Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
Anat Rec (Hoboken) ; 306(8): 2015-2029, 2023 08.
Article in English | MEDLINE | ID: mdl-35778853

ABSTRACT

This study's objective was to investigate obtaining high-resolution micro-computed tomography (CT) imaging of the injected arterial circulation of the brains of the dogfish (Squalus acanthias), American bullfrog (Rana catesbeiana), and green iguana (Iguana iguana). No micro-CT images of the arterial morphology of the brains of these vertebrates were previously published. Micro-CT imaging was performed on brains that had the cerebral arterial and ventricular systems injected with a radiopaque barium-gelatin compound in the early 1970s. These specimens were dissected and placed in a preservative fluid for 35 years, until imaged with micro-CT. The obtained micro-CT images were processed with a software program that provided 3D rotational motion rendering, and sequential display of 2D renderings of the micro-CT data. The anatomic information provided by the high-resolution micro-CT is not reproducible by any other radiopaque contrast currently available, without tissue removal corrosion, and enhanced the dissection information. The digital videos of the micro-CT 3D rotational motion rendering and sequential display of 2D renderings of the dogfish, bullfrog, and green iguana, demonstrate the extent of the arterial network within the brain, the arterial segments obscured by overlying structures such as nerves, and identified in the bullfrog the venous cerebral circulation resulting from the centrifugal leptomeningeal arterial capillaries. The rotational 3D images separated superimposed arterial structures, and the sequential display of the 2D renderings clarifies the relationship of cut or overlapped arterial branches. Comparing the brain and arterial morphology of the dogfish, bullfrog, and green iguana demonstrates some of the evolutionary modifications in these vertebrates.


Subject(s)
Iguanas , Squalus acanthias , Animals , Rana catesbeiana , Dogfish , X-Ray Microtomography
2.
PLoS One ; 12(4): e0174849, 2017.
Article in English | MEDLINE | ID: mdl-28369073

ABSTRACT

ErbB2/HER2/Neu is a receptor tyrosine kinase that is overexpressed in 25-30% of human breast cancers, usually associated with amplification of the ERBB2 gene. HER2 has no recognized ligands and heterodimers between HER2 and EGFR (ErbB1/HER1) or HER2 and ErbB3/HER3 are important in breast cancer. Unlike other ErbB family members, HER2 is resistant to internalization and degradation, and remains at the cell surface to signal for prolonged periods after it is activated. Although the mechanisms underlying retention of HER2 at the cell surface are not fully understood, prior studies have shown that, in order to avoid internalization, HER2 must interact with the chaperone, HSP90, and the calcium pump, PMCA2, within specific plasma membrane domains that protrude from the cell surface. In this report, we demonstrate that HER2 signaling, itself, is important for the formation and maintenance of membrane protrusions, at least in part, by maintaining PMCA2 expression and preventing increased intracellular calcium concentrations. Partial genetic knockdown of HER2 expression or pharmacologic inhibition of HER2 signaling causes the depletion of membrane protrusions and disruption of the interactions between HER2 and HSP90. This is associated with the ubiquitination of HER2, its internalization with EGFR or HER3, and its degradation. These results suggest a model by which some threshold of HER2 signaling is required for the formation and/or maintenance of multi-protein signaling complexes that reinforce and prolong HER2/EGFR or HER2/HER3 signaling by inhibiting HER2 ubiquitination and internalization.


Subject(s)
Cell Membrane/metabolism , Receptor, ErbB-2/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , ErbB Receptors/metabolism , Gene Knockdown Techniques , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Lapatinib , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/metabolism , Quinazolines/pharmacology , RNA, Small Interfering , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-3/metabolism , Ubiquitination
3.
J Biol Chem ; 292(16): 6555-6568, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28235801

ABSTRACT

We examined whether the scaffolding protein sodium-hydrogen exchanger regulatory factor 1 (NHERF1) interacts with the calcium pump PMCA2 and the tyrosine kinase receptor ErbB2/HER2 in normal mammary epithelial cells and breast cancer cells. NHERF1 interacts with the PDZ-binding motif in PMCA2 in both normal and malignant breast cells. NHERF1 expression is increased in HER2-positive breast cancers and correlates with HER2-positive status in human ductal carcinoma in situ (DCIS) lesions and invasive breast cancers as well as with increased mortality in patients. NHERF1 is part of a multiprotein complex that includes PMCA2, HSP90, and HER2 within specific actin-rich and lipid raft-rich membrane signaling domains. Knocking down NHERF1 reduces PMCA2 and HER2 expression, inhibits HER2 signaling, dissociates HER2 from HSP90, and causes the internalization, ubiquitination, and degradation of HER2. These results demonstrate that NHERF1 acts with PMCA2 to regulate HER2 signaling and membrane retention in breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Phosphoproteins/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Receptor, ErbB-2/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Motifs , Animals , Apoptosis , Breast Neoplasms/genetics , Calcium/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Microscopy, Fluorescence , RNA, Messenger/metabolism , Signal Transduction
4.
Cancer Res ; 76(18): 5348-60, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27450451

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.


Subject(s)
Breast Neoplasms/pathology , Parathyroid Hormone-Related Protein/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Breast Neoplasms/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , Mice , Mice, Knockout , Tissue Array Analysis
5.
Proc Natl Acad Sci U S A ; 113(3): E282-90, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26729871

ABSTRACT

In the lactating mammary gland, the plasma membrane calcium ATPase2 (PMCA2) transports milk calcium. Its expression is activated in breast cancers, where high tumor levels predict increased mortality. We find that PMCA2 expression correlates with HER2 levels in breast cancers and that PMCA2 interacts with HER2 in specific actin-rich membrane domains. Knocking down PMCA2 increases intracellular calcium, disrupts interactions between HER2 and HSP-90, inhibits HER2 signaling, and results in internalization and degradation of HER2. Manipulating PMCA2 levels regulates the growth of breast cancer cells, and knocking out PMCA2 inhibits the formation of tumors in mouse mammary tumor virus (MMTV)-Neu mice. These data reveal previously unappreciated molecular interactions regulating HER2 localization, membrane retention, and signaling, as well as the ability of HER2 to generate breast tumors, suggesting that interactions between PMCA2 and HER2 may represent therapeutic targets for breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Animals , Breast Neoplasms/pathology , Calcium/pharmacology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cell Survival , Endocytosis/drug effects , Female , Fluorescent Antibody Technique , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Intracellular Space/metabolism , Mammary Neoplasms, Animal , Mice , Protein Binding , Protein Transport , Survival Analysis
6.
Endocrinology ; 156(8): 2762-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25961842

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.


Subject(s)
Bone Resorption/prevention & control , Calcium/metabolism , Lactation/drug effects , Milk/drug effects , Milk/metabolism , Osteoprotegerin/therapeutic use , Animals , Animals, Suckling , Bone Density/drug effects , Calcium, Dietary/pharmacology , Female , Lactation/physiology , Mice , Mothers , Osteoprotegerin/pharmacology , Weaning
7.
PLoS One ; 9(5): e90418, 2014.
Article in English | MEDLINE | ID: mdl-24785493

ABSTRACT

Parathyroid hormone-related protein (PTHrP) can be secreted from cells and interact with its receptor, the Type 1 PTH/PTHrP Receptor (PTHR1) in an autocrine, paracrine or endocrine fashion. PTHrP can also remain inside cells and be transported into the nucleus, where its functions are unclear, although recent experiments suggest that it may broadly regulate cell survival and senescence. Disruption of either the PTHrP or PTHR1 gene results in many abnormalities including a failure of embryonic mammary gland development in mice and in humans. In order to examine the potential functions of nuclear PTHrP in the breast, we examined mammary gland development in PTHrP (1-84) knock-in mice, which express a mutant form of PTHrP that lacks the C-terminus and nuclear localization signals and which can be secreted but cannot enter the nucleus. Interestingly, we found that PTHrP (1-84) knock-in mice had defects in mammary mesenchyme differentiation and mammary duct outgrowth that were nearly identical to those previously described in PTHrP-/- and PTHR1-/- mice. However, the mammary buds in PTHrP (1-84) knock-in mice had severe reductions in mutant PTHrP mRNA levels, suggesting that the developmental defects were due to insufficient production of PTHrP by mammary epithelial cells and not loss of PTHrP nuclear function. Examination of the effects of nuclear PTHrP in the mammary gland in vivo will require the development of alternative animal models.


Subject(s)
Gene Deletion , Mammary Glands, Animal/embryology , Mammary Glands, Animal/metabolism , Nuclear Localization Signals/genetics , Parathyroid Hormone-Related Protein/biosynthesis , Parathyroid Hormone-Related Protein/genetics , Protein Interaction Domains and Motifs , Animals , Biomarkers/metabolism , Female , Gene Expression Regulation, Developmental , Mammary Glands, Animal/growth & development , Mice , Mice, Transgenic , Parathyroid Hormone-Related Protein/chemistry , Peptide Fragments/genetics , Sex Characteristics
8.
J Anat ; 225(1): 71-82, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24762197

ABSTRACT

The modeling of long bone surfaces during linear growth is a key developmental process, but its regulation is poorly understood. We report here that parathyroid hormone-related peptide (PTHrP) expressed in the fibrous layer of the periosteum (PO) drives the osteoclastic (OC) resorption that models the metaphyseal-diaphyseal junction (MDJ) in the proximal tibia and fibula during linear growth. PTHrP was conditionally deleted (cKO) in the PO via Scleraxis gene targeting (Scx-Cre). In the lateral tibia, cKO of PTHrP led to a failure of modeling, such that the normal concave MDJ was replaced by a mound-like deformity. This was accompanied by a failure to induce receptor activator of NF-kB ligand (RANKL) and a 75% reduction in OC number (P ≤ 0.001) on the cortical surface. The MDJ also displayed a curious threefold increase in endocortical osteoblast mineral apposition rate (P ≤ 0.001) and a thickened cortex, suggesting some form of coupling of endocortical bone formation to events on the PO surface. Because it fuses distally, the fibula is modeled only proximally and does so at an extraordinary rate, with an anteromedial cortex in CD-1 mice that was so moth-eaten that a clear PO surface could not be identified. The cKO fibula displayed a remarkable phenotype, with a misshapen club-like metaphysis and an enlargement in the 3D size of the entire bone, manifest as a 40-45% increase in the PO circumference at the MDJ (P ≤ 0.001) as well as the mid-diaphysis (P ≤ 0.001). These tibial and fibular phenotypes were reproduced in a Scx-Cre-driven RANKL cKO mouse. We conclude that PTHrP in the fibrous PO mediates the modeling of the MDJ of long bones during linear growth, and that in a highly susceptible system such as the fibula this surface modeling defines the size and shape of the entire bone.


Subject(s)
Bone Development/physiology , Fibula/growth & development , Parathyroid Hormone-Related Protein/physiology , Periosteum/physiology , Tibia/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors , Gene Deletion , Mice , Mice, Knockout , RANK Ligand/genetics
9.
J Anat ; 224(4): 490-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24266550

ABSTRACT

The developing cortical surfaces of long bones are sculpted and modeled by periosteal osteoclasts and osteoblasts. These surfaces also receive the insertions of tendons and ligaments, and these insertion sites too are modeled to form the root systems that anchor them into the cortical bone. The regulatory molecules that control modeling are poorly understood, but recent evidence suggests that parathyroid hormone-related protein (PTHrP) participates in this process. PTHrP functions principally as a paracrine regulatory molecule, and is known to be induced by mechanical loading in a number of sites. The most curious example of developmental modeling of the cortex is the migration of insertion sites such as that of the medial collateral ligament (MCL) along the bone surface during long-bone growth. We report here the mechanisms that mediate MCL migration using a combination of genetic, imaging and histological techniques. We describe a MCL migratory complex that comprises two components. The first is the MCL insertion site itself, which is a prototypical fibrous insertion site with coupled osteoclast and osteoblast activities, and its key feature is that it is anchored early in development, well before initiation of the long-bone growth spurt. Above the insertion site the periosteum is excavated by osteoclasts to form a migratory tract; this is mediated by wholly uncoupled osteoclastic bone resorption and remains as an unmineralized canal on the cortical surface in the adult. Load-induction of PTHrP appears to regulate the osteoclastic activity in both the insertion site and migratory tract.


Subject(s)
Medial Collateral Ligament, Knee/growth & development , Animals , Chondrocytes/cytology , Knee Joint/cytology , Knee Joint/growth & development , Mice , Osteoclasts/cytology , X-Ray Microtomography
10.
Best Pract Res Clin Endocrinol Metab ; 27(3): 403-14, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23856268

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.


Subject(s)
Breast/metabolism , Lactation/metabolism , Mammary Glands, Human/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Epithelial Cells/metabolism , Female , Humans , Mammary Glands, Animal/metabolism , Parathyroid Hormone-Related Protein/metabolism , Receptors, Calcium-Sensing/genetics , Signal Transduction/physiology
11.
Endocrinology ; 154(9): 3031-42, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23782944

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.


Subject(s)
Bone Development , Calcium/metabolism , Lactation/metabolism , Mammary Glands, Animal/metabolism , Parathyroid Hormone-Related Protein/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Animals, Newborn , Biological Transport , Bone Resorption/metabolism , Calcium/blood , Calcium/urine , Crosses, Genetic , Female , Gene Expression Regulation , Lactation/blood , Lactation/urine , Lactoglobulins/genetics , Lactoglobulins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Milk/metabolism , Parathyroid Hormone/blood , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Parathyroid Hormone-Related Protein/blood , Parathyroid Hormone-Related Protein/genetics , RNA, Messenger/metabolism , Receptors, Calcium-Sensing/genetics
12.
J Bone Miner Res ; 28(3): 598-607, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23109045

ABSTRACT

The sites that receive ligament and tendon insertions (entheses) on the cortical surfaces of long bones are poorly understood, particularly regarding modeling and regulation. Entheses are classified as either fibrocartilaginous or fibrous based on their structures. Fibrous entheses typically insert into the metaphysis or diaphysis of a long bone, bear a periosteal component, and are modeled during long-bone growth. This modeling forms a root system by which the insertions attach to the cortical surface. In the case of the medial collateral ligament, modeling drives actual migration of the ligament along the cortical surface in order to accommodate linear growth, whereas in other sites modeling may excavate a deep cortical root system (eg, the teres major insertion) or a shallow root system with a large footprint (eg, the latissimus dorsi insertion). We report here that conditionally deleting parathyroid hormone-related protein (PTHrP) in fibrous entheses via Scleraxis-Cre targeting causes modeling to fail in these three iterations of osteoclast-driven enthesis excavation or migration. These iterations appear to represent formes frustes of a common modeling strategy, presumably differing from each other as a consequence of differences in biomechanical control. In sites in which PTHrP is not induced, either physiologically or because of conditional deletion, modeling does not take place and fibrocartilage is induced. These findings represent the initial genetic evidence that PTHrP regulates periosteal/intramembranous bone cell activity on cortical bone surfaces and indicate that PTHrP serves as a load-induced modeling tool in fibrous insertion sites during linear growth.


Subject(s)
Bone Development/physiology , Fibrocartilage/growth & development , Models, Biological , Parathyroid Hormone-Related Protein/physiology , Animals , Mice
13.
J Mammary Gland Biol Neoplasia ; 17(2): 167-88, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22752723

ABSTRACT

This paper resulted from a conference entitled "Lactation and Milk: Defining and refining the critical questions" held at the University of Colorado School of Medicine from January 18-20, 2012. The mission of the conference was to identify unresolved questions and set future goals for research into human milk composition, mammary development and lactation. We first outline the unanswered questions regarding the composition of human milk (Section I) and the mechanisms by which milk components affect neonatal development, growth and health and recommend models for future research. Emerging questions about how milk components affect cognitive development and behavioral phenotype of the offspring are presented in Section II. In Section III we outline the important unanswered questions about regulation of mammary gland development, the heritability of defects, the effects of maternal nutrition, disease, metabolic status, and therapeutic drugs upon the subsequent lactation. Questions surrounding breastfeeding practice are also highlighted. In Section IV we describe the specific nutritional challenges faced by three different populations, namely preterm infants, infants born to obese mothers who may or may not have gestational diabetes, and infants born to undernourished mothers. The recognition that multidisciplinary training is critical to advancing the field led us to formulate specific training recommendations in Section V. Our recommendations for research emphasis are summarized in Section VI. In sum, we present a roadmap for multidisciplinary research into all aspects of human lactation, milk and its role in infant nutrition for the next decade and beyond.


Subject(s)
Breast Feeding , Child Development , Lactation , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Milk, Human/metabolism , Morphogenesis , Adult , Animals , Animals, Newborn , Biomedical Research/trends , Disease Susceptibility , Female , Humans , Infant , Infant, Newborn , Intestines/growth & development , Intestines/microbiology , Mammary Glands, Animal , Metabolic Diseases/etiology , Metabolic Diseases/prevention & control , Milk/metabolism
14.
Proc Natl Acad Sci U S A ; 109(14): 5190-5, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22431609

ABSTRACT

Magnetic resonance imaging (MRI) of solids is rarely attempted. One of the main reasons is that the broader MR linewidths, compared to the narrow resonance of the hydrogen ((1)H) in free water, limit both the attainable spatial resolution and the signal-to-noise ratio. Basic physics research, stimulated by the quest to build a quantum computer, gave rise to a unique MR pulse sequence that offers a solution to this long-standing problem. The "quadratic echo" significantly narrows the broad MR spectrum of solids. Applying field gradients in sync with this line-narrowing sequence offers a fresh approach to carry out MRI of hard and soft solids with high spatial resolution and with a wide range of potential uses. Here we demonstrate that this method can be used to carry out three-dimensional MRI of the phosphorus ((31)P) in ex vivo bone and soft tissue samples.


Subject(s)
Magnetic Resonance Imaging/methods , Bone and Bones/metabolism , Phosphorus Isotopes , Protons
15.
J Bone Miner Res ; 27(4): 865-75, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22189918

ABSTRACT

Despite the dramatic bone loss that occurs during lactation, bone mineral density rapidly recovers after offspring are weaned and milk production stops. The goal of this study is to quantify site-specific changes in bone quantity and quality during and after lactation in a mouse model. We used micro computed tomography (µCT), individual trabecula segmentation (ITS), digital topological analysis (DTA)-based tissue mineral density (TMD) analysis, and micro finite element analysis (µFEA) to quantify the effects of lactation and weaning on bone microarchitecture, mineralization, and stiffness at the spine, tibia, and femur. We found a significant decrease in trabecular plate microarchitecture, tissue mineralization of the trabecular surface, trabecular central skeleton, and intervening envelopes, and whole bone stiffness in lactating versus nulliparous mice at all three sites. In recovered mice, all these different aspects of bone quality were comparable to nulliparous mice at the spine. In contrast, trabecular plate microarchitecture and whole bone stiffness at the tibia and femur in recovered mice were lower than nulliparous mice, as were central trabecular tissue mineralization and cortical structure at the femur. These findings are consistent with clinical observations of partial recovery of femoral bone mineral density BMD after lactation in humans. The observed differences in trabecular surface tissue mineralization in nulliparous, lactating, and recovered mice are consistent with prior observations that maternal bone turnover shifts from resorption to formation at the time of pup weaning. The significant differences in trabecular central tissue mineralization during these three states suggest that osteocytes may contribute to the reversible loss of mineral during and after lactation. Future studies are necessary to determine whether differing functions of various bone cells at individual skeletal sites cause site-specific skeletal changes during and after lactation.


Subject(s)
Bone and Bones/anatomy & histology , Bone and Bones/physiology , Calcification, Physiologic/physiology , Lactation/physiology , Weaning , Animals , Biomechanical Phenomena/physiology , Bone Density/physiology , Bone and Bones/diagnostic imaging , Female , Finite Element Analysis , Imaging, Three-Dimensional , Mice , X-Ray Microtomography
16.
PLoS One ; 6(11): e27278, 2011.
Article in English | MEDLINE | ID: mdl-22087279

ABSTRACT

PTHrP is necessary for the formation of the embryonic mammary gland and, in its absence, the embryonic mammary bud fails to form the neonatal duct system. In addition, PTHrP is produced by the breast during lactation and contributes to the regulation of maternal calcium homeostasis during milk production. In this study, we examined the role of PTHrP during post-natal mammary development. Using a PTHrP-lacZ transgenic mouse, we surveyed the expression of PTHrP in the developing post-natal mouse mammary gland. We found that PTHrP expression is restricted to the basal cells of the gland during pubertal development and becomes expressed in milk secreting alveolar cells during pregnancy and lactation. Based on the previous findings that overexpression of PTHrP in cap and myoepithelial cells inhibited ductal elongation during puberty, we predicted that ablation of native PTHrP expression in the post-natal gland would result in accelerated ductal development. To address this hypothesis, we generated two conditional models of PTHrP-deficiency specifically targeted to the postnatal mammary gland. We used the MMTV-Cre transgene to ablate the floxed PTHrP gene in both luminal and myoepithelial cells and a tetracycline-regulated K14-tTA;tetO-Cre transgene to target PTHrP expression in just myoepithelial and cap cells. In both models of PTHrP ablation, we found that mammary development proceeds normally despite the absence of PTHrP. We conclude that PTHrP signaling is not required for normal ductal or alveolar development.


Subject(s)
Mammary Glands, Animal/growth & development , Parathyroid Hormone-Related Protein/physiology , Animals , Female , Gene Expression , Lactation , Mice , Mice, Transgenic , Parathyroid Hormone-Related Protein/analysis , Parathyroid Hormone-Related Protein/deficiency , Pregnancy , Tissue Distribution
17.
Calcif Tissue Int ; 89(2): 151-62, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21643724

ABSTRACT

Patients with X-linked hypophosphatemia (XLH) develop enthesophytes and osteophytes secondary to articular cartilage degeneration and together are the primary cause of morbidity in adult patients so afflicted. We have previously characterized the enthesopathy in Hyp mice, a murine model of XLH. We now extend these studies to the synovial joint in order to characterize potential cellular changes in articular cartilage that may predispose patients to the osteoarthropathy of XLH. We report that, despite highly elevated levels of alkaline phosphatase activity throughout articular cartilage, there is a complete loss in the mineralized zone of articular cartilage as assessed by von Kossa staining of mineral and as quantified by EPIC-microCT analysis and evidence of vascular invasion. We also identify the downregulation of extracellular matrix (ECM) factors identified as regulators of terminally differentiated mineralizing articular chondrocytes. There is also a striking increase in the histochemical staining of sulfated proteoglycans, a change that may reflect the loss of a transitional tissue that reduces mechanical stress at the interface between cartilage and subchondral bone. The failure of mineralizing articular chondrocytes to develop in the hypophosphatemic state suggests that phosphate may be a key regulator of chondrocyte mineralization. Accordingly, we find that the appropriate zonal arrangement and phenotypic markers of articular cartilage are significantly reestablished by phosphate-replacement therapy. Given the turnover and maintenance of articular cartilage ECM, the identification of early and abnormal cellular changes unique to XLH will undoubtedly aid in a more effective management of this disease to minimize the onset of degenerative osteoarthropathy.


Subject(s)
Calcification, Physiologic , Cartilage, Articular/pathology , Disease Models, Animal , Familial Hypophosphatemic Rickets/complications , Genetic Diseases, X-Linked , Mice, Mutant Strains , Osteoarthritis/etiology , Animals , Bone Density/physiology , Calcification, Physiologic/physiology , Cartilage, Articular/blood supply , Cartilage, Articular/metabolism , Cartilage, Articular/physiopathology , Familial Hypophosphatemic Rickets/pathology , Humans , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/pathology , Osteoarthritis/pathology
18.
Endocrinology ; 151(12): 5591-601, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21047946

ABSTRACT

During lactation, calcium is mobilized from the maternal skeleton to supply the breast for milk production. This results in rapid but fully reversible bone loss. Prior studies have suggested that PTHrP, secreted from the breast, and estrogen deficiency, due to suckling-induced central hypogonadism, combine to trigger bone resorption. To determine whether this combination was sufficient to explain bone loss during lactation, we raised PTHrP levels and decreased levels of estrogens in nulliparous mice. PTHrP was infused via osmotic minipumps and estrogens were decreased either by using leuprolide, a long-acting GnRH agonist, or by surgical ovariectomy (OVX). Bone mineral density declined by 23.2 ± 1.3% in the spine and 16.8 ± 1.9% in the femur over 10 d of lactation. This was accompanied by changes in trabecular architecture and an increase in both osteoblast and osteoclast numbers. OVX and PTHrP infusion both induced a modest decline in bone mineral density over 10 d, but leuprolide treatment did not. The combination of OVX and PTHrP was more effective than either treatment alone, but there was no interaction between PTHrP and leuprolide. None of the treatments reproduced the same degree of bone loss caused by lactation. However, both forms of estrogen deficiency led to an increase in osteoclasts, whereas infusion of PTHrP increased both osteoblasts and osteoclasts. Therefore, although the combination of PTHrP and estrogen deficiency contributes to bone loss, it is insufficient to reproduce the full response of the skeleton to lactation, suggesting that other factors also regulate bone metabolism during this period.


Subject(s)
Bone Density/drug effects , Bone and Bones/drug effects , Estrogens/metabolism , Lactation/physiology , Parathyroid Hormone-Related Protein/pharmacology , Animals , Bone Density/physiology , Bone and Bones/physiology , Female , Fertility Agents, Female/administration & dosage , Fertility Agents, Female/pharmacology , Leuprolide/administration & dosage , Leuprolide/pharmacology , Mice , Ovariectomy , Parathyroid Hormone-Related Protein/administration & dosage
19.
Proc Natl Acad Sci U S A ; 107(25): 11405-10, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20534448

ABSTRACT

After lactation, weaning causes mammary epithelial cell (MEC) apoptosis. MECs express the plasma membrane calcium-ATPase 2 (PMCA2), which transports calcium across the apical surface of the cells into milk. Here we show that PMCA2 is down-regulated early in mammary involution associated with changes in MEC shape. We demonstrate that loss of PMCA2 expression raises intracellular calcium levels and sensitizes MECs to apoptosis. In contrast, overexpression of PMCA2 in T47D breast cancer cells lowers intracellular calcium and protects them from apoptosis. Finally, we show that high PMCA2 expression in breast cancers is associated with poor outcome. We conclude that loss of PMCA2 expression at weaning triggers apoptosis by causing cellular calcium crisis. PMCA2 overexpression, on the other hand, may play a role in breast cancer progression by conferring resistance to apoptosis.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Animals , Disease Progression , Humans , Mammary Glands, Animal/pathology , Mice , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Treatment Outcome
20.
J Biol Chem ; 283(36): 24435-47, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18621740

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.


Subject(s)
Breast Neoplasms/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Neoplasm Proteins/metabolism , Parathyroid Hormone-Related Protein/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Hypercalcemia , Lactation/metabolism , MAP Kinase Signaling System , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Metastasis , Pregnancy , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL