Network


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

Hotspot


Dive into the research topics where Christopher Breuler is active.

Publication


Featured researches published by Christopher Breuler.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Inhibition of Hif1α prevents both trauma-induced and genetic heterotopic ossification

Shailesh Agarwal; Shawn Loder; Cameron Brownley; David Cholok; Laura Mangiavini; John Li; Christopher Breuler; Hsiao H. Sung; Shuli Li; Kavitha Ranganathan; Joshua Peterson; Ronald G. Tompkins; David N. Herndon; Wenzhong Xiao; Dolrudee Jumlongras; Björn Olsen; Thomas A. Davis; Yuji Mishina; Ernestina Schipani; Benjamin Levi

Significance Heterotopic ossification (HO) is a debilitating condition in which bone forms inappropriately within soft tissues. Two vastly different patient populations are at risk for developing HO: those with musculoskeletal trauma or severe burns and those with a genetic mutation in the bone morphogenetic protein receptor ACVR1 (Activin type 1 receptor). In this study, we demonstrate that both forms of HO share a common signaling pathway through hypoxia inducible factor-1α, and that pharmacologic inhibition or genetic knockout of this signaling pathway can mitigate and even abolish HO formation. These findings pave the way for pharmacologic inhibitors of hypoxia inducible factor-1α as therapeutic options for heterotopic ossification. Pathologic extraskeletal bone formation, or heterotopic ossification (HO), occurs following mechanical trauma, burns, orthopedic operations, and in patients with hyperactivating mutations of the type I bone morphogenetic protein receptor ACVR1 (Activin type 1 receptor). Extraskeletal bone forms through an endochondral process with a cartilage intermediary prompting the hypothesis that hypoxic signaling present during cartilage formation drives HO development and that HO precursor cells derive from a mesenchymal lineage as defined by Paired related homeobox 1 (Prx). Here we demonstrate that Hypoxia inducible factor-1α (Hif1α), a key mediator of cellular adaptation to hypoxia, is highly expressed and active in three separate mouse models: trauma-induced, genetic, and a hybrid model of genetic and trauma-induced HO. In each of these models, Hif1α expression coincides with the expression of master transcription factor of cartilage, Sox9 [(sex determining region Y)-box 9]. Pharmacologic inhibition of Hif1α using PX-478 or rapamycin significantly decreased or inhibited extraskeletal bone formation. Importantly, de novo soft-tissue HO was eliminated or significantly diminished in treated mice. Lineage-tracing mice demonstrate that cells forming HO belong to the Prx lineage. Burn/tenotomy performed in lineage-specific Hif1α knockout mice (Prx-Cre/Hif1αfl:fl) resulted in substantially decreased HO, and again lack of de novo soft-tissue HO. Genetic loss of Hif1α in mesenchymal cells marked by Prx-cre prevents the formation of the mesenchymal condensations as shown by routine histology and immunostaining for Sox9 and PDGFRα. Pharmacologic inhibition of Hif1α had a similar effect on mesenchymal condensation development. Our findings indicate that Hif1α represents a promising target to prevent and treat pathologic extraskeletal bone.


Stem Cells | 2017

Scleraxis‐Lineage Cells Contribute to Ectopic Bone Formation in Muscle and Tendon

Shailesh Agarwal; Shawn Loder; David Cholok; Joshua Peterson; John Li; Christopher Breuler; R. Cameron Brownley; Hsiao Hsin Sung; Michael T. Chung; Nobuhiro Kamiya; Shuli Li; Bin Zhao; Vesa Kaartinen; Thomas A. Davis; Ammar T. Qureshi; Ernestina Schipani; Yuji Mishina; Benjamin Levi

The pathologic development of heterotopic ossification (HO) is well described in patients with extensive trauma or with hyperactivating mutations of the bone morphogenetic protein (BMP) receptor ACVR1. However, identification of progenitor cells contributing to this process remains elusive. Here we show that connective tissue cells contribute to a substantial amount of HO anlagen caused by trauma using postnatal, tamoxifen‐inducible, scleraxis‐lineage restricted reporter mice (Scx‐creERT2/tdTomatofl/fl). When the scleraxis‐lineage is restricted specifically to adults prior to injury marked cells contribute to each stage of the developing HO anlagen and coexpress markers of endochondral ossification (Osterix, SOX9). Furthermore, these adult preinjury restricted cells coexpressed mesenchymal stem cell markers including PDGFRα, Sca1, and S100A4 in HO. When constitutively active ACVR1 (caACVR1) was expressed in scx‐cre cells in the absence of injury (Scx‐cre/caACVR1fl/fl), tendons and joints formed HO. Postnatal lineage‐restricted, tamoxifen‐inducible caACVR1 expression (Scx‐creERT2/caACVR1fl/fl) was sufficient to form HO after directed cardiotoxin‐induced muscle injury. These findings suggest that cells expressing scleraxis within muscle or tendon contribute to HO in the setting of both trauma or hyperactive BMP receptor (e.g., caACVR1) activity. Stem Cells 2017;35:705–710


Scientific Reports | 2016

Local and Circulating Endothelial Cells Undergo Endothelial to Mesenchymal Transition (EndMT) in Response to Musculoskeletal Injury

Shailesh Agarwal; Shawn Loder; David Cholok; Joshua Peterson; John Li; David Fireman; Christopher Breuler; Hsiao Sung Hsieh; Kavitha Ranganathan; Charles Hwang; James Drake; Shuli Li; Charles K. Chan; Michael T. Longaker; Benjamin Levi

Endothelial-to-mesenchymal transition (EndMT) has been implicated in a variety of aberrant wound healing conditions. However, unambiguous evidence of EndMT has been elusive due to limitations of in vitro experimental designs and animal models. In vitro experiments cannot account for the myriad ligands and cells which regulate differentiation, and in vivo tissue injury models may induce lineage-independent endothelial marker expression in mesenchymal cells. By using an inducible Cre model to mark mesenchymal cells (Scx-creERT/tdTomato + ) prior to injury, we demonstrate that musculoskeletal injury induces expression of CD31, VeCadherin, or Tie2 in mesenchymal cells. VeCadherin and Tie2 were expressed in non-endothelial cells (CD31−) present in marrow from uninjured adult mice, thereby limiting the specificity of these markers in inducible models (e.g. VeCadherin- or Tie2-creERT). However, cell transplantation assays confirmed that endothelial cells (ΔVeCadherin/CD31+/CD45−) isolated from uninjured hindlimb muscle tissue undergo in vivo EndMT when transplanted directly into the wound without intervening cell culture using PDGFRα, Osterix (OSX), SOX9, and Aggrecan (ACAN) as mesenchymal markers. These in vivo findings support EndMT in the presence of myriad ligands and cell types, using cell transplantation assays which can be applied for other pathologies implicated in EndMT including tissue fibrosis and atherosclerosis. Additionally, endothelial cell recruitment and trafficking are potential therapeutic targets to prevent EndMT.


PLOS ONE | 2015

Characterization of Heterotopic Ossification Using Radiographic Imaging: Evidence for a Paradigm Shift.

R. Cameron Brownley; Shailesh Agarwal; Shawn Loder; Oluwatobi Eboda; John Li; Joshua Peterson; Charles Hwang; Christopher Breuler; Vesa Kaartinen; Bin Zhou; Yuji Mishina; Benjamin Levi

Heterotopic ossification (HO) is the growth of extra-skeletal bone which occurs following trauma, burns, and in patients with genetic bone morphogenetic protein (BMP) receptor mutations. The clinical and laboratory evaluation of HO is dependent on radiographic imaging to identify and characterize these lesions. Here we show that despite its inadequacies, plain film radiography and single modality microCT continue to serve as a primary method of HO imaging in nearly 30% of published in vivo literature. Furthermore, we demonstrate that detailed microCT analysis is superior to plain film and single modality microCT radiography specifically in the evaluation of HO formed through three representative models due to its ability to 1) define structural relationships between growing extra-skeletal bone and normal, anatomic bone, 2) provide accurate quantification and growth rate based on volume of the space-occupying lesion, thereby facilitating assessments of therapeutic intervention, 3) identify HO at earlier times allowing for evaluation of early intervention, and 4) characterization of metrics of bone physiology including porosity, tissue mineral density, and cortical and trabecular volume. Examination of our trauma model using microCT demonstrated two separate areas of HO based on anatomic location and relationship with surrounding, normal bone structures. Additionally, microCT allows HO growth rate to be evaluated to characterize HO progression. Taken together, these data demonstrate the need for a paradigm shift in the evaluation of HO towards microCT as a standard tool for imaging.


JCI insight | 2016

mTOR inhibition and BMP signaling act synergistically to reduce muscle fibrosis and improve myofiber regeneration

Shailesh Agarwal; David Cholok; Shawn Loder; John Li; Christopher Breuler; Michael T. Chung; Hsiao Hsin Sung; Kavitha Ranganathan; Joe Habbouche; James Drake; Joshua Peterson; Caitlin Priest; Shuli Li; Yuji Mishina; Benjamin Levi

Muscle trauma is highly morbid due to intramuscular scarring, or fibrosis, and muscle atrophy. Studies have shown that bone morphogenetic proteins (BMPs) reduce muscle atrophy. However, increased BMP signaling at muscle injury sites causes heterotopic ossification, as seen in patients with fibrodysplasia ossificans progressiva (FOP), or patients with surgically placed BMP implants for bone healing. We use a genetic mouse model of hyperactive BMP signaling to show the development of intramuscular fibrosis surrounding areas of ectopic bone following muscle injury. Rapamycin, which we have previously shown to eliminate ectopic ossification in this model, also eliminates fibrosis without reducing osteogenic differentiation, suggesting clinical value for patients with FOP and with BMP implants. Finally, we use reporter mice to show that BMP signaling is positively associated with myofiber cross-sectional area. These findings underscore an approach in which 2 therapeutics (rapamycin and BMP ligand) can offset each other, leading to an improved outcome.


Bone | 2018

High-frequency spectral ultrasound imaging (SUSI) visualizes early post-traumatic heterotopic ossification (HO) in a mouse model

Kavitha Ranganathan; Xiaowei Hong; David Cholok; Joe Habbouche; Caitlin Priest; Christopher Breuler; Michael Chung; John Li; Arminder Kaura; Hsiao Hsin Sung Hsieh; Jonathan Butts; Serra Ucer; Ean Schwartz; Steven R. Buchman; Jan P. Stegemann; Cheri X. Deng; Benjamin Levi

PURPOSE Early treatment of heterotopic ossification (HO) is currently limited by delayed diagnosis due to limited visualization at early time points. In this study, we validate the use of spectral ultrasound imaging (SUSI) in an animal model to detect HO as early as one week after burn tenotomy. METHODS Concurrent SUSI, micro CT, and histology at 1, 2, 4, and 9weeks post-injury were used to follow the progression of HO after an Achilles tenotomy and 30% total body surface area burn (n=3-5 limbs per time point). To compare the use of SUSI in different types of injury models, mice (n=5 per group) underwent either burn/tenotomy or skin incision injury and were imaged using a 55MHz probe on VisualSonics VEVO 770 system at one week post injury to evaluate the ability of SUSI to distinguish between edema and HO. Average acoustic concentration (AAC) and average scatterer diameter (ASD) were calculated for each ultrasound image frame. Micro CT was used to calculate the total volume of HO. Histology was used to confirm bone formation. RESULTS Using SUSI, HO was visualized as early as 1week after injury. HO was visualized earliest by 4weeks after injury by micro CT. The average acoustic concentration of HO was 33% more than that of the control limb (n=5). Spectroscopic foci of HO present at 1week that persisted throughout all time points correlated with the HO present at 9weeks on micro CT imaging. CONCLUSION SUSI visualizes HO as early as one week after injury in an animal model. SUSI represents a new imaging modality with promise for early diagnosis of HO.


Plastic and reconstructive surgery. Global open | 2017

Abstract 46. Modulating TAK1 Signaling to Enhance Scaffold and Cell-Free Calvarial Healing

Shailesh Agarwal; Michael T. Chung; Shawn Loder; David Cholok; Hsiao Hsung Hsieh; Joseph Habbouche; Christopher Breuler; John Butts; Arminder Kaura; John Li; Kavitha Ranganathan; Serra Ucer; Caitlin Priest; Shuli Li; Jonathan Reimer; Yuji Mishina; Steven R. Buchman; Benjamin H. Levi

T ueday, M arch 8, 2017 METHODS: One hundred fifty-eight patients with 277 expanded skin cases during 2010 to 2014 were reviewed and photograph-evaluated for the expanded skin texture and regenerative condition. Overall texture of the expanded skin flaps (Good, Fair, Poor) were evaluated and documented by senior attending surgeons. The occurrence of five indications of skin regeneration limitation, including skin thickness, skin color, stretch mark, vessel varicose and skin lesion, during skin expansion were recorded. The correlation of indications to overall skin regeneration condition was statistically analyzed.


Plastic and reconstructive surgery. Global open | 2017

Abstract 96: Identification and Therapeutic Targeting of a Central DNA-Based Mechanism Through Which Movement Augments Inflammation

Shailesh Agarwal; Shawn Loder; David Cholok; Michael Chung; Arminder Kaura; John Li; Kavitha Ranganathan; Christopher Breuler; Joseph Habbouche; John Butts; Hsiao Hsieh Hsung; Shuli Li; Yuji Mishina; Benjamin H. Levi

RESULTS: Flow cytometry demonstrated that genetic loss of scleraxis among mesenchymal cells significantly reduced the presence of macrophages (F4/80+) and neutrophils (Cd11b+Ly6G+) at the injury site within 48 hours after injury. The presence of PDGFRa+ mesenchymal cells was also significantly reduced based on both flow cytometry and histologic analysis. These findings were confirmed with ciprofloxacin treatment. Furthermore, genetic loss of Scleraxis and ciprofloxacin both corresponded with a significant reduction in mesenchymal cell proliferation. Ciprofloxacin treatment led to reduced chondrogenic differentiation and aggrecan expression. Genetic loss of Scleraxis reduced ectopic cartilage formation when compared with wild type controls.


Plastic and reconstructive surgery. Global open | 2017

Abstract 2: Macrophage-specific TGF-B is a Targetable Cytokine to Prevent Heterotopic Ossification

David Cholok; Shailesh Agarwal; Shawn Loder; Michael Chung; Ramkumar Tiruvannamalai Annamalai; Joseph Habbouche; Caitlin Priest; Beau Carson; Christopher Breuler; Kavitha Ranganathan; John Li; John Butts; Arminder Kaura; Hsiao Hsieh Hsung; Shuli Li; Yuji Mishina; Benjamin H. Levi

PURPOSE: Lymphedema is a common, life-long complication of cancer treatment that currently has no cure. Patients with lymphedema have decreased quality of life and suffer recurrent infections, while current treatments are merely palliative and designed to prevent disease progression. Accumulating evidence indicates that T cells play a key role in the pathology of lymphedema by inhibiting lymphangiogenesis and promoting tissue fibrosis. Because the pathophysiology of lymphedema involves primarily the skin and subcutaneous tissues, it may be possible to target T cells locally using topical medications such as tacrolimus without inducing systemic immunosuppression. The purpose of this study was therefore to study the efficacy of topical tacrolimus for prevention and treatment of lymphedema using preclinical mouse models.


Plastic and reconstructive surgery. Global open | 2017

Abstract 95: Inhibition of Scleraxis Signaling Provides a Target to Reduce Mesenchymal Cell Inflammation During Wound Healing

Arminder Kaura; Michael Chung; David Cholok; Shawn Loder; Christopher Breuler; John Butts; Joseph Habbouche; John Li; Shuli Li; Shailesh Agarwal; Benjamin H. Levi

PURPOSE: Impaired wound healing is a serious complication and impacts patient’s quality of life adversely. Among extracellular matrix (ECM) molecules, hyaluronan (HA) has been suggested to play a critical role in all phases of wound healing as it modulates cell behavior including adhesion, migration, proliferation, metabolism and differentiation. HA is produced by 3 distinct hyaluronan synthesis (Has) and exists in varying sizes. It is widely accepted that large molecular weight HA is involved in structural and anti-inflammatory functions, whereas small molecular weight HA is angiogenetic and pro-inflammatory.

Collaboration


Dive into the Christopher Breuler's collaboration.

Top Co-Authors

Avatar

John Li

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shawn Loder

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge