging demonstrated extensive bony lytic damage, with "floating teeth" in the mandible. Normalisation of calcium levels and bony reformation of the mandible occurred following chemotherapy; PTH levels decreased initially but remained above normal levels. Imaging did not demonstrate any evidence of parathyroid abnormality. Tumour ectopic PTH secretion is a very rare cause of hypercalcaemia of malignancy in children. Hypercalcaemia with an elevated PTH, in the absence of parathyroid-related cause, should prompt investigation for underlying malignancy.


Subject(s)
Alveolar Bone Loss/blood , Gene Expression Regulation, Neoplastic , Hypercalcemia/blood , Parathyroid Hormone/biosynthesis , Rhabdomyosarcoma, Alveolar/blood , Adolescent , Humans , Male
6.
Cell Physiol Biochem ; 42(2): 551-563, 2017.
Article in English | MEDLINE | ID: mdl-28578352

ABSTRACT

BACKGROUND/AIMS: Endogenous parathyroid hormone (PTH) plays an important role in fracture healing. This study investigated whether endogenous PTH regulates fracture healing by bone morphogenetic protein (BMP) and/or the transforming growth factor-ß (TGF-ß) signaling pathway. METHODS: Eight-week-old wild-type (WT) and PTH-knockout (PTH KO) male mice were selected, and models of open right-femoral fracture were constructed. Fracture healing and callus characteristics of mice in the two groups were compared by X-ray, micro-computed tomography, histological, and immunohistochemical examinations. Bone marrow mesenchymal stem cells (BMMSCs) of 8-week-old WT and PTHKO male mice were obtained and induced into osteoblasts and chondrocytes. RESULTS: We found that expression levels of Runt-related transcription factor (RUNX2), bone morphogenetic protein-receptor-type Ⅱ (BMPR2), phosphorylated Smad 1/5/8, and phosphorylated cyclic adenosine monophosphate-responsive element binding protein (CREB) in the callus of PTHKO mice were significantly decreased, whereas no significant difference in expression of SOX9, TGF-ßR2,or pSMAD2/3 was observed between PTHKO and WT mice. Additionally, the activity of osteoblast alkaline phosphatase was low at 7 days post-induction, and was upregulated by addition of PTH or dibutyryl cyclic adenosine monophosphate (dbcAMP) to the cell culture. Furthermore, H89 (protein kinase A inhibitor)eliminated the simulating effects of PTH and dbcAMP, and a low concentration of cyclic adenosine monophosphate (cAMP) was observed in PTHKO mouse BMMSCs. CONCLUSION: These results suggested that endogenous PTH enhanced BMPR2 expression by a cAMP/PKA/CREB pathway in osteoblasts, and increased RUNX2 expression through transduction of the BMP/pSMAD1/5/8 signaling pathway.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/biosynthesis , Fracture Healing/genetics , Fractures, Open/genetics , Parathyroid Hormone/genetics , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/genetics , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Fractures, Open/pathology , Fractures, Open/therapy , Gene Expression Regulation/drug effects , Humans , Isoquinolines/administration & dosage , Mice , Mice, Knockout , Osteoblasts , Parathyroid Hormone/biosynthesis , Signal Transduction/genetics , Smad Proteins/genetics , Sulfonamides/administration & dosage
7.
J Pept Sci ; 19(8): 504-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23794508

ABSTRACT

The parathyroid hormone, PTH, is responsible for calcium and phosphate ion homeostasis in the body. The first 34 amino acids of the peptide maintain the biological activity of the hormone and is currently marketed for calcium imbalance disorders. Although several methods for the production of recombinant PTH(1-34) have been reported, most involve the use of cleavage conditions that result in a modified peptide or unfavorable side products. Herein, we detail the recombinant production of (15) N-enriched human parathyroid hormone, (15) N PTH(1-34), generated via a plasmid vector that gives reasonable yield, low-cost protease cleavage (leaving the native N-terminal serine in its amino form), and purification by affinity and size exclusion chromatography. We characterize the product by multidimensional, heteronuclear NMR, circular dichroism, and LC/MS.


Subject(s)
Endopeptidases/chemistry , Parathyroid Hormone/biosynthesis , Amino Acid Sequence , Base Sequence , Chromatography, Affinity , Escherichia coli , Humans , Molecular Sequence Data , Parathyroid Hormone/chemistry , Parathyroid Hormone/isolation & purification , Protein Structure, Secondary , Proteolysis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification
8.
J Ind Microbiol Biotechnol ; 40(6): 589-99, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23529666

ABSTRACT

Human serum albumin (HSA) and human parathyroid hormone (1-34) [PTH (1-34)] fusion protein [HSA/PTH (1-34)] is a promising long-acting form of PTH (1-34) for osteoporosis treatment. Secretory expression of intact HSA/PTH (1-34) in Pichia pastoris GS115 was accompanied by two degradation fragments, with molecular weights around 66 kDa, in addition to the well-known ~45 kDa HSA-truncated fragment, resulting in a low yield of intact protein. In this study, two internal cleavage sites were identified in the PTH (1-34) portion of the fusion protein by Western Blot analysis. To minimize proteolytic cleavages, several protease genes including PEP4 (encoding proteinase A), PRB1 (proteinase B) and seven YPSs genes (yapsin family members) were knocked out respectively by disruption of the individual genes and the selective combinations. Reduced degradation was observed by single disruption of either PEP4 gene or YPS1 gene, and the lowest level of degradation was observed in a pep4△yps1△ double disruptant. After 72 h of induction, more than 80 % of the HSA/PTH (1-34) secreted by the pep4△yps1△ double disruptant remained intact, in comparison to only 30 % with the wild-type strain.


Subject(s)
Aspartic Acid Endopeptidases/deficiency , Genes, Fungal/genetics , Parathyroid Hormone/metabolism , Pichia/genetics , Pichia/metabolism , Proteolysis , Recombinant Fusion Proteins/metabolism , Serum Albumin/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Batch Cell Culture Techniques , Bioreactors , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Fermentation , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Mass Spectrometry , Mutation/genetics , Parathyroid Hormone/biosynthesis , Parathyroid Hormone/genetics , Pichia/classification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Serum Albumin/biosynthesis , Serum Albumin/genetics
9.
PLoS Genet ; 6(12): e1001251, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21203493

ABSTRACT

In mammals, parathyroid hormone (PTH) is a key regulator of extracellular calcium and inorganic phosphorus homeostasis. Although the parathyroid glands were thought to be the only source of PTH, extra-parathyroid PTH production in the thymus, which shares a common origin with parathyroids during organogenesis, has been proposed to provide an auxiliary source of PTH, resulting in a higher than expected survival rate for aparathyroid Gcm2⁻/⁻ mutants. However, the developmental ontogeny and cellular identity of these "thymic" PTH-expressing cells is unknown. We found that the lethality of aparathyroid Gcm2⁻/⁻ mutants was affected by genetic background without relation to serum PTH levels, suggesting a need to reconsider the physiological function of thymic PTH. We identified two sources of extra-parathyroid PTH in wild-type mice. Incomplete separation of the parathyroid and thymus organs during organogenesis resulted in misplaced, isolated parathyroid cells that were often attached to the thymus; this was the major source of thymic PTH in normal mice. Analysis of thymus and parathyroid organogenesis in human embryos showed a broadly similar result, indicating that these results may provide insight into human parathyroid development. In addition, medullary thymic epithelial cells (mTECs) express PTH in a Gcm2-independent manner that requires TEC differentiation and is consistent with expression as a self-antigen for negative selection. Genetic or surgical removal of the thymus indicated that thymus-derived PTH in Gcm2⁻/⁻ mutants did not provide auxiliary endocrine function. Our data show conclusively that the thymus does not serve as an auxiliary source of either serum PTH or parathyroid function. We further show that the normal process of parathyroid organogenesis in both mice and humans leads to the generation of multiple small parathyroid clusters in addition to the main parathyroid glands, that are the likely source of physiologically relevant "thymic PTH."


Subject(s)
Parathyroid Glands/metabolism , Parathyroid Hormone/biosynthesis , Thymus Gland/metabolism , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Organogenesis , Parathyroid Glands/embryology , Parathyroid Glands/immunology , Parathyroid Hormone/blood , Parathyroid Hormone/immunology , Thymus Gland/embryology , Thymus Gland/immunology , Transcription Factors/genetics , Transcription Factors/immunology
10.
Clin Oral Investig ; 17(3): 989-97, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22777389

ABSTRACT

OBJECTIVES: Recent studies indicate that high mobility group box protein 1 (HMGB1) can be released by necrotic and damaged cells and functions as an alarmin that is recognized by the innate immune system. Little is known about the role of HMGB1 within the periodontal ligament (PDL). Therefore, we examined HMGB1 expression by PDL cells in vitro and compared the findings to an in vivo model of orthodontically induced tooth root resorption. In addition, we addressed the question of whether a potentially anabolic intermittent administration of parathyroid hormone (iPTH) would modulate the expression of HMGB1. MATERIALS AND METHODS: In confluent PDL cell cultures, HMGB1 messenger RNA (mRNA) expression was quantified by real-time polymerase chain reaction. In a rat model comprising 25 animals, mechanical loading for 5 days was followed by administration of either iPTH (1-34) systemically or sham injections for up to 56 days. HMGB1 expression was determined by means of immunohistochemistry and histomorphometry. RESULTS: The in vitro experiments revealed an inhibitory effect of iPTH on basal HMGB1 mRNA expression in confluent PDL cells. In vivo, the mechanical force-induced enhanced HMGB1 protein expression declined time dependently. Intermittent PTH further inhibited HMGB1 expression. The significantly higher basal HMGB1 protein expression in the former compression side was followed by a more pronounced time- and iPTH-dependent decline in the same area. CONCLUSIONS: These data indicate a major role for HMGB1 in the regulation of PDL wound healing following mechanical load-induced tissue injury. CLINICAL RELEVANCE: The findings point to the potential benefit of iPTH in the attempt to support these immune-associated reparative processes.


Subject(s)
Alveolar Bone Loss/surgery , Bone Regeneration/physiology , HMGB1 Protein/physiology , Parathyroid Hormone/pharmacology , Periodontal Ligament/physiology , Tooth Movement Techniques , Adolescent , Alveolar Bone Loss/etiology , Animals , Bone Regeneration/drug effects , Bone Regeneration/genetics , Child , Compressive Strength/physiology , Dental Stress Analysis , HMGB1 Protein/biosynthesis , HMGB1 Protein/genetics , Humans , Male , Osteoprotegerin/biosynthesis , Parathyroid Hormone/biosynthesis , Parathyroid Hormone/genetics , Parathyroid Hormone/physiology , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , RANK Ligand/biosynthesis , Rats , Rats, Wistar , Signal Transduction , Stress, Mechanical , Tensile Strength/physiology , Tooth Movement Techniques/adverse effects
11.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33912936

ABSTRACT

Secondary hyperparathyroidism (SHPT) in uremic patients is characterized by parathyroid gland (PTG) hyperplasia and parathyroid hormone (PTH) elevation. Previously, we demonstrated that NF-κB activation contributed to parathyroid cell proliferation in rats with chronic kidney disease. Although vitamin D inhibits inflammation and ameliorates SHPT, the contribution of vitamin D deficiency to SHPT via local NF-κB activation remains to be clarified. PTGs collected from 10 uremic patients with advanced SHPT were used to test the expressions of vitamin D receptor (VDR), NF-κB, and proliferating cell nuclear antigen (PCNA). Freshly excised PTG tissues were incubated for 24 hours in vitro with VDR activator (VDRA) calcitriol or NF-κB inhibitor pyrrolidine thiocarbamate (PDTC). Chromatin immunoprecipitation (ChIP) and luciferase reporter assays were performed to investigate the regulation of PTH transcription by NF-κB. We found higher levels of activated NF-κB and lower expression of VDR in nodular hyperplastic PTGs than in diffuse hyperplasia. In cultured PTG tissues, treatment with VDRA or PDTC inhibited NF-κB activation and PCNA expression, and downregulated preproPTH mRNA and intact PTH levels. ChIP assays demonstrated the presence of NF-κB binding sites in PTH promoter. Furthermore, in luciferase reporter assays, addition of exogenous p65 significantly increased PTH luciferase activity by 2.4-fold (P < 0.01), while mutation of NF-κB binding site at position -908 of the PTH promoter suppressed p65-induced PTH reporter activity (P < 0.01). In summary, local NF-κB activation contributes to SHPT and mediates the transcriptional activation of PTH directly in uremic patients. Vitamin D deficiency may be involved in SHPT via the activation of NF-κB pathway.


Subject(s)
NF-kappa B/physiology , Parathyroid Glands/metabolism , Parathyroid Hormone/metabolism , Uremia/metabolism , Calcitriol/administration & dosage , Female , Humans , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/metabolism , Hyperparathyroidism, Secondary/pathology , Hyperplasia , Male , Middle Aged , NF-kappa B/antagonists & inhibitors , Parathyroid Glands/chemistry , Parathyroid Glands/pathology , Parathyroid Hormone/biosynthesis , Parathyroid Hormone/genetics , Proliferating Cell Nuclear Antigen/analysis , Pyrrolidines/administration & dosage , Receptors, Calcitriol/analysis , Receptors, Calcitriol/drug effects , Receptors, Calcitriol/metabolism , Thiocarbamates/administration & dosage , Tissue Culture Techniques , Transcription Factor RelA/analysis , Transcription, Genetic/drug effects , Uremia/complications , Uremia/pathology
12.
J Clin Invest ; 117(12): 4003-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17992255

ABSTRACT

Phosphate homeostasis is maintained by a counterbalance between efflux from the kidney and influx from intestine and bone. FGF23 is a bone-derived phosphaturic hormone that acts on the kidney to increase phosphate excretion and suppress biosynthesis of vitamin D. FGF23 signals with highest efficacy through several FGF receptors (FGFRs) bound by the transmembrane protein Klotho as a coreceptor. Since most tissues express FGFR, expression of Klotho determines FGF23 target organs. Here we identify the parathyroid as a target organ for FGF23 in rats. We show that the parathyroid gland expressed Klotho and 2 FGFRs. The administration of recombinant FGF23 led to an increase in parathyroid Klotho levels. In addition, FGF23 activated the MAPK pathway in the parathyroid through ERK1/2 phosphorylation and increased early growth response 1 mRNA levels. Using both rats and in vitro rat parathyroid cultures, we show that FGF23 suppressed both parathyroid hormone (PTH) secretion and PTH gene expression. The FGF23-induced decrease in PTH secretion was prevented by a MAPK inhibitor. These data indicate that FGF23 acts directly on the parathyroid through the MAPK pathway to decrease serum PTH. This bone-parathyroid endocrine axis adds a new dimension to the understanding of mineral homeostasis.


Subject(s)
Fibroblast Growth Factors/metabolism , Gene Expression Regulation/physiology , Homeostasis/physiology , Parathyroid Glands/metabolism , Parathyroid Hormone/biosynthesis , Phosphates/metabolism , Animals , Bone and Bones/metabolism , Cells, Cultured , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation/drug effects , Glucuronidase/biosynthesis , Glucuronidase/genetics , Homeostasis/drug effects , Humans , Intestinal Mucosa/metabolism , Kidney/metabolism , Klotho Proteins , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Parathyroid Glands/cytology , Parathyroid Hormone/genetics , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Vitamin D/metabolism
13.
Biochem Biophys Res Commun ; 391(1): 768-72, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19944067

ABSTRACT

Polarized salivary epithelial cells can sort secretory proteins towards either the basolateral or apical pole. Transgenic human parathyroid hormone (hPTH) exclusively sorts apically in rat submandibular glands. To help understand this specific process we modified the hPTH cDNA sequence and delivered the cDNAs to glands in vivo using adenoviral (Ad) vectors. The Ad vectors encoded: (1) the native form of hPTH (Ad.pre-pro-hPTH1-84), (2) the native sequence, but with the pro-segment deleted (Ad.pre-hPTH1-84), and (3) a sequence containing the pre-segment followed by the first 34 amino acids of hPTH (Ad.pre-hPTH1-34). hPTH production and sorting were studied after two days. All constructs were effectively transcribed in targeted glands. However, the pre-hPTH1-84 modification led to reduced hPTH secretion and production, while no immunoreactive hPTH resulted from pre-hPTH1-34 cDNA infusion. The pre-hPTH1-84 modification had no effect on apical sorting. These in vivo results show that the signal responsible for hPTH's apical sorting does not reside in the pro-segment and that deleting both the pro-segment and the carboxyl-terminal region severely impairs post-translational processing of hPTH.


Subject(s)
Parathyroid Hormone/biosynthesis , Recombinant Proteins/biosynthesis , Salivary Glands/metabolism , Adenoviridae , Animals , Genetic Vectors , Humans , Male , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Protein Transport , Rats , Rats, Transgenic , Rats, Wistar , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transduction, Genetic
14.
Nephron Clin Pract ; 116(4): c263-8, 2010.
Article in English | MEDLINE | ID: mdl-20639672

ABSTRACT

Recent observational studies of patients with stage 3-5 chronic kidney disease (CKD) not undergoing dialysis have shown that even slight increases in parathyroid hormone (PTH) levels are associated with an increased cardiovascular risk, regardless of the serum levels of calcium and phosphorus and vitamin D therapy. These studies suggest paying particular attention to monitoring PTH levels from the early stages of CKD, and preventing any mineral metabolism disorders that may trigger the excessive synthesis and secretion of PTH. However, it is not easy to determine when an appropriate response becomes maladaptive and requires the pharmacological suppression of the parathyroid gland because the gland's adaptive response can vary widely from one person to another. Furthermore, PTH levels are not always a good predictor of bone turnover and current PTH assays have various methodological limitations. Treating the early mineral metabolism abnormalities of CKD may help prevent the cardiovascular complications whose frequency, costs and mortality have a profound effect on society as a whole. For this reason, there is great interest in establishing adequate target ranges for PTH at different stages of CKD, and determining the most appropriate strategies for reaching them.


Subject(s)
Kidney Failure, Chronic/blood , Parathyroid Hormone/blood , Biomarkers/blood , Clinical Trials as Topic/standards , Humans , Kidney Failure, Chronic/diagnosis , Parathyroid Hormone/biosynthesis , Parathyroid Hormone/metabolism
15.
Endocr J ; 57(2): 161-70, 2010.
Article in English | MEDLINE | ID: mdl-19952463

ABSTRACT

Elevation of serum parathyroid hormone (PTH) in patients with medullary thyroid cancer (MTC) is usually found in multiple endocrine neoplasia type 2A (MEN2A). However, ectopic production of PTH is rare and its molecular etiology remains largely uninvestigated. We report a case of ectopic production of PTH by a sporadic MTC. The etiology of ectopic PTH gene expression was examined, focusing on GCM2 which has a crucial role in developing parathyroid glands. We observed ectopic expression of the PTH and GCM2 genes in tissues from the tumor and metastatic lymph nodes. However, GCM2 gene expression was also detected in adjacent thyroid tissue and lymphoblasts, in which PTH gene expression was absent. Hypomethylation of the PTH promoter, which is reportedly associated with ectopic production of PTH, was not seen in either the tumor tissue or metastatic lymph nodes. Meanwhile, DNA hypomethylation was seen in a CpG island identified in the GCM2 promoter region, regardless of whether or not the GCM2 gene was expressed. We showed that transcriptional activity of the CpG island sequences cloned into a reporter plasmid was dependent upon DNA methylation. Finally, we present the first report of a PTH-producing MTC. There was no apparent association between ectopic PTH and GCM2 gene expression, despite co-expression of the two genes. Neither genomic rearrangement nor DNA hypomethylation in the PTH gene appeared responsible for ectopic production of PTH. Although DNA hypomethylation may be necessary for the GCM2 gene expression, ectopic expression of GCM2 won't be possible by DNA hypomethylation alone.


Subject(s)
Hormones, Ectopic/biosynthesis , Parathyroid Hormone/biosynthesis , Thyroid Neoplasms/metabolism , Adult , DNA Methylation , Female , Humans , Lymphatic Metastasis/physiopathology , Nuclear Proteins , Thyroid Gland/metabolism , Thyroid Neoplasms/pathology , Transcription Factors
16.
Kidney Int ; 75(5): 462-4, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19219003

ABSTRACT

Kanai et al. used antisense technology to reduce excessive PTH production. The authors have overcome technical difficulties to demonstrate that, by strategies of RNA interference, a steady reduction of PTH secretion can be induced in cultured parathyroid-cell spheroids and in athymic nude mice with hyperplastic parathyroid cells transplanted from patients with secondary hyperparathyroidism.


Subject(s)
Cell Transplantation , Hyperparathyroidism, Secondary/therapy , Parathyroid Glands/cytology , Animals , Cell Culture Techniques , Down-Regulation/genetics , Humans , Mice , Parathyroid Hormone/biosynthesis , Parathyroid Hormone/genetics , RNA Interference
17.
Semin Nephrol ; 29(2): 97-104, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19371800

ABSTRACT

As kidney function declines, chronic kidney disease (CKD) becomes an increasingly systemic disorder. Most patients with CKD eventually develop subclinical or clinical abnormalities in bone and mineral metabolism. Recent observational and basic scientific studies have led to a new emphasis on the changes in phosphorus and calcium metabolism, parathyroid hormone, and vitamin D that lead to this complex systemic bone/mineral disorder (CKD/BMD). At the center of the disorder are relationships among all 4 factors that conspire to create a perfect storm, leading to secondary hyperparathyroidism (SHPT). Some key current issues that are reviewed here are as follows: (1) factors promoting SHPT, (2) the role of fibroblast growth factor-23 in CKD/BMD, (3) molecular mechanisms of SHPT, (4) mechanisms of vascular calcification, and (5) medical management of the disorder, including calcimimetics. Current therapies directed at correcting the primary abnormalities (ie, improve conditions to an imperfect storm) and minimizing the consequences of CKD/BMD are discussed.


Subject(s)
Bone Diseases, Metabolic/etiology , Bone and Bones/metabolism , Kidney Failure, Chronic/metabolism , Minerals/metabolism , Bone Density , Bone Diseases, Metabolic/metabolism , Humans , Hyperparathyroidism, Secondary/complications , Hyperparathyroidism, Secondary/metabolism , Kidney Failure, Chronic/complications , Parathyroid Hormone/biosynthesis , Risk Factors
18.
J Cell Biol ; 80(3): 715-31, 1979 Mar.
Article in English | MEDLINE | ID: mdl-457765

ABSTRACT

The formation of parathyroid hormone (PTH) in the parathyroid gland occurs via two successive proteolytic cleavages from larger biosynthetic precursors. The initial product coded for by PTH mRNA is pre-proparathyroid hormone (PreProPTH), a polypeptide of 115 amino acids. Within 1 min of synthesis, the polypeptide, proparathyroid hormone (ProPTH), is formed as a result of the proteolytic removal of the NH2-terminal 25 amino acids from Pre-ProPTH. After a delay of 15-20 min, the NH2-terminal six-amino acid sequence of ProPTH is removed to give PTH of 84 amino acids. To investigate the subcellular sites in the parathyroid cell where the biosynthetic precursors undergo specific proteolytic cleavages, we examined, by electron microscopy autoradiography, the spatiotemporal migration of autoradiographic grains and, by electrophoresis, the kinetics of the disappearance of labeled Pre-ProPTH and the conversion of labeled ProPTH to PTH in bovine parathyroid gland slices incubated with [3H]leucine for 5 min (pulse incubation) followed by incubations with unlabeled leucine for periods up to 85 min (chase incubations). By 5 min, 85% of the autoradiographic grains were confined to the rough endoplasmic reticulum (RER). Autoradiographic grains increased rapidly in number in the Golgi region after 15 min of incubation; from 15 to 30 min they migrated within secretory vesicles still in the Golgi region and then migrated to mature secretory granules outside the Golgi area. Electrophoretic analyses showed that Pre-ProPTH disappeared rapidly (by 5 min) and that conversion of ProPTH to PTH was first detectable at 15 min and was completed by 30 min. At later times of incubation (30-90 min), autoradiographic grains within the secretion glanules migrated to the periphery of the cell and to the plasma membrane, in correlation with the release of PTH first detected by 30 min. We conclude that proteolytic conversion of Pre-ProPTH to ProPTH takes place in the RER and that subsequent conversion of ProPTH to PTH occurs in the Golgi complex.


Subject(s)
Parathyroid Glands/metabolism , Parathyroid Hormone/biosynthesis , Protein Precursors/metabolism , Animals , Autoradiography/methods , Cattle , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Microscopy, Electron
19.
J Cell Biol ; 85(2): 292-8, 1980 May.
Article in English | MEDLINE | ID: mdl-7372710

ABSTRACT

Early events in the cellular synthesis and subsequent transfer into membrane-limited compartments of pre-proparathyroid hormone (pre-proPTH) and proparathyroid hormone (proPTH) were investigated by electrophoretic analyses of newly synthesized proteins in subcellular fractions of parthyroid gland slices pulse-labeled for 0.5-5 min with [(35)S] methionine. During these short times of incubation, both pre-proPTH and proPTH were confined to the microsomal fraction. Labeled pre-proPTH and proPTH were detected in a 30-s interval between 0.5 and 1.0 min of incubation. The radioactivity in proPTH became relatively constant between 3 and 5 min, whereas the radioactivity in ProPTH increased markedly over this period. When corrected for the known content of methionine in the prohormone and the prohormone, we found four times as much radiolabeled prohormone as prehormone between 0.5 and 1.0 min of synthesis. Sequestration of labeled prohomrone into endoplasmic reticulum compartments was shown by treatment of the microsomal fraction with chymotrypsin and trypsin, which resulted in the degradation of the prehormone but not of the prohormones. Approximately 50 percent of pre-prohormone and 25 percent of prohormone were released from the microsomes by their extraction with 1.0 M KCl, whereas 80-90 percent of both was released by treatment with Triton X-100. These results in intact cells support the signal hypothesis proposed by Blobel and his co-workers in studies utilizing cell-free systems, inasmuch as the results indicate transfer of prohormone into the cisternal space of the rough endoplasmic reticulum concomitant with the growth of the nascent polypeptide chain. Appearance of membrane-sequestered proPTH takes place without entry of pre-proPTH into the cisternal space, suggesting that proteolytic removal of the leader peptide occurs during transfer of the polypeptide through the lipid bilayer. Further evidence in support of this process is that pre-proPTH is only partly extracted from the microsomes by treatment with 1.0 M KCl, suggesting that a substantial fraction of the nascent pre-proPTH is integrally inserted into the membranes before it is cleaved to form proPTH.


Subject(s)
Parathyroid Glands/metabolism , Parathyroid Hormone/biosynthesis , Protein Precursors/biosynthesis , Animals , Cattle , Cytosol/metabolism , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum/metabolism , Microsomes/metabolism , Models, Biological , Parathyroid Glands/ultrastructure , Polyethylene Glycols/pharmacology , Polyribosomes/metabolism , Potassium Chloride/pharmacology
20.
J Cell Biol ; 72(1): 1-10, 1977 Jan.
Article in English | MEDLINE | ID: mdl-556615

ABSTRACT

We previously suggested that after synthesis, proparathormone is transferred from rough endoplasmic reticulum to the Golgi region where its conversion to parathormone occurs. We have attempted to define more closely this transfer process. In the first type of study, bovine parathyroid slices were incubated with [3H]leucine for 10 min and then radioisotope labeling was restricted by addition of a large excess of nonradioactive leucine. Under these conditions, more than 90% of the initially labeled proparathormone was converted to parathormone in 40 min. Lowered temperature in the chase period markedly inhibited the conversion. Several chemical agents were employed individually in the chase period to examine their effect on the conversion process. Antimycin A, dinitrophenol, oligomycin, and anaerobiosis (N2) inhibited the conversion, whereas sodium flouride and cycloheximide had no effect. In the second type of study, parathyroid slices were incubated with [3H]leucine for the entire incubation period. Lowered temperature and inhibitors of energy metabolism and microtubular function all lengthened the interval (lag) between the initial synthesis of [3H]parathormone. Cycloheximide, Tris, and chloroquine decreased the rates of protein synthesis and conversion, respectively, but none had any effect on the lag. We interpret the lag to represent the time of transit for proparathormone from rough endoplasmic reticulum to the Golgi region. We conclude that this transfer process is independent of the synthesis of the prohormone and its conversion to the hormone. Moreover, this translocation requires metabolic energy and appears to be mediated by microtubules.


Subject(s)
Parathyroid Glands/metabolism , Parathyroid Hormone/metabolism , Anaerobiosis , Animals , Antimycin A/pharmacology , Cattle , Chloroquine/pharmacology , Colchicine/pharmacology , Cycloheximide/pharmacology , Cytochalasin B/pharmacology , Deoxyglucose/pharmacology , Deuterium/pharmacology , Dinitrophenols/pharmacology , In Vitro Techniques , Kinetics , Oligomycins/pharmacology , Parathyroid Hormone/biosynthesis , Rotenone/pharmacology , Temperature , Tromethamine/pharmacology , Vinblastine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL