Network


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

Hotspot


Dive into the research topics where Daniel J. Horan is active.

Publication


Featured researches published by Daniel J. Horan.


Science Translational Medicine | 2013

Sclerostin inhibition reverses skeletal fragility in an Lrp5-deficient mouse model of OPPG syndrome.

Rajendra Kedlaya; Shreya Veera; Daniel J. Horan; Rachel E. Moss; Ugur M. Ayturk; Christina M. Jacobsen; Margot E. Bowen; Chris Paszty; Matthew L. Warman; Alexander G. Robling

Humans with osteoporosis pseudoglioma syndrome might benefit from sclerostin neutralization therapies. Building Stronger Bones Osteoporosis pseudoglioma syndrome (OPPG) is a rare genetic condition caused by an autosomal recessive mutation in LRP5, which contributes to regulation of bone mineral density. This mutation results in severely thinner, brittle bones—osteoporosis. Most therapies for osteoporosis aim at inhibiting bone loss; however, in OPPG patients, bone resorption is normal but bone formation is markedly reduced, which suggests that anabolic therapies that promote bone formation may be more beneficial. Now, Kedlaya et al. examine the effects of the anabolic therapy sclerostin neutralization in an OPPG animal model. Sclerostin inhibits bone formation by binding to LRP5/6. Thus, although neutralizing sclerostin seemed a promising anabolic track for general osteoporosis patients, it was predicted to be less effective for OPPG patients with mutated LRP5. The authors tested this hypothesis in an LRP5-deficient mouse model. They found through both genetic and therapeutic experiments that sclerostin neutralization can improve bone mineral density even in the absence of functional LRP5. These data support the advent of clinical trials for sclerostin neutralization in OPPG patients. Osteoporosis pseudoglioma syndrome (OPPG) is a rare genetic disease that produces debilitating effects in the skeleton. OPPG is caused by mutations in LRP5, a WNT co-receptor that mediates osteoblast activity. WNT signaling through LRP5, and also through the closely related receptor LRP6, is inhibited by the protein sclerostin (SOST). It is unclear whether OPPG patients might benefit from the anabolic action of sclerostin neutralization therapy (an approach currently being pursued in clinical trials for postmenopausal osteoporosis) in light of their LRP5 deficiency and consequent osteoblast impairment. To assess whether loss of sclerostin is anabolic in OPPG, we measured bone properties in a mouse model of OPPG (Lrp5−/−), a mouse model of sclerosteosis (Sost−/−), and in mice with both genes knocked out (Lrp5−/−;Sost−/−). Lrp5−/−;Sost−/− mice have larger, denser, and stronger bones than do Lrp5−/− mice, indicating that SOST deficiency can improve bone properties via pathways that do not require LRP5. Next, we determined whether the anabolic effects of sclerostin depletion in Lrp5−/− mice are retained in adult mice by treating 17-week-old Lrp5−/− mice with a sclerostin antibody for 3 weeks. Lrp5+/+ and Lrp5−/− mice each exhibited osteoanabolic responses to antibody therapy, as indicated by increased bone mineral density, content, and formation rates. Collectively, our data show that inhibiting sclerostin can improve bone mass whether LRP5 is present or not. In the absence of LRP5, the anabolic effects of SOST depletion can occur via other receptors (such as LRP4/6). Regardless of the mechanism, our results suggest that humans with OPPG might benefit from sclerostin neutralization therapies.


Antimicrobial Agents and Chemotherapy | 2011

Investigation of the Efficacy of Micafungin in the Treatment of Histoplasmosis Using Two North American Strains of Histoplasma capsulatum

Chadi A. Hage; Patricia Connolly; Daniel J. Horan; Michelle Durkin; Melinda Smedema; Robert Zarnowski; Patricia Smith; L. Joseph Wheat

ABSTRACT Micafungin alone and combined with liposomal amphotericin B was evaluated against two strains of Histoplasma capsulatum. Micafungin was active in vitro against the mold but not the yeast form but was ineffective in vivo. Micafungin appears to be ineffective in treatment of histoplasmosis.


Endocrinology | 2016

Adult-Onset Deletion of β-Catenin in 10kbDmp1-Expressing Cells Prevents Intermittent PTH-Induced Bone Gain

Rajendra Kedlaya; Kyung Shin Kang; Jung Min Hong; Vidya Bettagere; Kyung Eun Lim; Daniel J. Horan; Paola Divieti-Pajevic; Alexander G. Robling

β-Catenin (βcat) is a major downstream signaling node in canonical Wingless-related integration site (Wnt) signaling pathway, and its activity is crucial for canonical Wnt signal transduction. Wnt signaling has recently been implicated in the osteo-anabolic response to PTH, a potent calcium-regulating factor. We investigated whether βcat is essential for the anabolic action of intermittent PTH by generating male mice with adult-onset deletion of βcat in a subpopulation of bone cells (osteocytes and late-stage osteoblasts), treating them with an anabolic regimen of PTH, and measuring the skeletal responses. Male (10kb)Dmp1-CreERt2 transgenic mice that also harbored floxed loss-of-function βcat alleles (βcat(f/f)) were induced for Cre activity using tamoxifen, then injected daily with human PTH 1-34 (30 μg/kg) or vehicle for 5 weeks. Mice in which βcat was deleted showed either total lack of bone mineral density (BMD) gain, or BMD loss, and did not respond to PTH treatment. However, bone mass measurements in the trabecular compartment of the femur and spine revealed PTH-induced bone gain whether βcat was deleted or not. PTH-stimulated increases in periosteal and cancellous bone formation rates were not impaired by βcat deletion, but resorption markers and cortical porosity were significantly increased in induced mice, particularly induced mice treated with PTH. These results suggest that βcat is required for net-positive BMD effects of PTH therapy but that the anabolic effects per se of PTH treatment might not require osteocytic/osteoblastic βcat.


Medical Mycology | 2013

Histoplasma capsulatum preferentially induces IDO in the lung

Chadi A. Hage; Daniel J. Horan; Michelle Durkin; Patricia Connolly; Zeruesenay Desta; Todd C. Skaar; Kenneth S. Knox; L. Joseph Wheat

Indoleamine 2,3 dioxygenase (IDO) plays an important role in immunoregulation as it is involved in downregulating immune responses to infections. We sought to characterize IDO activity in histoplasmosis and to do so, C57Bl6 mice were infected intranasally with Histoplasma capsulatum. After infection, lung and spleen IDO activity was assessed by HPLC and IDO expression by qRT-PCR. The distribution of IDO was determined by immunohistochemical staining. Cytokine levels were measured in lung and spleen homogenates using cytokine bead array. Fungal burden was quantified by culture. Subcutaneous pellets containing methyltryptophane (1-MT) were employed to inhibit IDO in vivo. Histoplasma infection strongly induced functional lung IDO, with activity at its highest at weeks 1 and 2 and then decreased thereafter as the mice cleared the infection. Lung IDO activity positively correlated with the fungal burden (Rho = 0.845), interferon-γ (Rho = 0.839) and tumor necrosis factor-α (Rho = 0.791) levels, P < 0.001. In contrast, spleen IDO activity was not induced despite high infection burden and cytokine levels. IDO expressing cells were predominately located at the ring edge of Histoplasma-induced granulomas. IDO inhibition prior to infection reduced fungal burdens and inflammation in lungs and spleen. Histoplasma preferentially induces lung IDO, as early as one week after infection. IDO appears to modulate the immune response to Histoplasma infection.


Journal of Bone and Mineral Research | 2018

Conditional Deletion of Sost in MSC-Derived Lineages Identifies Specific Cell-Type Contributions to Bone Mass and B-Cell Development.

Cristal S. Yee; Jennifer O. Manilay; Jiun C. Chang; Nicholas R. Hum; Deepa K. Murugesh; Jamila Bajwa; Melanie Mendez; Aris E. Economides; Daniel J. Horan; Alexander G. Robling; Gabriela G. Loots

Sclerostin (Sost) is a negative regulator of bone formation and blocking its function via antibodies has shown great therapeutic promise by increasing both bone mass in humans and animal models. Sclerostin deletion in Sost KO mice (Sost−/−) causes high bone mass (HBM) similar to sclerosteosis patients. Sost−/− mice have been shown to display an up to 300% increase in bone volume/total volume (BV/TV), relative to age‐matched controls. It has been postulated that the main source of skeletal sclerostin is the osteocyte. To understand the cell‐type specific contributions to the HBM phenotype described in Sost−/− mice, as well as to address the endocrine and paracrine mode of action of sclerostin, we examined the skeletal phenotypes of conditional Sost loss‐of‐function (SostiCOIN/iCOIN) mice with specific deletions in (1) the limb mesenchyme (Prx1‐Cre; targets osteoprogenitors and their progeny); (2) midstage osteoblasts and their progenitors (Col1‐Cre); (3) mature osteocytes (Dmp1‐Cre); and (4) hypertrophic chondrocytes and their progenitors (ColX‐Cre). All conditional alleles resulted in significant increases in bone mass in trabecular bone in both the femur and lumbar vertebrae, but only Prx1‐Cre deletion fully recapitulated the amplitude of the HBM phenotype in the appendicular skeleton and the B‐cell defect described in the global KO. Despite WT expression of Sost in the axial skeleton of Prx1‐Cre deleted mice, these mice also had a significant increase in bone mass in the vertebrae, but the sclerostin released in circulation by the axial skeleton did not affect bone parameters in the appendicular skeleton. Also, both Col1 and Dmp1 deletion resulted in a similar 80% significant increase in trabecular bone mass, but only Col1 and Prx1 deletion resulted in a significant increase in cortical thickness. We conclude that several cell types within the Prx1‐osteoprogenitor‐derived lineages contribute significant amounts of sclerostin protein to the paracrine pool of Sost in bone.


JCI insight | 2018

Sclerostin neutralization unleashes the osteoanabolic effects of Dkk1 inhibition

Phillip C. Witcher; Sara E. Miner; Daniel J. Horan; Whitney A. Bullock; Kyung-Eun Lim; Kyung Shin Kang; Alison L. Adaniya; Ryan D. Ross; Gabriela G. Loots; Alexander G. Robling

The WNT pathway has become an attractive target for skeletal therapies. High-bone-mass phenotypes in patients with loss-of-function mutations in the LRP5/6 inhibitor Sost (sclerosteosis), or in its downstream enhancer region (van Buchem disease), highlight the utility of targeting Sost/sclerostin to improve bone properties. Sclerostin-neutralizing antibody is highly osteoanabolic in animal models and in human clinical trials, but antibody-based inhibition of another potent LRP5/6 antagonist, Dkk1, is largely inefficacious for building bone in the unperturbed adult skeleton. Here, we show that conditional deletion of Dkk1 from bone also has negligible effects on bone mass. Dkk1 inhibition increases Sost expression, suggesting a potential compensatory mechanism that might explain why Dkk1 suppression lacks anabolic action. To test this concept, we deleted Sost from osteocytes in, or administered sclerostin neutralizing antibody to, mice with a Dkk1-deficient skeleton. A robust anabolic response to Dkk1 deletion was manifest only when Sost/sclerostin was impaired. Whole-body DXA scans, μCT measurements of the femur and spine, histomorphometric measures of femoral bone formation rates, and biomechanical properties of whole bones confirmed the anabolic potential of Dkk1 inhibition in the absence of sclerostin. Further, combined administration of sclerostin and Dkk1 antibody in WT mice produced a synergistic effect on bone gain that greatly exceeded individual or additive effects of the therapies, confirming the therapeutic potential of inhibiting multiple WNT antagonists for skeletal health. In conclusion, the osteoanabolic effects of Dkk1 inhibition can be realized if sclerostin upregulation is prevented. Anabolic therapies for patients with low bone mass might benefit from a strategy that accounts for the compensatory milieu of WNT inhibitors in bone tissue.


Bone | 2017

Clcn7F318L/+ as a new mouse model of Albers-Schönberg disease

J. Caetano-Lopes; S.G. Lessard; S. Hann; K. Espinoza; Kyung Shin Kang; K.E. Lim; Daniel J. Horan; H.R. Noonan; D. Hu; Roland Baron; Alexander G. Robling; Matthew L. Warman

Dominant negative mutations in CLCN7, which encodes a homodimeric chloride channel needed for matrix acidification by osteoclasts, cause Albers-Schönberg disease (also known as autosomal dominant osteopetrosis type 2). More than 25 different CLCN7 mutations have been identified in patients affected with Albers-Schönberg disease, but only one mutation (Clcn7G213R) has been introduced in mice to create an animal model of this disease. Here we describe a mouse with a different osteopetrosis-causing mutation (Clcn7F318L). Compared to Clcn7+/+ mice, 12-week-old Clcn7F318L/+ mice have significantly increased trabecular bone volume, consistent with Clcn7F318L acting as a dominant negative mutation. Clcn7F318L/F318L and Clcn7F318L/G213R mice die by 1month of age and resemble Clcn7 knockout mice, which indicate that p.F318L mutant protein is non-functional and p.F318L and p.G213R mutant proteins do not complement one another. Since it has been reported that treatment with interferon gamma (IFN-G) improves bone properties in Clcn7G213R/+ mice, we treated Clcn7F318L/+ mice with IFN-G and observed a decrease in osteoclast number and mineral apposition rate, but no overall improvement in bone properties. Our results suggest that the benefits of IFN-G therapy in patients with Albers-Schönberg disease may be mutation-specific.


Bone | 2018

Induction of Lrp5 HBM-causing mutations in Cathepsin-K expressing cells alters bone metabolism

Kyung Shin Kang; Jung Min Hong; Daniel J. Horan; Kyung-Eun Lim; Whitney A. Bullock; Angela Bruzzaniti; Steven Hann; Matthew L. Warman; Alexander G. Robling

High-bone-mass (HBM)-causing missense mutations in the low density lipoprotein receptor-related protein-5 (Lrp5) are associated with increased osteoanabolic action and protection from disuse- and ovariectomy-induced osteopenia. These mutations (e.g., A214V and G171V) confer resistance to endogenous secreted Lrp5/6 inhibitors, such as sclerostin (SOST) and Dickkopf homolog-1 (DKK1). Cells in the osteoblast lineage are responsive to canonical Wnt stimulation, but recent work has indicated that osteoclasts exhibit both indirect and direct responsiveness to canonical Wnt. Whether Lrp5-HBM receptors, expressed in osteoclasts, might alter osteoclast differentiation, activity, and consequent net bone balance in the skeleton, is not known. To address this, we bred mice harboring heterozygous Lrp5 HBM-causing conditional knock-in alleles to Ctsk-Cre transgenic mice and studied the phenotype using DXA, μCT, histomorphometry, serum assays, and primary cell culture. Mice with HBM alleles induced in Ctsk-expressing cells (TG) exhibited higher bone mass and architectural properties compared to non-transgenic (NTG) counterparts. In vivo and in vitro measurements of osteoclast activity, population density, and differentiation yielded significant reductions in osteoclast-related parameters in female but not male TG mice. Droplet digital PCR performed on osteocyte enriched cortical bone tubes from TG and NTG mice revealed that ~8-17% of the osteocyte population (depending on sex) underwent recombination of the conditional Lrp5 allele in the presence of Ctsk-Cre. Further, bone formation parameters in the midshaft femur cortex show a small but significant increase in anabolic action on the endocortical but not periosteal surface. These findings suggest that Wnt/Lrp5 signaling in osteoclasts affects osteoclastogenesis and activity in female mice, but also that some of the changes in bone mass in TG mice might be due to Cre expression in the osteocyte population.


Scientific Reports | 2017

Quick and inexpensive paraffin-embedding method for dynamic bone formation analyses

Amy Porter; Regina Irwin; Josselyn Miller; Daniel J. Horan; Alexander G. Robling; Laura R. McCabe

We have developed a straightforward method that uses paraffin-embedded bone for undemineralized thin sectioning, which is amenable to subsequent dynamic bone formation measurements. Bone has stiffer material properties than paraffin, and therefore has hereforto usually been embedded in plastic blocks, cured and sectioned with a tungsten carbide knife to obtain mineralized bone sections for dynamic bone formation measures. This process is expensive and requires special equipment, experienced personnel, and time for the plastic to penetrate the bone and cure. Our method utilizes a novel way to prepare mineralized bone that increases its compliance so that it can be embedded and easily section in paraffin blocks. The approach is simple, quick, and costs less than 10% of the price for plastic embedded bone sections. While not effective for static bone measures, this method allows dynamic bone analyses to be readily performed in laboratories worldwide which might not otherwise have access to traditional (plastic) equipment and expertise.


The FASEB Journal | 2018

Skeletal cell YAP and TAZ combinatorially promote bone development.

Christopher D. Kegelman; Devon E. Mason; James H. Dawahare; Daniel J. Horan; Genevieve D. Vigil; Scott S. Howard; Alexander G. Robling; Teresita Bellido; Joel D. Boerckel

Collaboration


Dive into the Daniel J. Horan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kyung Shin Kang

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Matthew L. Warman

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gabriela G. Loots

Lawrence Livermore National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge