Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Dermatologie (Heidelb) ; 75(3): 218-224, 2024 Mar.
Artículo en Alemán | MEDLINE | ID: mdl-38351374

RESUMEN

The pathogenesis of fibrosing alterations in the skin and other organ systems is not yet sufficiently understood and current therapeutic options are limited. Fibrosing diseases of the skin lead to a loss of function, which can subsequently be accompanied by serious impairments in quality of life, increased morbidity and ultimately increased mortality. There are currently only a few pharmacological and therapeutic approaches approved to prevent or ameliorate fibrosing diseases. Furthermore, tissue-specific versus common, non-organ-specific pathophysiological cellular and molecular mechanisms are not resolved. The development of new, cause-based and therefore likely more efficient therapeutic approaches is urgently needed. This represents a major challenge, but also opens up the opportunity for special contributions to improve this medically unsolved problem. Here we present important findings from recent years with a focus on the role of the immune response in fibrogenesis.


Asunto(s)
Calidad de Vida , Enfermedades de la Piel , Humanos , Fibrosis , Enfermedades de la Piel/etiología , Piel/patología
2.
J Invest Dermatol ; 144(1): 152-164.e7, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37516311

RESUMEN

Cells of the monocyte/macrophage lineage are an integral component of the body's innate ability to restore tissue function after injury. In parallel to mounting an inflammatory response, clearance of monocytes/macrophages from the wound site is critical to re-establish tissue functionality and integrity during the course of healing. The role of regulated cell death in macrophage clearance from damaged tissue and its implications for the outcome of the healing response is little understood. In this study, we explored the role of macrophage-specific FADD-mediated cell death on Ripk3-/- background in a mechanical skin injury model in mice. We found that combined inhibition of RIPK3-mediated necroptosis and FADD-caspase-8-mediated apoptosis in macrophages profoundly delayed wound healing. Importantly, RIPK3 deficiency alone did not considerably alter the wound healing process and macrophage population dynamics, arguing that inhibition of FADD-caspase-8-dependent death of macrophages is primarily responsible for delayed wound closure. Notably, TNF blockade reversed the accumulation of Ly6Chigh macrophages induced by combined deficiency of FADD and RIPK3, indicating a critical dual role of TNF-mediated prosurvival and cell death signaling, particularly in this highly proinflammatory macrophage subset. Our findings reveal a previously uncharacterized cross-talk of inflammatory and cell death signaling in macrophages in regulating repair processes in the skin.


Asunto(s)
Apoptosis , Macrófagos , Animales , Ratones , Caspasa 8/metabolismo , Macrófagos/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Piel/metabolismo
3.
STAR Protoc ; 4(4): 102765, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38060383

RESUMEN

The role of dermal white adipose tissue in regulating skin homeostasis and self-renewal processes has recently attracted interest. However, the isolation of proteins from dermal adipocytes for biochemical analysis is challenging. Here, we provide a protocol for the isolation of murine dermal adipocytes. We describe steps for inducing adipocyte-specific gene deletion, adipocyte isolation, protein purification, and western blot analysis. The reliability of the protocol is demonstrated by verifying efficient adipocyte-specific Atgl gene deletion in a tamoxifen-inducible Cre/loxP-based mouse model. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2019).1.


Asunto(s)
Adipocitos , Tejido Adiposo Blanco , Ratones , Animales , Reproducibilidad de los Resultados , Adipocitos/metabolismo , Tejido Adiposo Blanco/metabolismo , Piel
4.
Proc Natl Acad Sci U S A ; 120(40): e2215421120, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37756334

RESUMEN

Externalized histones erupt from the nucleus as extracellular traps, are associated with several acute and chronic lung disorders, but their implications in the molecular pathogenesis of interstitial lung disease are incompletely defined. To investigate the role and molecular mechanisms of externalized histones within the immunologic networks of pulmonary fibrosis, we studied externalized histones in human and animal bronchoalveolar lavage (BAL) samples of lung fibrosis. Neutralizing anti-histone antibodies were administered in bleomycin-induced fibrosis of C57BL/6 J mice, and subsequent studies used conditional/constitutive knockout mouse strains for TGFß and IL-27 signaling along with isolated platelets and cultured macrophages. We found that externalized histones (citH3) were significantly (P < 0.01) increased in cell-free BAL fluids of patients with idiopathic pulmonary fibrosis (IPF; n = 29) as compared to healthy controls (n = 10). The pulmonary sources of externalized histones were Ly6G+CD11b+ neutrophils and nonhematopoietic cells after bleomycin in mice. Neutralizing monoclonal anti-histone H2A/H4 antibodies reduced the pulmonary collagen accumulation and hydroxyproline concentration. Histones activated platelets to release TGFß1, which signaled through the TGFbRI/TGFbRII receptor complex on LysM+ cells to antagonize macrophage-derived IL-27 production. TGFß1 evoked multiple downstream mechanisms in macrophages, including p38 MAPK, tristetraprolin, IL-10, and binding of SMAD3 to the IL-27 promotor regions. IL-27RA-deficient mice displayed more severe collagen depositions suggesting that intact IL-27 signaling limits fibrosis. In conclusion, externalized histones inactivate a safety switch of antifibrotic, macrophage-derived IL-27 by boosting platelet-derived TGFß1. Externalized histones are accessible to neutralizing antibodies for improving the severity of experimental pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , Interleucina-27 , Humanos , Ratones , Animales , Ratones Endogámicos C57BL , Histonas , Plaquetas , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética
5.
J Invest Dermatol ; 143(12): 2456-2467.e5, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37295491

RESUMEN

Type 2 immune responses have been increasingly linked with tissue maintenance, regeneration, and metabolic homeostasis. The molecular basis of regulator and effector mechanisms of type 2 immunity in skin regeneration and homeostasis is still lacking. In this study, we analyzed the role of IL-4Rα signaling in the regeneration of diverse cellular compartments in the skin. Mutants with global IL-4Rα deficiency showed two major phenotypes: first, a pronounced atrophy of the interfollicular epidermis, and second, a significant increase in dermal white adipose tissue thickness in mice aged 3 weeks (postnatal day 21) compared with littermate controls. Notably, IL-4Rα deficiency decreased the activation of hormone-sensitive lipase, a rate-limiting step in lipolysis. Immunohistochemical and FACS analysis in IL-4/enhanced GFP reporter mice showed that IL-4 expression peaked on postnatal day 21 and that eosinophils are the predominant IL-4-expressing cells. Eosinophil-deficient mice recapitulated the lipolytic-defective dermal white adipose tissue phenotype of Il4ra-deficient mice, showing that eosinophils are necessary for dermal white adipose tissue lipolysis. Collectively, we provide mechanistic insights into the regulation of interfollicular epidermis and hormone-sensitive lipase-mediated lipolysis in dermal white adipose tissue in early life by IL-4Rα, and our findings show that eosinophils play a critical role in this process.


Asunto(s)
Interleucina-4 , Esterol Esterasa , Animales , Ratones , Epidermis , Piel , Tejido Adiposo Blanco
6.
Exp Mol Med ; 55(3): 565-573, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36869067

RESUMEN

The osmosensitive transcription factor nuclear factor of activated T cells 5 (NFAT5; or tonicity-responsive enhancer binding protein; TonEBP) plays a key role in macrophage-driven regulation of cutaneous salt and water balance. In the immune-privileged and transparent cornea, disturbances in fluid balance and pathological edema result in corneal transparency loss, which is one of the main causes of blindness worldwide. The role of NFAT5 in the cornea has not yet been investigated. We analyzed the expression and function of NFAT5 in naive corneas and in an established mouse model of perforating corneal injury (PCI), which causes acute corneal edema and transparency loss. In uninjured corneas, NFAT5 was mainly expressed in corneal fibroblasts. In contrast, after PCI, NFAT5 expression was highly upregulated in recruited corneal macrophages. NFAT5 deficiency did not alter corneal thickness in steady state; however, loss of NFAT5 led to accelerated resorption of corneal edema after PCI. Mechanistically, we found that myeloid cell-derived NFAT5 is crucial for controlling corneal edema, as edema resorption after PCI was significantly enhanced in mice with conditional loss of NFAT5 in the myeloid cell lineage, presumably due to increased pinocytosis of corneal macrophages. Collectively, we uncovered a suppressive role for NFAT5 in corneal edema resorption, thereby identifying a novel therapeutic target to combat edema-induced corneal blindness.


Asunto(s)
Edema Corneal , Ratones , Animales , Edema Corneal/etiología , Regulación de la Expresión Génica , Factores de Transcripción NFATC/genética , Factores de Transcripción
7.
Front Immunol ; 13: 1012647, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36248909

RESUMEN

To facilitate the recovery process of chronic and hard-to-heal wounds novel pro-resolving treatment options are urgently needed. We investigated the pro-regenerative properties of soluble CD83 (sCD83) on cutaneous wound healing, where sCD83 accelerated wound healing not only after systemic but also after topical application, which is of high therapeutic interest. Cytokine profile analyses revealed an initial upregulation of inflammatory mediators such as TNFα and IL-1ß, followed by a switch towards pro-resolving factors, including YM-1 and IL-10, both expressed by tissue repair macrophages. These cells are known to mediate resolution of inflammation and stimulate wound healing processes by secretion of growth factors such as epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF), which promote vascularization as well as fibroblast and keratinocyte differentiation. In conclusion, we have found strong wound healing capacities of sCD83 beyond the previously described role in transplantation and autoimmunity. This makes sCD83 a promising candidate for the treatment of chronic- and hard-to-heal wounds.


Asunto(s)
Interleucina-10 , Factor de Necrosis Tumoral alfa , Factor de Crecimiento Epidérmico , Mediadores de Inflamación/metabolismo , Interleucina-10/metabolismo , Macrófagos , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/fisiología
8.
Acta Derm Venereol ; 102: adv00834, 2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36250733

RESUMEN

Venous leg ulcers represent a clinical challenge and impair the quality of life of patients. This study examines impaired wound healing in venous leg ulcers at the molecular level. Protein expression patterns for biomarkers were analysed in venous leg ulcer wound fluids from 57 patients treated with a protease-modulating polyacrylate wound dressing for 12 weeks, and compared with exudates from 10 acute split-thickness wounds. Wound healing improved in the venous leg ulcer wounds: 61.4% of the 57 patients with venous leg ulcer achieved a relative wound area reduction of ≥ 40%, and 50.9% of the total 57 patients achieved a relative wound area reduction of ≥ 60%. Within the first 14 days, abundances of S100A8, S100A9, neutrophil elastase, matrix metalloproteinase-2, and fibronectin in venous leg ulcer exudates decreased significantly and remained stable, yet higher than in acute wounds. Interleukin-1ß, tumour necrosis factor alpha, and matrix metalloproteinase-9 abundance ranges were similar in venous leg ulcers and acute wound fluids. Collagen (I) α1 abundance was higher in venous leg ulcer wound fluids and was not significantly regulated. Overall, significant biomarker changes occurred in the first 14 days before a clinically robust healing response in the venous leg ulcer cohort.


Asunto(s)
Úlcera de la Pierna , Úlcera Varicosa , Humanos , Metaloproteinasa 2 de la Matriz , Péptido Hidrolasas , Trasplante de Piel , Calidad de Vida , Úlcera Varicosa/diagnóstico , Úlcera Varicosa/terapia , Úlcera Varicosa/metabolismo , Úlcera de la Pierna/diagnóstico , Úlcera de la Pierna/terapia
9.
Artículo en Inglés | MEDLINE | ID: mdl-36041784

RESUMEN

Monocytes/macrophages are key components of the body's innate ability to restore tissue function after injury. In most tissues, both embryo-derived tissue-resident macrophages and recruited blood monocyte-derived macrophages contribute to the injury response. The developmental origin of injury-associated macrophages has a major impact on the outcome of the healing process. Macrophages are abundant at all stages of repair and coordinate the progression through the different phases of healing. They are highly plastic cells that continuously adapt to their environment and acquire phase-specific activation phenotypes. Advanced omics methodologies have revealed a vast heterogeneity of macrophage activation phenotypes and metabolic status at injury sites in different organs. In this review, we highlight the role of the developmental origin, the link between the wound phase-specific activation state and metabolic reprogramming as well as the fate of macrophages during the resolution of the wounding response.


Asunto(s)
Activación de Macrófagos , Macrófagos , Macrófagos/metabolismo , Cicatrización de Heridas/fisiología
11.
Biology (Basel) ; 11(6)2022 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-35741452

RESUMEN

The skin epidermis, with its capacity for lifelong self-renewal and rapid repairing response upon injury, must maintain an active status in metabolism. Mechanistic target of rapamycin (mTOR) signaling is a central controller of cellular growth and metabolism that coordinates diverse physiological and pathological processes in a variety of tissues and organs. Recent evidence with genetic mouse models highlights an essential role of the mTOR signaling network in epidermal morphogenesis and barrier formation. In this review, we focus on the recent advances in understanding how mTOR signaling networks, including upstream inputs, kinases and downstream effectors, regulate epidermal morphogenesis and skin barrier formation. Understanding the details of the metabolic signaling will be critical for the development of novel pharmacological approaches to promote skin barrier regeneration and to treat epidermal barrier defect-associated diseases.

12.
STAR Protoc ; 3(2): 101337, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35496783

RESUMEN

Understanding macrophage heterogeneity in tissue repair is a major challenge. Here, we describe a protocol that combines isolation of immune cells from skin wounds with subsequent flow-cytometry-based sorting of wound macrophages and single-cell RNA sequencing. We use a modified version of the original Smart-seq2 protocol to increase speed and accuracy. This protocol is useful for analyzing the pronounced heterogeneity of activation phenotypes in wound macrophages and might be adapted to other experimental models of skin inflammation. For complete details on the use and execution of this protocol, please refer to Willenborg et al. (2021).


Asunto(s)
Macrófagos , Cicatrización de Heridas , Animales , Citometría de Flujo , Recuento de Leucocitos , Ratones , Análisis de Secuencia de ARN
13.
Sci Immunol ; 7(70): eabl7482, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35427180

RESUMEN

Macrophages populate every organ during homeostasis and disease, displaying features of tissue imprinting and heterogeneous activation. The disconnected picture of macrophage biology that has emerged from these observations is a barrier for integration across models or with in vitro macrophage activation paradigms. We set out to contextualize macrophage heterogeneity across mouse tissues and inflammatory conditions, specifically aiming to define a common framework of macrophage activation. We built a predictive model with which we mapped the activation of macrophages across 12 tissues and 25 biological conditions, finding a notable commonality and finite number of transcriptional profiles, in particular among infiltrating macrophages, which we modeled as defined stages along four conserved activation paths. These activation paths include a "phagocytic" regulatory path, an "inflammatory" cytokine-producing path, an "oxidative stress" antimicrobial path, or a "remodeling" extracellular matrix deposition path. We verified this model with adoptive cell transfer experiments and identified transient RELMɑ expression as a feature of monocyte-derived macrophage tissue engraftment. We propose that this integrative approach of macrophage classification allows the establishment of a common predictive framework of monocyte-derived macrophage activation in inflammation and homeostasis.


Asunto(s)
Activación de Macrófagos , Macrófagos , Animales , Citocinas/metabolismo , Homeostasis , Inflamación/metabolismo , Ratones
14.
Life Sci Alliance ; 5(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35027468

RESUMEN

Anti-TNF therapies are a core anti-inflammatory approach for chronic diseases such as rheumatoid arthritis and Crohn's Disease. Previously, we and others found that TNF blocks the emergence and function of alternative-activated or M2 macrophages involved in wound healing and tissue-reparative functions. Conceivably, anti-TNF drugs could mediate their protective effects in part by an altered balance of macrophage activity. To understand the mechanistic basis of how TNF regulates tissue-reparative macrophages, we used RNAseq, scRNAseq, ATACseq, time-resolved phospho-proteomics, gene-specific approaches, metabolic analysis, and signaling pathway deconvolution. We found that TNF controls tissue-reparative macrophage gene expression in a highly gene-specific way, dependent on JNK signaling via the type 1 TNF receptor on specific populations of alternative-activated macrophages. We further determined that JNK signaling has a profound and broad effect on activated macrophage gene expression. Our findings suggest that TNF's anti-M2 effects evolved to specifically modulate components of tissue and reparative M2 macrophages and TNF is therefore a context-specific modulator of M2 macrophages rather than a pan-M2 inhibitor.


Asunto(s)
Macrófagos , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Inhibidores del Factor de Necrosis Tumoral/farmacología
15.
Cell Metab ; 33(12): 2398-2414.e9, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34715039

RESUMEN

Wound healing is a coordinated process that initially relies on pro-inflammatory macrophages, followed by a pro-resolution function of these cells. Changes in cellular metabolism likely dictate these distinct activities, but the nature of these changes has been unclear. Here, we profiled early- versus late-stage skin wound macrophages in mice at both the transcriptional and functional levels. We found that glycolytic metabolism in the early phase is not sufficient to ensure productive repair. Instead, by combining conditional disruption of the electron transport chain with deletion of mitochondrial aspartyl-tRNA synthetase, followed by single-cell sequencing analysis, we found that a subpopulation of early-stage wound macrophages are marked by mitochondrial ROS (mtROS) production and HIF1α stabilization, which ultimately drives a pro-angiogenic program essential for timely healing. In contrast, late-phase, pro-resolving wound macrophages are marked by IL-4Rα-mediated mitochondrial respiration and mitohormesis. Collectively, we identify changes in mitochondrial metabolism as a critical control mechanism for macrophage effector functions during wound healing.


Asunto(s)
Macrófagos , Cicatrización de Heridas , Animales , Macrófagos/metabolismo , Ratones , Mitocondrias/metabolismo
16.
Sci Rep ; 11(1): 17926, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34504132

RESUMEN

The extracellular matrix architecture is composed of supramolecular fibrillar networks that define tissue specific cellular microenvironments. Hemicentins (Hmcn1 and Hmcn2) are ancient and very large members (> 600 kDa) of the fibulin family, whose short members are known to guide proper morphology and functional behavior of specialized cell types predominantly in elastic tissues. However, the tissue distribution and function of Hemicentins within the cellular microenvironment of connective tissues has remained largely unknown. Performing in situ hybridization and immunofluorescence analyses, we found that mouse Hmcn1 and Hmcn2 show a complementary distribution throughout different tissues and developmental stages. In postnatal dermal-epidermal junctions (DEJ) and myotendinous junctions (MTJ), Hmcn1 is primarily produced by mesenchymal cells (fibroblasts, tenocytes), Hmcn2 by cells of epithelial origin (keratinocytes, myocytes). Hmcn1-/- mice are viable and show no overt phenotypes in tissue tensile strength and locomotion tests. However, transmission electron microscopy revealed ultrastructural basement membrane (BM) alterations at the DEJ and MTJ of Hmcn1-/- mice, pointing to a thus far unknown role of Hmcn1 for BM and connective tissue boundary integrity.


Asunto(s)
Dermis/metabolismo , Epidermis/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Transducción de Señal/genética , Tendones/metabolismo , Animales , Células Cultivadas , Tejido Conectivo/metabolismo , Desarrollo Embrionario/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Fibroblastos/metabolismo , Queratinocitos/metabolismo , Locomoción/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Resistencia a la Tracción
17.
Cell Metab ; 33(9): 1726-1743, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34384520

RESUMEN

Wound healing requires cooperation between different cell types, among which macrophages play a central role. In particular, inflammatory macrophages are engaged in the initial response to wounding, and alternatively activated macrophages are essential for wound closure and the resolution of tissue repair. The links between temporal activation-induced changes in the metabolism of such macrophages and the influence this has on their functional states, along with the realization that metabolites play both intrinsic and extrinsic roles in the cells that produce them, has focused attention on the metabolism of wound healing. Here, we discuss macrophage metabolism during distinct stages of normal healing and its related pathologic processes, such as during cancer and fibrosis. Further, we frame these insights in a broader context of the current understanding of macrophage metabolic reprogramming linked to cellular activation and function. Finally, we discuss parallels between the metabolism of macrophages and fibroblasts, the latter being a key stromal cell type in wound healing, and consider the importance of the metabolic interplay between different cell types in the wound microenvironment.


Asunto(s)
Macrófagos , Cicatrización de Heridas , Fibroblastos , Fibrosis , Humanos , Macrófagos/metabolismo
18.
Front Immunol ; 12: 667830, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897716

RESUMEN

Macrophages are critical mediators of tissue vascularization both in health and disease. In multiple tissues, macrophages have been identified as important regulators of both blood and lymphatic vessel growth, specifically following tissue injury and in pathological inflammatory responses. In development, macrophages have also been implicated in limiting vascular growth. Hence, macrophages provide an important therapeutic target to modulate tissue vascularization in the clinic. However, the molecular mechanisms how macrophages mediate tissue vascularization are still not entirely resolved. Furthermore, mechanisms might also vary among different tissues. Here we review the role of macrophages in tissue vascularization with a focus on their role in blood and lymphatic vessel formation in the barrier tissues cornea and skin. Comparing mechanisms of macrophage-mediated hem- and lymphangiogenesis in the angiogenically privileged cornea and the physiologically vascularized skin provides an opportunity to highlight similarities but also tissue-specific differences, and to understand how macrophage-mediated hem- and lymphangiogenesis can be exploited for the treatment of disease, including corneal wound healing after injury, graft rejection after corneal transplantation or pathological vascularization of the skin.


Asunto(s)
Vasos Sanguíneos/metabolismo , Córnea/irrigación sanguínea , Neovascularización de la Córnea , Linfangiogénesis , Vasos Linfáticos/metabolismo , Macrófagos/metabolismo , Neovascularización Fisiológica , Piel/irrigación sanguínea , Animales , Vasos Sanguíneos/inmunología , Vasos Sanguíneos/patología , Humanos , Vasos Linfáticos/inmunología , Vasos Linfáticos/patología , Macrófagos/inmunología , Macrófagos/patología , Fenotipo , Transducción de Señal , Cicatrización de Heridas
19.
J Invest Dermatol ; 141(4S): 1063-1070, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33558058

RESUMEN

An effective healing response is critical to promote and ensure healthy aging. Major discoveries in both fields-repair and aging-have led to a better understanding of the mechanisms regulating the healing response and of the complexity of the aging process. It will now be important to translate and connect those findings to improve our insights into the decline of regeneration in the elderly. Furthermore, we need to understand how this process can be stalled to maintain and promote tissue resilience. Furthermore, it remains to be explored how the findings in model organisms are conserved in human wounds and how these findings might be translated into the clinic.


Asunto(s)
Inmunosenescencia , Queratinocitos/patología , Piel/lesiones , Cicatrización de Heridas/inmunología , Animales , Movimiento Celular/inmunología , Proliferación Celular , Senescencia Celular/inmunología , Modelos Animales de Enfermedad , Humanos , Queratinocitos/inmunología , Piel/citología , Piel/inmunología , Piel/patología
20.
Matrix Biol ; 94: 57-76, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32890632

RESUMEN

Skin integrity and function depends to a large extent on the composition of the extracellular matrix, which regulates tissue organization. Collagen XII is a homotrimer with short collagenous domains that confer binding to the surface of collagen I-containing fibrils and extended flexible arms, which bind to non-collagenous matrix components. Thereby, collagen XII helps to maintain collagen suprastructure and to absorb stress. Mutant or absent collagen XII leads to reduced muscle and bone strength and lax skin, whereas increased collagen XII amounts are observed in tumor stroma, scarring and fibrosis. This study aimed at uncovering in vivo mechanisms by which collagen XII may achieve these contrasting outcomes. We analyzed skin as a model tissue that contains abundant fibrils, composed of collagen I, III and V with collagen XII decorating their surface, and which is subject to mechanical stress. The impact of different collagen XII levels was investigated in collagen XII-deficient (Col12-KO) mice and in mice with collagen XII overexpression in the dermis (Col12-OE). Unchallenged skin of these mice was histologically inconspicuous, but at the ultrastructural level revealed distinct aberrations in collagen network suprastructure. Repair of excisional wounds deviated from controls in both models by delayed healing kinetics, which was, however, caused by completely different mechanisms in the two mouse lines. The disorganized matrix in Col12-KO wounds failed to properly sequester TGFß, resulting in elevated numbers of myofibroblasts. These are, however, unable to contract and remodel the collagen XII-deficient matrix. Excess of collagen XII, in contrast, promotes persistence of M1-like macrophages in the wound bed, thereby stalling the wounds in an early inflammatory stage of the repair process and delaying healing. Taken together, we demonstrate that collagen XII is a key component that assists in orchestrating proper skin matrix structure, controls growth factor availability and regulates cellular composition and function. Together, these functions are pivotal for re-establishing homeostasis after injury.


Asunto(s)
Colágeno Tipo XII/genética , Piel/crecimiento & desarrollo , Factor de Crecimiento Transformador beta/genética , Cicatrización de Heridas/genética , Animales , Colágeno Tipo I/genética , Matriz Extracelular , Fibroblastos/metabolismo , Fibroblastos/patología , Homeostasis/genética , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados/genética , Miofibroblastos/metabolismo , Piel/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...