Network


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

Hotspot


Dive into the research topics where Brian C. Wulff is active.

Publication


Featured researches published by Brian C. Wulff.


Cancer Research | 2007

Gender Differences in UVB-Induced Skin Carcinogenesis, Inflammation, and DNA Damage

Jennifer M. Thomas-Ahner; Brian C. Wulff; Kathleen L. Tober; Donna F. Kusewitt; Judy Riggenbach; Tatiana M. Oberyszyn

The American Cancer Society reports the incidence of squamous cell carcinoma in males to be thrice the incidence in females. This increased squamous cell carcinoma incidence has been attributed to men accumulating more sun exposure and using less sun protection than women. To date, there have been no controlled studies examining the effect of gender on skin tumor development following equal doses of UVB. Gender differences in UVB-induced skin carcinogenesis were examined using the Skh-1 mouse model. After chronic exposure to equal doses of UVB, male mice developed tumors earlier and had more tumors than female mice; tumors in male mice tended to be larger, and the total tumor burden was greater than in females. In addition, tumors in males were of more advanced histologic grade compared with those of female mice. To evaluate the contribution of differences in inflammation and DNA damage to differences in skin carcinogenesis, male and female Skh-1 mice were exposed once to 2,240 J/m(2) UVB and examined 48 h after exposure. Surprisingly, male mice developed less of an inflammatory response, as determined by skin fold thickness and myeloperoxidase activity, compared with females. Interestingly, male mice showed more cutaneous oxidative DNA damage than the females and lower antioxidant levels. These results show a gender bias in skin carcinogenesis and suggest that the gender difference in tumor development is more influenced by the extent of oxidative DNA damage and antioxidant capacities than by inflammatory response.


Journal of Investigative Dermatology | 2012

Mast Cells Contribute to Scar Formation during Fetal Wound Healing

Brian C. Wulff; Allison E. Parent; Melissa A. Meleski; Luisa A. DiPietro; Megan Schrementi; Traci A. Wilgus

Scar formation is a potentially detrimental process of tissue restoration in adults, affecting organ form and function. During fetal development, cutaneous wounds heal without inflammation or scarring at early stages of development, but begin to heal with significant inflammation and scarring as the skin becomes more mature. One possible cell type that could regulate the change from scarless to fibrotic healing is the mast cell. We show here that dermal mast cells in scarless wounds generated at embryonic day 15 (E15) are fewer in number, less mature and do not degranulate in response to wounding as effectively as mast cells of fibrotic wounds made at embryonic day 18 (E18). Differences were also observed between cultured mast cells from E15 and E18 skin with regard to degranulation and preformed cytokine levels. Injection of mast cell lysates into E15 wounds disrupted scarless healing, suggesting that mast cells interfere with scarless repair. Finally, wounds produced at E18, which normally heal with a scar, healed with significantly smaller scars in mast cell-deficient KitW/W-v mice compared to Kit+/+ littermates. Together, these data suggest that mast cells enhance scar formation, and that these cells may mediate the transition from scarless to fibrotic healing during fetal development.


Journal of Investigative Dermatology | 2008

Sirolimus reduces the incidence and progression of UVB-induced skin cancer in SKH mice even with co-administration of cyclosporine A.

Brian C. Wulff; Donna F. Kusewitt; Anne M. VanBuskirk; Jennifer M. Thomas-Ahner; Duncan F. Jason; Tatiana M. Oberyszyn

Transplant immunosuppressants have been implicated in the increased incidence of non-melanoma skin cancer in transplant recipients, most of whom harbor considerable UVB-induced DNA damage in their skin prior to transplantation. This study was designed to evaluate the effects of two commonly used immunosuppressive drugs, cyclosporine A (CsA) and sirolimus (SRL), on the development and progression of UVB-induced non-melanoma skin cancer. SKH-1 hairless mice were exposed to UVB alone for 15 weeks, and then were treated with CsA, SRL, or CsA+SRL for 9 weeks following cessation of UVB treatment. Compared with vehicle, CsA treatment resulted in enhanced tumor size and progression. In contrast, mice treated with SRL or CsA+SRL had decreased tumor multiplicity, size, and progression compared with vehicle-treated mice. CsA, but not SRL or combined treatment, increased dermal mast cell numbers and TGF-beta1 levels in the skin. These findings demonstrate that specific immunosuppressive agents differentially alter the cutaneous tumor microenvironment, which in turn may contribute to enhanced development of UVB-induced skin cancer in transplant recipients. Furthermore, these results suggest that CsA alone causes enhanced growth and progression of skin cancer, whereas co-administration of SRL with CsA causes the opposite effect. JID JOURNAL CLUB ARTICLE: For questions, answers, and open discussion about this article please go to http://network.nature.com/group/jidclub


American Journal of Transplantation | 2007

Clinically relevant immunosuppressants influence UVB-induced tumor size through effects on inflammation and angiogenesis.

F J. Duncan; Brian C. Wulff; Kathleen L. Tober; Amy K. Ferketich; Jason Martin; Jennifer M. Thomas-Ahner; Stephanie D. Allen; Donna F. Kusewitt; Tatiana M. Oberyszyn; Anne M. VanBuskirk

Immunosuppressive therapies allow long‐term patient and transplant survival, but are associated with increased development of UV‐induced skin cancers, particularly squamous cell carcinomas. The mechanisms by which CsA, MMF, tacrolimus (TAC) or sirolimus (SRL), alone or in dual combinations, influence tumor development and progression are not completely understood. In the current study, chronically UV‐exposed mice treated with SRL alone or in combination with CsA or TAC developed more tumors than mice treated with vehicle or other immunosuppressants, but the tumors were significantly smaller and less advanced. Mice treated with CsA or TAC developed significantly larger tumors than vehicle‐treated mice, and a larger percentage in the CsA group were malignant. The addition of MMF to CsA, but not to TAC, significantly reduced tumor size. Immunosuppressant effects on UVB‐induced inflammation and tumor angiogenesis may explain these findings. CsA enhanced both UVB‐induced inflammation and tumor blood vessel density, while MMF reduced inflammation. Addition of MMF to CsA reduced tumor size and vascularity. SRL did not affect inflammation, but significantly reduced tumor vascularity. Thus the choice of immunosuppressants has important implications for tumor number, size and progression, likely due to the influence of immunosuppressants on UVB‐induced inflammation and angiogenesis.


Experimental Dermatology | 2013

Mast cell activity in the healing wound: More than meets the eye?

Brian C. Wulff; Traci A. Wilgus

Mast cells (MCs) are an important part of the innate immune system and are abundant in barrier organs such as the skin. They are known primarily for initiating allergic reactions, but many other biological functions have now been described for these cells. Studies have indicated that during wound repair, MCs enhance acute inflammation, stimulate reepithelialization and angiogenesis, and promote scarring. MCs have also been linked to abnormal healing, with high numbers of MCs observed in chronic wounds, hypertrophic scars and keloids. Although MCs have gained attention in the wound healing field, several unique features of MCs have yet to be examined in the context of cutaneous repair. These include the ability of MCs to: (i) produce anti‐inflammatory mediators; (ii) release mediators without degranulating; and (iii) change their phenotype. Recent findings highlight the complexity of MCs and suggest that more information is needed to understand their complete range of activities during repair.


Photochemistry and Photobiology | 2008

Topical treatment with OGG1 enzyme affects UVB-induced skin carcinogenesis.

Brian C. Wulff; Jonathan Schick; Jennifer M. Thomas-Ahner; Donna F. Kusewitt; Daniel B. Yarosh; Tatiana M. Oberyszyn

Nonmelanoma skin cancer resulting from UVB exposure is a large and growing problem in the United States. Production of reactive oxygen species (ROS) during the UVB‐induced inflammatory response results in the formation of oxidative DNA adducts such as 8‐hydroxy‐2‐deoxyguanine (8‐oxo‐dG), which have been shown to contribute to the development of this cancer. The 8‐oxoguanine DNA glycosylase (OGG1) enzyme repairs 8‐oxo‐dG adducts, suggesting that enhancing its activity in the skin might increase 8‐oxo‐dG repair thus preventing skin cancer development. We therefore used the SKH‐1 murine model to examine the effect of topically applied OGG1 on UVB‐induced skin cancer development. Mice were exposed three times weekly to UVB followed immediately by topical treatment with a formulation of liposome‐encapsulated OGG1 enzyme for 25 weeks. While this treatment did not affect UVB‐induced tumor multiplicity, it did reduce tumor size and dramatically reduced tumor progression, as indicated by tumor grade. These results suggest that oxidative DNA damage contributes to the progression of UVB‐induced skin tumors and that a topical formulation containing OGG1, perhaps in conjunction with other DNA repair enzymes such as T4 endonuclease V, could be used in populations at high risk for skin cancer development.


Wound Repair and Regeneration | 2013

The alarmin HMGB-1 influences healing outcomes in fetal skin wounds

Adrienne Dardenne; Brian C. Wulff; Traci A. Wilgus

In mice, cutaneous wounds generated early in development (embryonic day 15, E15) heal scarlessly, while wounds generated late in gestation (embryonic day 18, E18) heal with scar formation. Even though both types of wounds are generated in the same sterile uterine environment, scarless fetal wounds heal without inflammation, but a strong inflammatory response is observed in scar‐forming fetal wounds. We hypothesized that altered release of alarmins, endogenous molecules that trigger inflammation in response to damage, may be responsible for the age‐related changes in inflammation and healing outcomes in fetal skin. The purpose of this study was to determine whether the alarmin high‐mobility group box‐1 (HMGB‐1) is involved in fetal wound repair. Immunohistochemical analysis showed that in unwounded skin, E18 keratinocytes expressed higher levels of HMGB‐1 compared with E15 keratinocytes. After injury, HMGB‐1 was released to a greater extent from keratinocytes at the margin of scar‐forming E18 wounds, compared with scarless E15 wounds. Furthermore, instead of healing scarlessly, E15 wounds healed with scars when treated with HMGB‐1. HMGB‐1‐injected wounds also had more fibroblasts, blood vessels, and macrophages compared with control wounds. Together, these data suggest that extracellular HMGB‐1 levels influence the quality of healing in cutaneous wounds.


Archives of Dermatological Research | 2013

Ultraviolet light exposure stimulates HMGB1 release by keratinocytes.

Kelly E. Johnson; Brian C. Wulff; Tatiana M. Oberyszyn; Traci A. Wilgus

The primary cause of non-melanoma skin cancer is ultraviolet (UV) light from the sun. Many studies have demonstrated that cutaneous inflammation resulting from UV exposure is important for the development of skin cancer. In fact, anti-inflammatory drugs have been shown to be effective in preventing skin cancer in animal models and in clinical trials. One new class of inflammatory mediators that could regulate UV-induced inflammation and skin carcinogenesis is alarmins. Alarmins are endogenous molecules that act as potent pro-inflammatory mediators when they are released by cells or accumulate extracellularly. The purpose of the current studies was to examine the expression and release of the alarmin high mobility group box 1 (HMGB1) after acute and chronic UV irradiation. Acute UV exposure stimulated the release of HMGB1 in cultured human keratinocytes and epidermal keratinocytes in murine skin. HMGB1 release correlated with pro-inflammatory cytokine production in vitro and inflammatory cell infiltration in vivo. HMGB1 was also examined in tumors arising in chronically irradiated murine skin. HMGB1 protein expression in low grade, benign papillomas was similar to adjacent skin. However, HMGB1 staining was more widespread with a higher number of HMGB1-positive cells observed in high grade papillomas and malignant tumors. Overall, the data suggest that HMGB1 may be an important regulator of UV-induced cutaneous inflammation and tumor formation. Additional studies are needed to assess whether targeting HMGB1 would be a useful strategy to prevent tumors from developing in response to chronic UV exposure.


International Journal of Cancer | 2010

Celecoxib reduces the effects of acute and chronic UVB exposure in mice treated with therapeutically relevant immunosuppressive drugs.

Brian C. Wulff; Jennifer M. Thomas-Ahner; Jonathan Schick; Tatiana M. Oberyszyn

Solid organ transplant recipients have a greatly increased risk for the development of non‐melanoma skin cancers. We have previously shown in our mouse model that sirolimus given in combination with cyclosporine A resulted in fewer and smaller tumors than cyclosporine A alone. In the current study, we tested the hypothesis that an anti‐inflammatory agent celecoxib applied topically after UVB exposure would further reduce UVB induced skin cancer in mice treated with cyclosporine A and sirolimus. The effect of celecoxib treatment on acute inflammation, initiation/promotion and tumor development was examined through a set of four experiments. Delayed tumor onset was observed in both tumor development experiments. Reduced tumor size and number compared to vehicle was observed when CX was administered concurrently with UVB and when CX was administered after cessation of UVB treatments, respectively. Prostaglandin E2 was confirmed to be significantly reduced in the dorsal skin of mice concurrently treated with immunosuppressants, CX and UVB for 13 weeks, suggesting a reduction in the inflammatory response could be the mechanism by which CX reduced tumorigenesis. Furthermore, topical celecoxib treatment following acute UVB exposure reduced dermal neutrophil number and activity compared to vehicle. In all of these experiments, unirradiated and vehicle treated mice were utilized as controls. In conclusion, these data suggest that even in the presence of cyclosporine A and sirolimus, topical celecoxib treatment can result in reduced inflammation, tumor number and size; properties which may be beneficial in the therapeutic reduction of skin cancer development in solid organ transplant recipients.


Wound Repair and Regeneration | 2013

Novel differences in the expression of inflammation-associated genes between mid- and late-gestational dermal fibroblasts.

Brian C. Wulff; Lianbo Yu; Allison E. Parent; Traci A. Wilgus

While cutaneous wounds of late‐gestational fetuses and on through adulthood result in scar formation, wounds incurred early in gestation have been shown to heal scarlessly. Unique properties of fetal fibroblasts are believed to mediate this scarless healing process. In this study, microarray analysis was used to identify differences in the gene expression profiles of cultured fibroblasts from embryonic day 15 (E15; midgestation) and embryonic day 18 (E18; late‐gestation) skin. Sixty‐two genes were differentially expressed and 12 of those genes are associated with inflammation, a process that correlates with scar formation in fetal wounds. One of the differentially expressed inflammatory genes was cyclooxygenase‐1 (COX‐1). COX‐1 was more highly expressed in E18 fibroblasts than in E15 fibroblasts, and these differences were confirmed at the gene and protein level. Differences in COX‐1 protein expression were also observed in fetal skin by immunohistochemical and immunofluorescence staining. The baseline differences in gene expression found in mid‐ and late‐gestational fetal fibroblasts suggest that developmental alterations in fibroblasts could be involved in the transition from scarless to fibrotic fetal wound healing. Furthermore, baseline differences in the expression of inflammatory genes by fibroblasts in E15 and E18 skin may contribute to inflammation and scar formation late in gestation.

Collaboration


Dive into the Brian C. Wulff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donna F. Kusewitt

University of Texas MD Anderson Cancer Center

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