Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Semin Cell Dev Biol ; 128: 130-136, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35400564

RESUMEN

Systemic sclerosis (SSc, scleroderma) is a complex disease with a pathogenic triad of autoimmunity, vasculopathy, and fibrosis involving the skin and multiple internal organs [1]. Because fibrosis accounts for as much as 45% of all deaths worldwide and appears to be increasing in prevalence [2], understanding its pathogenesis and progression is an urgent scientific challenge. Fibroblasts and myofibroblasts are the key effector cells executing physiologic tissue repair on one hand, and pathological fibrogenesis leading to chronic fibrosing conditions on the other. Recent studies identify innate immune signaling via toll-like receptors (TLRs) as a key driver of persistent fibrotic response in SSc. Repeated injury triggers the in-situ generation of "damage-associated molecular patterns" (DAMPs) or danger signals. Sensing of these danger signals by TLR4 on resident cells elicits potent stimulatory effects on fibrotic gene expression and myofibroblast differentiation triggering the self-limited tissue repair response to self-sustained pathological fibrosis characteristic of SSc. Our unbiased survey for DAMPs associated with SSc identified extracellular matrix glycoprotein tenascin-C as one of the most highly up-regulated ECM proteins in SSc skin and lung biopsies [3,4]. Furthermore, tenascin C is responsible for driving sustained fibroblasts activation, thereby progression of fibrosis [3]. This review summarizes recent studies examining the regulation and complex functional role of tenascin C, presenting tenascin-TLR4 axis in pathological fibrosis, and novel anti-fibrotic approaches targeting their signaling.


Asunto(s)
Esclerodermia Sistémica , Tenascina , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrosis , Humanos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Esclerodermia Sistémica/genética , Piel/metabolismo , Tenascina/genética , Receptor Toll-Like 4/metabolismo
2.
Neurobiol Dis ; 116: 60-68, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29705186

RESUMEN

Gliosis and fibrosis after spinal cord injury (SCI) lead to formation of a scar that is an impediment to axonal regeneration. Fibrotic scarring is characterized by the accumulation of fibronectin, collagen, and fibroblasts at the lesion site. The mechanisms regulating fibrotic scarring after SCI and its effects on axonal elongation and functional recovery are not well understood. In this study, we examined the effects of eliminating an isoform of fibronectin containing the Extra Domain A domain (FnEDA) on both fibrosis and on functional recovery after contusion SCI using male and female FnEDA-null mice. Eliminating FnEDA did not reduce the acute fibrotic response but markedly diminished chronic fibrotic scarring after SCI. Glial scarring was unchanged after SCI in FnEDA-null mice. We found that FnEDA was important for the long-term stability of the assembled fibronectin matrix during both the subacute and chronic phases of SCI. Motor functional recovery was significantly improved, and there were increased numbers of axons in the lesion site compared to wildtype mice, suggesting that the chronic fibrotic response is detrimental to recovery. Our data provide insight into the mechanisms of fibrosis after SCI and suggest that disruption of fibronectin matrix stability by targeting FnEDA represents a potential therapeutic strategy for promoting recovery after SCI.


Asunto(s)
Cicatriz/metabolismo , Cicatriz/patología , Fibronectinas/deficiencia , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Animales , Femenino , Fibronectinas/genética , Fibrosis/metabolismo , Fibrosis/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Recuperación de la Función/fisiología
3.
Curr Rheumatol Rep ; 17(1): 474, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25604573

RESUMEN

Pathological fibrosis is a distinguishing hallmark of systemic sclerosis (SSc) as well as a number of more common conditions. Fibrosis is a complex and dynamic process associated with immune dysregulation, vasculopathy, and uncontrolled extracellular matrix production leading to intractable scar formation in the skin and internal organs. Persistent or recurrent chemical, infectious, mechanical, or autoimmune injury in genetically predisposed individuals causes sustained fibroblasts activation. Innate immune signaling via toll-like receptors (TLRs) is increasingly recognized as a key player driving the persistent fibrotic response in SSc. In particular, expression of TLR4 as well as its endogenous ligands are elevated in lesional tissue from patients with SSc. Ligand-induced TLR4 activation elicits potent stimulatory effects on fibrotic gene expression and myofibroblast differentiation. Furthermore, TLR4 appears to sensitize fibroblasts to the profibrotic stimulatory effect of transforming growth factor-ß. This review highlights recent advances and emerging paradigms for understanding the regulation, complex functional roles, and therapeutic potential of TLRs in SSc pathogenesis.


Asunto(s)
Esclerodermia Sistémica/inmunología , Receptores Toll-Like/inmunología , Fibroblastos/inmunología , Fibrosis , Humanos , Inmunidad Innata , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología
4.
J Immunol ; 191(6): 2956-66, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23956427

RESUMEN

Activation of TLR3 by exogenous microbial ligands or endogenous injury-associated ligands leads to production of type I IFN. Scleroderma patients with progressive skin fibrosis display an IFN-regulated gene signature, implicating TLR3 signaling in the disease. In this study, we show that TLR3 expression was detected on foreskin, adult skin, and lung fibroblasts, and TLR3 levels were significantly elevated in a subset of scleroderma skin biopsies. In explanted skin and lung fibroblasts, the synthetic TLR3 ligand polyinosinic-polycytidylic acid (poly(I:C)), a dsRNA analog, caused dose- and time-dependent stimulation of IFN-ß production and generation of an IFN-response gene signature that was accompanied by substantial downregulation of collagen and α-smooth muscle actin gene expression. Furthermore, poly(I:C) abrogated TGF-ß-induced fibrotic responses and blocked canonical Smad signaling via upregulation of inhibitory Smad7. Surprisingly, the inhibitory effects of poly(I:C) in fibroblasts were independent of TLR3 and were mediated by the cytosolic receptors retinoic acid-inducible gene 1 and melanoma differentiation-associated gene 5, and involved signaling via the IFN receptor. Taken together, these results demonstrate that induction of a fibroblast IFN response gene signature triggered by dsRNA is associated with potent TLR3-independent anti-fibrotic effects. The characteristic IFN response gene signature seen in scleroderma lesions might therefore signify a tissue-autonomous protective attempt to restrict fibroblast activation during injury.


Asunto(s)
Comunicación Autocrina/fisiología , Fibroblastos/metabolismo , Interferones/metabolismo , Esclerodermia Sistémica/metabolismo , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Adulto , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/citología , Fibrosis/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Ligandos , Ratones , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Esclerodermia Sistémica/patología , Transducción de Señal/fisiología , Transcriptoma , Transfección
5.
Am J Pathol ; 183(4): 1197-1208, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23906810

RESUMEN

Members of the early growth response (Egr) gene family of transcription factors have nonredundant biological functions. Although Egr-3 is implicated primarily in neuromuscular development and immunity, its regulation and role in tissue repair and fibrosis has not been studied. We now show that in normal skin fibroblasts, Egr-3 was potently induced by transforming growth factor-ß via canonical Smad3. Moreover, transient Egr-3 overexpression was sufficient to stimulate fibrotic gene expression, whereas deletion of Egr-3 resulted in substantially attenuated transforming growth factor-ß responses. Genome-wide expression profiling in fibroblasts showed that genes associated with tissue remodeling and wound healing were prominently up-regulated by Egr-3. Notably, <5% of fibroblast genes regulated by Egr-1 or Egr-2 were found to be coregulated by Egr-3, revealing substantial functional divergence among these Egr family members. In a mouse model of scleroderma, development of dermal fibrosis was accompanied by accumulation of Egr-3-positive myofibroblasts in the lesional tissue. Moreover, skin biopsy samples from patients with scleroderma showed elevated Egr-3 levels in the dermis, and Egr-3 mRNA levels correlated with the extent of skin involvement. These results provide the first evidence that Egr-3, a functionally distinct member of the Egr family with potent effects on inflammation and immunity, is up-regulated in scleroderma and is necessary and sufficient for profibrotic responses, suggesting important and distinct roles in the pathogenesis of fibrosis.


Asunto(s)
Proteína 3 de la Respuesta de Crecimiento Precoz/metabolismo , Piel/metabolismo , Piel/patología , Factor de Crecimiento Transformador beta/farmacología , Adulto , Animales , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 3 de la Respuesta de Crecimiento Precoz/genética , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Piel/efectos de los fármacos , Proteínas Smad/metabolismo
6.
Am J Pathol ; 182(1): 192-205, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23141927

RESUMEN

Because recent studies implicate Toll-like receptors (TLRs) in the pathogenesis of fibrosis, we sought to investigate the in vitro and in vivo role and mechanism of TLR4-mediated fibroblast responses in fibrogenesis. We found that TLR4 was constitutively expressed, and accumulation of endogenous TLR4 ligands significantly elevated, in lesional skin and lung tissues from patients with scleroderma. Activation of TLR4 signaling in explanted fibroblasts resulted in enhanced collagen synthesis and increased expression of multiple genes involved in tissue remodeling and extracellular matrix homeostasis. Moreover, TLR4 dramatically enhanced the sensitivity of fibroblasts to the stimulatory effect of transforming growth factor-ß1. These profibrotic responses were abrogated by both genetic and pharmacological disruption of TLR4 signaling in vitro, and skin fibrosis induced by bleomycin in vivo was attenuated in mice harboring a mutated TLR4. Activation of TLR4 in fibroblasts augmented the intensity of canonical Smad signaling, and was accompanied by suppression of anti-fibrotic microRNA expression. Together, these results suggest a novel model to account for persistent fibrogenesis in scleroderma, in which activation of fibroblast TLR4 signaling, triggered by damage-associated endogenous TLR4 ligands, results in augmented transforming growth factor-ß1 sensitivity with increased matrix production and progressive connective tissue remodeling. Under these conditions, fibroblast TLR4 serves as the switch for converting self-limited tissue repair into intractable fibrosis.


Asunto(s)
Esclerodermia Sistémica/metabolismo , Receptor Toll-Like 4/fisiología , Factor de Crecimiento Transformador beta/farmacología , Adulto , Anciano , Animales , Biopsia , Bleomicina , Células Cultivadas , Colágeno/biosíntesis , Matriz Extracelular/fisiología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Ligandos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Mutantes , MicroARNs/biosíntesis , MicroARNs/genética , Persona de Mediana Edad , Esclerodermia Sistémica/inducido químicamente , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología , Transducción de Señal/fisiología , Piel/metabolismo , Piel/patología , Receptor Toll-Like 4/antagonistas & inhibidores
7.
J Pathol ; 229(2): 286-97, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23132749

RESUMEN

Fibroblasts and myofibroblasts are the key effector cells executing physiological tissue repair leading to regeneration on the one hand, and pathological fibrogenesis leading to chronic fibrosing conditions on the other. Recent studies identify the multifunctional transcription factor early growth response-1(Egr-1) as an important mediator of fibroblast activation triggered by diverse stimuli. Egr-1 has potent stimulatory effects on fibrotic gene expression, and aberrant Egr-1 expression or function is associated with animal models of fibrosis and human fibrotic disorders, including emphysema, pulmonary fibrosis, pulmonary hypertension and systemic sclerosis. Pharmacological suppression or genetic targeting of Egr-1 blocks fibrotic responses in vitro and ameliorates experimental fibrosis in the skin and lung. In contrast, Egr-1 appears to act as a negative regulator of hepatic fibrosis in mouse models, suggesting a context-dependent role in fibrosis. The Egr-1-binding protein Nab2 is an endogenous inhibitor of Egr-1-mediated signalling and abrogates the stimulation of fibrotic responses induced by transforming growth factor-ß (TGFß). Moreover, mice deficient in Nab2 show excessive collagen accumulation in the skin. These observations highlight a previously unsuspected fundamental physiological function for the Egr-1-Nab2 signalling axis in regulating fibrogenesis, and suggest that Egr-1 may be a potential novel therapeutic target in human diseases complicated by fibrosis. This review summarizes recent advances in understanding the regulation and complex functional role of Egr-1 and its related proteins and inhibitors in pathological fibrosis.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Fibroblastos/metabolismo , Regeneración , Transducción de Señal , Animales , Colágeno/metabolismo , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/química , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Retroalimentación Fisiológica , Fibroblastos/patología , Fibrosis , Humanos , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Conformación Proteica , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Factores de Transcripción p300-CBP/metabolismo
9.
J Invest Dermatol ; 143(10): 1877-1885, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37452808

RESUMEN

Damage-associated molecular patterns (DAMPs) are intracellular molecules released under cellular stress or recurring tissue injury, which serve as endogenous ligands for toll-like receptors (TLRs). Such DAMPs are either actively secreted by immune cells or passively released into the extracellular environment from damaged cells or generated as alternatively spliced mRNA variants of extracellular matrix (ECM) glycoproteins. When recognized by pattern recognition receptors (PRRs) such as TLRs, DAMPs trigger innate immune responses. Currently, the best-characterized PRRs include, in addition to TLRs, nucleotide-binding oligomerization domain-like receptors, RIG-I-like RNA helicases, C-type lectin receptors, and many more. Systemic sclerosis (SSc) is a chronic autoimmune condition characterized by inflammation and progressive fibrosis in multiple organs. Using an unbiased survey for SSc-associated DAMPs, we have identified the ECM glycoproteins fibronectin-containing extra domain A and tenascin C as the most highly upregulated in SSc skin and lung biopsies. These DAMPs activate TLR4 on resident stromal cells to elicit profibrotic responses and sustained myofibroblasts activation resulting in progressive fibrosis. This review summarizes the current understanding of the complex functional roles of DAMPs in the progression and failure of resolution of fibrosis in general, with a particular focus on SSc, and considers viable therapeutic approaches targeting DAMPs.


Asunto(s)
Esclerodermia Sistémica , Transducción de Señal , Humanos , Receptores Toll-Like , Receptores de Reconocimiento de Patrones , Fibrosis , Matriz Extracelular , Alarminas , Glicoproteínas
10.
JCI Insight ; 8(14)2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37306632

RESUMEN

Multiorgan fibrosis in systemic sclerosis (SSc) accounts for substantial mortality and lacks effective therapies. Lying at the crossroad of TGF-ß and TLR signaling, TGF-ß-activated kinase 1 (TAK1) might have a pathogenic role in SSc. We therefore sought to evaluate the TAK1 signaling axis in patients with SSc and to investigate pharmacological TAK1 blockade using a potentially novel drug-like selective TAK1 inhibitor, HS-276. Inhibiting TAK1 abrogated TGF-ß1 stimulation of collagen synthesis and myofibroblasts differentiation in healthy skin fibroblasts, and it ameliorated constitutive activation of SSc skin fibroblasts. Moreover, treatment with HS-276 prevented dermal and pulmonary fibrosis and reduced the expression of profibrotic mediators in bleomycin-treated mice. Importantly, initiating HS-276 treatment even after fibrosis was already established prevented its progression in affected organs. Together, these findings implicate TAK1 in the pathogenesis of SSc and identify targeted TAK1 inhibition using a small molecule as a potential strategy for the treatment of SSc and other fibrotic diseases.


Asunto(s)
Fibrosis Pulmonar , Esclerodermia Sistémica , Ratones , Animales , Fibrosis , Esclerodermia Sistémica/patología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/prevención & control , Fibrosis Pulmonar/metabolismo , Fibroblastos/metabolismo
11.
Am J Pathol ; 178(5): 2077-90, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21514423

RESUMEN

Although the early growth response-2 (Egr-2, alias Krox20) protein shows structural and functional similarities to Egr-1, these two related early-immediate transcription factors are nonredundant. Egr-2 plays essential roles in peripheral nerve myelination, adipogenesis, and immune tolerance; however, its regulation and role in tissue repair and fibrosis remain poorly understood. We show herein that transforming growth factor (TGF)-ß induced a Smad3-dependent sustained stimulation of Egr2 gene expression in normal fibroblasts. Overexpression of Egr-2 was sufficient to stimulate collagen gene expression and myofibroblast differentiation, whereas these profibrotic TGF-ß responses were attenuated in Egr-2-depleted fibroblasts. Genomewide transcriptional profiling revealed that multiple genes associated with tissue remodeling and wound healing were up-regulated by Egr-2, but the Egr-2-regulated gene expression profile overlapped only partially with the Egr-1-regulated gene profile. Levels of Egr-2 were elevated in lesional tissue from mice with bleomycin-induced scleroderma. Moreover, elevated Egr-2 was noted in biopsy specimens of skin and lung from patients with systemic sclerosis. These results provide the first evidence that Egr-2 is a functionally distinct transcription factor that is both necessary and sufficient for TGF-ß-induced profibrotic responses and is aberrantly expressed in lesional tissue in systemic sclerosis and in a murine model of scleroderma. Together, these findings suggest that Egr-2 plays an important nonredundant role in the pathogenesis of fibrosis. Targeting Egr-2 might represent a novel therapeutic strategy to control fibrosis.


Asunto(s)
Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Western Blotting , Células Cultivadas , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Fibrosis , Humanos , Inmunohistoquímica , Ratones , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esclerodermia Sistémica/genética , Transcripción Genética , Transfección , Regulación hacia Arriba
12.
Clin Exp Rheumatol ; 30(2 Suppl 71): S86-96, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22691216

RESUMEN

OBJECTIVES: Systemic sclerosis (SSc) is a heterogeneous multifactorial disease dominated by progressive skin and internal organ fibrosis that is driven in part by transforming growth factor-beta (TGF-ß). An important downstream target of TGF-ß is the Abelson (c-Abl) tyrosine kinase, and its inhibition by imatinib mesylate (Gleevec) attenuates fibrosis in mice. Here we examined the effect of c-Abl activation and blockade in explanted healthy control and SSc fibroblasts. METHODS: Skin biopsies and explanted fibroblasts from healthy subjects and patients with SSc were studied. Changes in genome-wide expression patterns in imatinib-treated control and SSc fibroblasts were analysed by DNA microarray. RESULTS: Treatment of control fibroblasts with TGF-ß resulted in activation of c-Abl and stimulation of fibrotic gene expression that was prevented by imatinib. Moreover, imatinib reduced basal collagen gene expression in SSc but not control fibroblasts. No significant differences in tissue levels of c-Abl and phospho-c-Abl were detected between SSc and control skin biopsies. In vitro, imatinib induced dramatic changes in the expression of genes involved in fibrosis, cardiovascular disease, inflammation, and lipid and cholesterol metabolism. Remarkably, of the 587-imatinib-responsive genes, 91% showed significant change in SSc fibroblasts, but only 12% in control fibroblasts. CONCLUSIONS: c-Abl plays a key role in fibrotic responses. Imatinib treatment results in dramatic changes in gene expression in SSc fibroblasts but has only modest effects in control fibroblasts. These data provide novel insights into the mechanisms underlying the antifibrotic effect of imatinib in SSc.


Asunto(s)
Fibroblastos/efectos de los fármacos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Pirimidinas/farmacología , Esclerodermia Sistémica/genética , Piel/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Animales , Benzamidas , Biopsia , Estudios de Casos y Controles , Células Cultivadas , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mesilato de Imatinib , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Proteínas Proto-Oncogénicas c-abl/deficiencia , Proteínas Proto-Oncogénicas c-abl/genética , Esclerodermia Sistémica/enzimología , Esclerodermia Sistémica/patología , Transducción de Señal/efectos de los fármacos , Piel/enzimología , Piel/patología , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo
13.
JCI Insight ; 7(21)2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36136452

RESUMEN

Activation of TLR4 by its cognate damage-associated molecular patterns (DAMPs) elicits potent profibrotic effects and myofibroblast activation in systemic sclerosis (SSc), while genetic targeting of TLR4 or its DAMPs in mice accelerates fibrosis resolution. To prevent aberrant DAMP/TLR4 activity, a variety of negative regulators evolved to dampen the magnitude and duration of the signaling. These include radioprotective 105 kDa (RP105), a transmembrane TLR4 homolog that competitively inhibits DAMP recognition of TLR4, blocking TLR4 signaling in immune cells. The role of RP105 in TLR4-dependent fibrotic responses in SSc is unknown. Using unbiased transcriptome analysis of skin biopsies, we found that levels of both TLR4 and its adaptor protein MD2 were elevated in SSc skin and significantly correlated with each other. Expression of RP105 was negatively associated with myofibroblast differentiation in SSc. Importantly, RP105-TLR4 association was reduced, whereas TLR4-TLR4 showed strong association in fibroblasts from patients with SSc, as evidenced by PLA assays. Moreover, RP105 adaptor MD1 expression was significantly reduced in SSc skin biopsies and explanted SSc skin fibroblasts. Exogenous RP105-MD1 abrogated, while loss of RP105 exaggerated, fibrotic cellular responses. Importantly, ablation of RP105 in mice was associated with augmented TLR4 signaling and aggravated skin fibrosis in complementary disease models. Thus, we believe RP105-MD1 to be a novel cell-intrinsic negative regulator of TLR4-MD2-driven sustained fibroblast activation, representing a critical regulatory network governing the fibrotic process. Impaired RP105 function in SSc might contribute to persistence of progression of the disease.


Asunto(s)
Esclerodermia Sistémica , Receptor Toll-Like 4 , Ratones , Animales , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Fibrosis , Fibroblastos/metabolismo , Transducción de Señal , Alarminas/metabolismo
14.
JCI Insight ; 7(5)2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35104243

RESUMEN

Systemic sclerosis (SSc) is a chronic, multisystem orphan disease with a highly variable clinical course, high mortality rate, and a poorly understood complex pathogenesis. We have identified an important role for a subpopulation of monocytes and macrophages characterized by surface expression of the scavenger receptor macrophage receptor with collagenous structure (MARCO) in chronic inflammation and fibrosis in SSc and in preclinical disease models. We show that MARCO+ monocytes and macrophages accumulate in lesional skin and lung in topographic proximity to activated myofibroblasts in patients with SSc and in the bleomycin-induced mouse model of SSc. Short-term treatment of mice with a potentially novel nanoparticle, poly(lactic-co-glycolic) acid (PLG), which is composed of a carboxylated, FDA-approved, biodegradable polymer and modulates activation and trafficking of MARCO+ inflammatory monocytes, markedly attenuated bleomycin-induced skin and lung inflammation and fibrosis. Mechanistically, in isolated cells in culture, PLG nanoparticles inhibited TGF-dependent fibrotic responses in vitro. Thus, MARCO+ monocytes are potent effector cells of skin and lung fibrosis and can be therapeutically targeted in SSc using PLG nanoparticles.


Asunto(s)
Nanopartículas , Esclerodermia Sistémica , Animales , Bleomicina/toxicidad , Fibroblastos/metabolismo , Fibrosis , Humanos , Ratones , Monocitos/metabolismo , Receptores Inmunológicos/metabolismo , Esclerodermia Sistémica/tratamiento farmacológico
15.
Nat Commun ; 13(1): 6358, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36289219

RESUMEN

In addition to autoimmune and inflammatory diseases, variants of the TNFAIP3 gene encoding the ubiquitin-editing enzyme A20 are also associated with fibrosis in systemic sclerosis (SSc). However, it remains unclear how genetic factors contribute to SSc pathogenesis, and which cell types drive the disease due to SSc-specific genetic alterations. We therefore characterize the expression, function, and role of A20, and its negative transcriptional regulator DREAM, in patients with SSc and disease models. Levels of A20 are significantly reduced in SSc skin and lungs, while DREAM is elevated. In isolated fibroblasts, A20 mitigates ex vivo profibrotic responses. Mice haploinsufficient for A20, or harboring fibroblasts-specific A20 deletion, recapitulate major pathological features of SSc, whereas DREAM-null mice with elevated A20 expression are protected. In DREAM-null fibroblasts, TGF-ß induces the expression of A20, compared to wild-type fibroblasts. An anti-fibrotic small molecule targeting cellular adiponectin receptors stimulates A20 expression in vitro in wild-type but not A20-deficient fibroblasts and in bleomycin-treated mice. Thus, A20 has a novel cell-intrinsic function in restraining fibroblast activation, and together with DREAM, constitutes a critical regulatory network governing the fibrotic process in SSc. A20 and DREAM represent novel druggable targets for fibrosis therapy.


Asunto(s)
Receptores de Adiponectina , Esclerodermia Sistémica , Animales , Ratones , Bleomicina , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis , Ratones Noqueados , Receptores de Adiponectina/metabolismo , Esclerodermia Sistémica/metabolismo , Transducción de Señal/genética , Piel/patología , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitinas/metabolismo
16.
iScience ; 24(1): 101902, 2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33385109

RESUMEN

The processes underlying synchronous multiple organ fibrosis in systemic sclerosis (SSc) remain poorly understood. Age-related pathologies are associated with organismal decline in nicotinamide adenine dinucleotide (NAD+) that is due to dysregulation of NAD+ homeostasis and involves the NADase CD38. We now show that CD38 is upregulated in patients with diffuse cutaneous SSc, and CD38 levels in the skin associate with molecular fibrosis signatures, as well as clinical fibrosis scores, while expression of key NAD+-synthesizing enzymes is unaltered. Boosting NAD+ via genetic or pharmacological CD38 targeting or NAD+ precursor supplementation protected mice from skin, lung, and peritoneal fibrosis. In mechanistic experiments, CD38 was found to reduce NAD+ levels and sirtuin activity to augment cellular fibrotic responses, while inhibiting CD38 had the opposite effect. Thus, we identify CD38 upregulation and resulting disrupted NAD+ homeostasis as a fundamental mechanism driving fibrosis in SSc, suggesting that CD38 might represent a novel therapeutic target.

17.
Curr Opin Rheumatol ; 22(6): 671-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20693905

RESUMEN

PURPOSE OF REVIEW: Progressive organ fibrosis and pulmonary arterial hypertension (PAH) are the leading causes of death in patients with systemic sclerosis (SSc). However, the pathogenesis and the link between these two processes remain obscure. A better understanding of these events is needed in order to facilitate the discovery and development of effective therapies for SSc. RECENT FINDINGS: Recent reports provide evidence that the orphan receptor peroxisome proliferator-activated receptor γ (PPARγ), better known for its pivotal role in metabolism, has potent effects on inflammation, fibrogenesis and vascular remodeling and is important in the pathogenesis of fibrosis and PAH, and as a potential therapeutic target in SSc. The studies discussed in this review indicate that ligands of PPARγ potently modulate connective tissue turnover and suggest that aberrant expression or function of PPARγ is associated with, and very likely contributes to, the progression of pathological fibrosis and vascular remodeling. These observations are of particularly relevance because FDA-approved drugs of the thiazolidinedione class currently used for the treatment of obesity-associated type 2 diabetes activate PPARγ signaling. Moreover, novel PPARγ ligands with selective activity are under development or in clinical trials for inflammatory diseases, asthma, Alzheimer disease and cancer. SUMMARY: Drugs targeting the PPARγ pathway might be effective for the control of fibrosis as well as pathological vascular remodeling underlying PAH and, therefore, might have a therapeutic potential in SSc. A greater understanding of the mechanisms underlying the antifibrogenic and vascular remodeling activities of PPARγ ligands will be necessary in order to advance these drugs into clinical use.


Asunto(s)
Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/fisiología , PPAR gamma/antagonistas & inhibidores , PPAR gamma/fisiología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/tendencias , Fibrosis , Humanos , Esclerodermia Sistémica/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
18.
Am J Pathol ; 175(3): 1041-55, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19679873

RESUMEN

The early growth response gene (Egr-1) codes for a zinc finger transcription factor that has important roles in the regulation of cell growth, differentiation, and survival. Aberrant Egr-1 expression is implicated in carcinogenesis, inflammation, atherosclerosis, and ischemic injury. We reported previously that normal fibroblasts stimulated by transforming growth factor-ss showed rapid and transient induction of Egr-1. Moreover, we observed that tissue expression of Egr-1 was elevated in patients with scleroderma, which suggests that Egr-1 may be involved in tissue repair and fibrosis. Here, we investigated matrix remodeling and wound healing in mice harboring gain of function or loss of function mutations of Egr-1. Using the model of bleomycin-induced scleroderma, we found that the early influx of inflammatory cells into the skin and lungs, and the subsequent development of fibrosis in these organs, were markedly attenuated in Egr-1 null mice. Furthermore, full-thickness incisional skin wound healing was impaired, and skin fibroblasts lacking Egr-1 showed reduced migration and myofibroblast transdifferentiation in vitro. In contrast, transgenic mice with fibroblast-specific Egr-1 overexpression showed exuberant tissue repair, with enhanced collagen accumulation and increased tensile strength of incisional wounds. Together, these results point to the fundamental role that Egr-1 plays in the regulation of transforming growth factor-ss-dependent physiological and pathological matrix remodeling.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Fibroblastos/metabolismo , Enfermedades Pulmonares/patología , Enfermedades de la Piel/patología , Cicatrización de Heridas/genética , Animales , Bleomicina/efectos adversos , Diferenciación Celular/genética , Movimiento Celular/genética , Células Cultivadas , Colágeno , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Femenino , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Inmunohistoquímica , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/genética , Masculino , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Enfermedades de la Piel/inducido químicamente , Enfermedades de la Piel/genética
19.
FASEB J ; 23(9): 2968-77, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19395477

RESUMEN

Ligands of peroxisome proliferator-activated receptor-gamma (PPAR-gamma) abrogate the stimulation of collagen gene transcription induced by transforming growth factor-beta (TGF-beta). Here, we delineate the mechanisms underlying this important novel physiological function for PPAR-gamma in connective tissue homeostasis. First, we demonstrated that antagonistic regulation of TGF-beta activity by PPAR-gamma ligands involves cellular PPAR-gamma, since 15-deoxy-Delta12,14-prostaglandin J(2) (15d-PGJ(2)) failed to block TGF-beta-induced responses in either primary cultures of PPAR-gamma-null murine embryonic fibroblasts, or in normal human skin fibroblasts with RNAi-mediated knockdown of PPAR-gamma. Next, we examined the molecular basis underlying the abrogation of TGF-beta signaling by PPAR-gamma in normal human fibroblasts in culture. The results demonstrated that Smad-dependent transcriptional responses were blocked by PPAR-gamma without preventing Smad2/3 activation. In contrast, the interaction between activated Smad2/3 and the transcriptional coactivator and histone acetyltransferase p300 induced by TGF-beta, and the accumulation of p300 on consensus Smad-binding DNA sequences and histone H4 hyperacetylation at the COL1A2 locus, were all prevented by PPAR-gamma. Wild-type p300, but not a mutant form of p300 lacking functional histone acetyltransferase, was able to restore TGF-beta-induced stimulation of COL1A2 in the presence of PPAR-gamma ligands. Collectively, these results indicate that PPAR-gamma blocked Smad-mediated transcriptional responses by preventing p300 recruitment and histone H4 hyperacetylation, resulting in the inhibition of TGF-beta-induced collagen gene expression. Pharmacological activation of PPAR-gamma thus may represent a novel therapeutic approach to target p300-dependent TGF-beta profibrotic responses such as stimulation of collagen gene expression.


Asunto(s)
Colágeno/genética , PPAR gamma/fisiología , Proteínas Smad/fisiología , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Histonas/metabolismo , Humanos , Ratones , Transporte de Proteínas , Activación Transcripcional , Factor de Crecimiento Transformador beta/fisiología , Factores de Transcripción p300-CBP/genética
20.
Comp Funct Genomics ; : 246738, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20454581

RESUMEN

Antheraea mylitta is one of the wild nonmulberry silkworms, which produces tasar silk. An EST project has been undertaken to understand the gene expression profile of A. mylitta silk gland. Two cDNA libraries, one from the whole bodies of one-day-old larvae and the other from the silkglands of fifth instar larvae, were constructed and sequenced. A total of 2476 good-quality ESTs (1239 clones) were obtained and grouped into 648 clusters containing 390 contigs and 258 singletons to represent 467 potential unigenes. Forty-five sequences contained putative coding region, and represented potentially novel genes. Among the 648 clusters, 241 were categorized according to Gene Ontology hierarchy and showed presence of several silk and immune-related genes. The A. mylitta ESTs have been organized into a freely available online database "AmyBASE". These data provide an initial insight into the A. mylitta transcriptome and help to understand the molecular mechanism of silk protein production in a Lepidopteran species.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA