Network


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

Hotspot


Dive into the research topics where Julia M. Hum is active.

Publication


Featured researches published by Julia M. Hum.


Current Osteoporosis Reports | 2014

Hypophosphatemic Rickets: Revealing Novel Control Points for Phosphate Homeostasis

Kenneth E. White; Julia M. Hum; Michael J. Econs

Rapid and somewhat surprising advances have recently been made toward understanding the molecular mechanisms causing heritable disorders of hypophosphatemia. The results of clinical, genetic, and translational studies have interwoven novel concepts underlying the endocrine control of phosphate metabolism, with far-reaching implications for treatment of both rare Mendelian diseases as well as common disorders of blood phosphate excess such as chronic kidney disease (CKD). In particular, diseases caused by changes in the expression and proteolytic control of the phosphaturic hormone fibroblast growth factor-23 (FGF23) have come to the forefront in terms of directing new models explaining mineral metabolism. These hypophosphatemic disorders as well as others resulting from independent defects in phosphate transport or metabolism will be reviewed herein, and implications for emerging therapeutic strategies based upon these new findings will be discussed.


PLOS ONE | 2011

Non-overlapping functions for Pyk2 and FAK in osteoblasts during fluid shear stress-induced mechanotransduction

Suzanne R.L. Young; Julia M. Hum; Eric J. Rodenberg; Charles H. Turner; Fredrick M. Pavalko

Mechanotransduction, the process by which cells convert external mechanical stimuli such as fluid shear stress (FSS) into biochemical changes, plays a critical role in maintenance of the skeleton. We have proposed that mechanical stimulation by FSS across the surfaces of bone cells results in formation of unique signaling complexes called mechanosomes that are launched from sites of adhesion with the extracellular matrix and with other bone cells [1]. Deformation of adhesion complexes at the cell membrane ultimately results in alteration of target gene expression. Recently, we reported that focal adhesion kinase (FAK) functions as a part of a mechanosome complex that is required for FSS-induced mechanotransduction in bone cells. This study extends this work to examine the role of a second member of the FAK family of non-receptor protein tyrosine kinases, proline-rich tyrosine kinase 2 (Pyk2), and determine its role during osteoblast mechanotransduction. We use osteoblasts harvested from mice as our model system in this study and compared the contributions of Pyk2 and FAK during FSS induced mechanotransduction in osteoblasts. We exposed Pyk2+/+ and Pyk2−/− primary calvarial osteoblasts to short period of oscillatory fluid flow and analyzed downstream activation of ERK1/2, and expression of c-fos, cyclooxygenase-2 and osteopontin. Unlike FAK, Pyk2 was not required for fluid flow-induced mechanotransduction as there was no significant difference in the response of Pyk2+/+ and Pyk2−/− osteoblasts to short periods of fluid flow (FF). In contrast, and as predicted, FAK−/− osteoblasts were unable to respond to FF. These data indicate that FAK and Pyk2 have distinct, non-redundant functions in launching mechanical signals during osteoblast mechanotransduction. Additionally, we compared two methods of generating FF in both cell types, oscillatory pump method and another orbital platform method. We determined that both methods of generating FF induced similar responses in both primary calvarial osteoblasts and immortalized calvarial osteoblasts.


Journal of Bone and Mineral Research | 2016

Conditional Deletion of Murine Fgf23: Interruption of the Normal Skeletal Responses to Phosphate Challenge and Rescue of Genetic Hypophosphatemia.

Erica L. Clinkenbeard; Taryn A. Cass; Pu Ni; Julia M. Hum; Teresita Bellido; Matthew R. Allen; Kenneth E. White

The transgenic and knockout (KO) animals involving Fgf23 have been highly informative in defining novel aspects of mineral metabolism, but are limited by shortened lifespan, inability of spatial/temporal FGF23 control, and infertility of the global KO. To more finely test the role of systemic and genetic influences in FGF23 production, a mouse was developed that carried a floxed (“f”)‐Fgf23 allele (exon 2 floxed) which demonstrated in vivo recombination when bred to global‐Cre transgenic mice (eIIa‐cre). Mice homozygous for the recombined allele (“Δ”) had undetectable serum intact FGF23, elevated serum phosphate (p < 0.05), and increased kidney Cyp27b1 mRNA (p < 0.05), similar to global Fgf23‐KO mice. To isolate cellular FGF23 responses during phosphate challenge, Fgf23Δ/f mice were mated with early osteoblast type Iα1 collagen 2.3‐kb promoter‐cre mice (Col2.3‐cre) and the late osteoblast/early osteocyte Dentin matrix protein‐1‐cre (Dmp1‐cre). Fgf23Δ/f/Col2.3‐cre+ and Fgf23Δ/f/Dmp1‐cre+ exhibited reduced baseline serum intact FGF23 versus controls. After challenge with high‐phosphate diet Cre– mice had 2.1‐fold to 2.5‐fold increased serum FGF23 (p < 0.01), but Col2.3‐cre+ mice had no significant increase, and Dmp1‐cre+ mice had only a 37% increase (p < 0.01) despite prevailing hyperphosphatemia in both models. The Fgf23Δ/f/Col2.3‐cre was bred onto the Hyp (murine X‐linked hypophosphatemia [XLH] model) genetic background to test the contribution of osteoblasts and osteocytes to elevated FGF23 and Hyp disease phenotypes. Whereas Hyp mice maintained inappropriately elevated FGF23 considering their marked hypophosphatemia, Hyp/Fgf23Δ/f/Col2.3‐cre+ mice had serum FGF23 <4% of Hyp (p < 0.01), and this targeted restriction normalized serum phosphorus and ricketic bone disease. In summary, deleting FGF23 within early osteoblasts and osteocytes demonstrated that both cell types contribute to baseline circulating FGF23 concentrations, and that targeting osteoblasts/osteocytes for FGF23 production can modify systemic responses to changes in serum phosphate concentrations and rescue the Hyp genetic syndrome.


Journal of The American Society of Nephrology | 2017

Chronic Hyperphosphatemia and Vascular Calcification Are Reduced by Stable Delivery of Soluble Klotho

Julia M. Hum; Linda Maureen O'Bryan; Arun K. Tatiparthi; Taryn A. Cass; Erica L. Clinkenbeard; Martin S. Cramer; Manoj Bhaskaran; Robert L. Johnson; Jonathan M. Wilson; Rosamund C. Smith; Kenneth E. White

αKlotho (αKL) regulates mineral metabolism, and diseases associated with αKL deficiency are characterized by hyperphosphatemia and vascular calcification (VC). αKL is expressed as a membrane-bound protein (mKL) and recognized as the coreceptor for fibroblast growth factor-23 (FGF23) and a circulating soluble form (cKL) created by endoproteolytic cleavage of mKL. The functions of cKL with regard to phosphate metabolism are unclear. We tested the ability of cKL to regulate pathways and phenotypes associated with hyperphosphatemia in a mouse model of CKD-mineral bone disorder and αKL-null mice. Stable delivery of adeno-associated virus (AAV) expressing cKL to diabetic endothelial nitric oxide synthase-deficient mice or αKL-null mice reduced serum phosphate levels. Acute injection of recombinant cKL downregulated the renal sodium-phosphate cotransporter Npt2a in αKL-null mice supporting direct actions of cKL in the absence of mKL. αKL-null mice with sustained AAV-cKL expression had a 74%-78% reduction in aorta mineral content and a 72%-77% reduction in mineral volume compared with control-treated counterparts (P<0.01). Treatment of UMR-106 osteoblastic cells with cKL + FGF23 increased the phosphorylation of extracellular signal-regulated kinase 1/2 and induced Fgf23 expression. CRISPR/Cas9-mediated deletion of fibroblast growth factor receptor 1 (FGFR1) or pretreatment with inhibitors of mitogen-activated kinase kinase 1 or FGFR ablated these responses. In summary, sustained cKL treatment reduced hyperphosphatemia in a mouse model of CKD-mineral bone disorder, and it reduced hyperphosphatemia and prevented VC in mice without endogenous αKL. Furthermore, cKL stimulated Fgf23 in an FGFR1-dependent manner in bone cells. Collectively, these findings indicate that cKL has mKL-independent activity and suggest the potential for enhancing cKL activity in diseases of hyperphosphatemia with associated VC.


Journal of Cellular Physiology | 2011

Blockade of TNFR1 signaling: A role of oscillatory fluid shear stress in osteoblasts

Haifang Wang; Suzanne R. Young; Rita Gerard-O'Riley; Julia M. Hum; Zhouqi Yang; Joseph P. Bidwell; Fredrick M. Pavalko

Fluid shear stress protects cells from TNF‐α‐induced apoptosis. Oscillatory fluid shear stress (OFSS) is generally perceived as physiologically relevant biophysical signal for bone cells. Here we identify several cellular mechanisms responsible for mediating the protective effects of OFSS against TNF‐α‐induced apoptosis in vitro. We found that exposure of MC3T3‐E1 osteoblast‐like cells to as little as 5 min of OFSS suppressed TNF‐α‐induced activation of caspase‐3, cleavage of PARP and phosphorylation of histone. In contrast, H2O2‐induced apoptosis was not inhibited by OFSS suggesting that OFSS might not be protecting cells from TNF‐α‐induced apoptosis via stimulation of global pro‐survival signaling pathways. In support of this speculation, OFSS inhibition of TNF‐α‐induced apoptosis was unaffected by inhibitors of several pro‐survival signaling pathways including pI3‐kinase (LY294002), MAPK/ERK kinase (PD98059 or U0126), intracellular Ca2+ release (U73122), NO production (L‐NAME), or protein synthesis (cycloheximide) that were applied to cells during exposure to OFSS and during TNF‐α treatment. However, TNF‐α‐induced phosphorylation and degradation of IκBα was blocked by pre‐exposure of cells to OFSS suggesting a more specific effect of OFSS on TNF‐α signaling. We therefore focused on the mechanism of OFSS regulation of TNF‐receptor 1 (TNFR1) signaling and found that OFSS (1) reduced the amount of receptor on the cell surface, (2) prevented the association of ubiquitinated RIP in TNFR1 complexes with TRADD and TRAF2, and (3) reduced TNF‐α‐induced IL‐8 promoter activity in the nucleus. We conclude that the anti‐apoptotic effect of OFSS is not mediated by activation of universal pro‐survival signaling pathways. Rather, OFSS inhibits TNF‐α‐induced pro‐apoptotic signaling which can be explained by the down‐regulation of TNFR1 on the cell surface and blockade of TNFR1 downstream signaling by OFSS. J. Cell. Physiol. 226: 1044–1051, 2011.


Endocrinology | 2017

Acute Parathyroid Hormone Injection Increases C-Terminal but Not Intact Fibroblast Growth Factor 23 Levels

Vanessa M. Knab; Braden Corbin; Olena Andrukhova; Julia M. Hum; Pu Ni; Seham M. Rabadi; Akira Maeda; Kenneth E. White; Reinhold G. Erben; Harald Jüppner; Marta Christov

The acute effects of parathyroid hormone (PTH) on fibroblast growth factor 23 (FGF23) in vivo are not well understood. After a single subcutaneous PTH (1-34) injection (50 nmol/kg) in mice, FGF23 levels were assessed in plasma using assays that measure either intact alone (iFGF23) or intact/C-terminal FGF23 (cFGF23). Furthermore, FGF23 messenger RNA (mRNA) and protein levels were assessed in bone. In addition, we examined the effects of PTH treatment on FGF23 production in vitro using differentiated calvarial osteocyte-like cells. cFGF23 levels increased by three- to fivefold within 2 hours following PTH injection, which returned to baseline by 4 hours. In contrast, iFGF23 levels remained unchanged for the first 2 hours, yet declined to ∼60% by 6 hours and remained suppressed before returning to baseline after 24 hours. Using homozygous mice for an autosomal dominant hypophosphatemic rickets-FGF23 mutation or animals treated with a furin inhibitor, we showed that cFGF23 and iFGF23 levels increased equivalently after PTH injection. These findings are consistent with increased FGF23 production in bone, yet rapid cleavage of the secreted intact protein. Using primary osteocyte-like cell cultures, we showed that PTH increased FGF23 mRNA expression through cyclic adenosine monophosphate/protein kinase A, but not inositol triphosphate/protein kinase C signaling; PTH also increased furin protein levels. In conclusion, PTH injection rapidly increases FGF23 production in bone in vivo and in vitro. However, iFGF23 is rapidly degraded. At later time points through an unidentified mechanism, a sustained decrease in FGF23 production occurs.


Haematologica | 2017

Erythropoietin stimulates murine and human fibroblast growth factor-23, revealing novel roles for bone and bone marrow.

Erica L. Clinkenbeard; Mark R. Hanudel; Keith R. Stayrook; Hitesh Appaiah; Emily Farrow; Taryn A. Cass; Lelia J. Summers; Colin S. Ip; Julia M. Hum; Joseph Thomas; Mircea Ivan; Briana M. Richine; Rebecca J. Chan; Thomas L. Clemens; Ernestina Schipani; Yves Sabbagh; Linlin Xu; Edward F. Srour; Marta B. Alvarez; Melissa A. Kacena; Isidro B. Salusky; Tomas Ganz; Elizabeta Nemeth; Kenneth E. White

Early stages of chronic kidney disease (CKD) are characterized by development of progressive anemia as well as concurrent marked elevation of the phosphaturic hormone fibroblast growth factor 23 (FGF23). As kidney function declines, FGF23 further increases and anemia worsens, due to either inadequate production of renal erythropoietin (EPO) or incidence of hypoferremia. Moreover, in CKD, anemia and elevated FGF23 levels are associated with left ventricular hypertrophy (LVH), CKD progression, and mortality. Treatment of CKD-related anemia involves iron repletion and erythropoietin (EPO) administration. EPO is one of the most extensively used medications in CKD, but its administration is associated with increased risks of cardiovascular disease and mortality. Although FGF23 levels increase early in CKD, the pathophysiological regulation of FGF23 is still not completely understood. Phosphate, 1,25-dihydroxyvitamin D (1,25D), parathyroid hormone, and calcium affect FGF23 production; however, these factors are still within normal ranges when bone and circulating FGF23 increase. Recent studies demonstrate intriguing associations between hypoxia, iron deficiency, and FGF23 upregulation. Indeed, in the settings of normal and impaired kidney function, iron deficiency potently increases bone Fgf23 expression. However, other anemia-related factors, including EPO, could potentially contribute to elevated FGF23 production. As both EPO therapy and FGF23 are associated with adverse outcomes in CKD, we explored the hypothesis that EPO is a previously unrecognized regulator of this phosphaturic hormone. Collectively, our pre-clinical findings suggest that modulating EPO exposure in CKD patients may lower FGF23 and thereby decrease its adverse effects. To examine whether exogenous EPO stimulates FGF23 in vivo, wild-type C57BL/6 mice at 6-8 weeks of age were injected with increasing doses of recombinant human EPO (25-250 U/g of body weight). A 3-day regimen induced a dose-dependent, 40-fold maximal increase in whole bone Fgf23 mRNA expression (Figure 1A), paralleled by increased serum total FGF23 as measured with an ELISA that detects both C-terminal FGF23 fragments (‘cFGF23’) and bioactive intact FGF23 (‘iFGF23’) (Figure


PLOS ONE | 2014

Mechanical loading in osteocytes induces formation of a Src/Pyk2/MBD2 complex that suppresses anabolic gene expression

Julia M. Hum; Richard N. Day; Joseph P. Bidwell; Yingxiao Wang; Fredrick M. Pavalko

Mechanical stimulation of the skeleton promotes bone gain and suppresses bone loss, ultimately resulting in improved bone strength and fracture resistance. The molecular mechanisms directing anabolic and/or anti-catabolic actions on the skeleton during loading are not fully understood. Identifying molecular mechanisms of mechanotransduction (MTD) signaling cascades could identify new therapeutic targets. Most research into MTD mechanisms is typically focused on understanding the signaling pathways that stimulate new bone formation in response to load. However, we investigated the structural, signaling and transcriptional molecules that suppress the stimulatory effects of loading. The high bone mass phenotype of mice with global deletion of either Pyk2 or Src suggests a role for these tyrosine kinases in repression of bone formation. We used fluid shear stress as a MTD stimulus to identify a novel Pyk2/Src-mediated MTD pathway that represses mechanically-induced bone formation. Our results suggest Pyk2 and Src function as molecular switches that inhibit MTD in our mechanically stimulated osteocyte culture experiments. Once activated by oscillatory fluid shear stress (OFSS), Pyk2 and Src translocate to and accumulate in the nucleus, where they associate with a protein involved in DNA methylation and the interpretation of DNA methylation patterns –methyl-CpG-binding domain protein 2 (MBD2). OFSS-induced Cox-2 and osteopontin expression was enhanced in Pyk2 KO osteoblasts, while inhibition of Src enhanced osteocalcin expression in response to OFSS. We found that Src kinase activity increased in the nucleus of osteocytes in response to OFSS and an interaction activated between Src (Y418) and Pyk2 (Y402) increased in response to OFSS. Thus, as a mechanism to prevent an over-reaction to physical stimulation, mechanical loading may induce the formation of a Src/Pyk2/MBD2 complex in the nucleus that functions to suppress anabolic gene expression.


Bone reports | 2017

The metabolic bone disease associated with the Hyp mutation is independent of osteoblastic HIF1α expression

Julia M. Hum; Erica L. Clinkenbeard; Colin S. Ip; Taryn A. Cass; Matthew R. Allen; Kenneth E. White

Fibroblast growth factor-23 (FGF23) controls key responses to systemic phosphate increases through its phosphaturic actions on the kidney. In addition to stimulation by phosphate, FGF23 positively responds to iron deficiency anemia and hypoxia in rodent models and in humans. The disorder X-linked hypophosphatemia (XLH) is characterized by elevated FGF23 in concert with an intrinsic bone mineralization defect. Indeed, the Hyp mouse XLH model has disturbed osteoblast to osteocyte differentiation with altered expression of a wide variety of genes, including FGF23. The transcription factor Hypoxia inducible factor-1α (HIF1α) has been implicated in regulating FGF23 production and plays a key role in proper bone cell differentiation. Thus the goals of this study were to determine whether HIF1α activation could influence FGF23, and to test osteoblastic HIF1α production on the Hyp endocrine and skeletal phenotypes in vivo. Treatment of primary cultures of osteoblasts/osteocytes and UMR-106 cells with the HIF activator AG490 resulted in rapid HIF1α stabilization and increased Fgf23 mRNA (50–100 fold; p < 0.01–0.001) in a time- and dose-dependent manner. Next, the Phex gene deletion in the Hyp mouse was bred onto mice with a HIF1α/Osteocalcin (OCN)-Cre background. Although HIF1α effects on bone could be detected, FGF23-related phenotypes due to the Hyp mutation were independent of HIF1α in vivo. In summary, FGF23 can be driven by ectopic HIF1α activation under normal iron conditions in vitro, but factors independent of HIF1α activity after mature osteoblast formation are responsible for the disease phenotypes in Hyp mice in vivo.


Bone | 2017

Novel functions of circulating Klotho

Julia M. Hum; Linda Maureen O'Bryan; Rosamund C. Smith; Kenneth E. White

Collaboration


Dive into the Julia M. Hum's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pu Ni

Indiana University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge