Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
JAMA Dermatol ; 159(2): 192-197, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36598766

ABSTRACT

Importance: Cutaneous T-cell lymphoma (CTCL) is a group of rare, complex cutaneous malignant neoplasms associated with significant disease burden on patients and the health care system. Currently, the population of patients with CTCL admitted to the hospital remains largely uncharacterized and poorly understood. Objective: To characterize the clinical characteristics, course of hospitalization, and mortality outcomes of an inpatient CTCL cohort. Design, Setting, and Participants: This multicenter retrospective cohort study reviewed medical records for adult patients (age ≥18 years) with a CTCL diagnosis per National Comprehensive Cancer Network guidelines admitted for inpatient hospitalization at 5 US academic medical centers with inpatient dermatology consult services and CTCL clinics between August 2016 and August 2020. Main Outcomes and Measures: Patient demographics, clinical history and findings, hospitalization courses, and mortality outcomes. Results: A total of 79 hospitalized patients with CTCL were identified, including 52 (70.3%) men and 22 (29.7%) women, with a median (IQR) age at hospitalization of 62.9 (27-92) years. The majority of admitted patients with CTCL were White (65 patients [82.3%]), had disease classified as mycosis fungoides (48 patients [61.5%]), and had advanced-stage disease (≥IIB, 70 patients [89.7%]). Most hospitalizations were complicated by infection (45 patients [57.0%]) and required intravenous antibiotic therapy (45 patients [57.0%]). In-hospital mortality occurred in 6 patients (7.6%) and was associated with higher body mass index (36.5 vs 25.3), history of thromboembolic disease (50.0% vs 12.3%), and diagnosis of sepsis on admission (66.7% vs 20.5%). At 1-year postdischarge, 36 patients (49.3%) patients had died, and mortality was associated with history of solid organ cancers (27.8% vs 10.8%), wound care as the reason for dermatology consultation (58.3% vs 24.3%), and presence of large cell transformation (58.3% vs 22.9%). Conclusions and Relevance: The findings of this cohort study improve the understanding of hospitalized patients with CTCL and lend valuable insight into identifying factors associated with both in-hospital and long-term mortality outcomes. This refined understanding of the inpatient CTCL population provides a foundation for larger, more robust studies to identify causal risk factors associated with mortality, development of prognostic scoring systems to estimate the probability of hospital mortality. Overall, the findings may prompt physicians caring for patients with CTCL to implement preventive strategies to diminish hospitalization and improve clinical management across this unique disease spectrum.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Skin Neoplasms , Adult , Male , Humans , Female , Adolescent , Middle Aged , Aged , Aged, 80 and over , Cohort Studies , Retrospective Studies , Aftercare , Skin Neoplasms/diagnosis , Skin Neoplasms/epidemiology , Skin Neoplasms/therapy , Patient Discharge , Lymphoma, T-Cell, Cutaneous/diagnosis , Lymphoma, T-Cell, Cutaneous/epidemiology , Lymphoma, T-Cell, Cutaneous/therapy
3.
Wound Repair Regen ; 28(2): 164-176, 2020 03.
Article in English | MEDLINE | ID: mdl-31674093

ABSTRACT

Venous leg ulcers (VLU) represent a major clinical unmet need, impairing quality of life for millions worldwide. The bioengineered bilayered living cell construct (BLCC) is the only FDA-approved therapy demonstrating efficacy in healing chronic VLU, yet its in vivo mechanisms of action are not well understood. Previously, we reported a BLCC-mediated acute wounding response at the ulcer edge; in this study we elucidated the BLCC-specific effects on the epidermis-free ulcer bed. We conducted a randomized controlled clinical trial (ClinicalTrials.gov NCT01327937) enrolling 30 subjects with nonhealing VLUs, and performed genotyping, genomic profiling, and functional analysis on wound bed biopsies obtained at baseline and 1 week after treatment with BLCC plus compression or compression therapy (control). The VLU bed transcriptome featured processes of chronic inflammation and was strikingly enriched for fibrotic/fibrogenic pathways and gene networks. BLCC application decreased expression of profibrotic TGFß1 gene targets and increased levels of TGFß inhibitor decorin. Surprisingly, BLCC upregulated metallothioneins and fibroblast-derived MMP8 collagenase, and promoted endogenous release of MMP-activating zinc to stimulate antifibrotic remodeling, a novel mechanism of cutaneous wound healing. By activating a remodeling program in the quiescent VLU bed, BLCC application shifts nonhealing to healing phenotype. As VLU bed fibrosis correlates with poor clinical healing, findings from this study identify the chronic VLU as a fibrotic skin disease and are first to support the development and application of antifibrotic therapies as a successful treatment approach.


Subject(s)
Collagen/therapeutic use , Fibrosis/genetics , Inflammation/genetics , Skin, Artificial , Varicose Ulcer/therapy , Wound Healing/genetics , Adult , Aged , Aged, 80 and over , Compression Bandages , Decorin/genetics , Female , Gene Expression Profiling , Humans , Male , Matrix Metalloproteinase 8/genetics , Metallothionein/genetics , Middle Aged , Phenotype , Transforming Growth Factor beta1/genetics , Treatment Outcome , Varicose Ulcer/genetics , Zinc/metabolism
4.
JCI Insight ; 4(23)2019 12 05.
Article in English | MEDLINE | ID: mdl-31661463

ABSTRACT

Diabetic foot ulcers (DFUs) are a life-threatening disease that often results in lower limb amputations and a shortened life span. Current treatment options are limited and often not efficacious, raising the need for new therapies. To investigate the therapeutic potential of topical statins to restore healing in patients with DFUs, we performed next-generation sequencing on mevastatin-treated primary human keratinocytes. We found that mevastatin activated and modulated the EGF signaling to trigger an antiproliferative and promigratory phenotype, suggesting that statins may shift DFUs from a hyperproliferative phenotype to a promigratory phenotype in order to stimulate healing. Furthermore, mevastatin induced a migratory phenotype in primary human keratinocytes through EGF-mediated activation of Rac1, resulting in actin cytoskeletal reorganization and lamellipodia formation. Interestingly, the EGF receptor is downregulated in tissue biopsies from patients with DFUs. Mevastatin restored EGF signaling in DFUs through disruption of caveolae to promote keratinocyte migration, which was confirmed by caveolin-1 (Cav1) overexpression studies. We conclude that topical statins may have considerable therapeutic potential as a treatment option for patients with DFUs and offer an effective treatment for chronic wounds that can be rapidly translated to clinical use.


Subject(s)
Caveolin 1/metabolism , ErbB Receptors/metabolism , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Signal Transduction/drug effects , Wound Healing/drug effects , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Diabetic Foot , Disease Models, Animal , Female , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Phenotype , Skin/pathology , Swine , Wound Healing/physiology
5.
Mol Ther ; 27(11): 1992-2004, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31409528

ABSTRACT

Chronic wounds-including diabetic foot ulcers, venous leg ulcers, and pressure ulcers-represent a major health problem that demands an urgent solution and new therapies. Despite major burden to patients, health care professionals, and health care systems worldwide, there are no efficacious therapies approved for treatment of chronic wounds. One of the major obstacles in achieving wound closure in patients is the lack of epithelial migration. Here, we used multiple pre-clinical wound models to show that Caveolin-1 (Cav1) impedes healing and that targeting Cav1 accelerates wound closure. We found that Cav1 expression is significantly upregulated in wound edge biopsies of patients with non-healing wounds, confirming its healing-inhibitory role. Conversely, Cav1 was absent from the migrating epithelium and is downregulated in acutely healing wounds. Specifically, Cav1 interacted with membranous glucocorticoid receptor (mbGR) and epidermal growth factor receptor (EGFR) in a glucocorticoid-dependent manner to inhibit cutaneous healing. However, pharmacological disruption of caveolae by MßCD or CRISPR/Cas9-mediated Cav1 knockdown resulted in disruption of Cav1-mbGR and Cav1-EGFR complexes and promoted epithelialization and wound healing. Our data reveal a novel mechanism of inhibition of epithelialization and wound closure, providing a rationale for pharmacological targeting of Cav1 as potential therapy for patients with non-healing chronic wounds.


Subject(s)
Caveolin 1/genetics , Gene Expression Regulation/drug effects , Re-Epithelialization/genetics , Wound Healing/drug effects , Wound Healing/genetics , Caveolin 1/metabolism , Cell Movement , Diabetic Foot/drug therapy , Diabetic Foot/etiology , Diabetic Foot/metabolism , Diabetic Foot/pathology , ErbB Receptors/metabolism , Gene Expression , Glucocorticoids/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Protein Binding , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism , Skin/pathology
6.
Wound Repair Regen ; 27(4): 360-365, 2019 07.
Article in English | MEDLINE | ID: mdl-30920083

ABSTRACT

The wound environment is a fertile ground for biofilm forming pathogens. Once biofilms form within the wound, they can be very challenging to eradicate. The purpose of this study was to examine the effect of a gelling fiber dressing with silver using a well-established porcine wound biofilm model. Deep partial thickness wounds were inoculated with Pseudomonas aeruginosa ATCC 27312 and covered with a polyurethane film dressing to promote biofilm formation. Wounds were then divided into treatment groups: gelling fiber dressing with silver, gelling fiber dressing without silver, hydrofiber dressing with silver, benzethonium chloride and ethylenediaminetetraacetic acid and compared to untreated control. Microbiological, biofilm, and histological wound assessments were performed on days 3, 5, and 7 postinfection. Treatment with gelling fiber dressing with silver resulted in significant reduction of P. aeruginosa biofilm when compared to all other treatment groups on every assessment time point. In addition, gelling fiber dressing with silver treatment resulted in detachment of biofilm from the wound, while wounds treated with gelling fiber dressing with and without silver showed more granulation tissue formation on day 3. Our data show that a new gelling fiber dressing with silver was effective in reducing biofilm associated P. aeruginosa in vivo. This study may have important clinical implications especially for wounds heavily colonized with gram-negative biofilm-forming bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bandages, Hydrocolloid , Pseudomonas Infections/drug therapy , Silver/pharmacology , Wound Healing/drug effects , Wound Infection/microbiology , Animals , Bacterial Physiological Phenomena , Biofilms/drug effects , Disease Models, Animal , Female , Gels , Swine , Wound Healing/physiology , Wound Infection/drug therapy
8.
J Cell Physiol ; 233(8): 5503-5512, 2018 08.
Article in English | MEDLINE | ID: mdl-29271488

ABSTRACT

Fibrosis can develop in nearly any tissue leading to a wide range of chronic fibrotic diseases. However, current treatment options are limited. In this study, we utilized an established aged mouse model of bleomycin-induced lung fibrosis (BLM) to test our hypothesis that fibrosis may develop simultaneously in multiple organs by evaluating skin fibrosis and wound healing. Fibrosis was induced in lung in aged (18-22-month-old) C57BL/6 male mice by intratracheal BLM administration. Allogeneic adipose-derived mesenchymal stromal cells (ASCs) or saline were injected intravenously 24 hr after BLM administration. Full thickness 8-mm punch wounds were performed 7 days later to study potential systemic anti-fibrotic and wound healing effects of intravenously delivered ASCs. Mice developed lung and skin fibrosis as well as delayed wound closure. Moreover, we observed similar changes in the expression of known pro-fibrotic factors in both lung and skin wound tissue, including miR-199 and protein expression of its corresponding target, caveolin-1, as well as phosphorylation of protein kinase B. Importantly, ASC-treated mice exhibited attenuation of BLM-induced lung and skin fibrosis and accelerated wound healing, suggesting that ASCs may prime injured tissues and prevent end-organ fibrosis.


Subject(s)
Lung/cytology , Mesenchymal Stem Cells/cytology , Pulmonary Fibrosis/prevention & control , Skin Diseases/prevention & control , Skin/cytology , Wound Healing/physiology , Animals , Bleomycin/pharmacology , Caveolin 1/metabolism , Disease Models, Animal , Lung/drug effects , Lung/metabolism , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Skin/drug effects , Skin/metabolism , Skin Diseases/chemically induced , Skin Diseases/metabolism , Wound Healing/drug effects
9.
Wound Repair Regen ; 25(6): 1017-1026, 2017 11.
Article in English | MEDLINE | ID: mdl-29235208

ABSTRACT

The ex vivo human skin wound model is a widely accepted model to study wound epithelialization. Due to a lack of animal models that fully replicate human conditions, the ex vivo model is a valuable tool to study mechanisms of wound reepithelialization, as well as for preclinical testing of novel therapeutics. The current standard for assessment of wound healing in this model is histomorphometric analysis, which is labor intensive, time consuming, and requires multiple biological and technical replicates in addition to assessment of different time points. Optical coherence tomography (OCT) is an emerging noninvasive imaging technology originally developed for noninvasive retinal scans that avoids the deleterious effects of tissue processing. This study investigated OCT as a novel method for assessing reepithelialization in the human ex vivo wound model. Excisional ex vivo wounds were created, maintained at air-liquid interface, and healing progression was assessed at days 4 and 7 with OCT and histology. OCT provided adequate resolution to identify the epidermis, the papillary and reticular dermis, and importantly, migrating epithelium in the wound bed. We have deployed OCT as a noninvasive tool to produce, longitudinal "optical biopsies" of ex vivo human wound healing process, and we established an optimal quantification method of re-epithelialization based on en face OCT images of the total wound area. Pairwise statistical analysis of OCT and histology based quantifications for the rate of epithelialization have shown the feasibility and superiority of OCT technology for noninvasive monitoring of human wound epithelialization. Furthermore, we have utilized OCT to evaluate therapeutic potential of allogeneic adipose stem cells revealing their ability to promote reepithelialization in human ex vivo wounds. OCT technology is promising for its applications in wound healing and evaluation of novel therapeutics in both the laboratory and the clinical settings.


Subject(s)
Re-Epithelialization , Skin/diagnostic imaging , Wounds and Injuries/diagnostic imaging , Adult , Dermis/diagnostic imaging , Dermis/pathology , Epidermis/diagnostic imaging , Epidermis/pathology , Humans , Middle Aged , Skin/injuries , Skin/pathology , Stem Cell Transplantation , Subcutaneous Fat/cytology , Tomography, Optical Coherence , Wounds and Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...