Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Dermatol ; 31(9): 1431-1442, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35620886

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) patients develop poorly healing skin wounds that are frequently colonized with microbiota. Because T cells play an important role in clearing such pathogens, we aimed to define the status of adaptive T cell-mediated immunity in RDEB wounds. Using a non-invasive approach for sampling of wound-associated constituents, we evaluated microbial contaminants in cellular fraction and exudates obtained from RDED wounds. Infectivity and intracellular trafficking of inactivated Staphylococcus aureus was accessed in RDEB keratinocytes. S. aureus and microbial antigen-specific activation of RDEB wound-derived T cells were investigated by fluorescence-activated cell sorting-based immune-phenotyping and T-cell functional assays. We found that RDEB wounds and epithelial cells are most frequently infected with Staphylococcus sp. and Pseudomonas sp. and that S. aureus essentially infects more RDEB keratinocytes and RDEB-derived squamous cell carcinoma cells than keratinocytes from healthy donors. The RDEB wound-associated T cells contain populations of CD4+ and CD8+ peripheral memory T cells that respond to soluble microbial antigens by proliferating and secreting interferon gamma (IFNγ). Moreover, CD8+ cytotoxic T lymphocytes recognize S. aureus-infected RDEB keratinocytes and respond by producing interleukin-2 (IL-2) and IFNγ and degranulating and cytotoxically killing infected cells. Prolonged exposure of RDEB-derived T cells to microbial antigens in vitro does not trigger PD-1-mediated T-cell exhaustion but induces differentiation of the CD4high population into CD4high CD25+ FoxP3+ regulatory T cells. Our data demonstrated that adaptive T cell-mediated immunity could clear infected cells from wound sites, but these effects might be inhibited by PD-1/Treg-mediated immuno-suppression in RDEB.


Asunto(s)
Infecciones Bacterianas , Epidermólisis Ampollosa Distrófica , Linfocitos T , Antígenos , Colágeno Tipo VII , Epidermólisis Ampollosa Distrófica/patología , Humanos , Queratinocitos/patología , Activación de Linfocitos , Receptor de Muerte Celular Programada 1 , Staphylococcus aureus , Linfocitos T/inmunología
2.
Exp Dermatol ; 30(12): 1724-1733, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34142388

RESUMEN

Hereditary epidermolysis bullosa (EB) is a mechanobullous skin fragility disorder characterized by defective epithelial adhesion, leading to mechanical stress-induced skin blistering. Based on the level of tissue separation within the dermal-epidermal junction, EB is categorized into simplex (EBS), junctional (JEB), dystrophic (DEB) and Kindler syndrome. There is no cure for EB, and painful chronic cutaneous wounds are one of the major complications in recessive (RDEB) patients. Although RDEB is considered a cutaneous disease, recent data support the underlying systemic immunological defects. Furthermore, chronic wounds are often colonized with pathogenic microbiota, leading to excessive inflammation and altered wound healing. Consequently, patients with RDEB suffer from a painful sensation of chronic, cutaneous itching/burning and an endless battle with bacterial infections. To improve their quality of life and life expectancy, it is important to prevent cutaneous infections, dampen chronic inflammation and stimulate wound healing. A clear scientific understanding of the immunological events underlying the maintenance of chronic poorly healing wounds in RDEB patients is necessary to improve disease management and better understand other wound healing disorders. In this review, we summarize current knowledge of the role of professional phagocytes, such as neutrophils, macrophages and dendritic cells, the role of T-cell-mediated immunity in lymphoid organs, and the association of microbiota with poor wound healing in RDEB. We conclude that RDEB patients have an underlying immunity defect that seems to affect antibacterial immunity.


Asunto(s)
Epidermólisis Ampollosa Distrófica/fisiopatología , Piel/patología , Cicatrización de Heridas , Epidermólisis Ampollosa Distrófica/inmunología , Humanos
3.
Exp Dermatol ; 30(10): 1428-1439, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33179358

RESUMEN

Pathogenic invasion of Staphylococcus aureus is a major concern in patients with chronic skin diseases like atopic dermatitis (AD), epidermolysis bullosa (EB), or chronic diabetic foot and venous leg ulcers, and can result in persistent and life-threatening chronic non-healing wounds. Staphylococcus aureus is generally recognized as extracellular pathogens. However, S. aureus can also invade, hide and persist in skin cells to contribute to wound chronicity. The intracellular life cycle of S. aureus is currently incompletely understood, although published studies indicate that its intracellular escape strategies play an important role in persistent cutaneous infections. This review provides current scientific knowledge about the intracellular life cycle of S. aureus in skin cells, which can be classified into professional and non-professional antigen-presenting cells, and its strategies to escape adaptive defense mechanisms. First, we discuss phenotypic switch of S. aureus, which affects intracellular routing and degradation. This review also evaluates potential intracellular escape mechanism of S. aureus to avoid intracellular degradation and antigen presentation, preventing an immune response. Furthermore, we discuss potential drug targets that can interfere with the intracellular life cycle of S. aureus. Taken together, this review aimed to increase scientific understanding about the intracellular life cycle of S. aureus into skin cells and its strategies to evade the host immune response, information that is crucial to reduce pathogenic invasion and life-threatening persistence of S. aureus in chronic cutaneous infections.


Asunto(s)
Enfermedades de la Piel/inmunología , Enfermedades de la Piel/microbiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Autofagia , Humanos , Staphylococcus aureus
4.
J Dermatol Sci ; 100(3): 209-216, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33143962

RESUMEN

BACKGROUND: Poorly healing wounds are one of the major complications in patients suffering from recessive dystrophic epidermolysis bullosa (RDEB). At present, there are no effective means to analyze changes in cellular and molecular networks occurring during RDEB wound progression to predict wound outcome and design betted wound management approaches. OBJECTIVES: To better define mechanisms influencing RDEB wound progression by evaluating changes in molecular and cellular networks. METHODS: We developed a non-invasive approach for sampling and analysis of wound-associated constituents using wound-covering bandages. Cellular and molecular components from seventy-six samples collected from early, established and chronic RDEB wounds were evaluated by FACS-based immuno-phenotyping and ELISA. RESULTS: Our cross-sectional analysis determined that progression of RDEB wounds to chronic state is associated with the accumulation (up to 90 %) of CD16+CD66b+ mature neutrophils, loss of CD11b+CD68+ macrophages, and a significant increase (up to 50 %) in a number of CD11c+CD80+CD86+ activated professional antigen presenting cells (APC). It was also marked by changes in activated T cells populations including a reduction of CD45RO+ peripheral memory T cells from 80 % to 30 % and an increase (up to 70 %) in CD45RA+ effector T cells. Significantly higher levels of MMP9, VEGF-A and cathepsin G were also associated with advancing of wounds to poorly healing state. CONCLUSIONS: Our data demonstrated that wound-covering bandages are useful for a non-invasive sampling and analysis of wound-associated constituents and that transition to poorly healing wounds in RDEB patients as associated with distinct changes in leukocytic infiltrates, matrix-remodeling enzymes and pro-angiogenic factors at wound sites.


Asunto(s)
Epidermólisis Ampollosa Distrófica/complicaciones , Leucocitos/inmunología , Piel/patología , Cicatrización de Heridas/inmunología , Adolescente , Adulto , Anciano , Niño , Preescolar , Estudios Transversales , Epidermólisis Ampollosa Distrófica/inmunología , Epidermólisis Ampollosa Distrófica/patología , Femenino , Humanos , Lactante , Leucocitos/metabolismo , Masculino , Persona de Mediana Edad , Receptores CCR2/metabolismo , Receptores de Interleucina-8B/metabolismo , Piel/citología , Piel/inmunología , Adulto Joven
5.
Stem Cell Res Ther ; 11(1): 463, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33138863

RESUMEN

BACKGROUND: Congenital muscular dystrophies (CMD) are a clinically and genetically heterogeneous group of neuromuscular disorders characterized by muscle weakness. The two most prevalent forms of CMD, collagen VI-related myopathies (COL6RM) and laminin α2 deficient CMD type 1A (MDC1A), are both caused by deficiency or dysfunction of extracellular matrix proteins. Previously, we showed that an intramuscular transplantation of human adipose-derived stem cells (ADSC) into the muscle of the Col6a1-/- mice results in efficient stem cell engraftment, migration, long-term survival, and continuous production of the collagen VI protein, suggesting the feasibility of the systemic cellular therapy for COL6RM. In order for this therapeutic approach to work however, stem cells must be efficiently targeted to the entire body musculature. Thus, the main goal of this study is to test whether muscle homing of systemically transplanted ADSC can be enhanced by employing muscle-specific chemotactic signals originating from CMD-affected muscle tissue. METHODS: Proteomic screens of chemotactic molecules were conducted in the skeletal muscles of COL6RM- and MDC1A-affected patients and CMD mouse models to define the inflammatory and immune activities, thus, providing potential markers of disease activity or treatment effect. Also using a pre-clinical animal model, recapitulating mild Ullrich congenital muscular dystrophy (UCMD), the therapeutic relevance of identified chemotactic pathways was investigated in vivo, providing a basis for future clinical investigations. RESULTS: Comprehensive proteomic screens evaluating relevant human and mouse skeletal muscle biopsies offered chemotactic axes to enhance directional migration of systemically transplanted cells into CMD-affected muscles, including CCL5-CCR1/3/5, CCL2-CCR2, CXCL1/2-CXCR1,2, and CXCL7-CXCR2. Also, the specific populations of ADSC selected with an affinity for the chemokines being released by damaged muscle showed efficient migration to injured site and presented their therapeutic effect. CONCLUSIONS: Collectively, identified molecules provided insight into the mechanisms governing directional migration and intramuscular trafficking of systemically infused stem cells, thus, permitting broad and effective application of the therapeutic adult stem cells for CMD treatment.


Asunto(s)
Células Madre Adultas , Distrofias Musculares , Animales , Quimiocinas , Humanos , Laminina , Ratones , Músculo Esquelético , Distrofias Musculares/genética , Distrofias Musculares/terapia , Proteómica
7.
J Invest Dermatol ; 137(11): 2298-2308, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28736230

RESUMEN

Hereditary epidermolysis bullosa (EB) is associated with skin blistering and the development of chronic nonhealing wounds. Although clinical studies have shown that cell-based therapies improve wound healing, the recruitment of therapeutic cells to blistering skin and to more advanced skin lesions remains a challenge. Here, we analyzed cytokines and chemokines in blister fluids of patients affected by dystrophic, junctional, and simplex EB. Our analysis revealed high levels of CXCR1, CXCR2, CCR2, and CCR4 ligands, particularly dominant in dystrophic and junctional EB. In vitro migration assays demonstrated the preferential recruitment of CCR4+ lymphocytes and CXCR1+, CXCR2+, and CCR2+ myeloid cells toward EB-derived blister fluids. Immunophenotyping of skin-infiltrating leukocytes confirmed substantial infiltration of EB-affected skin with resting (CD45RA+) and activated (CD45RO+) T cells and CXCR2+ CD11b+ cells, many of which were identified as CD16b+ neutrophils. Our studies also showed that abundance of CXCR2 ligand in blister fluids also creates a favorable milieu for the recruitment of the CXCR2+ stem cells, as validated by in vitro and in-matrix migration assays. Collectively, this study identified several chemotactic pathways that control the recruitment of leukocytes to the EB-associated skin lesions. These chemotactic axes could be explored for the refinement of the cutaneous homing of the therapeutic stem cells.


Asunto(s)
Quimiocinas/genética , Citocinas/genética , Epidermólisis Ampollosa/genética , Epidermólisis Ampollosa/patología , Receptores CXCR/genética , Vesícula/patología , Movimiento Celular/genética , Células Cultivadas , Progresión de la Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Biología Molecular , Pronóstico , Muestreo , Sensibilidad y Especificidad , Células Madre/metabolismo , Células Madre/patología
8.
J Invest Dermatol ; 137(5): 1126-1134, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28132854

RESUMEN

Generalized nonsegmental vitiligo is often associated with the activation of melanocyte-specific autoimmunity. Because chemokines play an important role in the maintenance of immune responses, we examined chemotactic signatures in cultured vitiligo melanocytes and skin samples of early (≤2 months) and advanced (≥6 months) vitiligo. Analysis showed that melanocytes in early lesions have altered expression of several chemotaxis-associated molecules, including elevated secretion of CXCL12 and CCL5. Higher levels of these chemokines coincided with prominent infiltration of the skin with antigen presenting cells (APCs) and T cells. Most of the intralesional APCs expressed the CD86 maturation marker and co-localized with T cells, particularly in early vitiligo lesions. These observations were confirmed by in vivo animal studies showing preferential recruitment of APCs and T cells to CXCL12- and CCL5-expressing transplanted melanocytes, immunotargeting of the chemokine-positive cells, continuous loss of the pigment-producing cells from the epidermis, and development of vitiligo-like lesions. Taken together, our studies show that melanocyte-derived CXCL12 and CCL5 support APC and T-cell recruitment, antigen acquisition, and T-cell activation in early vitiligo and reinforce the role of melanocyte-derived CXCL12 and CCL5 in activation of melanocyte-specific immunity and suggest inhibition of these chemotactic axes as a strategy for vitiligo stabilization.


Asunto(s)
Quimiocina CCL5/metabolismo , Quimiocina CXCL12/metabolismo , Melanocitos/metabolismo , Vitíligo/patología , Animales , Células Presentadoras de Antígenos/inmunología , Autoinmunidad , Línea Celular , Quimiocina CCL5/inmunología , Quimiocina CXCL12/inmunología , Quimiotaxis/inmunología , Progresión de la Enfermedad , Humanos , Melanocitos/inmunología , Ratones , Linfocitos T/inmunología , Vitíligo/inmunología
9.
Oncotarget ; 8(9): 14428-14442, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-28129639

RESUMEN

CXCR1 and CXCR2 chemokine receptors and their ligands (CXCL1/2/3/7/8) play an important role in tumor progression. Tested to date CXCR1/2 antagonists and chemokine-targeted antibodies were reported to affect malignant cells in vitro and in animal models. Yet, redundancy of chemotactic signals and toxicity hinder further clinical development of these approaches. In this pre-clinical study we investigated the capacity of a novel small molecule dual CXCR1/2 inhibitor, Ladarixin (LDX), to attenuate progression of experimental human melanomas. Our data showed that LDX-mediated inhibition of CXCR1/2 abrogated motility and induced apoptosis in cultured cutaneous and uveal melanoma cells and xenografts independently of the molecular defects associated with the malignant phenotype. These effects were mediated by the inhibition of AKT and NF-kB signaling pathways. Moreover, systemic treatment of melanoma-bearing mice with LDX also polarized intratumoral macrophages to M1 phenotype, abrogated intratumoral de novo angiogenesis and inhibited melanoma self-renewal. Collectively, these studies outlined the pre-requisites of the successful CXCR1/2 inhibition on malignant cells and demonstrated multifactorial effects of Ladarixin on cutaneous and uveal melanomas, suggesting therapeutic utility of LDX in treatment of various melanoma types.


Asunto(s)
Antineoplásicos/farmacología , Melanoma Experimental/tratamiento farmacológico , Receptores de Interleucina-8A/antagonistas & inhibidores , Receptores de Interleucina-8B/antagonistas & inhibidores , Sulfonamidas/farmacología , Microambiente Tumoral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiotaxis , Humanos , Interleucina-8/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Desnudos , FN-kappa B , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Stem Cell Res Ther ; 7(1): 124, 2016 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-27568180

RESUMEN

BACKGROUND: Dystrophic epidermolysis bullosa (DEB), a rare genodermatosis, is characterized by the formation of intra-epidermal blistering and the development of chronic nonhealing skin wounds. Recently, attempts have been made to develop cell-based therapies for this currently intractable disorder. The molecular mechanisms that govern directional migration of the adult stem cells, allowing their efficient and controlled homing to the skin affected with DEB, are poorly understood. The key mechanism that regulates recruitment of leukocytes and progenitor stem cells to distal anatomical tissues affected with disease is chemotaxis, which depends on the signaling molecules, chemokines, and acts primarily as part of the host defense and repair mechanism. METHODS: Comprehensive proteomic screening of chemokines in the blister fluids of DEB-affected mice was conducted to define the inflammatory and immune activities, thus providing potential to examine local biological mechanisms and define the protein signature within lesional skin as a potential marker of disease activity. Also, the therapeutic relevance of identified chemotactic pathways was investigated in vivo, providing a basis for future clinical investigations. RESULTS: Assessment of blister fluid-derived chemokines showed a persistent presence of several chemotactic molecules, including CXCL1 + 2 and CXCL5. The majority of blister-originated chemotactic signals were associated with preferential recruitment of CD45(+)CXCR2(+) and CD11b(+)CXCR2(+) leukocytes. Systemic transplantation of an enriched CXCR2 population of mouse adipose-derived stem cells (mADSC) into DEB-affected mice demonstrated effective recruitment of cells to the blistering skin under the influence of blister-derived ligands and deposition of therapeutic type VII collagen. CONCLUSIONS: Collectively, these studies demonstrate that recruitment of mADSC into DEB skin is tightly controlled by disease-site chemotactic activities and suggest a potential mechanism for effective application of therapeutic stem cells for DEB.


Asunto(s)
Células Madre Adultas/fisiología , Quimiotaxis/fisiología , Epidermólisis Ampollosa Distrófica/terapia , Células Madre Adultas/metabolismo , Animales , Vesícula/metabolismo , Vesícula/terapia , Movimiento Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Quimiocinas/metabolismo , Colágeno Tipo VII/metabolismo , Modelos Animales de Enfermedad , Epidermólisis Ampollosa Distrófica/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Piel/metabolismo , Cicatrización de Heridas/fisiología
11.
J Control Release ; 244(Pt B): 336-346, 2016 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-27288877

RESUMEN

Poly(ß-amino ester)s (PAEs) have emerged as a promising class of gene delivery vectors with performances that can even be compared to viruses. However, all of the transfection studies (over 2350 PAEs) have been limited to linear poly(ß-amino ester)s (LPAEs) despite increasing evidence that polymer structure significantly affects performance. Herein, we describe the development of highly branched poly(ß-amino ester)s (HPAEs) via a new "A2+B3+C2" Michael addition approach demonstrating 2 to 126-fold higher in vitro transfection efficiencies of different cell types in comparison to their linear LPAE counterparts as well as greatly out-performing the leading transfection reagents SuperFect and the "gold-standard" polyethyleneimine (PEI) - especially on skin epidermal cells. More importantly, the ability to correct a skin genetic defect is demonstrated in vivo utilizing a recessive dystrophic epidermolysis bullosa (RDEB) knockout mouse model. Our results provide evidence that the "A2+B3+C2" approach can be controlled and offers sufficient flexibility for the synthesis of HPAEs. The branched structures can significantly improve the transfection efficiency and safety of PAEs highlighting the great promise for the successful application of non-viral gene therapy in skin disease.


Asunto(s)
ADN/administración & dosificación , Epidermólisis Ampollosa Distrófica/terapia , Técnicas de Transferencia de Gen , Terapia Genética , Polímeros/administración & dosificación , Animales , Línea Celular , Células Cultivadas , Colágeno Tipo VII/genética , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Luciferasas/genética , Células Madre Mesenquimatosas , Ratones Noqueados , Piel
12.
J Biol Chem ; 290(35): 21443-59, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26178373

RESUMEN

Fibulin-4 is an extracellular matrix protein essential for elastic fiber formation. Frameshift and missense mutations in the fibulin-4 gene (EFEMP2/FBLN4) cause autosomal recessive cutis laxa (ARCL) 1B, characterized by loose skin, aortic aneurysm, arterial tortuosity, lung emphysema, and skeletal abnormalities. Homozygous missense mutations in FBLN4 are a prevalent cause of ARCL 1B. Here we generated a knock-in mouse strain bearing a recurrent fibulin-4 E57K homozygous missense mutation. The mutant mice survived into adulthood and displayed abnormalities in multiple organ systems, including loose skin, bent forelimb, aortic aneurysm, tortuous artery, and pulmonary emphysema. Biochemical studies of dermal fibroblasts showed that fibulin-4 E57K mutant protein was produced but was prone to dimer formation and inefficiently secreted, thereby triggering an endoplasmic reticulum stress response. Immunohistochemistry detected a low level of fibulin-4 E57K protein in the knock-in skin along with altered expression of selected elastic fiber components. Processing of a precursor to mature lysyl oxidase, an enzyme involved in cross-linking of elastin and collagen, was compromised. The knock-in skin had a reduced level of desmosine, an elastin-specific cross-link compound, and ultrastructurally abnormal elastic fibers. Surprisingly, structurally aberrant collagen fibrils and altered organization into fibers were characteristics of the knock-in dermis and forelimb tendons. Type I collagen extracted from the knock-in skin had decreased amounts of covalent intermolecular cross-links, which could contribute to the collagen fibril abnormalities. Our studies provide the first evidence that fibulin-4 plays a role in regulating collagen fibril assembly and offer a preclinical platform for developing treatments for ARCL 1B.


Asunto(s)
Vasos Sanguíneos/anomalías , Huesos/anomalías , Colágeno Tipo I/metabolismo , Cutis Laxo/patología , Tejido Elástico/anomalías , Proteínas de la Matriz Extracelular/genética , Técnicas de Sustitución del Gen , Piel/patología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Vasos Sanguíneos/patología , Huesos/patología , Colágeno Tipo I/ultraestructura , Reactivos de Enlaces Cruzados/metabolismo , Cutis Laxo/metabolismo , Modelos Animales de Enfermedad , Tejido Elástico/patología , Tejido Elástico/ultraestructura , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/enzimología , Fibroblastos/patología , Miembro Anterior/anomalías , Miembro Anterior/diagnóstico por imagen , Miembro Anterior/patología , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Biosíntesis de Proteínas , Multimerización de Proteína , Proteína-Lisina 6-Oxidasa/metabolismo , Radiografía , Tendones/anomalías , Tendones/patología , Tendones/ultraestructura
13.
Stem Cell Res Ther ; 5(1): 21, 2014 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-24522088

RESUMEN

INTRODUCTION: Congenital muscular dystrophies (CMD) are a clinically and genetically heterogeneous group of neuromuscular disorders characterized by muscle weakness within the first two years of life. Collagen VI-related muscle disorders have recently emerged as one of the most common types of CMD. COL6 CMD is caused by deficiency and/or dysfunction of extracellular matrix (ECM) protein collagen VI. Currently, there is no specific treatment for this disabling and life-threatening disease. The primary cellular targets for collagen VI CMD therapy are fibroblasts in muscle, tendon and skin, as opposed to muscle cells for other types of muscular dystrophies. However, recent advances in stem cell research have raised the possibility that use of adult stem cells may provide dramatic new therapies for treatment of COL6 CMD. METHODS: Here, we developed a procedure for isolation of human stem cells from the adipose layer of neonatal skin. The adipose-derived stem cells (ADSC) were examined for expression of ECM and related genes using gene expression array analysis. The therapeutic potential of ADSC was assessed after a single intramuscular transplantation in collagen VI-deficient mice. RESULTS: Analysis of primary cultures confirmed that established ADSC represent a morphologically homogenous population with phenotypic and functional features of adult mesenchymal stem cells. A comprehensive gene expression analysis showed that ADSC express a vast array of ECM genes. Importantly, it was observed that ADSC synthesize and secrete all three collagen VI chains, suggesting suitability of ADSC for COL6 CMD treatment. Furthermore, we have found that a single intramuscular transplantation of ADSC into Col6a1-/-Rag1-/- mice under physiological and cardiotoxin-induced injury/regeneration conditions results in efficient engraftment and migration of stem cells within the skeletal muscle. Importantly, we showed that ADSC can survive long-term and continuously secrete the therapeutic collagen VI protein missing in the mutant mice. CONCLUSIONS: Overall, our findings suggest that stem cell therapy can potentially provide a new avenue for the treatment of COL6 CMD and other muscular disorders and injuries.


Asunto(s)
Colágeno Tipo VI/genética , Trasplante de Células Madre Mesenquimatosas , Distrofias Musculares/terapia , Tejido Adiposo/citología , Animales , Diferenciación Celular , Células Cultivadas , Colágeno Tipo VI/metabolismo , Prepucio/citología , Humanos , Recién Nacido , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Distrofias Musculares/congénito
14.
Oncoimmunology ; 2(10): e26092, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24251079

RESUMEN

We have demonstrated that priming of intratumoral and intradermal vaccination sites with chemokines enhances cytotoxic immune response against established neoplasms. Additional insights into the molecular mechanisms that underlie these findings and the optimization of such an approach may lead to the development of cost-effective and generic immunotherapeutic regimens against cancer.

15.
Cytotherapy ; 15(2): 171-184.e1, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23321329

RESUMEN

BACKGROUND AIMS: Adult stem cells produce a plethora of extracellular matrix molecules and have a high potential as cell-based therapeutics for connective tissue disorders of the skin. However, the primary challenge of the stem cell-based approach is associated with the inefficient homing of systemically infused stem cells to the skin. METHODS: We examined chemotactic mechanisms that govern directional migration of mesenchymal stem cells (MSCs) into the skin by conducting a comprehensive expression analysis of chemotactic molecules in MSCs and defined cutaneous tissues from normal and hereditary epidermolysis bullosa (EB)-affected skin. RESULTS: Analysis of chemokine receptors in short-term and long-term MSC cultures showed tissue culture-dependent expression of several receptors. Assessment of epidermis-derived and dermis-derived chemokines showed that most chemotactic signals that originate from the skin preferentially recruit different sets of leukocytes rather than MSCs. Analysis of the chemotactic molecules derived from EB-affected non-blistered skin showed only minor changes in expression of selected chemokines and receptors. Nevertheless, the data allowed us to define the Ccl27-Ccr10 chemotactic axis as the most potent for the recruitment of MSCs to the skin. Our in vivo analysis demonstrated that uniform expression of Ccr10 on MSCs and alteration of Ccl27 level in the skin enhance extravasation of stem cells from circulation and facilitate their migration within cutaneous tissue. CONCLUSIONS: Collectively, our study provides a comprehensive analysis of chemotactic signals in normal and EB-affected skin and proof-of-concept data demonstrating that alteration of the chemotactic pathways can enhance skin homing of the therapeutic stem cells.


Asunto(s)
Células de la Médula Ósea/metabolismo , Quimiocina CCL27/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptores CCR10/metabolismo , Piel/metabolismo , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Quimiocina CCL27/genética , Quimiocinas/metabolismo , Epidermólisis Ampollosa , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/citología , Ratones , Receptores CCR10/genética , Receptores de Quimiocina/metabolismo , Piel/citología
16.
Cytotherapy ; 13(1): 30-45, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20854215

RESUMEN

BACKGROUND AIMS: Multiple studies have demonstrated that mesenchymal stromal cells (MSC) can be utilized therapeutically for various congenital and acquired disorders. The involvement of MSC in the maintenance of skin homeostasis and their curative application for the treatment of skin wounds have also been documented. However, it is not known whether MSC can commit to cutaneous lineages, produce structural proteins essential for the skin integrity or be used for hereditary skin disorders. METHODS: To address these questions, we conducted a comparative expression analysis between MSC and potentially adjacent cutaneous cells, fibroblasts and keratinocytes, with specific emphasis on extracellular matrix encoding and related genes. RESULTS: Our data demonstrated that MSC share many features with cutaneous fibroblasts. We also observed that under direct influence of cutaneous fibroblasts in vitro and fibroblast-derived matrix in vivo, MSC acquired a fibroblastic phenotype, suggesting that specific cell-cell interactions play a key regulatory role in the differentiation of MSC. Additionally, the observed fibroblastic transition of MSC was underlined by a significant up-regulation of several cutaneous-specific genes encoding lumican, decorin, type VII collagen, laminin and other structural proteins. As many of the identified genes have considerable therapeutic value for dermatologic afflictions, particularly type VII collagen, we evaluated further the therapeutic potential of congenic MSC in the skin of Col7a1-null mice recapitulating human recessive dystrophic epidermolysis bullosa (RDEB). Remarkably, MSC-derived type VII collagen was sufficient for restoration of the damaged dermal-epidermal junction and partial reversal of the RDEB phenotype. CONCLUSIONS: Collectively, our results suggest that MSC may offer promising therapeutics for the treatment of RDEB and potentially other genodermatoses.


Asunto(s)
Vesícula/genética , Vesícula/terapia , Células de la Médula Ósea/citología , Perfilación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Piel/metabolismo , Piel/patología , Animales , Vesícula/patología , Células de la Médula Ósea/metabolismo , Adhesión Celular , Forma de la Célula , Técnicas de Cocultivo , Colágeno Tipo VII/deficiencia , Matriz Extracelular/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Humanos , Inmunofenotipificación , Queratinocitos/metabolismo , Queratinocitos/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/ultraestructura , Trasplante Autólogo
17.
Aging Cell ; 9(4): 580-91, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20477760

RESUMEN

Werner syndrome (WS) is an autosomal recessive disorder, the hallmarks of which are premature aging and early onset of neoplastic diseases (Orren, 2006; Bohr, 2008). The gene, whose mutation underlies the WS phenotype, is called WRN. The protein encoded by the WRN gene, WRNp, has DNA helicase activity (Gray et al., 1997; Orren, 2006; Bohr, 2008; Opresko, 2008). Extensive evidence suggests that WRNp plays a role in DNA replication and DNA repair (Chen et al., 2003; Hickson, 2003; Orren, 2006; Turaga et al., 2007; Bohr, 2008). However, WRNp function is not yet fully understood. In this study, we show that WRNp is involved in de novo DNA methylation of the promoter of the Oct4 gene, which encodes a crucial stem cell transcription factor. We demonstrate that WRNp localizes to the Oct4 promoter during retinoic acid-induced differentiation of human pluripotent cells and associates with the de novo methyltransferase Dnmt3b in the chromatin of differentiating pluripotent cells. Depletion of WRNp does not affect demethylation of lysine 4 of the histone H3 at the Oct4 promoter, nor methylation of lysine 9 of H3, but it blocks the recruitment of Dnmt3b to the promoter and results in the reduced methylation of CpG sites within the Oct4 promoter. The lack of DNA methylation was associated with continued, albeit greatly reduced, Oct4 expression in WRN-deficient, retinoic acid-treated cells, which resulted in attenuated differentiation. The presented results reveal a novel function of WRNp and demonstrate that WRNp controls a key step in pluripotent stem cell differentiation.


Asunto(s)
Epigénesis Genética , Exodesoxirribonucleasas/metabolismo , Silenciador del Gen , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , RecQ Helicasas/metabolismo , Biomarcadores/metabolismo , Antígenos CD4/genética , Antígenos CD4/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Silenciador del Gen/efectos de los fármacos , Histonas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Modelos Biológicos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/metabolismo , Tretinoina/farmacología , Helicasa del Síndrome de Werner , Globinas beta/genética , Globinas beta/metabolismo , ADN Metiltransferasa 3B
18.
Methods Mol Biol ; 522: 3-14, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19247621

RESUMEN

The use of recombinant DNA has become a powerful tool in the analysis of functional and structural properties of the extracellular matrix proteins. During last decade, various procedures of plasmid DNA delivery using liposome-based or electroporation-based transfection have been developed. However, in many instances, these procedures were shown to be not effective in DNA transfer or toxic for the mammalian cells. On contrary, retrovirus-mediated infection represents a superior mode of gene delivery with a success rate and viability of the cells approaching 100% in in vitro conditions. The use of the retroviral system also allows permanent insertion of the gene of interest into the chromosome of the infected cell, resulting in efficient gene transfer in which most recipient cells will incorporate and express the transduced gene. In this chapter, we will describe several retrovirus-based systems and provide step-by-step protocols applicable for the production of the recombinant virus and efficient delivery of the ECM genes.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Vectores Genéticos , Retroviridae/genética , ADN Recombinante/genética
19.
Oligonucleotides ; 18(2): 111-22, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18637729

RESUMEN

Gene therapy using viral vectors for liver diseases, particularly congenital disorders, is besought with difficulties, particularly immunologic reactions to viral antigens. As a result, nonviral methods for gene transfer in hepatocytes have also been explored. Gene repair by small synthetic single-stranded oligodeoxynucleotides (ODNs) produces targeted alterations in the genome of mammalian cells and represents a great potential for nonviral gene therapy. To test the feasibility of ODN-mediated gene repair within chromosomal DNA in human hepatocytes, two new cell lines with stably integrated mutant reporter genes, namely neomycin and enhanced green fluorescent protein were established. Targeting theses cells with ODNs specifically designed for repair resulted in site-directed and permanent gene conversion of the single-point mutation of the reporter genes. Moreover, the frequency of gene alteration was highly dependent on the mitotic activity of the cells, indicating that the proliferative status is an important factor for successful targeting in human hepatocytes. cDNA array expression profiling of DNA repair genes under different cell culture conditions combined with RNA interference assay showed that mismatch repair (MMR) in actively growing hepatocytes imposes a strong barrier to efficient gene repair mediated by ODNs. Suppression of MSH2 activity in hepatocytes transduced with short hairpin RNAs (shRNAs) targeted to MSH2 mRNA resulted in 25- to 30-fold increase in gene repair rate, suggesting a negative effect of MMR on ODN-mediated gene repair. Taken together, these data suggest that under appropriate conditions nonviral chromosomal targeting may represent a feasible approach to gene therapy in liver disease.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Marcación de Gen/métodos , Terapia Genética/métodos , Hepatocitos/metabolismo , Oligonucleótidos/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Estudios de Factibilidad , Fluoresceína-5-Isotiocianato/metabolismo , Colorantes Fluorescentes/metabolismo , Técnicas de Transferencia de Gen , Genes Reporteros , Humanos , Liposomas , Neoplasias Hepáticas/metabolismo , Oligonucleótidos/administración & dosificación , Mutación Puntual , Transfección
20.
J Invest Dermatol ; 128(6): 1476-86, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18079746

RESUMEN

Junctional epidermolysis bullosa (JEB) is an inherited mechanobullous disease characterized by reduced adherence of the epidermal keratinocytes to the underlying dermis, and is often caused by the absence of functional laminin 332 due to the lack or dysfunction of its beta3 chain. As there are no specific therapies for JEB, we tested whether a protein replacement strategy could be applicable for the restoration of the laminin 332 assembly and reversion of the JEB phenotype in human keratinocytes that lack beta3 subunit. Here, we developed the protocol for production and purification of the biologically active recombinant beta3 chain. Next, we demonstrated that delivery of recombinant beta3 polypeptide into the endoplasmic reticulum of the immortalized beta3-null keratinocytes led to the restoration of the laminin 332 assembly, secretion, and deposition into the basement membrane zone, as confirmed by Western blot analysis, confocal immunofluorescent microscopy in vitro, and on cultured organotypic human JEB skin reconstructs. Although the amount of laminin 332 produced by protein-treated beta3-null keratinocytes is lower than that in normal human keratinocytes, our results demonstrate the applicability of the recombinant proteins for JEB treatment and open new perspectives for the development of novel therapeutics for this inherited, currently intractable, skin disorder.


Asunto(s)
Epidermólisis Ampollosa de la Unión/terapia , Integrina beta3/genética , Integrina beta3/fisiología , Queratinocitos/metabolismo , Laminina/química , Proteínas Recombinantes/uso terapéutico , Adhesión Celular , Retículo Endoplásmico/metabolismo , Humanos , Microscopía Confocal , Modelos Biológicos , Fenotipo , Transporte de Proteínas , Piel/patología , Ingeniería de Tejidos/métodos , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...