Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
2.
JAMA Dermatol ; 158(4): 439-443, 2022 04 01.
Article En | MEDLINE | ID: mdl-35234827

IMPORTANCE: Epidermolysis bullosa simplex with muscular dystrophy (EBS-MD) is an autosomal recessive disorder caused by pathogenic variants in PLEC1, which encodes plectin. It is characterized by mild mucocutaneous fragility and blistering and muscle weakness. Translational readthrough-inducing drugs, such as repurposed aminoglycoside antibiotics, may represent a valuable therapeutic alternative for untreatable rare diseases caused by nonsense variants. OBJECTIVE: To evaluate whether systemic gentamicin, at a dose of 7.5 mg/kg/d for 14 consecutive days, is clinically beneficial in a patient with EBS-MD. DESIGN, SETTING, AND PARTICIPANTS: A single patient in Madrid, Spain, received 2 treatment courses with gentamicin on July 2019 and February 2020 with a follow-up period of 120 and 150 days, respectively. RESULTS: In this case report of a woman in her 30s with EBS-MD, before gentamicin treatment, the patient had mucocutaneous involvement, skeletal and respiratory muscle weakness, and myalgia that negatively affected her quality of life. Outcomes were evaluated with extensive laboratory tests and clinical scales. No nephrotoxic or ototoxic effects were detected after intravenous gentamicin administration. Gentamicin treatment was followed by plectin expression in the skin for at least 5 months. Although minimal changes were noted in skeletal muscle function (as measured by the Hammersmith functional motor scale and its expanded version: 6/40 to 7/40 and from 10/66 to 11/66, respectively) and respiratory musculature (maximal inspiratory and expiratory pressures D0 vs D16, MIP: 2.86 vs 3.63 KPa and MEP: 2.93 vs 4.63 KPa), myalgia disappeared (VAS dropped from 6 to 0), and quality of life improved (EuroQoL-5D-3L pain and anxiety dropped from 2 to 1). CONCLUSIONS AND RELEVANCE: The findings of this single case report suggest that gentamicin treatment may help suppress PLEC1 premature termination codons and induce plectin expression in EBS-MD primary keratinocytes and skin. Our study suggests that gentamicin may play an important role in treating EBS-MD owing to nonsense variants.


Epidermolysis Bullosa Simplex , Muscular Dystrophies , Epidermolysis Bullosa Simplex/complications , Epidermolysis Bullosa Simplex/drug therapy , Epidermolysis Bullosa Simplex/genetics , Female , Gentamicins/therapeutic use , Humans , Muscular Dystrophies/complications , Muscular Dystrophies/diagnosis , Muscular Dystrophies/drug therapy , Muscular Dystrophies, Limb-Girdle , Myalgia , Plectin/genetics , Quality of Life
3.
J Invest Dermatol ; 142(8): 2238-2248.e8, 2022 08.
Article En | MEDLINE | ID: mdl-34979109

Chronic wounds represent a major health problem worldwide. Some of the available therapies based on recombinant proteins usually fail owing to the hostile environment found at the wound bed. Aptamers appear as an attractive alternative to recombinant factors owing in part to their stability, sensitivity, specificity, and low-cost production. In this study, the Cell-Systematic Evolution of Ligands by EXponential Enrichment technology was employed to generate aptamers that specifically recognize and modulate the function of the FPR2, a receptor expressed in a variety of cells involved in wound repair. Three aptamers were obtained that specifically bound to FPR2 stable transfectants generated in HaCaT cells. The targeted aptamers were shown to act as FPR2 agonists in different in vitro functional assays, including wound healing assays, and elicited a similar pattern of response to that obtained with other known FPR2 peptide agonists, such as the human LL37 cathelicidin. We have also obtained in vivo evidence for the prohealing activities of one of these FPR2 aptamers in a skin-humanized mouse model developed by us, previously shown to accurately recreate the main phases of physiological human wound repair process. In conclusion, we provide evidence of the potential therapeutic value of FPR2 aptamers for cutaneous repair.


Aptamers, Nucleotide , Receptors, Formyl Peptide , Animals , Humans , Ligands , Mice , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/agonists , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Wound Healing
4.
J Hepatol ; 76(3): 694-725, 2022 03.
Article En | MEDLINE | ID: mdl-34715263

As one of the most metabolically complex systems in the body, the liver ensures multi-organ homeostasis and ultimately sustains life. Nevertheless, during early postnatal development, the liver is highly immature and takes about 2 years to acquire and develop almost all of its functions. Different events occurring at the environmental and cellular levels are thought to mediate hepatic maturation and function postnatally. The crosstalk between the liver, the gut and its microbiome has been well appreciated in the context of liver disease, but recent evidence suggests that the latter could also be critical for hepatic function under physiological conditions. The gut-liver crosstalk is thought to be mediated by a rich repertoire of microbial metabolites that can participate in a myriad of biological processes in hepatic sinusoids, from energy metabolism to tissue regeneration. Studies on germ-free animals have revealed the gut microbiome as a critical contributor in early hepatic programming, and this influence extends throughout life, mediating liver function and body homeostasis. In this seminar, we describe the microbial molecules that have a known effect on the liver and discuss how the gut microbiome and the liver evolve throughout life. We also provide insights on current and future strategies to target the gut microbiome in the context of hepatology research.


Gastrointestinal Microbiome/physiology , Liver Function Tests/statistics & numerical data , Liver/growth & development , Homeostasis/immunology , Homeostasis/physiology , Humans , Liver/physiology , Liver Function Tests/methods
5.
Front Med (Lausanne) ; 7: 576558, 2020.
Article En | MEDLINE | ID: mdl-33324660

Recessive dystrophic epidermolysis bullosa (RDEB) is an incurable inherited mucocutaneous fragility disorder characterized by recurrent blisters, erosions, and wounds. Continuous blistering triggers overlapping cycles of never-ending healing and scarring commonly evolving to chronic systemic inflammation and fibrosis. The systemic treatment with allogeneic mesenchymal cells (MSC) from bone marrow has previously shown benefits in RDEB. MSC from adipose tissue (ADMSC) are easier to isolate. This is the first report on the use of systemic allogeneic ADMSC, correlating the clinical, inflammatory, and immunologic outcomes in RDEB indicating long-lasting benefits. We present the case of an RDEB patient harboring heterozygous biallelic COL7A1 gene mutations and with a diminished expression of C7. The patient presented with long-lasting refractory and painful oral ulcers distressing her quality of life. Histamine receptor antagonists, opioid analgesics, proton-pump inhibitors, and low-dose tricyclic antidepressants barely improved gastric symptoms, pain, and pruritus. Concomitantly, allogeneic ADMSC were provided as three separate intravenous injections of 106 cells/kg every 21 days. ADMSC treatment was well-tolerated. Improvements in wound healing, itch, pain and quality of life were observed, maximally at 6-9 months post-treatment, with the relief of symptoms still noticeable for up to 2 years. Remarkably, significant modifications in PBL participating in both the innate and adaptive responses, alongside regulation of levels of profibrotic factors, MCP-1/CCL2 and TGF-ß, correlated with the health improvement. This treatment might represent an alternative for non-responding patients to conventional management. It seems critical to elucidate the paracrine modulation of the immune system by MSC for their rational use in regenerative/immunoregulatory therapies.

6.
Regen Med ; 15(9): 2053-2065, 2020 09.
Article En | MEDLINE | ID: mdl-33245008

Venous leg ulcers (VLU) represent an uphill economic, health and social burden, aggravated in the elderly. Best-practice care interventions are often insufficient and alternative therapies need to be explored. Herein, we have treated for the first time a chronic VLU in an elderly patient by combining cell therapy and tissue engineering in the context of a compassionate use. The administration of allogeneic adipose-derived mesenchymal stromal cells (MSCs) embedded in a plasma-based bioengineered dermis covering the ulcer bed and also injected into the ulcer margins led to the complete closure of a 10-year recalcitrant VLU in an 85-year-old patient. Regenerative properties of MSCs might be boosted by the use of bioengineered matrices for their delivery.


Leg Ulcer , Mesenchymal Stem Cells , Varicose Ulcer , Adipose Tissue , Aged , Aged, 80 and over , Humans , Leg Ulcer/therapy , Tissue Engineering
7.
Cells ; 9(9)2020 09 16.
Article En | MEDLINE | ID: mdl-32947957

Recessive dystrophic epidermolysis bullosa (RDEB) is a severe skin disease caused by mutation of the COL7A1 gene. RDEB is associated with high levels of TGF-ß1, which is likely to be involved in the fibrosis that develops in this disease. Endoglin (CD105) is a type III coreceptor for TGF-ß1 and its overexpression in fibroblasts deregulates physiological Smad/Alk1/Alk5 signalling, repressing the synthesis of TGF-ß1 and extracellular matrix (ECM) proteins. Raloxifene is a specific estrogen receptor modulator designated as an orphan drug for hereditary hemorrhagic telangiectasia, a rare vascular disease. Raloxifene stimulates endoglin synthesis, which could attenuate fibrosis. By contrast, the antioxidant N-acetylcysteine may have therapeutic value to rectify inflammation, fibrosis and endothelial dysfunction. Thus, we present here a repurposing strategy based on the molecular and functional screening of fibroblasts from RDEB patients with these drugs, leading us to propose the repositioning of these two well-known drugs currently in clinical use, raloxifene and N-acetylcysteine, to counteract fibrosis and inflammation in RDEB. Both compounds modulate the profibrotic events that may ultimately be responsible for the clinical manifestations in RDEB, suggesting that these findings may also be relevant for other diseases in which fibrosis is an important pathophysiological event.


Acetylcysteine/pharmacology , Drug Repositioning , Epidermolysis Bullosa/genetics , Fibroblasts/drug effects , Raloxifene Hydrochloride/pharmacology , Transforming Growth Factor beta1/genetics , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Antioxidants/pharmacology , Case-Control Studies , Collagen Type VII/genetics , Collagen Type VII/metabolism , Endoglin/genetics , Endoglin/metabolism , Epidermolysis Bullosa/metabolism , Epidermolysis Bullosa/pathology , Estrogen Antagonists/pharmacology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Gene Expression Regulation , Humans , Inheritance Patterns , Primary Cell Culture , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Severity of Illness Index , Signal Transduction , Skin/drug effects , Skin/metabolism , Skin/pathology , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism
8.
Genes (Basel) ; 12(1)2020 12 31.
Article En | MEDLINE | ID: mdl-33396192

Defective healing leading to cutaneous ulcer formation is one of the most feared complications of diabetes due to its consequences on patients' quality of life and on the healthcare system. A more in-depth analysis of the underlying molecular pathophysiology is required to develop effective healing-promoting therapies for those patients. Major architectural and functional differences with human epidermis limit extrapolation of results coming from rodents and other small mammal-healing models. Therefore, the search for reliable humanized models has become mandatory. Previously, we developed a diabetes-induced delayed humanized wound healing model that faithfully recapitulated the major histological features of such skin repair-deficient condition. Herein, we present the results of a transcriptomic and functional enrichment analysis followed by a mechanistic analysis performed in such humanized wound healing model. The deregulation of genes implicated in functions such as angiogenesis, apoptosis, and inflammatory signaling processes were evidenced, confirming published data in diabetic patients that in fact might also underlie some of the histological features previously reported in the delayed skin-humanized healing model. Altogether, these molecular findings support the utility of such preclinical model as a valuable tool to gain insight into the molecular basis of the delayed diabetic healing with potential impact in the translational medicine field.


Diabetes Mellitus, Experimental/genetics , Metabolic Networks and Pathways/genetics , Skin Ulcer/genetics , Transcriptome , Wound Healing/genetics , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Humans , Mice , Mice, Nude , Microarray Analysis , Molecular Sequence Annotation , Principal Component Analysis , Signal Transduction , Skin/metabolism , Skin/pathology , Skin Transplantation , Skin Ulcer/chemically induced , Skin Ulcer/metabolism , Skin Ulcer/pathology , Streptozocin/administration & dosage , Tissue Engineering/methods , Transplantation, Heterologous
10.
J Invest Dermatol ; 135(12): 3133-3143, 2015 Dec.
Article En | MEDLINE | ID: mdl-26288350

Here we describe a spontaneous mutation in the Zdhhc13 (zinc finger, DHHC domain containing 13) gene (also called Hip14l), one of 24 genes encoding palmitoyl acyltransferase (PAT) enzymes in the mouse. This mutation (Zdhhc13luc) was identified as a nonsense base substitution, which results in a premature stop codon that generates a truncated form of the ZDHHC13 protein, representing a potential loss-of-function allele. Homozygous Zdhhc13luc/Zdhhc13luc mice developed generalized hypotrichosis, associated with abnormal hair cycle, epidermal and sebaceous gland hyperplasia, hyperkeratosis, and increased epidermal thickness. Increased keratinocyte proliferation and accelerated transit from basal to more differentiated layers were observed in mutant compared with wild-type (WT) epidermis in untreated skin and after short-term 12-O-tetradecanoyl-phorbol-13-acetate treatment and acute UVB exposure. Interestingly, this epidermal phenotype was associated with constitutive activation of NF-κB (RelA) and increased neutrophil recruitment and elastase activity. Furthermore, tumor multiplicity and malignant progression of papillomas after chemical skin carcinogenesis were significantly higher in mutant mice than WT littermates. To our knowledge, this is the first report of a protective role for PAT in skin carcinogenesis.


Acyltransferases/genetics , Genetic Predisposition to Disease , Mutation , Skin Neoplasms/genetics , Animals , Bromodeoxyuridine/metabolism , Codon, Terminator , Epidermal Cells , Keratinocytes/physiology , Leukocyte Elastase/metabolism , Mice , NF-kappa B/physiology , NIH 3T3 Cells , Neutrophil Infiltration , Phenotype , Skin Neoplasms/etiology
11.
Oncotarget ; 5(6): 1439-51, 2014 Mar 30.
Article En | MEDLINE | ID: mdl-24742402

Germline mutations in CDKN2A and/or red hair color variants in MC1R genes are associated with an increased susceptibility to develop cutaneous melanoma or non melanoma skin cancer. We studied the impact of the CDKN2A germinal mutation p.G101W and MC1R variants on gene expression and transcription profiles associated with skin cancer. To this end we set-up primary skin cell co-cultures from siblings of melanoma prone-families that were later analyzed using the expression array approach. As a result, we found that 1535 transcripts were deregulated in CDKN2A mutated cells, with over-expression of immunity-related genes (HLA-DPB1, CLEC2B, IFI44, IFI44L, IFI27, IFIT1, IFIT2, SP110 and IFNK) and down-regulation of genes playing a role in the Notch signaling pathway. 3570 transcripts were deregulated in MC1R variant carriers. In particular, genes related to oxidative stress and DNA damage pathways were up-regulated as well as genes associated with neurodegenerative diseases such as Parkinson's, Alzheimer and Huntington. Finally, we observed that the expression signatures indentified in phenotypically normal cells carrying CDKN2A mutations or MC1R variants are maintained in skin cancer tumors (melanoma and squamous cell carcinoma). These results indicate that transcriptome deregulation represents an early event critical for skin cancer development.


Biomarkers, Tumor/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genetic Predisposition to Disease , Melanoma/genetics , Mutation/genetics , Receptor, Melanocortin, Type 1/genetics , Skin Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Coculture Techniques , Gene Expression Profiling , Genotype , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Melanoma/pathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/pathology , Melanoma, Cutaneous Malignant
13.
Exp Dermatol ; 22(3): 195-201, 2013 Mar.
Article En | MEDLINE | ID: mdl-23489422

Cutaneous diabetic wounds greatly affect the quality of life of patients, causing a substantial economic impact on the healthcare system. The limited clinical success of conventional treatments is mainly attributed to the lack of knowledge of the pathogenic mechanisms related to chronic ulceration. Therefore, management of diabetic ulcers remains a challenging clinical issue. Within this context, reliable animal models that recapitulate situations of impaired wound healing have become essential. In this study, we established a new in vivo humanised model of delayed wound healing in a diabetic context that reproduces the main features of the human disease. Diabetes was induced by multiple low doses of streptozotocin in bioengineered human-skin-engrafted immunodeficient mice. The significant delay in wound closure exhibited in diabetic wounds was mainly attributed to alterations in the granulation tissue formation and resolution, involving defects in wound bed maturation, vascularisation, inflammatory response and collagen deposition. In the new model, a cell-based wound therapy consisting of the application of plasma-derived fibrin dermal scaffolds containing fibroblasts consistently improved the healing response by triggering granulation tissue maturation and further providing a suitable matrix for migrating keratinocytes during wound re-epithelialisation. The present preclinical wound healing model was able to shed light on the biological processes responsible for the improvement achieved, and these findings can be extended for designing new therapeutic approaches with clinical relevance.


Diabetes Mellitus, Experimental/physiopathology , Disease Models, Animal , Fibroblasts/physiology , Regeneration/physiology , Skin Physiological Phenomena , Wound Healing/physiology , Animals , Bioengineering/methods , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Female , Fibroblasts/cytology , Humans , Mice , Mice, Nude , Streptozocin/adverse effects , Time Factors , Tissue Scaffolds , Transplantation, Heterologous
14.
J Invest Dermatol ; 128(6): 1565-75, 2008 Jun.
Article En | MEDLINE | ID: mdl-18185534

Using a recently described skin-humanized model based on the engraftment of human bioengineered skin equivalents onto immunodeficient mice, we compared the efficacy of different in vivo gene transfer strategies aimed at delivering growth factors to promote skin wound healing. The approaches involving transient delivery of keratinocyte growth factor (KGF) to wounds performed in the engrafted human skin included (1) KGF gene transfer by intradermal adenoviral injection; (2) KGF gene transfer by adenoviral vector immobilized in a fibrin carrier; and (3) KGF-adenoviral gene-transferred human fibroblasts embedded in a fibrin matrix. All delivery systems achieved KGF protein overproduction at the wound site, with a concomitant re-epithelialization enhancement. However, although direct gene delivery strategies exhibited variability in terms of the number of successfully transduced humanized mice, the use of genetically modified fibroblast-containing matrix as an in situ protein bioreactor was highly reproducible, leading to a significant improvement of the overall healing process. This latter approach appeared to be the most reliable means to deliver growth factors to wounds and also avoided the potential danger of scoring cases of faulty administration as therapeutic failures and direct exposure to viral vectors. The combined use of cell and gene therapy appears a robust tool to aid healing in a clinical context.


Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Gene Transfer Techniques , Genetic Vectors , Skin/pathology , Wound Healing , Wounds and Injuries/therapy , Adenoviridae/genetics , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Keratinocytes/cytology , Mice , Mice, Nude , Retroviridae/genetics
15.
Mol Carcinog ; 46(8): 741-5, 2007 Aug.
Article En | MEDLINE | ID: mdl-17610222

Skin tissue engineering emerged as an experimental regenerative therapy motivated primarily by the critical need for early permanent coverage of extensive burn injuries in patients with insufficient sources of autologous skin for grafting. With time, the approach evolved toward a wider range of applications including disease modeling. We have established a skin-humanized mouse model system consisting in bioengineered human-skin-engrafted immunodeficient mice. This new model allows to performing regenerative medicine, gene therapy, genomics, and pathology studies in a human context on homogeneous samples. Starting from skin cells (keratinocytes and fibroblasts) isolated from normal donor skin or patient's biopsies, we have been able to deconstruct-reconstruct several inherited skin disorders including genodermatoses and cancer-prone diseases in a large number of skin humanized mice. In addition, the model allows conducting studies in normal human skin to gain further insight into physiological processes such as wound healing or UV-responses.


Epidermis/injuries , Skin Diseases/pathology , Skin, Artificial , Tissue Engineering , Wound Healing/physiology , Animals , Disease Models, Animal , Epidermal Cells , Epidermis/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Regeneration
...