Network


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

Hotspot


Dive into the research topics where Daniel Branstetter is active.

Publication


Featured researches published by Daniel Branstetter.


Molecular and Cellular Biology | 2007

RANK Overexpression in Transgenic Mice with Mouse Mammary Tumor Virus Promoter-Controlled RANK Increases Proliferation and Impairs Alveolar Differentiation in the Mammary Epithelia and Disrupts Lumen Formation in Cultured Epithelial Acini

Eva González-Suárez; Daniel Branstetter; Allison Armstrong; Huyen Dinh; Hal Blumberg; William C. Dougall

ABSTRACT RANK and RANKL, the key regulators of osteoclast differentiation and activation, also play an important role in the control of proliferation and differentiation of mammary epithelial cells during pregnancy. Here, we show that RANK protein expression is strictly regulated in a spatial and temporal manner during mammary gland development. RANK overexpression under the control of the mouse mammary tumor virus (MMTV) promoter in a transgenic mouse model results in increased mammary epithelial cell proliferation during pregnancy, impaired differentiation of lobulo-alveolar structures, decreased expression of the milk proteins β-casein and whey acidic protein, and deficient lactation. We also show that treatment of three-dimensional in vitro cultures of primary mammary cells from MMTV-RANK mice with RANKL results in increased proliferation and decreased apoptosis in the luminal area, resulting in bigger acini with filled lumens. Taken together, these results suggest that signaling through RANK not only promotes proliferation but also inhibits the terminal differentiation of mammary epithelial cells. Moreover, the increased proliferation and survival observed in a three-dimensional culture system suggests a role for aberrant RANK signaling during breast tumorigenesis.


Cancer Biology & Therapy | 2010

Conatumumab, a fully human agonist antibody to death receptor 5, induces apoptosis via caspase activation in multiple tumor types

Paula Kaplan-Lefko; Jonathan David Graves; Stephen J. Zoog; Yang Pan; Jason Wall; Daniel Branstetter; Jodi Moriguchi; Angela Coxon; Justin Huard; Ren Xu; Matthew L. Peach; Gloria Juan; Stephen Kaufman; Qing Chen; Allison Bianchi; Jennifer Joy Kordich; Mark Ma; Ian Foltz; Brian Gliniak

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) binds to death receptors 4 and 5 (DR4, DR5) to transduce apoptotic signals. Conatumumab (AMG 655) is an investigational, fully human monoclonal agonist antibody (IgG1) to human DR5, which induces apoptosis via caspase activation. In this study, we demonstrate that conatumumab binds to DR5, activating intracellular caspases in vitro in the presence of a cross-linker. We also show that conatumumab has activity in vivo and inhibits tumor growth in colon (Colo205 and HCT-15), lung (H2122), and pancreatic (MiaPaCa2/T2) xenograft models. Conatumumab also enhances the anti-tumor activity of chemotherapeutics in vivo. Caspase activation in Colo205 tumors is dose-dependent and correlated with serum concentrations of conatumumab. We demonstrate for the first time that increases in serum caspase-3/7 activity and levels of M30 (neoepitope of caspase-cleaved cytokeratin-18) are linked to activation of the extrinsic apoptotic pathway using conatumumab in a preclinical model. These data suggest that conatumumab has potential as a therapeutic agent for treating patients with multiple tumor types, and that serum caspase-3/7 and M30 levels may serve as biomarkers of conatumumab activity.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Intravital imaging reveals distinct responses of depleting dynamic tumor-associated macrophage and dendritic cell subpopulations.

Marja Lohela; Amy-Jo Casbon; Aleksandra Olow; Lynn Bonham; Daniel Branstetter; Ning Weng; Jeffrey S. Smith; Zena Werb

Significance Tumor-infiltrating myeloid cells fail to support antitumor immunity, and instead contribute to increased malignancy and poor prognosis in breast cancer. We used intravital microscopy in a model of breast cancer to provide unique insight into cellular composition and real-time dynamics of the stromal microenvironment. We characterized the effects of targeted therapy against CSF-1R, an important myeloid cell mitogen receptor. We demonstrate that by blocking accumulation and compromising survival, anti–CSF-1R treatment depletes a cell population sharing characteristics of tumor-associated macrophages and dendritic cells, which further comprises subgroups with different endocytic and matrix metalloproteinase activities. However, the resulting relatively modest delay in tumor growth and metastasis suggests that other cells, such as neutrophils or fibroblasts, may maintain the tumor trophic microenvironment. Tumor-infiltrating inflammatory cells comprise a major part of the stromal microenvironment and support cancer progression by multiple mechanisms. High numbers of tumor myeloid cells correlate with poor prognosis in breast cancer and are coupled with the angiogenic switch and malignant progression. However, the specific roles and regulation of heterogeneous tumor myeloid populations are incompletely understood. CSF-1 is a major myeloid cell mitogen, and signaling through its receptor CSF-1R is also linked to poor outcomes. To characterize myeloid cell function in tumors, we combined confocal intravital microscopy with depletion of CSF-1R–dependent cells using a neutralizing CSF-1R antibody in the mouse mammary tumor virus long-terminal region-driven polyoma middle T antigen breast cancer model. The depleted cells shared markers of tumor-associated macrophages and dendritic cells (M-DCs), matching the phenotype of tumor dendritic cells that take up antigens and interact with T cells. We defined functional subgroups within the M-DC population by imaging endocytic and matrix metalloproteinase activity. Anti–CSF-1R treatment altered stromal dynamics and impaired both survival of M-DCs and accumulation of new M-DCs, but did not deplete Gr-1+ neutrophils or block doxorubicin-induced myeloid cell recruitment, and had a minimal effect on lung myeloid cells. Nevertheless, prolonged treatment led to delayed tumor growth, reduced vascularity, and decreased lung metastasis. Because the myeloid infiltrate in metastatic lungs differed significantly from that in mammary tumors, the reduction in metastasis may result from the impact on primary tumors. The combination of functional analysis by intravital imaging with cellular characterization has refined our understanding of the effects of experimental targeted therapies on the tumor microenvironment.


Journal of Immunology | 2007

Receptor Activator of NF-κB Ligand Inhibition Suppresses Bone Resorption and Hypercalcemia but Does Not Affect Host Immune Responses to Influenza Infection

Robert E. Miller; Daniel Branstetter; Allison P. Armstrong; Bryan Kennedy; Jon Jones; Laine Cowan; Jeanine Bussiere; William C. Dougall

Receptor activator of NF-κB (RANK) and its ligand (RANKL) are essential for osteoclast formation, function, and survival. Osteoprotegerin (OPG) inhibits RANK signaling by sequestering RANKL. This study evaluated the antiosteoclast and immunoregulatory effects of mouse rRANK-Fc, which, similar to OPG, can bind RANKL. The effect of RANKL inhibition by RANK-Fc on osteoclast function was determined by inhibition of vitamin D3 (1,25(OH)2D3)-induced hypercalcemia. Mice were injected with a single dose of 0, 10, 100, 500, or 1000 μg of RANK-Fc; 100 μg of OPG-Fc; or 5 μg of zoledronate 2 h before 1,25(OH)2D3 challenge on day 0, and sacrificed on days 1, 2, 4, 6, 8, 12, 16, and 20. RANK-Fc doses of 100 or 500 μg were tested in a mouse respiratory influenza virus host-resistance model. A single dose of RANK-Fc ≥100 μg suppressed elevation of serum calcium levels and suppressed the bone turnover marker serum pyridinoline at day 4 and later time points, similar to those observed with OPG-Fc and zoledronate (p ≤ 0.01 vs controls). By day 6, both immature and mature osteoclasts were depleted by high doses of RANK-Fc (500 and 1000 μg) or 100 μg of OPG-Fc. RANK-Fc doses of 100 or 500 μg had no detectable effect on immune responses to influenza infection, as measured by activation of cytotoxic T cell activity, influenza-specific IgG response, and virus clearance. RANK-Fc inhibition of RANKL has antiosteoclast activity at doses that have no detectable immunoregulatory activity, suggesting that RANKL inhibitors be further studied for their potential to treat excess bone loss.


Apoptosis | 2007

Expression of anti-apoptotic factors modulates Apo2L/TRAIL resistance in colon carcinoma cells

Mara S. Lippa; Laura Strockbine; Tiep Le; Daniel Branstetter; Craig A. Strathdee; Pamela M. Holland

Tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) selectively induces apoptosis in transformed cells. Normal cells and certain tumor cells can evade Apo2L/TRAIL induced cell death, but the determinants of Apo2L/TRAIL sensitivity are poorly understood. To better understand the factors that contribute to Apo2L/TRAIL resistance, we characterized two colon carcinoma lines with pronounced differences in Apo2L/TRAIL sensitivity. Colo205 cells are highly sensitive to Apo2L/TRAIL whereas Colo320 cells are unresponsive. Components of the DISC (death inducing signaling complex) could be immunoprecipitated from both cell lines in response to Apo2L/TRAIL. Sensitizing agents including a proteasome inhibitor conferred Apo2L/TRAIL sensitivity in Colo320 cells, indicating that the apoptotic machinery was intact and functional. We specifically suppressed the expression of Bcl-2, FLIP or XIAP in Colo320 cells. Downregulation of either FLIP or XIAP but not Bcl-2 restored sensitivity of Colo320 cells to Apo2L/TRAIL. Moreover, stable knockdown of XIAP expression in Colo320 subcutaneous tumors resulted in suppression of tumor growth and sensitivity to Apo2L/TRAIL in vivo. Our results indicate that only a specific subset of anti-apoptotic proteins can confer resistance to Apo2L/TRAIL in Colo320 cells. Elucidation of the factors that contribute to Apo2L/TRAIL resistance in tumor cells may provide insight into combination therapies with Apo2L/TRAIL in a clinical setting.


Breast Cancer Research | 2013

Progestin effects on cell proliferation pathways in the postmenopausal mammary gland

Charles E. Wood; Daniel Branstetter; Allison P. Jacob; J. Mark Cline; Thomas C. Register; Kathy Rohrbach; Li-Ya Huang; Hermina Borgerink; William C. Dougall

IntroductionMenopausal hormone therapies vary widely in their effects on breast cancer risk, and the mechanisms underlying these differences are unclear. The primary goals of this study were to characterize the mammary gland transcriptional profile of estrogen + progestin therapy in comparison with estrogen-alone or tibolone and investigate pathways of cell proliferation in a postmenopausal primate model.MethodsOvariectomized female cynomolgus macaque monkeys were randomized into the following groups: placebo (Con), oral conjugated equine estrogens (CEE), CEE with medroxyprogesterone acetate (MPA) (CEE + MPA), and tibolone given at a low or high dose (Lo or Hi Tib). All study treatment doses represented human clinical dose equivalents and were administered in the diet over a period of 2 years.ResultsTreatment with CEE + MPA had the greatest effect on global mRNA profiles and markers of mammary gland proliferation compared to CEE or tibolone treatment. Changes in the transcriptional patterns resulting from the addition of MPA to CEE were related to increased growth factors and decreased estrogen receptor (ER) signaling. Specific genes induced by CEE + MPA treatment included key members of prolactin receptor (PRLR)/signal transducer and activator of transcription 5 (STAT5), epidermal growth factor receptor (EGFR), and receptor activator of nuclear factor kappa B (RANK)/receptor activator of nuclear factor kappa B ligand (RANKL) pathways that were highly associated with breast tissue proliferation. In contrast, tibolone did not affect breast tissue proliferation but did elicit a mixed pattern of ER agonist activity.ConclusionOur findings indicate that estrogen + progestin therapy results in a distinct molecular profile compared to estrogen-alone or tibolone therapy, including upregulation of key growth factor targets associated with mammary carcinogenesis in mouse models. These changes may contribute to the promotional effects of estrogen + progestin therapy on breast cancer risk.


Breast Cancer Research and Treatment | 2012

RANKL inhibition combined with tamoxifen treatment increases anti-tumor efficacy and prevents tumor-induced bone destruction in an estrogen receptor-positive breast cancer bone metastasis model

Jude Canon; Rebecca Bryant; Martine Roudier; Daniel Branstetter; William C. Dougall

Tumor cells in bone can induce the activation of osteoclasts, which mediate bone resorption and release of growth factors and calcium from the bone matrix, resulting in a cycle of tumor growth and bone breakdown. Targeting the bone microenvironment by the inhibition of RANKL, an essential mediator of osteoclast function, not only prevents tumor-induced osteolysis but also decreases skeletal tumor burden in preclinical models. The inhibition of skeletal tumor progression after the inhibition of osteoclasts is via interruption of the “vicious cycle” of tumor/bone interactions. The majority of breast cancer patients at risk for bone metastases harbor estrogen receptor-positive (ER+) tumors. We developed a mouse model for ER+ breast cancer bone metastasis and evaluated the effect of RANKL inhibition on tumor-induced osteolysis and skeletal tumor growth both alone and in combination with tamoxifen. Luciferase-labeled MCF-7 cells (MCF-7Luc) formed metastatic foci in the hind limbs following intracardiac injection and caused mixed osteolytic/osteoblastic lesions. RANKL inhibition by OPG-Fc treatment blocked osteoclast activity and prevented tumor-induced osteolysis, as well as caused a marked decrease in skeletal tumor burden. Tamoxifen as a single agent reduced MCF-7Luc tumor growth in the hind limbs. In a combination experiment, OPG-Fc plus tamoxifen resulted in significantly greater tumor growth inhibition than either single agent alone. Histologic analysis revealed a decrease in the proliferation of tumor cells by both single agents, which was enhanced in the combination treatment. Upon treatment with OPG-Fc alone or in combination with tamoxifen, there was a complete absence of osteolytic lesions, demonstrating the ability of RANKL inhibition to prevent skeletal related morbidity in an ER+ model. The combination approach of targeting osteoclasts and the bone microenvironment by RANKL inhibition and the tumor directly via hormonal therapy may provide additional benefit to reducing skeletal tumor progression in ER+ breast cancer patients.


Breast Cancer Research and Treatment | 2014

RANKL expression in normal and malignant breast tissue responds to progesterone and is up-regulated during the luteal phase

Hong Hu; Jun Wang; Akash Gupta; Ali Shidfar; Daniel Branstetter; Oukseub Lee; David Ivancic; Megan E. Sullivan; Robert T. Chatterton; William C. Dougall; Seema A. Khan

The receptor activator of nuclear factor-κB ligand (RANKL) acts as a paracrine factor in progesterone-induced mammary epithelial proliferation and tumorigenesis. This evidence comes mainly from mouse models. Our aim was to examine whether RANKL expression in human normal and malignant breast is under the control of progesterone throughout the menstrual cycle. Breast epithelial samples were obtained by random fine needle aspiration (rFNA) of the contralateral unaffected breasts (CUB) of 18 breast cancer patients, with simultaneous serum hormone measurements. Genes correlated with serum progesterone levels were identified through Illumina microarray analysis. Validation was performed using qRT-PCR in rFNA samples from CUB of an additional 53 women and using immunohistochemistry in tissue microarrays of 61 breast cancer samples. Expression of RANKL, DIO2, and MYBPC1 was correlated with serum progesterone in CUB, and was significantly higher in luteal phase. RANKL and MYBPC1 mRNA expression were highly correlated between CUB and matched tumor samples. RANKL protein expression was also significantly increased in the luteal phase and highly correlated with serum progesterone levels in cancer samples, especially in hormone receptor positive tumors. The regulatory effects of progesterone on the expression of RANKL, DIO2, and MYBPC1 were confirmed in three-dimensional cultures of normal breast organoids. In normal breast and in breast cancer, RANKL mRNA and protein expression fluctuate with serum progesterone with highest levels in the luteal phase, suggesting that RANKL is a modulator of progesterone signaling in normal and malignant breast tissue and a potential biomarker of progesterone action and blockade.


Cancer Research | 2009

RANKL inhibition decreases the incidence of mammary adenocarcinomas in wild type (WT) and MMTV-RANK transgenic mice.

Daniel Branstetter; Allison P. Jacob; E Gonzalez-Suarez; Jon Jones; Ryan Erwert; M Chaisson-Blake; William C. Dougall

CTRC-AACR San Antonio Breast Cancer Symposium: 2008 Abstracts Abstract #4167 Purpose: RANK and its ligand (RANKL), key factors for bone remodeling and metastasis, are crucial for the development of mouse mammary gland during pregnancy. Upon treatment with hormone medroxyprogesterone (MPA) and a carcinogen (DMBA), transgenic mice overexpressing RANK in the mammary gland via the MMTV promoter exhibit a higher incidence of ductular hyperplasias, mammary intraepithelial neoplasias (MIN), and mammary adenocarcinomas than WT mice. This study assessed the expression of mouse RANK and RANKL in WT and MMTV-RANK during mammary tumor progression in this model and determined whether RANKL inhibition could inhibit mammary carcinogenesis. Methods: At 6 weeks of age WT and MMTV-RANK transgenic mice were implanted with 90-day release 50mg MPA pellets that were replaced at expiration (weeks 19 and 32). DMBA was administered orally at weeks 9, 10, 12, and 13. The expression of RANK and RANKL was determined by immunohistochemistry (IHC). The specificity of the anti-mouse RANKL and anti-mouse RANK antibodies were verified by their lack of reactivity in RANKL -/- or RANK -/- mouse tissues, respectively. Simultaneous with the first DMBA treatment, mice were treated with RANK-Fc (10 mg/kg, 3x/week) or vehicle and the onset of mammary tumor formation was measured. Mammary tumor formation was determined by palpation and confirmed by histologic examination. Preneoplastic lesions were counted by whole mount analysis of the mammary gland at 4 weeks after the last DMBA treatment. Epithelial proliferation was measured by BrdU labeling of the mammary epithelium at multiple timepoints following the last DMBA treatment (2 days after the first DMBA treatment; 4 and 7 weeks following the last). Results: In the MPA/DMBA-induced mammary tumor model, MMTV-RANK mice developed palpable mammary tumors earlier than WT mice with a median onset of 86 days for the MMTV-RANK cohort compared with 132 days for the WT mice. At early timepoints, the number of preneoplastic lesions and mammary epithelial proliferation were greater in MMTV-RANK mice relative to WT. IHC demonstrated that MPA increased the number of RANKL-positive epithelial cells and that RANKL was present in preneoplastic MIN lesions and adenocarcinomas in both WT and MMTV-RANK mice. RANK expression was observed in the epithelial component of hyperplasias, MIN lesions, and adenocarcinomas in both WT and MMTV-RANK mice. Inhibition of RANKL with RANK-Fc substantially decreased the presence of preneoplastic lesions and the proliferation of mammary epithelial cells in MMTV-RANK mice. In addition, treatment of either MMTV-RANK or WT mice with RANK-Fc delayed the onset of palpable mammary tumors. Treatment of the MMTV-RANK mice with RANK-Fc decreased the incidence of mammary adenocarcinoma. Conclusion: These data suggest an operative role for RANKL in progesterone-dependent mouse mammary tumor development in both transgenic MMTV-RANK and WT mice and support additional studies to determine whether RANKL inhibition will delay tumor progression and metastases. Citation Information: Cancer Res 2009;69(2 Suppl):Abstract nr 4167.


Journal of bone oncology | 2015

RANK and RANK ligand expression in primary human osteosarcoma

Daniel Branstetter; Kathy Rohrbach; Li-Ya Huang; Rosalia Soriano; Mark E. Tometsko; Michelle Blake; Allison P. Jacob; William C. Dougall

Receptor activator of nuclear factor kappa-B ligand (RANKL) is an essential mediator of osteoclast formation, function and survival. In patients with solid tumor metastasis to the bone, targeting the bone microenvironment by inhibition of RANKL using denosumab, a fully human monoclonal antibody (mAb) specific to RANKL, has been demonstrated to prevent tumor-induced osteolysis and subsequent skeletal complications. Recently, a prominent functional role for the RANKL pathway has emerged in the primary bone tumor giant cell tumor of bone (GCTB). Expression of both RANKL and RANK is extremely high in GCTB tumors and denosumab treatment was associated with tumor regression and reduced tumor-associated bone lysis in GCTB patients. In order to address the potential role of the RANKL pathway in another primary bone tumor, this study assessed human RANKL and RANK expression in human primary osteosarcoma (OS) using specific mAbs, validated and optimized for immunohistochemistry (IHC) or flow cytometry. Our results demonstrate RANKL expression was observed in the tumor element in 68% of human OS using IHC. However, the staining intensity was relatively low and only 37% (29/79) of samples exhibited≥10% RANKL positive tumor cells. RANK expression was not observed in OS tumor cells. In contrast, RANK expression was clearly observed in other cells within OS samples, including the myeloid osteoclast precursor compartment, osteoclasts and in giant osteoclast cells. The intensity and frequency of RANKL and RANK staining in OS samples were substantially less than that observed in GCTB samples. The observation that RANKL is expressed in OS cells themselves suggests that these tumors may mediate an osteoclastic response, and anti-RANKL therapy may potentially be protective against bone pathologies in OS. However, the absence of RANK expression in primary human OS cells suggests that any autocrine RANKL/RANK signaling in human OS tumor cells is not operative, and anti-RANKL therapy would not directly affect the tumor.

Collaboration


Dive into the Daniel Branstetter's collaboration.

Researchain Logo
Decentralizing Knowledge