Network


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

Hotspot


Dive into the research topics where Erin M. Wolf Horrell is active.

Publication


Featured researches published by Erin M. Wolf Horrell.


Molecular Cell | 2014

PKA-Mediated Phosphorylation of ATR Promotes Recruitment of XPA to UV-Induced DNA Damage

Stuart G. Jarrett; Erin M. Wolf Horrell; Perry A. Christian; Jillian C. Vanover; Mary C. Boulanger; Yue Zou; John A. D’Orazio

The melanocortin 1 receptor (MC1R), which signals through cAMP, is a melanocytic transmembrane receptor involved in pigmentation, adaptive tanning, and melanoma resistance. We report MC1R-mediated or pharmacologically-induced cAMP signaling promotes nucleotide excision repair (NER) in a cAMP-dependent protein kinase A (PKA)-dependent manner. PKA directly phosphorylates ataxia telangiectasia and Rad3-related protein (ATR) at Ser435, which actively recruits the key NER protein xeroderma pigmentosum complementation group A (XPA) to sites of nuclear UV photodamage, accelerating clearance of UV-induced photolesions and reducing mutagenesis. Loss of Ser435 within ATR prevents PKA-mediated ATR phosphorylation, disrupts ATR-XPA binding, delays recruitment of XPA to UV-damaged DNA, and elevates UV-induced mutagenesis. This study mechanistically links cAMP-PKA signaling to NER and illustrates potential benefits of cAMP pharmacological rescue to reduce UV mutagenesis in MC1R-defective, melanoma-susceptible individuals.


Journal of Investigative Dermatology | 2015

Defining the Contribution of MC1R Physiological Ligands to ATR Phosphorylation at Ser435, a Predictor of DNA Repair in Melanocytes

Stuart G. Jarrett; Erin M. Wolf Horrell; Mary C. Boulanger; John A. D'Orazio

The melanocortin 1 receptor (MC1R), a GS-coupled receptor that signals through cAMP and PKA, regulates pigmentation, adaptive tanning, and melanoma resistance. MC1R-cAMP signaling promotes PKA-mediated phosphorylation of ataxia telangiectasia and rad3-related (ATR) at Ser435 (ATR-pS435), a modification that enhances nucleotide excision repair (NER) by facilitating recruitment of the XPA protein to sites of UV-induced DNA damage. High-throughput methods were developed to quantify ATR-pS435, measure XPA-photodamage interactions and assess NER function. We report that melanocyte stimulating hormone (α-MSH) or adrenocorticotropic hormone (ACTH) induce ATR-pS435, enhance XPA’s association with UV-damaged DNA and optimize melanocytic NER. In contrast, MC1R antagonists agouti signaling protein (ASIP) or human β-defensin 3 (HBD3) interfere with ATR-pS435 generation, impair the XPA-DNA interaction and reduce DNA repair. Although ASIP and HBD3 each blocked α-MSH-mediated induction of the signaling pathway, only ASIP depleted basal ATR-pS435. Our findings confirm that ASIP diminishes agonist-independent MC1R basal signaling whereas HBD3 is a neutral MC1R antagonist that blocks activation by melanocortins. Furthermore, our data suggest that ATR-pS435 may be a useful biomarker for the DNA repair-deficient MC1R phenotype.


The Journal of Physiology | 2015

Intrinsic muscle clock is necessary for musculoskeletal health

Elizabeth A. Schroder; Brianna D. Harfmann; Xiping Zhang; Ratchakrit Srikuea; Jonathan H. England; Brian A. Hodge; Yuan Wen; Lance A. Riley; Qi Yu; Alexander Christie; Jeffrey D. Smith; Tanya Seward; Erin M. Wolf Horrell; Jyothi Mula; Charlotte A. Peterson; Timothy A. Butterfield; Karyn A. Esser

The endogenous molecular clock in skeletal muscle is necessary for maintenance of phenotype and function. Loss of Bmal1 solely from adult skeletal muscle (iMSBmal1−/−) results in reductions in specific tension, increased oxidative fibre type and increased muscle fibrosis with no change in feeding or activity. Disruption of the molecular clock in adult skeletal muscle is sufficient to induce changes in skeletal muscle similar to those seen in the Bmal1 knockout mouse (Bmal1−/−), a model of advanced ageing. iMSBmal1−/− mice develop increased bone calcification and decreased joint collagen, which in combination with the functional changes in skeletal muscle results in altered gait. This study uncovers a fundamental role for the skeletal muscle clock in musculoskeletal homeostasis with potential implications for ageing.


Frontiers in Genetics | 2016

Melanocortin 1 Receptor: Structure, Function, and Regulation

Erin M. Wolf Horrell; Mary C. Boulanger; John A. D’Orazio

The melanocortin 1 receptor (MC1R) is a melanocytic Gs protein coupled receptor that regulates skin pigmentation, UV responses, and melanoma risk. It is a highly polymorphic gene, and loss of function correlates with a fair, UV-sensitive, and melanoma-prone phenotype due to defective epidermal melanization and sub-optimal DNA repair. MC1R signaling, achieved through adenylyl cyclase activation and generation of the second messenger cAMP, is hormonally controlled by the positive agonist melanocortin, the negative agonist agouti signaling protein, and the neutral antagonist β-defensin 3. Activation of cAMP signaling up-regulates melanin production and deposition in the epidermis which functions to limit UV penetration into the skin and enhances nucleotide excision repair (NER), the genomic stability pathway responsible for clearing UV photolesions from DNA to avoid mutagenesis. Herein we review MC1R structure and function and summarize our laboratory’s findings on the molecular mechanisms by which MC1R signaling impacts NER.


Redox biology | 2014

Neutral sphingomyelinase-3 mediates TNF-stimulated oxidant activity in skeletal muscle

Jennifer S. Moylan; Jeffrey D. Smith; Erin M. Wolf Horrell; Julie B. McLean; Gergana M. Deevska; Mark R. Bonnell; Mariana Nikolova-Karakashian; Michael B. Reid

Aims Sphingolipid and oxidant signaling affect glucose uptake, atrophy, and force production of skeletal muscle similarly and both are stimulated by tumor necrosis factor (TNF), suggesting a connection between systems. Sphingolipid signaling is initiated by neutral sphingomyelinase (nSMase), a family of agonist-activated effector enzymes. Northern blot analyses suggest that nSMase3 may be a striated muscle-specific nSMase. The present study tested the hypothesis that nSMase3 protein is expressed in skeletal muscle and functions to regulate TNF-stimulated oxidant production. Results We demonstrate constitutive nSMase activity in skeletal muscles of healthy mice and humans and in differentiated C2C12 myotubes. nSMase3 (Smpd4 gene) mRNA is highly expressed in muscle. An nSMase3 protein doublet (88 and 85 kD) is derived from alternative mRNA splicing of exon 11. The proteins partition differently. The full-length 88 kD isoform (nSMase3a) fractionates with membrane proteins that are resistant to detergent extraction; the 85 kD isoform lacking exon 11 (nSMase3b) is more readily extracted and fractionates with detergent soluble membrane proteins; neither variant is detected in the cytosol. By immunofluorescence microscopy, nSMase3 resides in both internal and sarcolemmal membranes. Finally, myotube nSMase activity and cytosolic oxidant activity are stimulated by TNF. Both if these responses are inhibited by nSMase3 knockdown. Innovation These findings identify nSMase3 as an intermediate that links TNF receptor activation, sphingolipid signaling, and skeletal muscle oxidant production. Conclusion Our data show that nSMase3 acts as a signaling nSMase in skeletal muscle that is essential for TNF-stimulated oxidant activity.


Nucleic Acids Research | 2016

AKAP12 mediates PKA-induced phosphorylation of ATR to enhance nucleotide excision repair

Stuart G. Jarrett; Erin M. Wolf Horrell; John A. D'Orazio

Loss-of-function in melanocortin 1 receptor (MC1R), a GS protein-coupled receptor that regulates signal transduction through cAMP and protein kinase A (PKA) in melanocytes, is a major inherited melanoma risk factor. Herein, we report a novel cAMP-mediated response for sensing and responding to UV-induced DNA damage regulated by A-kinase-anchoring protein 12 (AKAP12). AKAP12 is identified as a necessary participant in PKA-mediated phosphorylation of ataxia telangiectasia mutated and Rad3-related (ATR) at S435, a post-translational event required for cAMP-enhanced nucleotide excision repair (NER). Moreover, UV exposure promotes ATR-directed phosphorylation of AKAP12 at S732, which promotes nuclear translocation of AKAP12–ATR-pS435. This complex subsequently recruits XPA to UV DNA damage and enhances 5′ strand incision. Preventing AKAP12s interaction with PKA or with ATR abrogates ATR-pS435 accumulation, delays recruitment of XPA to UV-damaged DNA, impairs NER and increases UV-induced mutagenesis. Our results define a critical role for AKAP12 as an UV-inducible scaffold for PKA-mediated ATR phosphorylation, and identify a repair complex consisting of AKAP12–ATR-pS435-XPA at photodamage, which is essential for cAMP-enhanced NER.


Frontiers in Physiology | 2015

Proteomic analysis of media from lung cancer cells reveals role of 14-3-3 proteins in cachexia

Julie B. McLean; Jennifer S. Moylan; Erin M. Wolf Horrell; Francisco H. Andrade

Aims: At the time of diagnosis, 60% of lung cancer patients present with cachexia, a severe wasting syndrome that increases morbidity and mortality. Tumors secrete multiple factors that contribute to cachectic muscle wasting, and not all of these factors have been identified. We used Orbitrap electrospray ionization mass spectrometry to identify novel cachexia-inducing candidates in media conditioned with Lewis lung carcinoma cells (LCM). Results: One-hundred and 58 proteins were confirmed in three biological replicates. Thirty-three were identified as secreted proteins, including 14-3-3 proteins, which are highly conserved adaptor proteins known to have over 200 binding partners. We confirmed the presence of extracellular 14-3-3 proteins in LCM via western blot and discovered that LCM contained less 14-3-3 content than media conditioned with C2C12 myotubes. Using a neutralizing antibody, we depleted extracellular 14-3-3 proteins in myotube culture medium, which resulted in diminished myosin content. We identified the proposed receptor for 14-3-3 proteins, CD13, in differentiated C2C12 myotubes and found that inhibiting CD13 via Bestatin also resulted in diminished myosin content. Conclusions: Our novel findings show that extracellular 14-3-3 proteins may act as previously unidentified myokines and may signal via CD13 to help maintain muscle mass.


F1000Research | 2014

UV-independent induction of beta defensin 3 in neonatal human skin explants

Erin M. Wolf Horrell; John A. D'Orazio

In order to determine the effect of UV radiation on β-defensin 3 (BD3) expression in human skin, freshly-isolated UV-naïve skin was obtained from newborn male infants undergoing planned circumcision. Skin explants sustained ex vivo dermis side down on RPMI media were exposed to 0.5 kJ/m 2 UVB, and biopsies were taken from the explant through 72 hours after radiation. mRNA expression was measured by qRTPCR and normalized to TATA-binding protein. BD3 expression at each time point was compared with an untreated control taken at time 0 within each skin sample. Extensive variability in both the timing and magnitude of BD3 induction across individuals was noted and was not predicted by skin pigment phenotype, suggesting that BD3 induction was not influenced by epidermal melanization. However, a mock-irradiated time course demonstrated UV-independent BD3 mRNA increases across multiple donors which was not further augmented by treatment with UV radiation, suggesting that factors other than UV damage promoted increased BD3 expression in the skin explants. We conclude that BD3 expression is induced in a UV-independent manner in human skin explants processed and maintained in standard culture conditions, and that neonatal skin explants are an inappropriate model with which to study the effects of UV on BD3 induction in whole human skin.


Experimental Dermatology | 2017

Divergence of cAMP signaling pathways mediating augmented nucleotide excision repair and pigment induction in melanocytes.

Erin M. Wolf Horrell; Stuart G. Jarrett; Katharine M Carter; John A. D'Orazio

Loss‐of‐function melanocortin 1 receptor (MC1R) polymorphisms are common in UV‐sensitive fair‐skinned individuals and are associated with blunted cAMP second messenger signalling and higher lifetime risk of melanoma because of diminished ability of melanocytes to cope with UV damage. cAMP signalling positions melanocytes to resist UV injury by upregulating synthesis of UV‐blocking eumelanin pigment and by enhancing the repair of UV‐induced DNA damage. cAMP enhances melanocyte nucleotide excision repair (NER), the genome maintenance pathway responsible for the removal of mutagenic UV photolesions, through cAMP‐activated protein kinase (protein kinase A)‐mediated phosphorylation of the ataxia telangiectasia‐mutated and Rad3‐related (ATR) protein on the S435 residue. We investigated the interdependence of cAMP‐mediated melanin upregulation and cAMP‐enhanced DNA repair in primary human melanocytes and a melanoma cell line. We observed that the ATR‐dependent molecular pathway linking cAMP signalling to the NER pathway is independent of MITF activation. Similarly, cAMP‐mediated upregulation of pigment synthesis is independent of ATR, suggesting that the key molecular events driving MC1R‐mediated enhancement of genome maintenance (eg PKA‐mediated phosphorylation of ATR) and MC1R‐induced pigment induction (eg MITF activation) are distinct.


Archive | 2016

Melanocortin 1 Receptor (MC1R) as a Global Regulator of Cutaneous UV Responses: Molecular Interactions and Opportunities for Melanoma Prevention

Erin M. Wolf Horrell; John A. D’Orazio

UV radiation is a pervasive environmental agent that affects the skin in complex ways. It benefits human health by its contribution to the biosynthesis of vitamin D in the skin, however it also is a major carcinogen responsible for millions of skin cancers diagnosed each year. One of the most important physiologic responses recruited with UV exposure is the melanocortin signaling axis. This pathway, initiated by melanocortins such as melanocyte stimulating hormone (α-MSH) or adrenocorticotropic hormone (ACTH), is dependent on the signaling function of the melanocortin 1 receptor (MC1R), a Gs protein-coupled cell surface receptor found on melanocytes in the skin. MC1R mediates its downstream UV-protective responses through activation of adenylyl cyclase and production of the second messenger cAMP. In melanocytes, cAMP stimulation leads to improved survival and UV-defensive sequelae. Here, we review how MC1R signaling protects melanocytes from UV-induced malignant degeneration, focusing on recent insights into molecular links between MC1R signaling and the nucleotide excision repair (NER) genome maintenance pathway. Finally, we highlight how insights into the MC1R UV protective response may facilitate the development of rational melanoma-protective strategies.

Collaboration


Dive into the Erin M. Wolf Horrell'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
Top Co-Authors

Avatar

Yue Zou

East Tennessee State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge