Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Patricia W. M. Ho is active.

Publication


Featured researches published by Patricia W. M. Ho.


Journal of Cellular Physiology | 1996

Expression of parathyroid hormone‐related protein in cells of osteoblast lineage

Naoto Suda; Matthew T. Gulespie; Kathy Traianedes; Hong Zhou; Patricia W. M. Ho; Daphne K. Hards; Elizabeth H. Allan; T. John Martin; Jane M. Moseley

The expression of parathyroid hormone‐related protein (PTHrP) was studied in a range of cell cultures representative of the osteoblast lineage and in rat calvarial sections. Primary newborn rat calvarial cells, a rat preosteoblastic cell line (UMR 201), a mouse stromal cell line (ST 2), a mouse calvaria‐derived osteoblastic cell line (KS 4), and rat osteosarcoma cell lines (UMR 106‐01 and ‐06), all expressed PTHrP when examined by reverse transcription polymerase chain reaction (RT‐PCR). Using a radioimmunoassay we also demonstrated PTHrP in the conditioned medium of the cultured cells, with the exception of UMR 106‐01 and ‐06 cells. Treatment of UMR 201 cells with all‐trans‐retinoic acid which induces them to acquire a more differentiated phenotype, also led to a time‐dependent decrease in PTHrP mRNA levels as determined by RT‐PCR, Northern blot analysis, and in situ hybridization. Decreased PTHrP levels in the conditioned medium of the treated cells was also observed. These results suggested that PTHrP production might be greater in less mature osteoblasts. Examination of the populations obtained from newborn rat calvariae by sequential collagenase digestion revealed that the early digests exhibited low ALP activity, low expression of PTH/PTHrP receptor mRNA, and no adenylate cyclase response to PTHrP(1–34). These populations showed the highest level of mRNA and production of PTHrP. Cells from later digests, the “osteoblast‐rich” populations, had reduced PTHrP expression. Immunohistochemistry and in situ hybridization in sections of newborn rat calvariae showed PTHrp expression in cuboidal osteoblasts located adjacent to bone and in spindle‐shaped cells in the periosteal region. It is concluded that PTHrP is produced by cells of the osteoblast lineage, supporting the hypothesis that PTHrP may function physiologically as a paracrine factor in bone.


Journal of Cellular Physiology | 1997

PTHrP and cell division: Expression and localization of PTHrP in a keratinocyte cell line (HaCaT) during the cell cycle

M. H. C. Lam; S. L. Olsen; W. A. Rankin; Patricia W. M. Ho; T. J. Martin; Matthew T. Gillespie; Jane M. Moseley

Parathyroid hormone‐related protein (PTHrP) is highly expressed in normal skin keratinocytes, and its involvement in growth and differentiation processes in these cells has been implicated by several lines of evidence which include the use of antisense PTHrP (Kaiser et al., 1994, Mol. Endocrinol., 8:139–147). In this study, we have investigated whether PTHrP expression and its subcellular localization is linked to cell cycle progression in a human keratinocyte cell line (HaCaT), which constitutively expresses and secretes PTHrP. PTHrP mRNA and immunoreactive PTHrP were assessed in asynchronous dividing cells and in cells blocked at G1 or G2 + M phases of the cell cycle using several different protocols. The response of PTHrP mRNA expression was examined following readdition of serum in the continued presence of cycle blockers, and after release from cell cycle block, or from cell synchronization by serum deprivation. PTHrP expression was greatest in actively dividing cells when cells were in S and G2 + M phases of the cell cycle and were lowest in quiescent G1 cells. Most notable were the high levels of PTHrP mRNA and protein in cells at G2 + M phase of the cell cycle at division. Furthermore, PTHrP was localized to the nucleolus in quiescent cells, but redistributed to the cytoplasm when cells were actively dividing. Taken together, these results support a role for PTHrP in cell division in keratinocytes. In asynchronously growing cells, PTHrP expression fell as cells became confluent at a time when cell growth is inhibited and cells begin to differentiate. Mitogen stimulation of HaCaT cells resulted in a rapid increase in PTHrP mRNA expression, but was dependent upon cells being in the G1 phase of the cell cycle. Cells blocked in G1 responded to mitogen both in the continued presence of aphidicolin or when released from block. Cells blocked at G2 + M with colcemid expressed high levels of PTHrP mRNA and protein, and PTHrP mRNA did not respond further to mitogen in the continued presence of blocker. However, in cells released from block at G2 + M by addition of serum, an increase in PTHrP expression was seen coincident with the progression of cells into G1. In contrast, in a squamous cancer cell line (COLO16), basal PTHrP expression was high and was not altered during the cell cycle or by cell cycle block, consistent with association of its dysregulated expression in malignant cells. The results of this study suggest that PTHrP may have two roles in the cell cycle; one in G1 in response to mitogen, and a second at cell division when its expression is high and it is relocated from the nucleolus to the cytoplasm. J. Cell. Physiol. 173:433–446, 1997.


Journal of Cellular Biochemistry | 2003

Differentiation potential of a mouse bone marrow stromal cell line.

Elizabeth H. Allan; Patricia W. M. Ho; Akihro Umezawa; Jun-ichi Hata; Fusao Makishima; Matthew T. Gillespie; T. John Martin

In order to study osteoblast differentiation we subcloned a cell derived from a mouse a bone marrow stromal cell line, Kusa O, and obtained a number of clones representative of three different phenotypes. One that neither differentiated into osteoblasts nor into adipocytes, a second that differentiated into osteoblasts but not adipocytes, and a third that differentiated into both osteoblasts and adipocytes. Four subclones were selected for further characterization according to their ability to mineralize and/or differentiate into adipocytes. The non‐mineralizing clone had no detectable alkaline phosphatase activity although some alkaline phosphatase mRNA was detected after 21 days in osteoblast differentiating medium. Alkaline phosphatase activity and mRNA in the three mineralizing clones were comparable with the parent clones. Osteocalcin mRNA and protein levels in the non‐mineralizing clone were low and non‐detectable, respectively, while both were elevated in the parent cells and mineralizing subclones after 21 days in differentiating medium. PTH receptor mRNA and activity increased in the four subclones and parent cells with differentiation. mRNA for two other osteoblast phenotypic markers, osteopontin and bone sialoprotein, were similarly expressed in the parent cells and subclones while mRNAs for the transcription factors, Runx2 and osterix, were detectable in both parent and subclone cells. Runx2 was unchanged with differentiation while osterix was increased. Interestingly, PPARγ mRNA expression did not correlate with cell line potential to differentiate into adipocytes. Indian hedgehog mRNA and its receptor (patched) mRNA levels both increased with differentiation while mRNA levels of the Wnt pathway components β‐catenin and dickkopf also increased with differentiation. Although we have focussed on characterizing these clones from the osteoblast perspective it is clear that they may be useful for studying both osteoblast and adipocyte differentiation as well as their transdifferentiation. J. Cell. Biochem. 90: 158–169, 2003.


Journal of Bone and Mineral Research | 2013

EphrinB2/EphB4 inhibition in the osteoblast lineage modifies the anabolic response to parathyroid hormone.

Farzin M. Takyar; Stephen Tonna; Patricia W. M. Ho; Blessing Crimeen-Irwin; Emma K. Baker; T. John Martin; Natalie A. Sims

Previous reports indicate that ephrinB2 expression by osteoblasts is stimulated by parathyroid hormone (PTH) and its related protein (PTHrP) and that ephrinB2/EphB4 signaling between osteoblasts and osteoclasts stimulates osteoblast differentiation while inhibiting osteoclast differentiation. To determine the role of the ephrinB2/EphB4 interaction in the skeleton, we used a specific inhibitor, soluble EphB4 (sEphB4), in vitro and in vivo. sEphB4 treatment of cultured osteoblasts specifically inhibited EphB4 and ephrinB2 phosphorylation and reduced mRNA levels of late markers of osteoblast/osteocyte differentiation (osteocalcin, dentin matrix protein‐1 [DMP‐1], sclerostin, matrix‐extracellular phosphoglycoprotein [MEPE]), while substantially increasing RANKL. sEphB4 treatment in vivo in the presence and absence of PTH increased osteoblast formation and mRNA levels of early osteoblast markers (Runx2, alkaline phosphatase, Collagen 1α1, and PTH receptor [PTHR1]), but despite a substantial increase in osteoblast numbers, there was no significant change in bone formation rate or in late markers of osteoblast/osteocyte differentiation. Rather, in the presence of PTH, sEphB4 treatment significantly increased osteoclast formation, an effect that prevented the anabolic effect of PTH, causing instead a decrease in trabecular number. This enhancement of osteoclastogenesis by sEphB4 was reproduced in vitro but only in the presence of osteoblasts. These data indicate that ephrinB2/EphB4 signaling within the osteoblast lineage is required for late stages of osteoblast differentiation and, further, restricts the ability of osteoblasts to support osteoclast formation, at least in part by limiting RANKL production. This indicates a key role for the ephrinB2/EphB4 interaction within the osteoblast lineage in osteoblast differentiation and support of osteoclastogenesis.


Bone | 2013

Modeling distinct osteosarcoma subtypes in vivo using Cre:lox and lineage-restricted transgenic shRNA

Anthony J. Mutsaers; Emma K. Baker; Megan Russell; Alistair M. Chalk; Meaghan Wall; Brain J.J. Liddicoat; Patricia W. M. Ho; John Slavin; Ankita Goradia; T. John Martin; Louise E. Purton; Ross A. Dickins; Carl R. Walkley

Osteosarcoma is the most common primary cancer of bone and one that predominantly affects children and adolescents. Osteoblastic osteosarcoma represents the major subtype of this tumor, with approximately equal representation of fibroblastic and chondroblastic subtypes. We and others have previously described murine models of osteosarcoma based on osteoblast-restricted Cre:lox deletion of Trp53 (p53) and Rb1 (Rb), resulting in a phenotype most similar to fibroblastic osteosarcoma in humans. We now report a model of the most prevalent form of human osteosarcoma, the osteoblastic subtype. In contrast to other osteosarcoma models that have used Cre:lox mediated gene deletion, this model was generated through shRNA-based knockdown of p53. As is the case with the human disease the shRNA tumors most frequently present in the long bones and preferentially disseminate to the lungs; feature less consistently modeled using Cre:lox approaches. Our approach allowed direct comparison of the in vivo consequences of targeting the same genetic drivers using two different technologies, Cre:lox and shRNA. This demonstrated that the effects of Cre:lox and shRNA mediated knock-down are qualitatively different, at least in the context of osteosarcoma, and yielded distinct subtypes of osteosarcoma. Through the use of complementary genetic modification strategies we have established a model of the most common clinical subtype of osteosarcoma that was not previously represented and more fully recapitulated the clinical spectrum of this cancer.


The FASEB Journal | 2014

EphrinB2 signaling in osteoblasts promotes bone mineralization by preventing apoptosis

Stephen Tonna; Farzin M. Takyar; Christina Vrahnas; Blessing Crimeen-Irwin; Patricia W. M. Ho; Ingrid J. Poulton; Holly J. Brennan; Narelle E. McGregor; Elizabeth H. Allan; Huynh Nguyen; Mark R. Forwood; Liliana Tatarczuch; Eleanor J. Mackie; T. John Martin; Natalie A. Sims

Cells that form bone (osteoblasts) express both ephrinB2 and EphB4, and previous work has shown that pharmacological inhibition of the eph‐rinB2/EphB4 interaction impairs osteoblast differentiation in vitro and in vivo. The purpose of this study was to determine the role of ephrinB2 signaling in the osteoblast lineage in the process of bone formation. Cultured osteoblasts from mice with osteoblast‐specific ablation of ephrinB2 showed delayed expression of osteoblast differentiation markers, a finding that was reproduced by ephrinB2, but not EphB4, RNA interference. Microcomputed tomography, histomorphometry, and mechanical testing of the mice lacking ephrinB2 in osteoblasts revealed a 2‐fold delay in bone mineralization, a significant reduction in bone stiffness, and a 50% reduction in osteoblast differentiation induced by anabolic parathyroid hormone (PTH) treatment, compared to littermate sex‐ and age‐matched controls. These defects were associated with significantly lower mRNA levels of late osteoblast differentiation markers and greater levels of osteoblast and osteocyte apoptosis, indicated by TUNEL staining and transmission electron microscopy of bone samples, and a 2‐fold increase in annexin V staining and 7‐fold increase in caspase 8 activation in cultured ephrinB2 deficient osteoblasts. We conclude that osteoblast differentiation and bone strength are maintained by antiapoptotic actions of ephrinB2 signaling within the osteoblast lineage.—Tonna, S., Takyar, F. M., Vrahnas, C., Crimeen‐Irwin, B., Ho, P. W. M., Poulton, I. J., Brennan, H. J., McGregor, N. E., Allan, E. H., Nguyen, H., Forwood, M. R., Tatarczuch, L., Mackie, E. J., Martin, T. J., Sims, N. A., EphrinB2 signaling in osteoblasts promotes bone mineralization by preventing apoptosis. FASEB J. 28, 4482–4496 (2014). www.fasebj.org


Journal of Bone and Mineral Research | 2012

Sustained RANKL response to parathyroid hormone in oncostatin M receptor‐deficient osteoblasts converts anabolic treatment to a catabolic effect in vivo

Emma C. Walker; Ingrid J. Poulton; Narelle E. McGregor; Patricia W. M. Ho; Elizabeth H. Allan; Julie M. Quach; T. John Martin; Natalie A. Sims

Parathyroid hormone (PTH) is the only approved anabolic agent for osteoporosis treatment. It acts via osteoblasts to stimulate both osteoclast formation and bone formation, with the balance between these two activities determined by the mode of administration. Oncostatin M (OSM), a gp130‐dependent cytokine expressed by osteoblast lineage cells, has similar effects and similar gene targets in the osteoblast lineage. In this study, we investigated whether OSM might participate in anabolic effects of PTH. Microarray analysis and quantitative real‐time polymerase chain reaction (qPCR) of PTH‐treated murine stromal cells and primary calvarial osteoblasts identified significant regulation of gp130 and gp130‐dependent coreceptors and ligands, including a significant increase in OSM receptor (OSMR) expression. To determine whether OSMR signaling is required for PTH anabolic action, 6‐week‐old male Osmr−/− mice and wild‐type (WT) littermates were treated with hPTH(1–34) for 3 weeks. In WT mice, PTH increased trabecular bone volume and trabecular thickness. In contrast, the same treatment had a catabolic effect in Osmr−/− mice, reducing both trabecular bone volume and trabecular number. This was not explained by any alteration in the increased osteoblast formation and mineral apposition rate in response to PTH in Osmr−/− compared with WT mice. Rather, PTH treatment doubled osteoclast surface in Osmr−/− mice, an effect not observed in WT mice. Consistent with this finding, when osteoclast precursors were cultured in the presence of osteoblasts, more osteoclasts were formed in response to PTH when Osmr−/− osteoblasts were used. Neither PTH1R mRNA levels nor cAMP response to PTH were modified in Osmr−/− osteoblasts. However, RANKL induction in PTH‐treated Osmr−/− osteoblasts was sustained at least until 24 hours after PTH exposure, an effect not observed in WT osteoblasts. These data indicate that the transient RANKL induction by intermittent PTH administration, which is associated with its anabolic action, is changed to a prolonged induction in OSMR‐deficient osteoblasts, resulting in bone destruction.


Reproduction, Fertility and Development | 1995

Parathyroid hormone-related protein (PTHrP) concentrations in human amniotic fluid during gestation and at the time of labour

Mary E. Wlodek; Patricia W. M. Ho; Gregory E. Rice; Jane M. Moseley; T.J. Martin; Shaun P. Brennecke

To establish the changes associated with gestational age and labour status in parathyroid hormone-related protein (PTHrP) concentrations in the amniotic fluid, human amniotic fluid was collected from non-labouring and labouring women at < 37 weeks of gestation (preterm) and at term (> or = 37 weeks). PTHrP was assayed by a specific N-terminal radioimmunoassay. PTHrP concentrations in amniotic fluid obtained from non-labouring women were significantly lower at preterm (15-36 weeks; 14.1 +/- 2.5 pmol L(-1); n = 11) than at term (37-42 weeks; 39.3 +/- 7.6 pmol L(-1); n = 16; P < 0.0009). Concentrations of PTHrP in amniotic fluid obtained from labouring women were also significantly lower at preterm (27-36 week; 12.2 +/- 4.7 pmol L(-1); n = 4; P < 0.01) than at term (37-42 weeks; 63. 8 +/- 19.6 pmol L(-1); n = 9). There were no significant changes in concentration associated with labour status, either at preterm or at term. The physiological significance of elevated amniotic fluid concentrations of PTHrP has yet to be established, but the data are consistent with the suggestion that PTHrP plays a role in fetal membrane function during late gestation.


Journal of Cellular Physiology | 2008

BMP-2 regulation of PTHrP and osteoclastogenic factors during osteoblast differentiation of C2C12 cells

Antonio R. G. Susperregui; Francesc Viñals; Patricia W. M. Ho; Matthew T. Gillespie; T. John Martin; Francesc Ventura

Bone morphogenetic protein‐2 (BMP‐2) is strongly involved in the induction of osteoblast differentiation from mesenchymal cell precursors, as well as in enhancing bone matrix production by osteoblastic cells. Likewise, the osteoporotic phenotype of PTHrP deficient mice makes clear the importance of this paracrine regulator in bone physiology. Here, we report that BMP‐2 rapidly down‐regulated PTHrP gene expression through a transcriptional mechanism in pluripotent mesenchymal C2C12 cells, whereas BMP‐2 increased expression of PTHrP receptor. PTHrP did not significantly alter the BMP‐dependent Smad transcriptional pathway. Similarly, PTHrP did not significantly modify the BMP‐regulated expression of RANKL or OPG, cytokines involved in osteoclastogenesis. More importantly, addition of PTHrP, through the PKA signaling pathway, partially prevented the BMP‐dependent induction of some osteogenic markers such as Runx2 and Osterix in C2C12 cells. Our data suggest that BMP‐2 down‐regulation of PTHrP could facilitate terminal differentiation of osteoblasts. J. Cell. Physiol. 216: 144–152, 2008.


Oncogene | 2015

Knockdown of PTHR1 in osteosarcoma cells decreases invasion and growth and increases tumor differentiation in vivo

Patricia W. M. Ho; Ankita Goradia; Megan Russell; Alistair M. Chalk; K.M. Milley; Emma K. Baker; Janine A. Danks; John Slavin; Mannu K. Walia; Blessing Crimeen-Irwin; Ross A. Dickins; T. J. Martin; Carl R. Walkley

Osteosarcoma (OS) is the most common cancer of bone. Parathyroid hormone (PTH) regulates calcium homeostasis and bone development, while the paracrine/autocrine PTH-related protein (PTHrP) has central roles in endochondral bone formation and bone remodeling. Using a murine OS model, we found that OS cells express PTHrP and the common PTH/PTHrP receptor (PTHR1). To investigate the role of PTHR1 signaling in OS cell behavior, we used shRNA to reduce PTHR1 expression. This only mildly inhibited proliferation in vitro, but markedly reduced invasion through collagen and reduced expression of RANK ligand (RANKL). Administration of PTH(1–34) did not stimulate OS proliferation in vivo but, strikingly, PTHR1 knockdown resulted in a profound growth inhibition and increased differentiation/mineralization of the tumors. Treatment with neutralizing antibody to PTHrP did not recapitulate the knockdown of PTHR1. Consistent with this lack of activity, PTHrP was predominantly intracellular in OS cells. Knockdown of PTHR1 resulted in increased expression of late osteoblast differentiation genes and upregulation of Wnt antagonists. RANKL production was reduced in knockdown tumors, providing for reduced homotypic signaling through the receptor, RANK. Loss of PTHR1 resulted in the coordinated loss of gene signatures associated with the polycomb repressive complex 2 (PRC2). Using Ezh2 inhibitors, we demonstrate that the increased expression of osteoblast maturation markers is in part mediated by the loss of PRC2 activity. Collectively these results demonstrate that PTHR1 signaling is important in maintaining OS proliferation and undifferentiated state. This is in part mediated by intracellular PTHrP and through regulation of the OS epigenome.

Collaboration


Dive into the Patricia W. M. Ho's collaboration.

Top Co-Authors

Avatar

T. John Martin

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Natalie A. Sims

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jane M. Moseley

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

T. J. Martin

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Blessing Crimeen-Irwin

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ingrid J. Poulton

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Gillespie

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Carl R. Walkley

St. Vincent's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Narelle E. McGregor

St. Vincent's Institute of Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge