1
|
Fricke HP, Hernandez LL. The Serotonergic System and Bone Metabolism During Pregnancy and Lactation and the Implications of SSRI Use on the Maternal-Offspring Dyad. J Mammary Gland Biol Neoplasia 2023; 28:7. [PMID: 37086330 PMCID: PMC10122632 DOI: 10.1007/s10911-023-09535-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
Lactation is a physiological adaptation of the class Mammalia and is a product of over 200 million years of evolution. During lactation, the mammary gland orchestrates bone metabolism via serotonin signaling in order to provide sufficient calcium for the offspring in milk. The role of serotonin in bone remodeling was first discovered over two decades ago, and the interplay between serotonin, lactation, and bone metabolism has been explored in the years following. It is estimated that postpartum depression affects 10-15% of the population, and selective serotonin reuptake inhibitors (SSRI) are often used as the first-line treatment. Studies conducted in humans, nonhuman primates, sheep, and rodents have provided evidence that there are consequences on both parent and offspring when serotonin signaling is disrupted during the peripartal period; however, the long-term consequences of disruption of serotonin signaling via SSRIs during the peripartal period on the maternal and offspring skeleton are not fully known. This review will focus on the relationship between the mammary gland, serotonin, and bone remodeling during the peripartal period and the skeletal consequences of the dysregulation of the serotonergic system in both human and animal studies.
Collapse
Affiliation(s)
- Hannah P Fricke
- Animal and Dairy Sciences Department, University of Wisconsin-Madison, Madison, WI, USA
| | - Laura L Hernandez
- Animal and Dairy Sciences Department, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
2
|
Athonvarangkul D, Wysolmerski JJ. Crosstalk within a brain-breast-bone axis regulates mineral and skeletal metabolism during lactation. Front Physiol 2023; 14:1121579. [PMID: 36875035 PMCID: PMC9979219 DOI: 10.3389/fphys.2023.1121579] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/27/2023] [Indexed: 02/18/2023] Open
Abstract
To support the increased calcium demands for milk production during lactation, a dramatic and reversible physiological response occurs to alter bone and mineral metabolism. This coordinated process involves a brain-breast-bone axis that integrates hormonal signals that allow for adequate calcium delivery to milk yet also protects the maternal skeletal from excessive bone loss or decreases in bone quality or function. Here, we review the current knowledge on the crosstalk between the hypothalamus, mammary gland, and skeleton during lactation. We discuss the rare entity of pregnancy and lactation associated osteoporosis and consider how the physiology of bone turnover in lactation may impact the pathophysiology of postmenopausal osteoporosis. Further understanding of the regulators of bone loss during lactation, particularly in humans, may provide insights into new therapies for osteoporosis and other diseases of excess bone loss.
Collapse
Affiliation(s)
- Diana Athonvarangkul
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | | |
Collapse
|
3
|
Solodneva EV, Kuznetsov SB, Velieva AE, Stolpovsky YA. Molecular-Genetic Bases of Mammary Gland Development Using the Example of Cattle and Other Animal Species: I. Embryonic and Pubertal Developmental Stage. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422080087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
4
|
Sheftel CM, Liu L, Field SL, Weaver SR, Vezina CM, Peñagaricano F, Hernandez LL. Impact of Fluoxetine Treatment and Folic Acid Supplementation on the Mammary Gland Transcriptome During Peak Lactation. Front Pharmacol 2022; 13:828735. [PMID: 35281892 PMCID: PMC8904566 DOI: 10.3389/fphar.2022.828735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
Serotonin is a key regulator of mammary gland homeostasis during lactation. Selective serotonin reuptake inhibitors (SSRIs) are commonly used to treat peripartum depression, but also modulates mammary gland serotonin concentrations and signaling in part through DNA methylation. The objective of this study was to determine mouse mammary transcriptome changes in response to the SSRI fluoxetine and how methyl donor supplementation, achieved by folic acid supplementation, affected the transcriptome. Female C57BL/6J mice were fed either breeder diet (containing 4 mg/kg folic acid) or supplemented diet (containing 24 mg/kg folic acid) beginning 2 weeks prior to mating, then on embryonic day 13 mice were injected daily with either saline or 20 mg/kg fluoxetine. Mammary glands were harvested at peak lactation, lactation day 10, for transcriptomic analysis. Fluoxetine but not folic acid altered circulating serotonin and calcium concentrations, and folic acid reduced mammary serotonin concentrations, however only fluoxetine altered genes in the mammary transcriptome. Fluoxetine treatment altered fifty-six genes. Elovl6 was the most significantly altered gene by fluoxetine treatment along with gene pathways involving fatty acid homeostasis, PPARγ, and adipogenesis, which are critical for milk fat synthesis. Enriched pathways in the mammary gland by fluoxetine revealed pathways including calcium signaling, serotonin receptors, milk proteins, and cellular response to cytokine stimulus which are important for lactation. Although folic acid did not impact specific genes, a less stringent pathway analysis revealed more diffuse effects where folic acid enriched pathways involving negative regulation of gene expression as expected, but additionally enriched pathways involving serotonin, glycolysis, and lactalbumin which are critical for lactation. In conclusion, peripartal SSRI use and folic acid supplementation altered critical genes related to milk synthesis and mammary gland function that are important to a successful lactation. However, folic acid supplementation did not reverse changes in the mammary gland transcriptome altered by peripartal SSRI treatment.
Collapse
Affiliation(s)
- Celeste M Sheftel
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Lihe Liu
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Sena L Field
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Francisco Peñagaricano
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Laura L Hernandez
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
5
|
Al-Khan AA, Al Balushi NR, Richardson SJ, Danks JA. Roles of Parathyroid Hormone-Related Protein (PTHrP) and Its Receptor (PTHR1) in Normal and Tumor Tissues: Focus on Their Roles in Osteosarcoma. Front Vet Sci 2021; 8:637614. [PMID: 33796580 PMCID: PMC8008073 DOI: 10.3389/fvets.2021.637614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/17/2021] [Indexed: 12/17/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumor and originates from bone forming mesenchymal cells and primarily affects children and adolescents. The 5-year survival rate for OS is 60 to 65%, with little improvement in prognosis during the last four decades. Studies have demonstrated the evolving roles of parathyroid hormone-related protein (PTHrP) and its receptor (PTHR1) in bone formation, bone remodeling, regulation of calcium transport from blood to milk, regulation of maternal calcium transport to the fetus and reabsorption of calcium in kidneys. These two molecules also play critical roles in the development, progression and metastasis of several tumors such as breast cancer, lung carcinoma, chondrosarcoma, squamous cell carcinoma, melanoma and OS. The protein expression of both PTHrP and PTHR1 have been demonstrated in OS, and their functions and proposed signaling pathways have been investigated yet their roles in OS have not been fully elucidated. This review aims to discuss the latest research with PTHrP and PTHR1 in OS tumorigenesis and possible mechanistic pathways. This review is dedicated to Professor Michael Day who died in May 2020 and was a very generous collaborator.
Collapse
Affiliation(s)
- Awf A Al-Khan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.,Department of Pathology, Sohar Hospital, Sohar, Oman
| | - Noora R Al Balushi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Samantha J Richardson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.,School of Science, RMIT University, Bundoora, VIC, Australia
| | - Janine A Danks
- School of Science, RMIT University, Bundoora, VIC, Australia.,The University of Melbourne, Department of Medicine, Austin Health, Heidelberg, VIC, Australia
| |
Collapse
|
6
|
Diao J, DeBono A, Josephs TM, Bourke JE, Capuano B, Gregory KJ, Leach K. Therapeutic Opportunities of Targeting Allosteric Binding Sites on the Calcium-Sensing Receptor. ACS Pharmacol Transl Sci 2021; 4:666-679. [PMID: 33860192 DOI: 10.1021/acsptsci.1c00046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Indexed: 01/24/2023]
Abstract
The CaSR is a class C G protein-coupled receptor (GPCR) that acts as a multimodal chemosensor to maintain diverse homeostatic functions. The CaSR is a clinical therapeutic target in hyperparathyroidism and has emerged as a putative target in several other diseases. These include hyper- and hypocalcaemia caused either by mutations in the CASR gene or in genes that regulate CaSR signaling and expression, and more recently in asthma. The development of CaSR-targeting drugs is complicated by the fact that the CaSR possesses many different binding sites for endogenous and exogenous agonists and allosteric modulators. Binding sites for endogenous and exogenous ligands are located throughout the large CaSR protein and are interconnected in ways that we do not yet fully understand. This review summarizes our current understanding of CaSR physiology, signaling, and structure and how the many different binding sites of the CaSR may be targeted to treat disease.
Collapse
Affiliation(s)
- Jiayin Diao
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Aaron DeBono
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Tracy M Josephs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jane E Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| | - Ben Capuano
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| |
Collapse
|
7
|
Grinman D, Athonvarungkul D, Wysolmerski J, Jeong J. Calcium Metabolism and Breast Cancer: Echoes of Lactation? ACTA ACUST UNITED AC 2020; 15:63-70. [PMID: 33299957 DOI: 10.1016/j.coemr.2020.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lactation requires a series of adaptations in maternal calcium and bone metabolism to ensure a steady supply of calcium to the lactating mammary gland. The alterations in systemic metabolism are accompanied by alterations in the expression of calcium receptors, channels, binding proteins, pumps and transporters in mammary epithelial cells to increase the uptake of calcium from the extracellular fluid and to transport it into milk. Intracellular calcium regulates signaling pathways that mediate changes in cell proliferation, differentiation and death and many of the molecules involved in supporting and coordinating calcium secretion into milk are re-expressed and redeployed to support malignant behavior in breast cancer cells. In this article, we review adaptations of systemic calcium homeostasis during lactation, as well as the mechanisms of milk calcium transport. We then discuss how reactivation of these pathways contributes to the pathophysiology of breast cancer.
Collapse
Affiliation(s)
- Diego Grinman
- Section of Endocrinology and Metabolism, Department of Medicine, Yale School of Medicine
| | - Diana Athonvarungkul
- Section of Endocrinology and Metabolism, Department of Medicine, Yale School of Medicine
| | - John Wysolmerski
- Section of Endocrinology and Metabolism, Department of Medicine, Yale School of Medicine
| | - Jaekwang Jeong
- Section of Endocrinology and Metabolism, Department of Medicine, Yale School of Medicine
| |
Collapse
|
8
|
Sheftel CM, Hernandez LL. Serotonin stimulated parathyroid hormone related protein induction in the mammary epithelia by transglutaminase-dependent serotonylation. PLoS One 2020; 15:e0241192. [PMID: 33095824 PMCID: PMC7584195 DOI: 10.1371/journal.pone.0241192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/10/2020] [Indexed: 12/26/2022] Open
Abstract
Mammary-derived serotonin has been implicated in breast-to-bone communication during lactation by increasing parathyroid hormone related-protein (PTHrP) in the mammary gland. It is well established that PTHrP acts on the bone to liberate calcium for milk synthesis during lactation; however, the mechanism of serotonin’s regulation of PTHrP has not been fully elucidated. Recently, serotonylation has been shown to be involved in a variety of physiological processes mediated by serotonin. Therefore, we investigated whether serotonylation is involved in serotonin’s regulation of PTHrP in the mammary gland using lactogenically differentiated mouse mammary epithelial cells. We investigated the effect of increased intracellular serotonin using the antidepressant fluoxetine or 5-hydroxytryptophan (serotonin precursor), with or without transglutaminase inhibition and the corresponding action on PTHrP induction and activity. Treatment with fluoxetine or 5-hydroxytryptophan significantly increased intracellular serotonin concentrations and subsequently increased PTHrP gene expression, which was reduced with transglutaminase inhibition. Furthermore, we determined that transglutaminase activity is increased with lactogenic differentiation and 5-hydroxytryptophan or fluoxetine treatment. We investigated whether RhoA, Rac1, and Rab4 were potential serotonylation target proteins. We speculate that RhoA is potentially a serotonylation target protein. Our data suggest that serotonin regulates PTHrP induction in part through the process of serotonylation under lactogenic conditions in mouse mammary epithelial cells.
Collapse
Affiliation(s)
- Celeste M. Sheftel
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Laura L. Hernandez
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
9
|
Jin L, Sun H, Dan S, Li S, Zhang C, Zhang C, Ren X, Shan D, Ling S. Serotonin regulates maternal calcium homeostasis during the perinatal period of sheep. J Anim Sci 2020; 97:5009-5015. [PMID: 31697833 DOI: 10.1093/jas/skz346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/06/2019] [Indexed: 01/29/2023] Open
Abstract
The goal of this experiment was to demonstrate the ability of an infusion of serotonin (5-HT; 5-hydroxytryptamine) precursors to increase 5-HT production during the transition from pregnancy to lactation and its effects on gene expression related to calcium (Ca) transporters in the mammary gland and bone resorption markers in the femur. Thirty pregnant Bamei mutton sheep were randomly assigned to 3 experimental groups. All groups received a daily intravenous infusion of saline (control group; n = 10), saline containing 0.178 mg of L-tryptophan/kg body weight (BW) (TRP group, n = 10) or 0.178 mg of 5-hydroxytryptophan/kg BW (5-HTP group, n = 10), beginning on day 7 of prepartum and continuing until delivery. Serum (pre- and postpartum), milk (postpartum), and femur and mammary gland tissue (day 9) were collected. Sheep infused with 5-HTP had a larger total serum Ca concentration on days 3, 6, 15, and 30 of lactation and total milk Ca concentration on days 3, 6, 12, and 15 of lactation compared with that of the control group. Sheep infused with 5-HTP and TRP increased blood and milk concentrations of 5-HT on days 3, 6, 9, and 30 of lactation and parathyroid hormone-related protein (PTHrP) on day 3 of prepartum and on days 3, 6, and 15 of lactation (P < 0.05). In addition, compared to that of the control group, the TRP or 5-HTP infusion upregulated PTHrP, a sodium/calcium exchanger, plasma membrane Ca2+ ATPase 2, secretory pathway Ca2+ ATPase 1, and calcium sensing receptor mRNA expression in mammary gland and receptor-activated nuclear factor kappa-B ligand mRNA expression in the femur, but had no effect on receptor-activated nuclear factor kappa-B and osteoprotegerin mRNA expression in the femur (P < 0.05). This suggests that 5-HT and PTHrP may be involved in regulating maternal Ca homeostasis during the transition from pregnancy to lactation in the sheep.
Collapse
Affiliation(s)
- Lu Jin
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| | - Haizhou Sun
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| | - Sang Dan
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| | - Shengli Li
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| | - Chongzhi Zhang
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| | - Chunhua Zhang
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| | - Xiaoping Ren
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| | - Dan Shan
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| | - Shuli Ling
- Institute for Animal Nutrition and Feed Research, Inner Mongolia Academy of Agriculture and Animal Sciences, Hohhot, China
| |
Collapse
|
10
|
Das S, Clézardin P, Kamel S, Brazier M, Mentaverri R. The CaSR in Pathogenesis of Breast Cancer: A New Target for Early Stage Bone Metastases. Front Oncol 2020; 10:69. [PMID: 32117726 PMCID: PMC7013091 DOI: 10.3389/fonc.2020.00069] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
The Ca2+-sensing receptor (CaSR) is a class-C G protein-coupled receptor which plays a pivotal role in calciotropic processes, primarily in regulating parathyroid hormone secretion to maintain systemic calcium homeostasis. Among its non-calciotropic roles, where the CaSR sits at the intersection of myriad processes, it has steadily garnered attention as an oncogene or tumor suppressor in different organs. In maternal breast tissues the CaSR promotes lactation but in breast cancer it acts as an oncoprotein and has been shown to drive the pathogenesis of skeletal metastases from breast cancer. Even though research has made great strides in treating primary breast cancer, there is an unmet need when it comes to treatment of metastatic breast cancer. This review focuses on how the CaSR leads to the pathogenesis of breast cancer by contrasting its role in healthy tissues and tumorigenesis, and by drawing brief parallels with the tissues where it has been implicated as an oncogene. A class of compounds called calcilytics, which are CaSR antagonists, have also been surveyed in the instances where they have been used to target the receptor in cancerous tissues and constitute a proof of principle for repurposing them. Current clinical therapies for treating bone metastases from breast cancer are limited to targeting osteoclasts and a deeper understanding of the CaSR signaling nexus in this context can bolster them or lead to novel therapeutic interventions.
Collapse
Affiliation(s)
- Souvik Das
- MP3CV, EA7517, CURS, University of Picardie Jules Verne, Amiens, France
| | - Philippe Clézardin
- INSERM, Research Unit UMR_S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, Lyon, France
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Said Kamel
- MP3CV, EA7517, CURS, University of Picardie Jules Verne, Amiens, France
- Department of Biochemistry, Amiens-Picardie University Hospital, Amiens, France
- Faculty of Pharmacy, University of Picardie Jules Verne, Amiens, France
| | - Michel Brazier
- MP3CV, EA7517, CURS, University of Picardie Jules Verne, Amiens, France
- Department of Biochemistry, Amiens-Picardie University Hospital, Amiens, France
- Faculty of Pharmacy, University of Picardie Jules Verne, Amiens, France
| | - Romuald Mentaverri
- MP3CV, EA7517, CURS, University of Picardie Jules Verne, Amiens, France
- Department of Biochemistry, Amiens-Picardie University Hospital, Amiens, France
- Faculty of Pharmacy, University of Picardie Jules Verne, Amiens, France
| |
Collapse
|
11
|
Gaignon P, Le Grand K, Laza-Knoerr AL, Hurtaud C, Boudon A. Effect of calcium intake and the dietary cation-anion difference during early lactation on the bone mobilization dynamics throughout lactation in dairy cows. PLoS One 2019; 14:e0218979. [PMID: 31774817 PMCID: PMC6881032 DOI: 10.1371/journal.pone.0218979] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/07/2019] [Indexed: 12/29/2022] Open
Abstract
This study investigated the consequences of a low supply of dietary Ca with or without a low dietary cation-anion difference (DCAD) during early lactation on bone mobilization and reconstitution during lactation and on the dynamics of milk Ca content. Fifteen multiparous Holstein cows were distributed among 3 treatments 5 weeks before their expected calving date. These treatments differed based on the provision of diets through the first 10 weeks of lactation. During this period, the control treatment (NCa) consisted of a diet providing 100% of the Ca requirement, with a DCAD of 200 mEq/kg dry matter (DM). The LCa (low Ca) and LCaLD (low Ca, low DCAD) treatments consisted of diets providing 70% of the Ca requirement, with a DCAD of 200 and 0 mEq/kg DM, respectively. After 10 weeks, all cows received the same total mixed ration, which was formulated to meet 100% of the Ca requirement. LCa and LCaLD tended to decrease the body retention of Ca at 3 weeks of lactation compared with NCa but affected neither the dynamics of the blood biomarkers of bone formation and resorption during lactation nor the body retention of Ca at 17 weeks of lactation. Cows almost entirely compensated for the decrease in Ca supply caused by LCa and LCaLD by increasing their apparent digestive absorption of Ca at 3 weeks of lactation, whereas their apparent digestive absorption was unaffected by the treatments at 17 weeks of lactation. Milk production tended to be lower throughout lactation with LCa and LCaLD compared with NCa, with a mean difference of 2 kg/d. The results of this study also indicated that measuring the dynamics of milk Ca content during lactation cannot be considered effective for indirectly estimating the dynamics of bone mobilization in cows. The results also suggested that limited Ca intake at the beginning of lactation may have deleterious effects on milk production.
Collapse
Affiliation(s)
| | | | | | | | - Anne Boudon
- PEGASE, Agrocampus Ouest, INRA, Saint-Gilles, France
- * E-mail:
| |
Collapse
|
12
|
Han J, Shao J, Chen Q, Sun H, Guan L, Li Y, Liu J, Liu H. Transcriptional changes in the hypothalamus, pituitary, and mammary gland underlying decreased lactation performance in mice under heat stress. FASEB J 2019; 33:12588-12601. [PMID: 31480864 DOI: 10.1096/fj.201901045r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Because of climate change, heat stress (HS) causes more and more impacts on dairy animals to decrease lactation performance. The neuroendocrine system is key in regulating systemic physiological processes and milk synthesis. However, the hypothalamic-pituitary axis response to HS is still unclear. In this study, a group of lactating mice underwent a daily 2-h heat treatment (36°C) for 14 d to explore possible cross-talk between the hypothalamic-pituitary axis and mammary gland under HS. Transcriptome analyses by multitissue RNA-Seq indicated the possible mechanisms of reduced lactation performance in animals under HS. In the hypothalamus, the cAMP signaling pathway was activated to resist neuronal death, and the expression of downstream genes was increased to promote cell survival under HS. Reduced food intake might be caused by down-regulated appetite-related peptide, whereas up-regulated neuropeptide Y acted to attenuate reduced food intake. In pituitary, energy stress from lower food intake might result in reduced secretion of prolactin and growth hormone. Under HS, the mammary gland may undergo hypoxic stress, causing mammary epithelial cell apoptosis. Together, these data showed systemic changes in tissues to accommodate the effects of HS on lactation.-Han, J., Shao, J., Chen, Q., Sun, H., Guan, L., Li, Y., Liu, J., Liu, H. Transcriptional changes in the hypothalamus, pituitary, and mammary gland underlying decreased lactation performance in mice under heat stress.
Collapse
Affiliation(s)
- Jialiang Han
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Juanjuan Shao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Qiong Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Huizeng Sun
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and.,Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Leluo Guan
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and.,Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Yongxin Li
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Jianxin Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Hongyun Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| |
Collapse
|
13
|
García M, Rodríguez-Hernández CJ, Mateo-Lozano S, Pérez-Jaume S, Gonçalves-Alves E, Lavarino C, Mora J, de Torres C. Parathyroid hormone-like hormone plays a dual role in neuroblastoma depending on PTH1R expression. Mol Oncol 2019; 13:1959-1975. [PMID: 31293052 PMCID: PMC6717746 DOI: 10.1002/1878-0261.12542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/21/2019] [Accepted: 07/08/2019] [Indexed: 12/15/2022] Open
Abstract
We have previously reported the expression of parathyroid hormone‐like hormone (PTHLH) in well‐differentiated, Schwannian stroma‐rich neuroblastic tumors. The aim of this study was to functionally assess the role of PTHLH and its receptor, PTH1R, in neuroblastoma. Stable knockdown of PTHLH and PTH1R was conducted in neuroblastoma cell lines to investigate the succeeding phenotype induced both in vitro and in vivo. Downregulation of PTHLH reduced MYCN expression and subsequently induced cell cycle arrest, senescence, and migration and invasion impairment in a MYCN‐amplified, TP53‐mutated neuroblastoma cell line. These phenotypes were associated with reduced tumorigenicity in a murine model. We also show that PTHLH expression is not under the control of the calcium‐sensing receptor in neuroblastoma. Conversely, its production is stimulated by epidermal growth factor receptor (EGFR). Accordingly, irreversible EGFR inhibition with canertinib abolished PTHLH expression. The oncogenic role of PTHLH appeared to be a consequence of its intracrine function, as downregulation of its receptor, PTH1R, increased anchorage‐independent growth and induced a more undifferentiated, invasive phenotype. Respectively, high PTH1R mRNA expression was found in MYCN nonamplified primary tumors and also significantly associated with other prognostic factors of good outcome. This study provides the first evidence of the dual role of PTHLH in the behavior of neuroblastomas. Moreover, the identification of EGFR as a transcriptional regulator of PTHLH in neuroblastoma provides a novel therapeutic opportunity to promote a less aggressive tumor phenotype through irreversible inhibition of EGFR tyrosine kinase activity.
Collapse
Affiliation(s)
- Marta García
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | | | - Silvia Mateo-Lozano
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Sara Pérez-Jaume
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Eliana Gonçalves-Alves
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Cinzia Lavarino
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain.,Department of Haematology and Oncology, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Spain
| | - Jaume Mora
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain.,Department of Haematology and Oncology, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Spain
| | - Carmen de Torres
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain.,Department of Haematology and Oncology, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Spain
| |
Collapse
|
14
|
Hannan FM, Kallay E, Chang W, Brandi ML, Thakker RV. The calcium-sensing receptor in physiology and in calcitropic and noncalcitropic diseases. Nat Rev Endocrinol 2018; 15:33-51. [PMID: 30443043 PMCID: PMC6535143 DOI: 10.1038/s41574-018-0115-0] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Ca2+-sensing receptor (CaSR) is a dimeric family C G protein-coupled receptor that is expressed in calcitropic tissues such as the parathyroid glands and the kidneys and signals via G proteins and β-arrestin. The CaSR has a pivotal role in bone and mineral metabolism, as it regulates parathyroid hormone secretion, urinary Ca2+ excretion, skeletal development and lactation. The importance of the CaSR for these calcitropic processes is highlighted by loss-of-function and gain-of-function CaSR mutations that cause familial hypocalciuric hypercalcaemia and autosomal dominant hypocalcaemia, respectively, and also by the fact that alterations in parathyroid CaSR expression contribute to the pathogenesis of primary and secondary hyperparathyroidism. Moreover, the CaSR is an established therapeutic target for hyperparathyroid disorders. The CaSR is also expressed in organs not involved in Ca2+ homeostasis: it has noncalcitropic roles in lung and neuronal development, vascular tone, gastrointestinal nutrient sensing, wound healing and secretion of insulin and enteroendocrine hormones. Furthermore, the abnormal expression or function of the CaSR is implicated in cardiovascular and neurological diseases, as well as in asthma, and the CaSR is reported to protect against colorectal cancer and neuroblastoma but increase the malignant potential of prostate and breast cancers.
Collapse
Affiliation(s)
- Fadil M Hannan
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Enikö Kallay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Wenhan Chang
- Endocrine Research Unit, Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Luisa Brandi
- Metabolic Bone Diseases Unit, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy.
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
15
|
Zang WJ, Li H, Zhang ZF, QuZhen R, CuoMu YZ, Zhang DK, Luo J, Loor JJ, Zheng HL. Serotonin induces parathyroid hormone-related protein in goat mammary gland. J Anim Sci 2018; 96:1010-1016. [PMID: 29617866 DOI: 10.1093/jas/skx023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Indexed: 12/11/2022] Open
Abstract
During lactation, large amounts of calcium are exported from the mammary gland into milk to ensure skeletal growth of the offspring. Recent studies revealed that serotonin (5-HT) is essential to stimulate skeletal calcium resorption for milk synthesis. Our objective was to explore the correlation between circulating 5-HT and serum calcium and parathyroid hormone-related protein (PTHrP) concentrations around parturition in dairy goats. We also investigated the effect of 5-HT on PTHrP expression in cultured primary goat mammary epithelial cells (GMEC). Blood samples of multiparous Guanzhong dairy goats were collected on day -5 to 3 postpartum for analysis of serum concentrations of calcium, 5-HT, and PTHrP. Results revealed that from day -3 to 0 postpartum serum calcium and 5-HT concentrations decreased progressively, but serum PTHrP concentration only had a sharp drop in the postpartum period sampled. Correlation analysis of circulating 5-HT and serum calcium and PTHrP concentrations on day 1 and 2 postpartum revealed that low serum 5-HT concentration was positively correlated with serum total calcium or PTHrP concentration. By knocking down tryptophan hydroxylase-1 (TPH1) or adding 5-hydroxytryptophan (5-HTP) to decrease or increase the levels of 5-HT in GMEC, we observed that 5-HTP increased PTHrP expression in a dose-dependent manner and siTPH1 decreased PTHrP protein expression. Furthermore, 5-HT increased mRNA abundance of calcium-sensing receptor (CaSR) in a dose-dependent manner and decreased the expression of plasma membrane Ca2+ ATPase-1 (PMCA1). Taken together, 5-HT seems to induce PTHrP expression in goat mammary cells during and after parturition. These findings suggest that increasing 5-HT biosynthesis could be a potential therapeutic target for prevention of hypocalcemia in dairy goats.
Collapse
Affiliation(s)
- W J Zang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - H Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Z F Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - R QuZhen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Y Z CuoMu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - D K Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - J Luo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - J J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL
| | - H L Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
16
|
Weaver SR, Hernandez LL. Could use of Selective Serotonin Reuptake Inhibitors During Lactation Cause Persistent Effects on Maternal Bone? J Mammary Gland Biol Neoplasia 2018; 23:5-25. [PMID: 29603039 DOI: 10.1007/s10911-018-9390-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/13/2018] [Indexed: 02/07/2023] Open
Abstract
The lactating mammary gland elegantly coordinates maternal homeostasis to provide calcium for milk. During lactation, the monoamine serotonin regulates the synthesis and release of various mammary gland-derived factors, such as parathyroid hormone-related protein (PTHrP), to stimulate bone resorption. Recent evidence suggests that bone mineral lost during prolonged lactation is not fully recovered following weaning, possibly putting women at increased risk of fracture or osteoporosis. Selective Serotonin Reuptake Inhibitor (SSRI) antidepressants have also been associated with reduced bone mineral density and increased fracture risk. Therefore, SSRI exposure while breastfeeding may exacerbate lactational bone loss, compromising long-term bone health. Through an examination of serotonin and calcium homeostasis during lactation, lactational bone turnover and post-weaning recovery of bone mineral, and the effect of peripartum depression and SSRI on the mammary gland and bone, this review will discuss the hypothesis that peripartum SSRI exposure causes persistent reductions in bone mineral density through mammary-derived PTHrP signaling with bone.
Collapse
Affiliation(s)
- Samantha R Weaver
- Endocrine and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Laura L Hernandez
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
17
|
Suarez-Bregua P, Guerreiro PM, Rotllant J. Stress, Glucocorticoids and Bone: A Review From Mammals and Fish. Front Endocrinol (Lausanne) 2018; 9:526. [PMID: 30250453 PMCID: PMC6139303 DOI: 10.3389/fendo.2018.00526] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
Glucocorticoids (GCs) are the final effector products of a neuroendocrine HPA/HPI axis governing energy balance and stress response in vertebrates. From a physiological point of view, basal GC levels are essential for intermediary metabolism and participate in the development and homeostasis of a wide range of body tissues, including the skeleton. Numerous mammalian studies have demonstrated that GC hormones exert a positive role during bone modeling and remodeling as they promote osteoblastogenesis to maintain the bone architecture. Although the pharmacological effect of the so-called stress hormones has been widely reported, the role of endogenous GCs on bone mineral metabolism as result of the endocrine stress response has been largely overlooked across vertebrates. In addition, stress responses are variable depending on the stressor (e.g., starvation, predation, and environmental change), life cycle events (e.g., migration and aging), and differ among vertebrate lineages, which react differently according to their biological, social and cognitive complexity (e.g., mineral demands, physical, and psychological stress). This review intends to summarize the endogenous GCs action on bone metabolism of mammals and fish under a variety of challenging circumstances. Particular emphasis will be given to the regulatory loop between GCs and the parathyroid hormone (PTH) family peptides, and other key regulators of mineral homeostasis and bone remodeling in vertebrates.
Collapse
Affiliation(s)
- Paula Suarez-Bregua
- Institute of Marine Research, Spanish National Research Council (IIM-CSIC), Vigo, Spain
| | | | - Josep Rotllant
- Institute of Marine Research, Spanish National Research Council (IIM-CSIC), Vigo, Spain
| |
Collapse
|
18
|
Nicolini A, Ferrari P, Diodati L, Carpi A. Recent Advances in Comprehending the Signaling Pathways Involved in the Progression of Breast Cancer. Int J Mol Sci 2017; 18:E2321. [PMID: 29099748 PMCID: PMC5713290 DOI: 10.3390/ijms18112321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 12/11/2022] Open
Abstract
This review describes recent advances in the comprehension of signaling pathways involved in breast cancer progression. Calcium sensing receptor (CaSR), caveolae signaling, signaling referred to hypoxia-inducing factors and disturbances in the apoptotic machinery are related to more general biological mechanisms and are considered first. The others refer to signaling pathways of more specific biological mechanisms, namely the heparin/heparin-sulfate interactome, over-expression of miRNA-378a-5p, restriction of luminal and basal epithelial cells, fatty-acid synthesis, molecular pathways related to epithelial to mesenchimal transition (EMT), HER-2/neu gene amplification and protein expression, and the expression of other members of the epithelial growth factor receptor family. This progress in basic research is fundamental to foster the ongoing efforts that use the new genotyping technologies, and aim at defining new prognostic and predictive biomarkers for a better personalized management of breast cancer disease.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Paola Ferrari
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Lucrezia Diodati
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Angelo Carpi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| |
Collapse
|
19
|
Suarez-Bregua P, Cal L, Cañestro C, Rotllant J. PTH Reloaded: A New Evolutionary Perspective. Front Physiol 2017; 8:776. [PMID: 29062283 PMCID: PMC5640766 DOI: 10.3389/fphys.2017.00776] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/22/2017] [Indexed: 11/23/2022] Open
Abstract
The parathyroid hormone (PTH) family is a group of structurally-related secreted peptides involved in bone mineral homeostasis and multitude of developmental processes in vertebrates. These peptides mediate actions through PTH receptors (PTHRs), which belong to the transmembrane G protein-coupled receptor group. To date, genes encoding for PTH and PTHR have only been identified in chordates, suggesting that this signaling pathway may be an evolutionary innovation of our phylum. In vertebrates, we found up to six PTH and three PTHR different paralogs, varying in number between mammals and teleost fishes due to the different rounds of whole-genome duplication and specific gene losses suffered between the two groups of animals. The diversification of the PTH gene family has been accompanied by both functional divergence and convergence, making sometimes difficult the comparison between PTH peptides of teleosts and mammals. Here, we review the roles of all Pth peptides in fishes, and based on the evolutionary history of PTH paralogs, we propose a new and simple nomenclature from PTH1 to PTH4. Moreover, the recent characterization of the Pth4 in zebrafish allows us to consider the prominent role of the brain-to-bone signaling pathway in the regulation of bone development and homeostasis. Finally, comparison between PTH peptides of fish and mammals allows us to discuss an evolutionary model for PTH functions related to bone mineral balance during the vertebrate transition from an aquatic to a terrestrial environment.
Collapse
Affiliation(s)
| | - Laura Cal
- Institute of Marine Research (IIM-CSIC), Vigo, Spain
| | - Cristian Cañestro
- Departament de Genètica, Microbiologia i Estadística, IRBio, Universitat de Barcelona, Barcelona, Spain
| | | |
Collapse
|
20
|
What Is Breast in the Bone? Int J Mol Sci 2016; 17:ijms17101764. [PMID: 27782069 PMCID: PMC5085788 DOI: 10.3390/ijms17101764] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/11/2016] [Accepted: 10/14/2016] [Indexed: 12/17/2022] Open
Abstract
The normal developmental program that prolactin generates in the mammary gland is usurped in the cancerous process and can be used out of its normal cellular context at a site of secondary metastasis. Prolactin is a pleiotropic peptide hormone and cytokine that is secreted from the pituitary gland, as well as from normal and cancerous breast cells. Experimental and epidemiologic data suggest that prolactin is associated with mammary gland development, and also the increased risk of breast tumors and metastatic disease in postmenopausal women. Breast cancer spreads to the bone in approximately 70% of cases with advanced breast cancer. Despite treatment, new bone metastases will still occur in 30%–50% of patients. Only 20% of patients with bone metastases survive five years after the diagnosis of bone metastasis. The breast cancer cells in the bone microenvironment release soluble factors that engage osteoclasts and/or osteoblasts and result in bone breakdown. The breakdown of the bone matrix, in turn, enhances the proliferation of the cancer cells, creating a vicious cycle. Recently, it was shown that prolactin accelerated the breast cancer cell-mediated osteoclast differentiation and bone breakdown by the regulation of breast cancer-secreted proteins. Interestingly, prolactin has the potential to affect multiple proteins that are involved in both breast development and likely bone metastasis, as well. Prolactin has normal bone homeostatic roles and, combined with the natural “recycling” of proteins in different tissues that can be used for breast development and function, or in bone function, increases the impact of prolactin signaling in breast cancer bone metastases. Thus, this review will focus on the role of prolactin in breast development, bone homeostasis and in breast cancer to bone metastases, covering the molecular aspects of the vicious cycle.
Collapse
|
21
|
Kim W, Wysolmerski JJ. Calcium-Sensing Receptor in Breast Physiology and Cancer. Front Physiol 2016; 7:440. [PMID: 27746743 PMCID: PMC5043011 DOI: 10.3389/fphys.2016.00440] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/16/2016] [Indexed: 12/31/2022] Open
Abstract
The calcium-sensing receptor (CaSR) is expressed in normal breast epithelial cells and in breast cancer cells. During lactation, activation of the CaSR in mammary epithelial cells increases calcium transport into milk and inhibits parathyroid hormone-related protein (PTHrP) secretion into milk and into the circulation. The ability to sense changes in extracellular calcium allows the lactating breast to actively participate in the regulation of systemic calcium and bone metabolism, and to coordinate calcium usage with calcium availability during milk production. Interestingly, as compared to normal breast cells, in breast cancer cells, the regulation of PTHrP secretion by the CaSR becomes rewired due to a switch in its G-protein usage such that activation of the CaSR increases instead of decreases PTHrP production. In normal cells the CaSR couples to Gαi to inhibit cAMP and PTHrP production, whereas in breast cancer cells, it couples to Gαs to stimulate cAMP and PTHrP production. Activation of the CaSR on breast cancer cells regulates breast cancer cell proliferation, death and migration, in part, by stimulating PTHrP production. In this article, we discuss the biology of the CaSR in the normal breast and in breast cancer, and review recent findings suggesting that the CaSR activates a nuclear pathway of PTHrP action that stimulates cellular proliferation and inhibits cell death, helping cancer cells adapt to elevated extracellular calcium levels. Understanding the diverse actions mediated by the CaSR may help us better understand lactation physiology, breast cancer progression and osteolytic bone metastases.
Collapse
Affiliation(s)
- Wonnam Kim
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine New Haven, CT, USA
| | - John J Wysolmerski
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine New Haven, CT, USA
| |
Collapse
|
22
|
Kim W, Takyar FM, Swan K, Jeong J, VanHouten J, Sullivan C, Dann P, Yu H, Fiaschi-Taesch N, Chang W, Wysolmerski J. Calcium-Sensing Receptor Promotes Breast Cancer by Stimulating Intracrine Actions of Parathyroid Hormone-Related Protein. Cancer Res 2016; 76:5348-60. [PMID: 27450451 DOI: 10.1158/0008-5472.can-15-2614] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 06/17/2016] [Indexed: 12/21/2022]
Abstract
Parathyroid hormone-related protein (PTHrP) contributes to the development and metastatic progression of breast cancer by promoting hypercalcemia, tumor growth, and osteolytic bone metastases, but it is not known how PTHrP is upregulated in breast tumors. Here we report a central role in this process for the calcium-sensing receptor, CaSR, which enables cellular responses to changes in extracellular calcium, through studies of CaSR-PTHrP interactions in the MMTV-PymT transgenic mouse model of breast cancer and in human breast cancer cells. CaSR activation stimulated PTHrP production by breast cancer cells in vitro and in vivo Tissue-specific disruption of the casr gene in mammary epithelial cells in MMTV-PymT mice reduced tumor PTHrP expression and inhibited tumor cell proliferation and tumor outgrowth. CaSR signaling promoted the proliferation of human breast cancer cell lines and tumor cells cultured from MMTV-PyMT mice. Further, CaSR activation inhibited cell death triggered by high extracellular concentrations of calcium. The actions of the CaSR appeared to be mediated by nuclear actions of PTHrP that decreased p27(kip1) levels and prevented nuclear accumulation of the proapoptotic factor apoptosis inducing factor. Taken together, our findings suggest that CaSR-PTHrP interactions might be a promising target for the development of therapeutic agents to limit tumor cell growth in bone metastases and in other microenvironments in which elevated calcium and/or PTHrP levels contribute to breast cancer progression. Cancer Res; 76(18); 5348-60. ©2016 AACR.
Collapse
Affiliation(s)
- Wonnam Kim
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven Connecticut
| | - Farzin M Takyar
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven Connecticut
| | - Karena Swan
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven Connecticut
| | - Jaekwang Jeong
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven Connecticut
| | - Joshua VanHouten
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven Connecticut
| | - Catherine Sullivan
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven Connecticut
| | - Pamela Dann
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven Connecticut
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii School of Medicine, Honolulu, Hawaii
| | - Nathalie Fiaschi-Taesch
- Section of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Wenhan Chang
- Endocrine Unit, San Francisco and Veteran Affairs Medical Center, University of California, San Francisco, California
| | - John Wysolmerski
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven Connecticut.
| |
Collapse
|
23
|
Ardeshirpour L, Dumitru C, Dann P, Sterpka J, VanHouten J, Kim W, Kostenuik P, Wysolmerski J. OPG Treatment Prevents Bone Loss During Lactation But Does Not Affect Milk Production or Maternal Calcium Metabolism. Endocrinology 2015; 156:2762-73. [PMID: 25961842 PMCID: PMC4511126 DOI: 10.1210/en.2015-1232] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lactation is associated with increased bone turnover and rapid bone loss, which liberates skeletal calcium used for milk production. Previous studies suggested that an increase in the skeletal expression of receptor activator of nuclear factor kappa-light-chain-enhancer of activated B cells ligand (RANKL) coupled with a decrease in osteoprotegerin (OPG) levels likely triggered bone loss during lactation. In this study, we treated lactating mice with recombinant OPG to determine whether bone loss during lactation was dependent on RANKL signaling and whether resorption of the maternal skeleton was required to support milk production. OPG treatment lowered bone resorption rates and completely prevented bone loss during lactation but, surprisingly, did not decrease osteoclast numbers. In contrast, OPG was quite effective at lowering osteoblast numbers and inhibiting bone formation in lactating mice. Furthermore, treatment with OPG during lactation prevented the usual anabolic response associated with reversal of lactational bone loss after weaning. Preventing bone loss had no appreciable effect on milk production, milk calcium levels, or maternal calcium homeostasis when mice were on a standard diet. However, when dietary calcium was restricted, treatment with OPG caused maternal hypocalcemia, maternal death, and decreased milk production. These studies demonstrate that RANKL signaling is a requirement for bone loss during lactation, and suggest that osteoclast activity may be required to increase osteoblast numbers during lactation in preparation for the recovery of bone mass after weaning. These data also demonstrate that maternal bone loss is not absolutely required to supply calcium for milk production unless dietary calcium intake is inadequate.
Collapse
Affiliation(s)
- Laleh Ardeshirpour
- Section of Endocrinology (L.A.), Department of Pediatrics, and Section of Endocrinology and Metabolism (C.D., P.D., J.S., J.V., W.K., J.W.), Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Metabolic Disorders (P.K.), Amgen, Inc, Thousand Oaks, California 91320
| | - Cristina Dumitru
- Section of Endocrinology (L.A.), Department of Pediatrics, and Section of Endocrinology and Metabolism (C.D., P.D., J.S., J.V., W.K., J.W.), Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Metabolic Disorders (P.K.), Amgen, Inc, Thousand Oaks, California 91320
| | - Pamela Dann
- Section of Endocrinology (L.A.), Department of Pediatrics, and Section of Endocrinology and Metabolism (C.D., P.D., J.S., J.V., W.K., J.W.), Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Metabolic Disorders (P.K.), Amgen, Inc, Thousand Oaks, California 91320
| | - John Sterpka
- Section of Endocrinology (L.A.), Department of Pediatrics, and Section of Endocrinology and Metabolism (C.D., P.D., J.S., J.V., W.K., J.W.), Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Metabolic Disorders (P.K.), Amgen, Inc, Thousand Oaks, California 91320
| | - Joshua VanHouten
- Section of Endocrinology (L.A.), Department of Pediatrics, and Section of Endocrinology and Metabolism (C.D., P.D., J.S., J.V., W.K., J.W.), Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Metabolic Disorders (P.K.), Amgen, Inc, Thousand Oaks, California 91320
| | - Wonnam Kim
- Section of Endocrinology (L.A.), Department of Pediatrics, and Section of Endocrinology and Metabolism (C.D., P.D., J.S., J.V., W.K., J.W.), Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Metabolic Disorders (P.K.), Amgen, Inc, Thousand Oaks, California 91320
| | - Paul Kostenuik
- Section of Endocrinology (L.A.), Department of Pediatrics, and Section of Endocrinology and Metabolism (C.D., P.D., J.S., J.V., W.K., J.W.), Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Metabolic Disorders (P.K.), Amgen, Inc, Thousand Oaks, California 91320
| | - John Wysolmerski
- Section of Endocrinology (L.A.), Department of Pediatrics, and Section of Endocrinology and Metabolism (C.D., P.D., J.S., J.V., W.K., J.W.), Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520; and Department of Metabolic Disorders (P.K.), Amgen, Inc, Thousand Oaks, California 91320
| |
Collapse
|
24
|
Peripheral serotonin regulates maternal calcium trafficking in mammary epithelial cells during lactation in mice. PLoS One 2014; 9:e110190. [PMID: 25299122 PMCID: PMC4192539 DOI: 10.1371/journal.pone.0110190] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/08/2014] [Indexed: 12/26/2022] Open
Abstract
Lactation is characterized by massive transcellular flux of calcium, from the basolateral side of the mammary alveolar epithelium (blood) into the ductal lumen (milk). Regulation of calcium transport during lactation is critical for maternal and neonatal health. The monoamine serotonin (5-HT) is synthesized by the mammary gland and functions as a homeostatic regulation of lactation. Genetic ablation of tryptophan hydroxylase 1 (Tph1), which encodes the rate-limiting enzyme in non-neuronal serotonin synthesis, causes a deficiency in circulating serotonin. As a consequence maternal calcium concentrations decrease, mammary epithelial cell morphology is altered, and cell proliferation is decreased during lactation. Here we demonstrate that serotonin deficiency decreases the expression and disrupts the normal localization of calcium transporters located in the apical (PMCA2) and basolateral (CaSR, ORAI-1) membranes of the lactating mammary gland. In addition, serotonin deficiency decreases the mRNA expression of calcium transporters located in intracellular compartments (SERCA2, SPCA1 and 2). Mammary expression of serotonin receptor isoform 2b and its downstream pathways (PLCβ3, PKC and MAP-ERK1/2) are also decreased by serotonin deficiency, which might explain the numerous phenotypic alterations described above. In most cases, addition of exogenous 5-hydroxy-L-tryptophan to the Tph1 deficient mice rescued the phenotype. Our data supports the hypothesis that serotonin is necessary for proper mammary gland structure and function, to regulate blood and mammary epithelial cell transport of calcium during lactation. These findings can be applicable to the treatment of lactation-induced hypocalcemia in dairy cows and can have profound implications in humans, given the wide-spread use of selective serotonin reuptake inhibitors as antidepressants during pregnancy and lactation.
Collapse
|
25
|
Ontsouka EC, Albrecht C. Cholesterol transport and regulation in the mammary gland. J Mammary Gland Biol Neoplasia 2014; 19:43-58. [PMID: 24510467 DOI: 10.1007/s10911-014-9316-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 01/22/2014] [Indexed: 01/28/2023] Open
Abstract
The milk-producing alveolar epithelial cells secrete milk that remains after birth the principal source of nutrients for neonates. Milk secretion and composition are highly regulated processes via integrated actions of hormones and local factors which involve specific receptors and downstream signal transduction pathways. Overall milk composition is similar among mammalian species, although the content of individual constituents such as lipids may significantly differ from one species to another. The milk lipid fraction is essentially composed of triglycerides, which represent more than 95 % of the total lipids in human and commercialized bovine milk. Though sterols, including cholesterol, which is the major milk sterol, represent less than 0.5 % of the total milk lipid fraction, they are of key importance for several biological processes. Cholesterol is required for the formation of biological membranes especially in rapidly growing organisms, and for the synthesis of sterol-based compounds. Cholesterol found in milk originates predominantly from blood uptake and, to a certain extent, from local synthesis in the mammary tissue. The present review summarizes current knowledge on cellular mechanisms and regulatory processes determining intra- and transcellular cholesterol transport in the mammary gland. Cholesterol exchanges between the blood, the mammary alveolar cells and the milk, and the likely role of active cholesterol transporters in these processes are discussed. In this context, the hormonal regulation and signal transduction pathways promoting active cholesterol transport as well as potential regulatory crosstalks are highlighted.
Collapse
Affiliation(s)
- Edgar C Ontsouka
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland
| | | |
Collapse
|
26
|
Cross BM, Breitwieser GE, Reinhardt TA, Rao R. Cellular calcium dynamics in lactation and breast cancer: from physiology to pathology. Am J Physiol Cell Physiol 2013; 306:C515-26. [PMID: 24225884 DOI: 10.1152/ajpcell.00330.2013] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Breast cancer is the second leading cause of cancer mortality in women, estimated at nearly 40,000 deaths and more than 230,000 new cases diagnosed in the U.S. this year alone. One of the defining characteristics of breast cancer is the radiographic presence of microcalcifications. These palpable mineral precipitates are commonly found in the breast after formation of a tumor. Since free Ca(2+) plays a crucial role as a second messenger inside cells, we hypothesize that these chelated precipitates may be a result of dysregulated Ca(2+) secretion associated with tumorigenesis. Transient and sustained elevations of intracellular Ca(2+) regulate cell proliferation, apoptosis and cell migration, and offer numerous therapeutic possibilities in controlling tumor growth and metastasis. During lactation, a developmentally determined program of gene expression controls the massive transcellular mobilization of Ca(2+) from the blood into milk by the coordinated action of calcium transporters, including pumps, channels, sensors and buffers, in a functional module that we term CALTRANS. Here we assess the evidence implicating genes that regulate free and buffered Ca(2+) in normal breast epithelium and cancer cells and discuss mechanisms that are likely to contribute to the pathological characteristics of breast cancer.
Collapse
Affiliation(s)
- Brandie M Cross
- Department of Physiology, The Johns Hopkins University, Baltimore, Maryland
| | | | | | | |
Collapse
|
27
|
O'Seaghdha CM, Wu H, Yang Q, Kapur K, Guessous I, Zuber AM, Köttgen A, Stoudmann C, Teumer A, Kutalik Z, Mangino M, Dehghan A, Zhang W, Eiriksdottir G, Li G, Tanaka T, Portas L, Lopez LM, Hayward C, Lohman K, Matsuda K, Padmanabhan S, Firsov D, Sorice R, Ulivi S, Brockhaus AC, Kleber ME, Mahajan A, Ernst FD, Gudnason V, Launer LJ, Mace A, Boerwinckle E, Arking DE, Tanikawa C, Nakamura Y, Brown MJ, Gaspoz JM, Theler JM, Siscovick DS, Psaty BM, Bergmann S, Vollenweider P, Vitart V, Wright AF, Zemunik T, Boban M, Kolcic I, Navarro P, Brown EM, Estrada K, Ding J, Harris TB, Bandinelli S, Hernandez D, Singleton AB, Girotto G, Ruggiero D, d'Adamo AP, Robino A, Meitinger T, Meisinger C, Davies G, Starr JM, Chambers JC, Boehm BO, Winkelmann BR, Huang J, Murgia F, Wild SH, Campbell H, Morris AP, Franco OH, Hofman A, Uitterlinden AG, Rivadeneira F, Völker U, Hannemann A, Biffar R, Hoffmann W, Shin S, Lescuyer P, Henry H, Schurmann C, Munroe PB, Gasparini P, Pirastu N, Ciullo M, Gieger C, März W, Lind L, Spector TD, Smith AV, Rudan I, Wilson JF, Polasek O, Deary IJ, Pirastu M, Ferrucci L, Liu Y, et alO'Seaghdha CM, Wu H, Yang Q, Kapur K, Guessous I, Zuber AM, Köttgen A, Stoudmann C, Teumer A, Kutalik Z, Mangino M, Dehghan A, Zhang W, Eiriksdottir G, Li G, Tanaka T, Portas L, Lopez LM, Hayward C, Lohman K, Matsuda K, Padmanabhan S, Firsov D, Sorice R, Ulivi S, Brockhaus AC, Kleber ME, Mahajan A, Ernst FD, Gudnason V, Launer LJ, Mace A, Boerwinckle E, Arking DE, Tanikawa C, Nakamura Y, Brown MJ, Gaspoz JM, Theler JM, Siscovick DS, Psaty BM, Bergmann S, Vollenweider P, Vitart V, Wright AF, Zemunik T, Boban M, Kolcic I, Navarro P, Brown EM, Estrada K, Ding J, Harris TB, Bandinelli S, Hernandez D, Singleton AB, Girotto G, Ruggiero D, d'Adamo AP, Robino A, Meitinger T, Meisinger C, Davies G, Starr JM, Chambers JC, Boehm BO, Winkelmann BR, Huang J, Murgia F, Wild SH, Campbell H, Morris AP, Franco OH, Hofman A, Uitterlinden AG, Rivadeneira F, Völker U, Hannemann A, Biffar R, Hoffmann W, Shin S, Lescuyer P, Henry H, Schurmann C, Munroe PB, Gasparini P, Pirastu N, Ciullo M, Gieger C, März W, Lind L, Spector TD, Smith AV, Rudan I, Wilson JF, Polasek O, Deary IJ, Pirastu M, Ferrucci L, Liu Y, Kestenbaum B, Kooner JS, Witteman JCM, Nauck M, Kao WHL, Wallaschofski H, Bonny O, Fox CS, Bochud M. Meta-analysis of genome-wide association studies identifies six new Loci for serum calcium concentrations. PLoS Genet 2013; 9:e1003796. [PMID: 24068962 PMCID: PMC3778004 DOI: 10.1371/journal.pgen.1003796] [Show More Authors] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/29/2013] [Indexed: 01/08/2023] Open
Abstract
Calcium is vital to the normal functioning of multiple organ systems and its serum concentration is tightly regulated. Apart from CASR, the genes associated with serum calcium are largely unknown. We conducted a genome-wide association meta-analysis of 39,400 individuals from 17 population-based cohorts and investigated the 14 most strongly associated loci in ≤ 21,679 additional individuals. Seven loci (six new regions) in association with serum calcium were identified and replicated. Rs1570669 near CYP24A1 (P = 9.1E-12), rs10491003 upstream of GATA3 (P = 4.8E-09) and rs7481584 in CARS (P = 1.2E-10) implicate regions involved in Mendelian calcemic disorders: Rs1550532 in DGKD (P = 8.2E-11), also associated with bone density, and rs7336933 near DGKH/KIAA0564 (P = 9.1E-10) are near genes that encode distinct isoforms of diacylglycerol kinase. Rs780094 is in GCKR. We characterized the expression of these genes in gut, kidney, and bone, and demonstrate modulation of gene expression in bone in response to dietary calcium in mice. Our results shed new light on the genetics of calcium homeostasis.
Collapse
Affiliation(s)
- Conall M. O'Seaghdha
- National Heart, Lung, and Blood Institute's Framingham Heart Study and Center for Population Studies, Framingham, Massachusetts, United States of America
- Renal Division, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Hongsheng Wu
- National Heart, Lung, and Blood Institute's Framingham Heart Study and Center for Population Studies, Framingham, Massachusetts, United States of America
- Department of Biostatistics, Boston University, Boston, Massachusetts, United States of America
- Department of Medical Biology, University of Split, School of Medicine, Split, Croatia
| | - Qiong Yang
- National Heart, Lung, and Blood Institute's Framingham Heart Study and Center for Population Studies, Framingham, Massachusetts, United States of America
- Department of Biostatistics, Boston University, Boston, Massachusetts, United States of America
| | - Karen Kapur
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Idris Guessous
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne, Switzerland
- Unit of Population Epidemiology, Division of Primary Care Medicine, Department of Community Medicine and Primary Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
- Geriatric Unit, Azienda Sanitaria Firenze (ASF), Florence, Italy
| | - Annie Mercier Zuber
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Anna Köttgen
- Renal Division, Freiburg University Hospital, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Candice Stoudmann
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Massimo Mangino
- King's College London, St. Thomas' Hospital Campus, London, United Kingdom
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Weihua Zhang
- Catheter Lab, Cardiology, Ealing Hospital, Southall, Middlesex, United Kingdom
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | | | - Guo Li
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America
| | - Toshiko Tanaka
- Clinical Research Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Laura Portas
- Institute of Population Genetics, CNR-Traversa La Crucca, Reg. Baldinca Li Punti, Sassari, Italy
| | - Lorna M. Lopez
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Caroline Hayward
- MRC Human Genetics Unit, MRC IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Kurt Lohman
- Cardiology Group, ClinPhenomics GmbH&Co KG, Frankfurt-Sachsenhausen, Germany
| | - Koichi Matsuda
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, Division of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - Dmitri Firsov
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Rossella Sorice
- Institute of Genetics and Biophysics ‘Adriano-Buzzati Traverso’, CNR, Napoli, Italy
| | - Sheila Ulivi
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - A. Catharina Brockhaus
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians University Munich, Munich, Germany
| | - Marcus E. Kleber
- Department of Internal Medicine II – Cardiology, University of Ulm Medical Centre, Ulm, Germany
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, United Kingdom
| | - Florian D. Ernst
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association Research Institute, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Lenore J. Launer
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Aurelien Mace
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Eric Boerwinckle
- University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dan E. Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Chizu Tanikawa
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yusuke Nakamura
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Morris J. Brown
- Cambridge Institute of Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Jean-Michel Gaspoz
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jean-Marc Theler
- Unit of Population Epidemiology, Division of Primary Care Medicine, Department of Community Medicine and Primary Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - David S. Siscovick
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America
- Departments of Medicine and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
- Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, Washington, United States of America
| | - Sven Bergmann
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Veronique Vitart
- MRC Human Genetics Unit, MRC IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan F. Wright
- MRC Human Genetics Unit, MRC IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Mladen Boban
- Department of Pharmacology, Faculty of Medicine, University of Split, Split, Croatia
| | - Ivana Kolcic
- Faculty of Medicine, University of Split, Split, Croatia
| | - Pau Navarro
- MRC Human Genetics Unit, MRC IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Edward M. Brown
- Division of Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Karol Estrada
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jingzhong Ding
- Division of Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Stefania Bandinelli
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Dena Hernandez
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andrew B. Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Giorgia Girotto
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics ‘Adriano-Buzzati Traverso’, CNR, Napoli, Italy
| | - Adamo Pio d'Adamo
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Antonietta Robino
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Computer Science and Networking, Wentworth Institute of Technology, Boston, Massachusetts, United States of America
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Gail Davies
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - John C. Chambers
- Catheter Lab, Cardiology, Ealing Hospital, Southall, Middlesex, United Kingdom
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Epidemiology and Biostatistics, Imperial College London, Norfolk Place, London, United Kingdom
| | - Bernhard O. Boehm
- Ulm University Medical Centre, Department of Internal Medicine I, Ulm University, Ulm, Germany
- LKC School of Medicine, Imperial College London and Nanyang Technological University, Singapore, Singapore
| | - Bernhard R. Winkelmann
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Jie Huang
- Division of Primary Care Medicine, Department of Community Medicine and Primary Care and Emergency Medicine , Geneva University Hospitals, Geneva, Switzerland
| | - Federico Murgia
- Institute of Population Genetics, CNR-Traversa La Crucca, Reg. Baldinca Li Punti, Sassari, Italy
| | - Sarah H. Wild
- Centre for Population Health Sciences, The University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - Harry Campbell
- Centre for Population Health Sciences, The University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, United Kingdom
| | - Oscar H. Franco
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andre G. Uitterlinden
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Fernando Rivadeneira
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Anke Hannemann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Reiner Biffar
- Department of Prosthetic Dentistry, Gerostomatology and Dental Materials, University Medicine Greifswald, Greifswald, Germany
| | - Wolfgang Hoffmann
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - So–Youn Shin
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Pierre Lescuyer
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Hughes Henry
- Clinical Chemistry Laboratory, Lausanne University Hospital, Lausanne, Switzerland
| | - Claudia Schurmann
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | | | | | - Patricia B. Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Paolo Gasparini
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Nicola Pirastu
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Marina Ciullo
- Institute of Genetics and Biophysics ‘Adriano-Buzzati Traverso’, CNR, Napoli, Italy
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Winfried März
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Synlab Centre of Laboratory Diagnostics, Heidelberg, Germany
| | - Lars Lind
- Institute of Medical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Tim D. Spector
- King's College London, St. Thomas' Hospital Campus, London, United Kingdom
| | - Albert V. Smith
- Icelandic Heart Association Research Institute, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Igor Rudan
- Centre for Population Health Sciences, The University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - James F. Wilson
- Centre for Population Health Sciences, The University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mario Pirastu
- Institute of Population Genetics, CNR-Traversa La Crucca, Reg. Baldinca Li Punti, Sassari, Italy
| | - Luigi Ferrucci
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Bryan Kestenbaum
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, Washington, United States of America
| | - Jaspal S. Kooner
- Catheter Lab, Cardiology, Ealing Hospital, Southall, Middlesex, United Kingdom
- Faculty of Medicine, National Heart & Lung Institute, Cardiovascular Science, Hammersmith Hospital, Hammersmith Campus, Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - W. H. Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Welch Center for Prevention, Epidemiology and Clinical Research, John Hopkins University, Baltimore, Maryland, United States of America
| | - Henri Wallaschofski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Olivier Bonny
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
- Service of Nephrology, Lausanne University Hospital, Lausanne, Switzerland
| | - Caroline S. Fox
- National Heart, Lung, and Blood Institute's Framingham Heart Study and Center for Population Studies, Framingham, Massachusetts, United States of America
- Division of Endocrinology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Murielle Bochud
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
28
|
Mamillapalli R, VanHouten J, Dann P, Bikle D, Chang W, Brown E, Wysolmerski J. Mammary-specific ablation of the calcium-sensing receptor during lactation alters maternal calcium metabolism, milk calcium transport, and neonatal calcium accrual. Endocrinology 2013; 154:3031-42. [PMID: 23782944 PMCID: PMC3749485 DOI: 10.1210/en.2012-2195] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
To meet the demands for milk calcium, the lactating mother adjusts systemic calcium and bone metabolism by increasing dietary calcium intake, increasing bone resorption, and reducing renal calcium excretion. As part of this adaptation, the lactating mammary gland secretes PTHrP into the maternal circulation to increase bone turnover and mobilize skeletal calcium stores. Previous data have suggested that, during lactation, the breast relies on the calcium-sensing receptor (CaSR) to coordinate PTHrP secretion and milk calcium transport with calcium availability. To test this idea genetically, we bred BLG-Cre mice with CaSR-floxed mice to ablate the CaSR specifically from mammary epithelial cells only at the onset of lactation (CaSR-cKO mice). Loss of the CaSR in the lactating mammary gland did not disrupt alveolar differentiation or milk production. However, it did increase the secretion of PTHrP into milk and decreased the transport of calcium from the circulation into milk. CaSR-cKO mice did not show accelerated bone resorption, but they did have a decrease in bone formation. Loss of the mammary gland CaSR resulted in hypercalcemia, decreased PTH secretion, and increased renal calcium excretion in lactating mothers. Finally, loss of the mammary gland CaSR resulted in decreased calcium accrual by suckling neonates, likely due to the combination of increased milk PTHrP and decreased milk calcium. These results demonstrate that the mammary gland CaSR coordinates maternal bone and calcium metabolism, calcium transport into milk, and neonatal calcium accrual during lactation.
Collapse
Affiliation(s)
- Ramanaiah Mamillapalli
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, TAC S131, Box 208020, New Haven, Connecticut 06520-8020, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Normal breast epithelial cells and breast cancer cells express the calcium-sensing receptor (CaSR), the master regulator of systemic calcium metabolism. During lactation, activation of the CaSR in mammary epithelial cells downregulates parathyroid hormone-related protein (PTHrP) levels in milk and in the circulation, and increases calcium transport into milk. In contrast, in breast cancer cells the CaSR upregulates PTHrP production. A switch in G-protein usage underlies the opposing effects of the CaSR on PTHrP expression in normal and malignant breast cells. During lactation, the CaSR in normal breast cells coordinates a feedback loop that matches the transport of calcium into milk and maternal calcium metabolism to the supply of calcium. A switch in CaSR G-protein usage during malignant transformation converts this feedback loop into a feed-forward cycle in breast cancer cells that may promote the growth of osteolytic skeletal metastases.
Collapse
Affiliation(s)
- Joshua N Vanhouten
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, TAC S131, Box 208020, New Haven, CT, USA.
| | | |
Collapse
|
30
|
Romero JR, Youte R, Brown EM, Pollak MR, Goltzman D, Karaplis A, Pong LC, Chien L, Chattopadhyay N, Rivera A. Parathyroid hormone ablation alters erythrocyte parameters that are rescued by calcium-sensing receptor gene deletion. Eur J Haematol 2013; 91:37-45. [PMID: 23528155 DOI: 10.1111/ejh.12110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2013] [Indexed: 11/29/2022]
Abstract
The mechanisms by which parathyroid hormone (PTH) produces anemia are unclear. Parathyroid hormone secretion is regulated by the extracellular Ca2+ -sensing receptor. We investigated the effects of ablating PTH on hematological indices and erythrocytes volume regulation in wild-type, PTH-null, and Ca2+ -sensing receptor-null/PTH-null mice. The erythrocyte parameters were measured in whole mouse blood, and volume regulatory systems were determined by plasma membrane K+ fluxes, and osmotic fragility was measured by hemoglobin determination at varying osmolarities. We observed that the absence of PTH significantly increases mean erythrocyte volume and reticulocyte counts, while decreasing erythrocyte counts, hemoglobin, hematocrit, and mean corpuscular hemoglobin concentration. These changes were accompanied by increases in erythrocyte cation content, a denser cell population, and increased K+ permeability, which were in part mediated by activation of the K+ /Cl- cotransporter and Gardos channel. In addition we observed that erythrocyte osmotic fragility in PTH-null compared with wild-type mice was enhanced. When Ca2+ -sensing receptor gene was deleted on the background of PTH-null mice, we observed that several of the alterations in erythrocyte parameters of PTH-null mice were largely rescued, particularly those related to erythrocyte volume, K+ fluxes and osmotic fragility, and became similar to those observed in wild-type mice. Our results demonstrate that Ca2+ -sensing receptor and parathyroid hormone are functionally coupled to maintain erythrocyte homeostasis.
Collapse
Affiliation(s)
- Jose R Romero
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
PTHrP was identified as a cause of hypercalcemia in cancer patients 25 yr ago. In the intervening years, we have learned that PTHrP and PTH are encoded by related genes that are part of a larger "PTH gene family." This evolutionary relationship permits them to bind to the same type 1 PTH/PTHrP receptor, which explains why humoral hypercalcemia of malignancy resembles hyperparathyroidism. This review will outline basic facts about PTHrP biology and its normal physiological functions, with an emphasis on new findings of the past 5-10 yr. The medical and research communities first became aware of PTHrP because of its involvement in a common paraneoplastic syndrome. Now, research into the basic biology of PTHrP has suggested previously unrecognized connections to a variety of disease states such as osteoporosis, osteoarthritis, and breast cancer and has highlighted how PTHrP itself might be used in therapy for osteoporosis and diabetes. Therefore, the story of this remarkable protein is a paradigm for translational research, having gone from bedside to bench and now back to bedside.
Collapse
Affiliation(s)
- John J Wysolmerski
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, TAC S131, Box 208020, New Haven, Connecticut 06520-8020, USA.
| |
Collapse
|
32
|
Ritter CS, Haughey BH, Miller B, Brown AJ. Differential gene expression by oxyphil and chief cells of human parathyroid glands. J Clin Endocrinol Metab 2012; 97:E1499-505. [PMID: 22585091 PMCID: PMC3591682 DOI: 10.1210/jc.2011-3366] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Parathyroid oxyphil cells, whose function is unknown, are thought to be derived from chief cells. Oxyphil cells increase in number in parathyroid glands of patients with chronic kidney disease (CKD) and are even more abundant in patients receiving treatment for hyperparathyroidism with calcitriol and/or the calcimimetic cinacalcet. OBJECTIVE We examined oxyphil and chief cells of parathyroid glands of CKD patients for differential expression of genes important to parathyroid function. DESIGN/SETTING/PARTICIPANTS Parathyroid tissue from CKD patients with refractory hyperparathyroidism was immunostained for gene expression studies. MAIN OUTCOME MEASURE Immunostaining for PTH, PTHrP, calcium-sensing receptor, glial cells missing 2, vitamin D receptor, 25-hydroxyvitamin D-1α-hydroxylase, and cytochrome c was quantified and expression reported for oxyphil and chief cells. RESULTS Expression of all proteins analyzed, except for the vitamin D receptor, was higher in oxyphil cells than in chief cells. CONCLUSION Human parathyroid oxyphil cells express parathyroid-relevant genes found in the chief cells and have the potential to produce additional autocrine/paracrine factors, such as PTHrP and calcitriol. Additional studies are warranted to define the secretory properties of these cells and clarify their role in parathyroid pathophysiology.
Collapse
Affiliation(s)
- Cynthia S Ritter
- Renal Division, Washington University School of Medicine, Barnes Jewish Hospital, St Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
33
|
Baio G, Fabbi M, Emionite L, Cilli M, Salvi S, Ghedin P, Prato S, Carbotti G, Tagliafico A, Truini M, Neumaier CE. In vivo imaging of human breast cancer mouse model with high level expression of calcium sensing receptor at 3T. Eur Radiol 2012; 22:551-558. [PMID: 21947485 DOI: 10.1007/s00330-011-2285-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/22/2011] [Accepted: 09/09/2011] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To demonstrate that manganese can visualise calcium sensing receptor (CaSR)-expressing cells in a human breast cancer murine model, as assessed by clinical 3T magnetic resonance (MR). METHODS Human MDA-MB-231-Luc or MCF7-Luc breast cancer cells were orthotopically grown in NOD/SCID mice to a minimum mass of 5 mm. Mice were evaluated on T1-weighted sequences before and after intravenous injection of MnCl(2). To block the CaSR-activated Ca(2+) channels, verapamil was injected at the tumour site 5 min before Mn(2+) administration. CaSR expression in vivo was studied by immunohistochemistry. RESULTS Contrast enhancement was observed at the tumour periphery 10 min after Mn(2+) administration, and further increased up to 40 min. In verapamil-treated mice, no contrast enhancement was observed. CaSR was strongly expressed at the tumour periphery. CONCLUSION Manganese enhanced magnetic resonance imaging can visualise CaSR-expressing breast cancer cells in vivo, opening up possibilities for a new MR contrast agent. KEY POINTS • Manganese contrast agents helped demonstrate breast cancer cells in an animal model. • Enhancement was most marked in cells with high calcium sensing receptor expression. • Manganese uptake was related to the distribution of CaSR within the tumour. • Manganese MRI may become useful to investigate human breast cancer.
Collapse
Affiliation(s)
- Gabriella Baio
- Department of Diagnostic Imaging, IST, National Cancer Institute, Largo Rosanna Benzi 10, 16132 Genoa, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
The calcium-sensing receptor is necessary for the rapid development of hypercalcemia in human lung squamous cell carcinoma. Neoplasia 2011; 13:428-38. [PMID: 21532883 DOI: 10.1593/neo.101620] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 02/09/2011] [Accepted: 02/10/2011] [Indexed: 11/18/2022] Open
Abstract
The calcium-sensing receptor (CaR) is responsible for the regulation of extracellular calcium (Ca(2+) (o)) homeostasis. CaR activation has been shown to increase proliferation in several cancer cell lines; however, its presence or function has never been documented in lung cancer. We report that Ca(2+) (o)-activated CaR results in MAPK-mediated stimulation of parathyroid hormone-related protein (PTHrP) production in human lung squamous cell carcinoma (SCC) lines and humoral hypercalcemia of malignancy (HHM) in vivo. Furthermore, a single nucleotide polymorphism in CaR identified from a hypercalcemia-inducing lung SCC reduced the receptor's activation threshold leading to increased PTHrP expression and secretion. Increasing the expression of either wild-type CaR or a CaR variant with a single nucleotide polymorphism in the cytoplasmic domain was both necessary and sufficient for lung SCC to induce HHM. Because lung cancer patients who frequently develop HHM and PTHrP expression in lung cancer has been only partially explained, the significance of our findings indicates that CaR variants may provide a positive feedback between PTHrP and calcium and result in the syndrome of HHM.
Collapse
|
35
|
Kirby BJ, Ardeshirpour L, Woodrow JP, Wysolmerski JJ, Sims NA, Karaplis AC, Kovacs CS. Skeletal recovery after weaning does not require PTHrP. J Bone Miner Res 2011; 26:1242-51. [PMID: 21308774 PMCID: PMC3179289 DOI: 10.1002/jbmr.339] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Revised: 11/28/2010] [Accepted: 01/13/2011] [Indexed: 12/12/2022]
Abstract
Mice lose 20% to 25% of trabecular bone mineral content (BMC) during lactation and restore it after weaning through unknown mechanisms. We found that tibial Pthrp mRNA expression was upregulated fivefold by 7 days after weaning versus end of lactation in wild-type (WT) mice. To determine whether parathyroid hormone-related protein (PTHrP) stimulates bone formation after weaning, we studied a conditional knockout in which PTHrP is deleted from preosteoblasts and osteoblasts by collagen I promoter-driven Cre (Cre(ColI) ). These mice are osteopenic as adults but have normal serum calcium, calcitriol, and parathyroid hormone (PTH). Pairs of Pthrp(flox/flox) ;Cre(ColI) (null) and WT;Cre(ColI) (WT) females were mated and studied through pregnancy, lactation, and 3 weeks of postweaning recovery. By end of lactation, both genotypes lost lumbar spine BMC: WT declined by 20.6% ± 3.3%, and null decreased by 22.5% ± 3.5% (p < .0001 versus baseline; p = NS between genotypes). During postweaning recovery, both restored BMC to baseline: WT to -3.6% ± 3.7% and null to 0.3% ± 3.7% (p = NS versus baseline or between genotypes). Similar loss and full recovery of BMC were seen at the whole body and hind limb. Histomorphometry confirmed that nulls had lower bone mass at baseline and that this was equal to the value achieved after weaning. Osteocalcin, propeptide of type 1 collagen (P1NP), and deoxypyridinoline increased equally during recovery in WT and null mice; PTH decreased and calcitriol increased equally; serum calcium was unchanged. Urine calcium increased during recovery but remained no different between genotypes. Although osteoblast-derived PTHrP is required to maintain adult bone mass and Pthrp mRNA upregulates in bone after weaning, it is not required for recovery of bone mass after lactation. The factors that stimulate postweaning bone formation remain unknown.
Collapse
Affiliation(s)
- Beth J Kirby
- Faculty of Medicine, Memorial University of NewfoundlandSt. John's, Newfoundland, Canada
| | - Laleh Ardeshirpour
- Faculty of Medicine, Yale University School of MedicineNew Haven, CT, USA
| | - Janine P Woodrow
- Faculty of Medicine, Memorial University of NewfoundlandSt. John's, Newfoundland, Canada
| | - John J Wysolmerski
- Faculty of Medicine, Yale University School of MedicineNew Haven, CT, USA
| | - Natalie A Sims
- St Vincent's Institute for Medical Research and Department of Medicine, St. Vincent's Hospital Melbourne, The University of MelbourneFitzroy, Victoria, Australia
| | - Andrew C Karaplis
- McGill University and Jewish General HospitalMontréal, Quebec, Canada
| | - Christopher S Kovacs
- Faculty of Medicine, Memorial University of NewfoundlandSt. John's, Newfoundland, Canada
| |
Collapse
|
36
|
Shu L, Ji J, Zhu Q, Cao G, Karaplis A, Pollak MR, Brown E, Goltzman D, Miao D. The calcium-sensing receptor mediates bone turnover induced by dietary calcium and parathyroid hormone in neonates. J Bone Miner Res 2011; 26:1057-71. [PMID: 21542007 PMCID: PMC3179300 DOI: 10.1002/jbmr.300] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
We have investigated, in neonates, whether the calcium-sensing receptor (CaR) mediates the effects of dietary calcium on bone turnover and/or modulates parathyroid hormone (PTH)-induced bone turnover. Wild-type (WT) pups and pups with targeted deletion of the Pth (Pth(-/-)) gene or of both Pth and CaR (Pth(-/-)CaR(-/-)) genes were nursed by dams on a normal or high-calcium diet. Pups nursed by dams on a normal diet received daily injections of vehicle or of PTH(1-34) (80 µg/kg) for 2 weeks starting from 1 week of age. In pups receiving vehicle and fed by dams on a normal diet, trabecular bone volume, osteoblast number, type 1 collagen-positive area, and mineral apposition rate, as well as the expression of bone-formation-related genes, all were reduced significantly in Pth(-/-) pups compared with WT pups and were decreased even more dramatically in Pth(-/-)CaR(-/-) pups. These parameters were increased in WT and Pth(-/-) pups but not in Pth(-/-)CaR(-/-) pups fed by dams on a high-calcium diet compared with pups fed by dams on a normal diet. These parameters also were increased in WT, Pth(-/-), and Pth(-/-)CaR(-/-) pups following exogenous PTH treatment; however, the percentage increase was less in Pth(-/-)CaR(-/-) pups than in WT and Pth(-/-) pups. In vehicle-treated pups fed by dams on either the normal or high-calcium diet and in PTH-treated pups fed by dams on a normal diet, the number and surfaces of osteoclasts and the ratio of RANKL/OPG were reduced significantly in Pth(-/-) pups and less significantly in Pth(-/-)CaR(-/-) pups compared with WT pups. These parameters were further reduced significantly in WT and Pth(-/-) pups from dams fed a high-calcium diet but did not decrease significantly in similarly treated Pth(-/-)CaR(-/-) pups, and they increased significantly in PTH-treated pups compared with vehicle-treated, genotype-matched pups fed by dams on the normal diet. These results indicate that in neonates, the CaR mediates alterations in bone turnover in response to changes in dietary calcium and modulates PTH-stimulated bone turnover.
Collapse
Affiliation(s)
- Lei Shu
- Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Parkash J, Asotra K. L-histidine sensing by calcium sensing receptor inhibits voltage-dependent calcium channel activity and insulin secretion in β-cells. Life Sci 2011; 88:440-6. [PMID: 21219913 PMCID: PMC3044179 DOI: 10.1016/j.lfs.2010.12.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2010] [Revised: 11/22/2010] [Accepted: 12/14/2010] [Indexed: 12/29/2022]
Abstract
AIMS Our goal was to test the hypothesis that the histidine-induced activation of calcium sensing receptor (CaR) can regulate calcium channel activity of L-type voltage dependent calcium channel (VDCC) due to increased spatial interaction between CaR and VDCC in β-cells and thus modulate glucose-induced insulin secretion. MAIN METHODS Rat insulinoma (RINr1046-38) insulin-producing β-cells were cultured in RPMI-1640 medium on 25 mm diameter glass coverslips in six-well culture plates in a 5% CO(2) incubator at 37°C. The intracellular calcium concentration, [Ca(2+)](i), was determined by ratio fluorescence microscopy using Fura-2AM. The spatial interactions between CaR and L-type VDCC in β-cells were measured by immunofluorescence confocal microscopy using a Nikon C1 laser scanning confocal microscope. The insulin release was determined by enzyme-linked immunosorbent assay (ELISA). KEY FINDINGS The addition of increasing concentrations of L-histidine along with 10 mM glucose resulted in 57% decrease in [Ca(2+)](i). The confocal fluorescence imaging data showed 5.59 to 8.62-fold increase in colocalization correlation coefficient between CaR and VDCC in β-cells exposed to L-histidine thereby indicating increased membrane delimited spatial interactions between these two membrane proteins. The insulin ELISA data showed 54% decrease in the 1st phase of glucose-induced insulin secretion in β-cells exposed to increasing concentrations of L-histidine. SIGNIFICANCE L-histidine-induced increased spatial interaction of CaR with VDCC can inhibit calcium channel activity of VDCC and consequently regulate glucose-induced insulin secretion by β-cells. The L-type VDCC could therefore be a potential therapeutic target in diabetes.
Collapse
Affiliation(s)
- Jai Parkash
- Robert Stempel College of Public Health and Social Work, Department of Environmental and Occupational Health, Florida International University, 11200 SW 8 Street, HLS-594, Miami, FL 33199, USA
| | - Kamlesh Asotra
- Cardiovascular Disease and General Biomedical Sciences, Tobacco-Related Disease Research Program, University of California Office of President, 300 Lakeside Drive, Oakland, CA 94612, USA, Tel: (510) 287-3366, FAX: (510) 835-4740,
| |
Collapse
|
38
|
Wysolmerski JJ. Interactions between breast, bone, and brain regulate mineral and skeletal metabolism during lactation. Ann N Y Acad Sci 2010; 1192:161-9. [PMID: 20392232 DOI: 10.1111/j.1749-6632.2009.05249.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mammalian reproduction requires that nursing mothers transfer large amounts of calcium to their offspring through milk. As a result, lactation is associated with dramatic alterations in bone and mineral metabolism, including reversible bone loss. One theme that has emerged from recent studies examining these adaptations is that the lactating breast actively participates in regulating bone and mineral metabolism. This review will detail our current knowledge of interactions between the breast, skeleton, and hypothalamus during lactation and will consider implications that this reproductive physiology has for the pathophysiology of osteoporosis and breast cancer.
Collapse
|
39
|
Mamillapalli R, Wysolmerski J. The calcium-sensing receptor couples to Galpha(s) and regulates PTHrP and ACTH secretion in pituitary cells. J Endocrinol 2010; 204:287-97. [PMID: 20032198 PMCID: PMC3777408 DOI: 10.1677/joe-09-0183] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The calcium-sensing receptor (CaR or CASR as listed in the MGI Database) is a G protein-coupled receptor that binds and signals in response to extracellular calcium and other polycations. It is highly expressed on parathyroid and kidney cells, where it participates in the regulation of systemic calcium homeostasis. It is also expressed on many other cell types and is involved in a wide array of biological functions such as cell growth and differentiation, ion transport, and hormone secretion. It has been described to couple to several different G proteins including Galpha(i/0), Galpha(q/11), and Galpha(12/13). Recently, it has also been shown to stimulate cAMP production by coupling to Galpha(s) in immortalized or malignant breast cells. The CaR is expressed on cells in the anterior pituitary and had previously been described to stimulate cAMP production in these cells. In this report, we examined signaling from the CaR in murine pituitary corticotroph-derived, AtT-20 cells. We found that CaR activation led to the stimulation of cAMP production, and PTH-related protein (PTHrP or PTHLH as listed in the MGI Database) and ACTH secretion from these cells. Furthermore, manipulation of cAMP levels was able to modulate PTHrP and ACTH secretion independent of changes in extracellular calcium. Finally, we demonstrated that the CaR couples to Galpha(s) in AtT-20 cells. Therefore, in pituitary corticotroph-like cells, as in breast cancer cells, the CaR utilizes Galpha(s) and activates cAMP production to stimulate hormone secretion.
Collapse
Affiliation(s)
- Ramanaiah Mamillapalli
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, TAC S131, New Haven, Connecticut 06520-8020, USA
| | | |
Collapse
|
40
|
Riccardi D, Brown EM. Physiology and pathophysiology of the calcium-sensing receptor in the kidney. Am J Physiol Renal Physiol 2010; 298:F485-99. [PMID: 19923405 PMCID: PMC2838589 DOI: 10.1152/ajprenal.00608.2009] [Citation(s) in RCA: 235] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 11/13/2009] [Indexed: 12/21/2022] Open
Abstract
The extracellular calcium-sensing receptor (CaSR) plays a major role in the maintenance of a physiological serum ionized calcium (Ca2+) concentration by regulating the circulating levels of parathyroid hormone. It was molecularly identified in 1993 by Brown et al. in the laboratory of Dr. Steven Hebert with an expression cloning strategy. Subsequent studies have demonstrated that the CaSR is highly expressed in the kidney, where it is capable of integrating signals deriving from the tubular fluid and/or the interstitial plasma. Additional studies elucidating inherited and acquired mutations in the CaSR gene, the existence of activating and inactivating autoantibodies, and genetic polymorphisms of the CaSR have greatly enhanced our understanding of the role of the CaSR in mineral ion metabolism. Allosteric modulators of the CaSR are the first drugs in their class to become available for clinical use and have been shown to treat successfully hyperparathyroidism secondary to advanced renal failure. In addition, preclinical and clinical studies suggest the possibility of using such compounds in various forms of hypercalcemic hyperparathyroidism, such as primary and lithium-induced hyperparathyroidism and that occurring after renal transplantation. This review addresses the role of the CaSR in kidney physiology and pathophysiology as well as current and in-the-pipeline treatments utilizing CaSR-based therapeutics.
Collapse
Affiliation(s)
- Daniela Riccardi
- School of Biosciences, Cardiff University, Cardiff, United Kingdom.
| | | |
Collapse
|
41
|
Kantham L, Quinn SJ, Egbuna OI, Baxi K, Butters R, Pang JL, Pollak MR, Goltzman D, Brown EM. The calcium-sensing receptor (CaSR) defends against hypercalcemia independently of its regulation of parathyroid hormone secretion. Am J Physiol Endocrinol Metab 2009; 297:E915-23. [PMID: 19797241 PMCID: PMC2763782 DOI: 10.1152/ajpendo.00315.2009] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The calcium-sensing receptor (CaSR) controls parathyroid hormone (PTH) secretion, which, in turn, via direct and indirect actions on kidney, bone, and intestine, maintains a normal extracellular ionized calcium concentration (Ca(2+)(o)). There is less understanding of the CaSR's homeostatic importance outside of the parathyroid gland. We have employed single and double knockout mouse models, namely mice lacking PTH alone (CaSR(+/+) PTH(-/-), referred to as C(+)P(-)), lacking both CaSR and PTH (CaSR(-/-) PTH(-/-), C(-)P(-)) or wild-type (CaSR(+/+) PTH(+/+), C(+)P(+)) mice to study CaSR-specific functions without confounding CaSR-mediated changes in PTH. The mice received three hypercalcemic challenges: an oral Ca(2+) load, injection or constant infusion of PTH via osmotic pump, or a phosphate-deficient diet. C(-)P(-) mice show increased susceptibility to developing hypercalcemia with all three challenges compared with the other two genotypes, whereas C(+)P(-) mice defend against hypercalcemia similarly to C(+)P(+) mice. Reduced renal Ca(2+) clearance contributes to the intolerance of the C(-)P(-) mice to Ca(2+) loads, as they excrete less Ca(2+) at any given Ca(2+)(o) than the other two genotypes, confirming the CaSR's direct role in regulating renal Ca(2+) handling. In addition, C(+)P(+) and C(+)P(-), but not C(-)P(-), mice showed increases in serum calcitonin (CT) levels during hypercalcemia. The level of 1,25(OH)(2)D(3) in C(-)P(-) mice, in contrast, was similar to those in C(+)P(-) and C(+)P(+) mice during an oral Ca(2+) load, indicating that increased 1,25(OH)(2)D(3) production cannot account for the oral Ca(2+)-induced hypercalcemia in the C(-)P(-) mice. Thus, CaSR-stimulated PTH release serves as a "floor" to defend against hypocalcemia. In contrast, high-Ca(2+)(o)-induced inhibition of PTH is not required for a robust defense against hypercalcemia, at least in mice, whereas high-Ca(2+)(o)-stimulated, CaSR-mediated CT secretion and renal Ca(2+) excretion, and perhaps other factors, serve as a "ceiling" to limit hypercalcemia resulting from various types of hypercalcemic challenges.
Collapse
Affiliation(s)
- Lakshmi Kantham
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Adult hematopoietic stem cells (HSCs) reside in the bone marrow in stable microenvironments known as the stem cell niche. One key component of the stem cell niche is cells of the osteoblastic lineage. Factors that are known to affect osteoblast activity, such as parathyroid hormone (PTH), have also been shown to affect the HSCs. Treatment of mice with PTH has led to beneficial effects on the HSC pool, which have led to clinical trials of PTH treatment to enhance HSC-based therapies.
Collapse
Affiliation(s)
- Narges Rashidi
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | |
Collapse
|
43
|
Galitzer H, Lavi-Moshayoff V, Nechama M, Meir T, Silver J, Naveh-Many T. The calcium-sensing receptor regulates parathyroid hormone gene expression in transfected HEK293 cells. BMC Biol 2009; 7:17. [PMID: 19397786 PMCID: PMC2681451 DOI: 10.1186/1741-7007-7-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Accepted: 04/27/2009] [Indexed: 11/10/2022] Open
Abstract
Background The parathyroid calcium receptor determines parathyroid hormone secretion and the response of parathyroid hormone gene expression to serum Ca2+ in the parathyroid gland. Serum Ca2+ regulates parathyroid hormone gene expression in vivo post-transcriptionally affecting parathyroid hormone mRNA stability through the interaction of trans-acting proteins to a defined cis element in the parathyroid hormone mRNA 3'-untranslated region. These parathyroid hormone mRNA binding proteins include AUF1 which stabilizes and KSRP which destabilizes the parathyroid hormone mRNA. There is no parathyroid cell line; therefore, we developed a parathyroid engineered cell using expression vectors for the full-length human parathyroid hormone gene and the human calcium receptor. Results Co-transfection of the human calcium receptor and the human parathyroid hormone plasmid into HEK293 cells decreased parathyroid hormone mRNA levels and secreted parathyroid hormone compared with cells that do not express the calcium receptor. The decreased parathyroid hormone mRNA correlated with decreased parathyroid hormone mRNA stability in vitro, which was dependent upon the 3'-UTR cis element. Moreover, parathyroid hormone gene expression was regulated by Ca2+ and the calcimimetic R568, in cells co-transfected with the calcium receptor but not in cells without the calcium receptor. RNA immunoprecipitation analysis in calcium receptor-transfected cells showed increased KSRP-parathyroid hormone mRNA binding and decreased binding to AUF1. The calcium receptor led to post-translational modifications in AUF1 as occurs in the parathyroid in vivo after activation of the calcium receptor. Conclusion The expression of the calcium receptor is sufficient to confer the regulation of parathyroid hormone gene expression to these heterologous cells. The calcium receptor decreases parathyroid hormone gene expression in these engineered cells through the parathyroid hormone mRNA 3'-UTR cis element and the balanced interactions of the trans-acting factors KSRP and AUF1 with parathyroid hormone mRNA, as in vivo in the parathyroid. This is the first demonstration that the calcium receptor can regulate parathyroid hormone gene expression in heterologous cells.
Collapse
Affiliation(s)
- Hillel Galitzer
- Minerva Center for Calcium and Bone Metabolism, Nephrology Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| | | | | | | | | | | |
Collapse
|
44
|
Mamillapalli R, VanHouten J, Zawalich W, Wysolmerski J. Switching of G-protein usage by the calcium-sensing receptor reverses its effect on parathyroid hormone-related protein secretion in normal versus malignant breast cells. J Biol Chem 2008; 283:24435-47. [PMID: 18621740 PMCID: PMC2528989 DOI: 10.1074/jbc.m801738200] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 05/26/2008] [Indexed: 12/20/2022] Open
Abstract
The calcium-sensing receptor (CaR) is a G-protein-coupled receptor that signals in response to extracellular calcium and regulates parathyroid hormone secretion. The CaR is also expressed on normal mammary epithelial cells (MMECs), where it has been shown to inhibit secretion of parathyroid hormone-related protein (PTHrP) and participate in the regulation of calcium and bone metabolism during lactation. In contrast to normal breast cells, the CaR has been reported to stimulate PTHrP production by breast cancer cells. In this study, we confirmed that the CaR inhibits PTHrP production by MMECs but stimulates PTHrP production by Comma-D cells (immortalized murine mammary cells) and MCF-7 human breast cancer cells. We found that changes in intracellular cAMP, but not phospholipase C or MAPK signaling, correlated with the opposing effects of the CaR on PTHrP production. Pharmacologic stimulation of cAMP accumulation increased PTHrP production by normal and transformed breast cells. Inhibition of protein kinase A activity mimicked the effects of CaR activation on inhibiting PTHrP secretion by MMECs and blocked the effects of the CaR on stimulating PTHrP production in Comma-D and MCF-7 cells. We found that the CaR coupled to Galphai in MMECs but coupled to Galphas in Comma-D and MCF-7 cells. Thus, the opposing effects of the CaR on PTHrP production are because of alternate G-protein coupling of the receptor in normal versus transformed breast cells. Because PTHrP contributes to hypercalcemia and bone metastases, switching of G-protein usage by the CaR may contribute to the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Ramanaiah Mamillapalli
- Section of Endocrinology and Metabolism,
Department of Internal Medicine, School of Medicine and
School of Nursing, Yale University, New Haven,
Connecticut 06520
| | - Joshua VanHouten
- Section of Endocrinology and Metabolism,
Department of Internal Medicine, School of Medicine and
School of Nursing, Yale University, New Haven,
Connecticut 06520
| | - Walter Zawalich
- Section of Endocrinology and Metabolism,
Department of Internal Medicine, School of Medicine and
School of Nursing, Yale University, New Haven,
Connecticut 06520
| | - John Wysolmerski
- Section of Endocrinology and Metabolism,
Department of Internal Medicine, School of Medicine and
School of Nursing, Yale University, New Haven,
Connecticut 06520
| |
Collapse
|
45
|
Bruder ED, Van Hoof J, Young JB, Raff H. Epidermal growth factor and parathyroid hormone-related peptide mRNA in the mammary gland and their concentrations in milk: effects of postpartum hypoxia in lactating rats. Horm Metab Res 2008; 40:446-53. [PMID: 18401831 PMCID: PMC2504024 DOI: 10.1055/s-2008-1058101] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The physiological adaptations of the neonatal rat to hypoxia from birth include changes in gastrointestinal function and intermediary metabolism. We hypothesized that the hypoxic lactating dam would exhibit alterations in mammary gland function leading to changes in the concentration of milk peptides that are important in neonatal gastrointestinal development. The present study assessed the effects of chronic hypoxia on peptides produced by the mammary glands and present in milk. Chronic hypoxia decreased the concentration of epidermal growth factor (EGF) in expressed milk and pup stomach contents and decreased maternal mammary gland EGF mRNA. The concentration of parathyroid hormone-related protein (PTHrp) was unchanged in milk and decreased in pup stomach contents; however, mammary PTHLH mRNA was increased by hypoxia. There was a significant increase in adiponectin concentrations in milk from hypoxic dams. Chronic hypoxia decreased maternal body weight, and pair feeding normoxic dams an amount of food equivalent to hypoxic dam food intake decreased body weight to an equivalent degree. Decreased food intake did not affect the expression of EGF, PTHLH, or LEP mRNA in mammary tissue. The results indicated that chronic hypoxia modulated mammary function independently of hypoxia-induced decreases in maternal food intake. Decreased EGF and increased adiponectin concentrations in milk from hypoxic dams likely affect the development of neonatal intestinal function.
Collapse
Affiliation(s)
- E. D. Bruder
- Endocrine Research Laboratory, Aurora St. Luke’s Medical Center, Milwaukee, Wisconsin, USA
| | - J. Van Hoof
- Endocrine Research Laboratory, Aurora St. Luke’s Medical Center, Milwaukee, Wisconsin, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - J. B. Young
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - H. Raff
- Endocrine Research Laboratory, Aurora St. Luke’s Medical Center, Milwaukee, Wisconsin, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
46
|
Breitwieser GE. Extracellular calcium as an integrator of tissue function. Int J Biochem Cell Biol 2008; 40:1467-80. [PMID: 18328773 PMCID: PMC2441573 DOI: 10.1016/j.biocel.2008.01.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 01/16/2008] [Accepted: 01/18/2008] [Indexed: 12/23/2022]
Abstract
The past several decades of research into calcium signaling have focused on intracellular calcium (Ca(i)(2+)), revealing both exquisite spatial and dynamic control of this potent second messenger. Our understanding of Ca(i)(2+) signaling has benefited from the evolution of cell culture methods, development of high affinity fluorescent calcium indicators (both membrane-permeant small molecules and genetically encoded proteins), and high-resolution fluorescence microscopy. As our understanding of single cell calcium dynamics has increased, translational efforts have attempted to push calcium signaling studies back into tissues, organs and whole animals. Emerging results from these more complicated, diffusion-limited systems have begun to define a role for extracellular calcium (Ca(o)(2+)) as an agonist, spurred by the cloning and characterization of a G protein-coupled receptor activated by Ca(o)(2+) (the calcium sensing receptor, CaR). Here, we review the current state-of-the art for measurement of Ca(o)(2+) fluctuations, and the evidence that fluctuations in Ca(o)(2+) can act as primary signals regulating cell function. Current results suggest that Ca(o)(2+) in bone and epidermis may act as a chemotactic homing signal, targeting cells to the appropriate tissue locations prior to initiation of the differentiation program. Ca(i)(2+) signaling-mediated Ca(o)(2+) fluctuations in interstitial spaces may integrate cell signaling responses in multicellular networks through activation of CaR. Appreciation of the importance of Ca(o)(2+) fluctuations in coordinating cell function will likely spur identification of additional, niche-specific Ca(2+) sensors, and provide unique insights into the regulation of multicellular signaling networks.
Collapse
Affiliation(s)
- Gerda E Breitwieser
- Weis Center for Research, Geisinger Clinic, 100 N. Academy Avenue, Danville, PA 17822, United States.
| |
Collapse
|
47
|
VanHouten JN, Neville MC, Wysolmerski JJ. The calcium-sensing receptor regulates plasma membrane calcium adenosine triphosphatase isoform 2 activity in mammary epithelial cells: a mechanism for calcium-regulated calcium transport into milk. Endocrinology 2007; 148:5943-54. [PMID: 17823248 PMCID: PMC7108505 DOI: 10.1210/en.2007-0850] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The calcium-sensing receptor (CaR) regulates transepithelial calcium transport into milk by mammary epithelial cells. Using a genome-wide screening strategy, we identified the plasma membrane calcium ATPase isoform 2 (PMCA2) as a potential downstream target of the CaR. We show that PMCA2 expression in the mouse mammary gland increases during lactation and that PMCA2 is localized solely to the apical plasma membrane of mammary epithelial cells. In milk from deafwaddler mice, which have mutations in the gene encoding PMCA2, calcium concentrations were reduced, confirming its importance in calcium transport into milk. Furthermore, in cultured primary and EpH4 mouse mammary epithelial cells, CaR stimulation up-regulated calcium-dependent ATPase activity in plasma membrane preparations. By small interfering RNA-mediated gene knockdown of PMCA2, we show that PMCA2 accounts for the preponderance of calcium-ATPase activity. We also show that reduction of CaR expression with small interfering RNA eliminates the ability of extracellular calcium to elicit an increase in calcium-dependent ATPase activity in EpH4 cell membranes. These results demonstrate that activation of the CaR increases PMCA2 activity in mouse mammary epithelial cells, providing a mechanism for the regulation of transepithelial calcium transport by calcium in the lactating mouse mammary gland.
Collapse
Affiliation(s)
- Joshua N VanHouten
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8020, USA
| | | | | |
Collapse
|
48
|
Page-Wilson G, Smith PC, Welt CK. Short-term prolactin administration causes expressible galactorrhea but does not affect bone turnover: pilot data for a new lactation agent. Int Breastfeed J 2007; 2:10. [PMID: 17650319 PMCID: PMC1950489 DOI: 10.1186/1746-4358-2-10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Accepted: 07/24/2007] [Indexed: 11/18/2022] Open
Abstract
Background Medications used to augment lactation increase prolactin secretion but can have intolerable side effects. We examined the biological activity of recombinant human prolactin (r-hPRL) as preliminary data for its use to augment lactation. Methods Healthy, non-postpartum women (n = 21) with regular menstrual cycles underwent a seven day randomized, double-blind, placebo-controlled trial of r-hPRL. Expressible galactorrhea, markers of bone turnover, calcium homeostasis and gonadal function were measured and side effects recorded. Results Prolactin levels increased during r-hPRL administration (20.0 ± 2.8 to 231.7 ± 48.9 μg/L at 6 hours; p < 0.05). Five of nine participants who received r-hPRL developed expressible galactorrhea (p < 0.001). Urinary deoxypyridinoline decreased and bone specific alkaline phosphatase increased in r-hPRL and placebo groups. Menstrual cycle lengths were not altered and side effects were similar between r-hPRL and placebo groups. Conclusion In summary, r-hPRL can cause expressible galactorrhea. Seven days of r-hPRL administration does not adversely affect bone turnover or menstrual cyclicity. Thus, r-hPRL may be a viable option for short-term lactation augmentation. Trial registration Clinical Trials.gov NCT00438490
Collapse
Affiliation(s)
- Gabrielle Page-Wilson
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, USA
- Harvard Medical School, Boston, USA
| | - Patricia C Smith
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, USA
| | - Corrine K Welt
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, USA
| |
Collapse
|
49
|
Anderson SM, Rudolph MC, McManaman JL, Neville MC. Key stages in mammary gland development. Secretory activation in the mammary gland: it's not just about milk protein synthesis! Breast Cancer Res 2007; 9:204. [PMID: 17338830 PMCID: PMC1851396 DOI: 10.1186/bcr1653] [Citation(s) in RCA: 287] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The transition from pregnancy to lactation is a critical event in the survival of the newborn since all the nutrient requirements of the infant are provided by milk. While milk contains numerous components, including proteins, that aid in maintaining the health of the infant, lactose and milk fat represent the critical energy providing elements of milk. Much of the research to date on mammary epithelial differentiation has focused upon expression of milk protein genes, providing a somewhat distorted view of alveolar differentiation and secretory activation. While expression of milk protein genes increases during pregnancy and at secretory activation, the genes whose expression is more tightly regulated at this transition are those that regulate lipid biosynthesis. The sterol regulatory element binding protein (SREBP) family of transcription factors is recognized as regulating fatty acid and cholesterol biosynthesis. We propose that SREBP1 is a critical regulator of secretory activation with regard to lipid biosynthesis, in a manner that responds to diet, and that the serine/threonine protein kinase Akt influences this process, resulting in a highly efficient lipid synthetic organ that is able to support the nutritional needs of the newborn.
Collapse
Affiliation(s)
- Steven M Anderson
- Department of Pathology, University of Colorado Health Sciences Center, East 17th Avenue, Aurora, CO 80045, USA
- Program in Molecular Biology, University of Colorado Health Sciences Center, East 17th Avenue, Aurora, CO 80045, USA
| | - Michael C Rudolph
- Department of Physiology and Biophysics, University of Colorado Health Sciences Center, East 17th Avenue, Aurora, CO 80045, USA
| | - James L McManaman
- Program in Molecular Biology, University of Colorado Health Sciences Center, East 17th Avenue, Aurora, CO 80045, USA
- Department of Physiology and Biophysics, University of Colorado Health Sciences Center, East 17th Avenue, Aurora, CO 80045, USA
- Department of Obstetrics and Gynecology, University of Colorado Health Sciences Center, East 17th Avenue, Aurora, CO 80045, USA
| | - Margaret C Neville
- Department of Physiology and Biophysics, University of Colorado Health Sciences Center, East 17th Avenue, Aurora, CO 80045, USA
- Department of Obstetrics and Gynecology, University of Colorado Health Sciences Center, East 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
50
|
Pi M, Faber P, Ekema G, Jackson PD, Ting A, Wang N, Fontilla-Poole M, Mays RW, Brunden KR, Harrington JJ, Quarles LD. Identification of a novel extracellular cation-sensing G-protein-coupled receptor. J Biol Chem 2005; 280:40201-9. [PMID: 16199532 PMCID: PMC1435382 DOI: 10.1074/jbc.m505186200] [Citation(s) in RCA: 246] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The C family G-protein-coupled receptors contain members that sense amino acid and extracellular cations, of which calcium-sensing receptor (CASR) is the prototypic extracellular calcium-sensing receptor. Some cells, such as osteoblasts in bone, retain responsiveness to extracellular calcium in CASR-deficient mice, consistent with the existence of another calcium-sensing receptor. We examined the calcium-sensing properties of GPRC6A, a newly identified member of this family. Alignment of GPRC6A with CASR revealed conservation of both calcium and calcimimetic binding sites. In addition, calcium, magnesium, strontium, aluminum, gadolinium, and the calcimimetic NPS 568 resulted in a dose-dependent stimulation of GPRC6A overexpressed in human embryonic kidney cells 293 cells. Also, osteocalcin, a calcium-binding protein highly expressed in bone, dose-dependently stimulated GPRC6A activity in the presence of calcium but inhibited the calcium-dependent activation of CASR. Coexpression of beta-arrestins 1 and 2, regulators of G-protein signaling RGS2 or RGS4, the RhoA inhibitor C3 toxin, the dominant negative Galpha(q)-(305-359) minigene, and pretreatment with pertussis toxin inhibited activation of GPRC6A by extracellular cations. Reverse transcription-PCR analyses showed that mouse GPRC6A is widely expressed in mouse tissues, including bone, calvaria, and the osteoblastic cell line MC3T3-E1. These data suggest that in addition to sensing amino acids, GPRC6A is a cation-, calcimimetic-, and osteocalcin-sensing receptor and a candidate for mediating extracellular calcium-sensing responses in osteoblasts and possibly other tissues.
Collapse
Affiliation(s)
- Min Pi
- From the Kidney Institute, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160 and
| | | | | | | | | | | | | | | | | | | | - L. Darryl Quarles
- From the Kidney Institute, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160 and
- To whom correspondence should be addressed: The Kidney Institute and Division of Nephrology, 3901 Rainbow Boulevard, 6018 Wahl Hall East, Kansas City, KS 66160., Tel.: 913-588-9255; Fax: 913-5889251; E-mail:
| |
Collapse
|