Network


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

Hotspot


Dive into the research topics where Brett Shook is active.

Publication


Featured researches published by Brett Shook.


Adipocyte | 2014

Characterization of Cre recombinase models for the study of adipose tissue

Elise Jeffery; Ryan Berry; Christopher D. Church; Songtao Yu; Brett Shook; Valerie Horsley; Evan D. Rosen; Matthew S. Rodeheffer

The study of adipose tissue in vivo has been significantly advanced through the use of genetic mouse models. While the aP2-CreBI and aP2-CreSalk lines have been widely used to target adipose tissue, the specificity of these lines for adipocytes has recently been questioned. Here we characterize Cre recombinase activity in multiple cell populations of the major adipose tissue depots of these and other Cre lines using the membrane-Tomato/membrane-GFP (mT/mG) dual fluorescent reporter. We find that the aP2-CreBI and aP2-CreSalk lines lack specificity for adipocytes within adipose tissue, and that the aP2-CreBI line does not efficiently target adipocytes in white adipose depots. Alternatively, the Adiponectin-CreERT line shows high efficiency and specificity for adipocytes, while the PdgfRα-CreERUCL and PdgfRα-CreERJHU lines do not efficiently target adipocyte precursor cells in the major adipose depots. Instead, we show that the PdgfRα-Cre line is preferable for studies targeting adipocyte precursor cells in vivo.


Cell Stem Cell | 2016

Skin Adipocyte Stem Cell Self-Renewal Is Regulated by a PDGFA/AKT-Signaling Axis.

Guillermo C. Rivera-Gonzalez; Brett Shook; Johanna Andrae; Brandon Holtrup; Katherine Bollag; Christer Betsholtz; Matthew S. Rodeheffer; Valerie Horsley

Tissue growth and maintenance requires stem cell populations that self-renew, proliferate, and differentiate. Maintenance of white adipose tissue (WAT) requires the proliferation and differentiation of adipocyte stem cells (ASCs) to form postmitotic, lipid-filled mature adipocytes. Here we use the dynamic adipogenic program that occurs during hair growth to uncover an unrecognized regulator of ASC self-renewal and proliferation, PDGFA, which activates AKT signaling to drive and maintain the adipogenic program in the skin. Pdgfa expression is reduced in aged ASCs and is required for ASC proliferation and maintenance in the dermis, but not in other WATs. Our molecular and genetic studies uncover PI3K/AKT2 as a direct PDGFA target that is activated in ASCs during WAT hyperplasia and is functionally required for dermal ASC proliferation. Our data therefore reveal active mechanisms that regulate ASC self-renewal in the skin and show that distinct regulatory mechanisms operate in different WAT depots.


Cold Spring Harbor Perspectives in Medicine | 2014

Adipocytes in Skin Health and Disease

Guillermo C. Rivera-Gonzalez; Brett Shook; Valerie Horsley

Adipocytes are intimately associated with the dermal compartment of the skin, existing in a specialized dermal depot and displaying dynamic changes in size during tissue homeostasis. However, the roles of adipocytes in cutaneous biology and disease are not well understood. Traditionally, adipocytes within tissues were thought to act as reservoirs of energy, as thermal, or as structural support. In this review, we discuss recent studies revealing the cellular basis of the dynamic development and regenerative capacity of dermal adipocytes associated with the hair cycle and following injury. We discuss and speculate on potential roles of dermal adipocytes in cutaneous biology with an emphasis on communication during hair follicle growth and wound healing. Finally, we explore how alterations in the dermal adipose tissue may support clinical manifestations of cutaneous diseases such as lipodystrophy, obesity, and alopecia.


Journal of Investigative Dermatology | 2016

CD301b+ Macrophages Are Essential for Effective Skin Wound Healing

Brett Shook; Eric Xiao; Yosuke Kumamoto; Akiko Iwasaki; Valerie Horsley

Regeneration of skins barrier function after injury requires temporally coordinated cellular interactions between multiple cell types. Macrophages are essential inflammatory cells in skin wound regeneration. These cells switch their phenotype from inflammatory in the early regenerative stages to anti-inflammatory in the midstages of healing to coordinate skin repair. However, little is known about how different subsets of anti-inflammatory macrophages contribute to skin wound healing. Here, we characterize midstage macrophages (CD45(+)/CD11b(+)/F4-80(+)) and identify two major populations: CD206(+)/CD301b(+) and CD206(+)/CD301b(-). The numbers of CD206(+)/CD301b(+) macrophages increased concomitantly with repair, when the anti-inflammatory phenotype switch occurs in midstage healing. Using diphtheria toxin-mediated depletion models in mice, we show that selective depletion of midstage CD301b-expressing macrophages phenocopied wound healing defects observed in mice where multiple myeloid lineages are depleted. Additionally, when FACS-isolated subpopulations of myeloid cells were transplanted into 3-day wounds of syngeneic mice, only CD206(+)/CD301b(+) macrophages significantly increased proliferation and fibroblast repopulation. These data show that the CD301b-expressing subpopulation of macrophages is critical for activation of reparative processes during the midstage of cutaneous repair.


Nature Communications | 2018

Adipocyte hypertrophy and lipid dynamics underlie mammary gland remodeling after lactation

Rachel Zwick; Michael C. Rudolph; Brett Shook; Brandon Holtrup; Eve Roth; Vivian Lei; Alexandra Van Keymeulen; Victoria Seewaldt; Stephanie L. Kwei; John J. Wysolmerski; Matthew S. Rodeheffer; Valerie Horsley

Adipocytes undergo pronounced changes in size and behavior to support diverse tissue functions, but the mechanisms that control these changes are not well understood. Mammary gland-associated white adipose tissue (mgWAT) regresses in support of milk fat production during lactation and expands during the subsequent involution of milk-producing epithelial cells, providing one of the most marked physiological examples of adipose growth. We examined cellular mechanisms and functional implications of adipocyte and lipid dynamics in the mouse mammary gland (MG). Using in vivo analysis of adipocyte precursors and genetic tracing of mature adipocytes, we find mature adipocyte hypertrophy to be a primary mechanism of mgWAT expansion during involution. Lipid tracking and lipidomics demonstrate that adipocytes fill with epithelial-derived milk lipid. Furthermore, ablation of mgWAT during involution reveals an essential role for adipocytes in milk trafficking from, and proper restructuring of, the mammary epithelium. This work advances our understanding of MG remodeling and tissue-specific roles for adipocytes.During mammary gland involution, the organ undergoes extensive remodeling. Here, the authors explore the role of mammary gland adipose tissue (mgWAT) in this process and demonstrate that adipocyte hypertrophy and lipid trafficking underlie mgWAT expansion and epithelial regression.


Stem cell investigation | 2017

PDGFA regulation of dermal adipocyte stem cells

Guillermo C. Rivera-Gonzalez; Brett Shook; Valerie Horsley

Adipose tissue is widely studied for its central role in regulating systemic metabolism and contribution to obesity-related diseases; however, skin resident dermal white adipose tissue (dWAT) also contributes to many aspects of skin function. Skin-resident mature adipocytes are thought to prevent hair growth activation through the secretion of BMP molecules (1). Furthermore, following S. aureus infection, adipose tissue in the skin expands and produces an antibacterial peptide (2). During wound healing, defects in adipogenesis can abrogate fibroblast recruitment into wound beds, leading to defects in extracellular matrix deposition and tissue repair (3). Migration of adipocyte precursors into the wounded area might also contribute to the mesenchymal repopulation of the wounded site (4) and in wounds where hair follicle neogenesis is observed fully functional adipocyte precursors are present (5). These findings highlight the breadth of the role of adipose tissue in the skin.


Annual Review of Cell and Developmental Biology | 2016

The Role of Adipocytes in Tissue Regeneration and Stem Cell Niches

Brett Shook; Guillermo Rivera Gonzalez; Sarah Ebmeier; Gabriella Grisotti; Rachel Zwick; Valerie Horsley


Journal of Investigative Dermatology | 2017

934 CD301b+ macrophages activate proliferation of CD29+;CD34+;Sca1+ fibroblasts through multiple secreted factors during wound healing

Brett Shook; G. Rivera Gonzalez; R. Wasko; Valerie Horsley


Journal of Investigative Dermatology | 2017

661 Mechanisms that regulate adipocyte stem cell behavior in the skin

G. Rivera Gonzalez; Brett Shook; B. Holtrup; Gleb Baida; Irina Budunova; M. Rodeheffer; Valerie Horsley


Cell Metabolism | 2017

Forecasting Fat Fibrosis

Brett Shook; Matthew S. Rodeheffer

Collaboration


Dive into the Brett Shook'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge