Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.344
Filtrar
1.
Int J Mol Sci ; 25(15)2024 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-39126067

RESUMEN

The keratinocyte carcinomas, basal cell carcinoma (BCC), and cutaneous squamous cell carcinoma (cSCC), are the most common cancers in humans. Recently, an increasing body of literature has investigated the role of miRNAs in keratinocyte carcinoma pathogenesis, progression and their use as therapeutic agents and targets, or biomarkers. However, there is very little consistency in the literature regarding the identity of and/or role of individual miRNAs in cSCC (and to a lesser extent BCC) biology. miRNA analyses that combine clinical evidence with experimental elucidation of targets and functional impact provide far more compelling evidence than studies purely based on clinical findings or bioinformatic analyses. In this study, we review the clinical evidence associated with miRNA dysregulation in KCs, assessing the quality of validation evidence provided, identify gaps, and provide recommendations for future studies based on relevant studies that investigated miRNA levels in human cSCC and BCC. Furthermore, we demonstrate how miRNAs contribute to the regulation of a diverse network of cellular functions, and that large-scale changes in tumor cell biology can be attributed to miRNA dysregulation. We highlight the need for further studies investigating the role of miRNAs as communicators between different cell types in the tumor microenvironment. Finally, we explore the clinical benefits of miRNAs as biomarkers of keratinocyte carcinoma prognosis and treatment.


Asunto(s)
Biomarcadores de Tumor , Carcinoma Basocelular , Carcinoma de Células Escamosas , Regulación Neoplásica de la Expresión Génica , Queratinocitos , MicroARNs , Neoplasias Cutáneas , Humanos , MicroARNs/genética , Queratinocitos/metabolismo , Queratinocitos/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Carcinoma Basocelular/genética , Carcinoma Basocelular/patología , Carcinoma Basocelular/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Biomarcadores de Tumor/genética , Microambiente Tumoral/genética , Pronóstico
2.
Arch Dermatol Res ; 316(8): 519, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39136792

RESUMEN

Vitiligo is an autoimmune disorder characterized by epidermal melanocyte damage, with the typical clinical manifestation of white patches of skin. Keratinocytes, which work in concert with melanocytes to maintain the structural and functional integrity of the skin, are implicated in the progression of vitiligo. Recent studies have reported abnormal keratinocyte proliferation and epidermal thickening in some patients with vitiligo; however, the relationship between these changes and the clinical characteristics of vitiligo remains unclear. We assessed the changes in epidermal thickness in patients with vitiligo and their correlation with clinical characteristics. Compared to the non-lesional skins, the stratum corneum, viable epidermis, and full epidermis in the lesional skins were all significantly thicker. The thickness of the stratum corneum in the head, neck, and trunk was greatly lower than that in the extremities. The thickness of the stratum corneum in the sun-exposed area was higher than that in the sun-protected area, whereas the thickness of the viable epidermis decreased. In conclusion, our study found that the epidermis in the lesional skins of patients with vitiligo was significantly thickened, especially in the sun-exposed areas and extremities.


Asunto(s)
Epidermis , Vitíligo , Humanos , Vitíligo/patología , Vitíligo/diagnóstico , Epidermis/patología , Masculino , Adulto , Femenino , Persona de Mediana Edad , Adulto Joven , Adolescente , Melanocitos/patología , Queratinocitos/patología , Niño , Luz Solar/efectos adversos , Anciano
3.
Cell Commun Signal ; 22(1): 386, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39090602

RESUMEN

BACKGROUND: T-LAK cell-oriented protein kinase (TOPK) strongly promotes the malignant proliferation of cancer cells and is recognized as a promising biomarker of tumor progression. Psoriasis is a common inflammatory skin disease featured by excessive proliferation of keratinocytes. Although we have previously reported that topically inhibiting TOPK suppressed psoriatic manifestations in psoriasis-like model mice, the exact role of TOPK in psoriatic inflammation and the underlying mechanism remains elusive. METHODS: GEO datasets were analyzed to investigate the association of TOPK with psoriasis. Skin immunohistochemical (IHC) staining was performed to clarify the major cells expressing TOPK. TOPK conditional knockout (cko) mice were used to investigate the role of TOPK-specific deletion in IMQ-induced psoriasis-like dermatitis in mice. Flow cytometry was used to analyze the alteration of psoriasis-related immune cells in the lesional skin. Next, the M5-induced psoriasis cell model was used to identify the potential mechanism by RNA-seq, RT-RCR, and western blotting. Finally, the neutrophil-neutralizing antibody was used to confirm the relationship between TOPK and neutrophils in psoriasis-like dermatitis in mice. RESULTS: We found that TOPK levels were strongly associated with the progression of psoriasis. TOPK was predominantly increased in the epidermal keratinocytes of psoriatic lesions, and conditional knockout of TOPK in keratinocytes suppressed neutrophils infiltration and attenuated psoriatic inflammation. Neutrophils deletion by neutralizing antibody greatly diminished the suppressive effect of TOPK cko in psoriasis-like dermatitis in mice. In addition, topical application of TOPK inhibitor OTS514 effectively attenuated already-established psoriasis-like dermatitis in mice. Mechanismly, RNA-seq revealed that TOPK regulated the expression of some genes in the IL-17 signaling pathway, such as neutrophils chemokines CXCL1, CXCL2, and CXCL8. TOPK modulated the expression of neutrophils chemokines via activating transcription factors STAT3 and NF-κB p65 in keratinocytes, thereby promoting neutrophils infiltration and psoriasis progression. CONCLUSIONS: This study identified a crucial role of TOPK in psoriasis by regulating neutrophils infiltration, providing new insights into the pathogenesis of psoriasis.


Asunto(s)
Queratinocitos , Quinasas de Proteína Quinasa Activadas por Mitógenos , Infiltración Neutrófila , Psoriasis , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Imiquimod , Queratinocitos/patología , Queratinocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Neutrófilos/patología , Psoriasis/patología , Psoriasis/genética , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Regulación hacia Arriba , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo
4.
Cell Commun Signal ; 22(1): 400, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143467

RESUMEN

A comprehensive understanding of the intricate cellular and molecular changes governing the complex interactions between cells within acne lesions is currently lacking. Herein, we analyzed early papules from six subjects with active acne vulgaris, utilizing single-cell and high-resolution spatial RNA sequencing. We observed significant changes in signaling pathways across seven different cell types when comparing lesional skin samples (LSS) to healthy skin samples (HSS). Using CellChat, we constructed an atlas of signaling pathways for the HSS, identifying key signal distributions and cell-specific genes within individual clusters. Further, our comparative analysis revealed changes in 49 signaling pathways across all cell clusters in the LSS- 4 exhibited decreased activity, whereas 45 were upregulated, suggesting that acne significantly alters cellular dynamics. We identified ten molecules, including GRN, IL-13RA1 and SDC1 that were consistently altered in all donors. Subsequently, we focused on the function of GRN and IL-13RA1 in TREM2 macrophages and keratinocytes as these cells participate in inflammation and hyperkeratinization in the early stages of acne development. We evaluated their function in TREM2 macrophages and the HaCaT cell line. We found that GRN increased the expression of proinflammatory cytokines and chemokines, including IL-18, CCL5, and CXCL2 in TREM2 macrophages. Additionally, the activation of IL-13RA1 by IL-13 in HaCaT cells promoted the dysregulation of genes associated with hyperkeratinization, including KRT17, KRT16, and FLG. These findings suggest that modulating the GRN-SORT1 and IL-13-IL-13RA1 signaling pathways could be a promising approach for developing new acne treatments.


Asunto(s)
Acné Vulgar , Piel , Humanos , Acné Vulgar/genética , Acné Vulgar/patología , Acné Vulgar/metabolismo , Piel/patología , Piel/metabolismo , Transducción de Señal/genética , Masculino , Macrófagos/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Subunidad alfa1 del Receptor de Interleucina-13/genética , Subunidad alfa1 del Receptor de Interleucina-13/metabolismo , Femenino
5.
Acta Derm Venereol ; 104: adv40601, 2024 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-39101340

RESUMEN

Keratinocyte-derived skin cancers comprise basal cell carcinoma, squamous cell carcinoma, its precursor actinic keratosis, and Bowen's disease. Historically, this group of neoplasms has been subsumed under the term non-melanoma skin cancer. However, the term non-melanoma skin cancer can be misleading and lacks precision. Therefore, more precise and reasonable terminology, valuing the relevance of keratinocyte-derived cancer, appears pertinent to meet its clinical and scientific significance. A group of experienced dermato-oncologists initiated a consensus approach to promote the use of the term "keratinocyte cancer" instead of "non-melanoma skin cancer" when referring to carcinomas and their precursors that are derived from keratinocytes. The vote among members of the consensus group indicated unanimous agreement on the consistent use of the term "keratinocyte cancer" instead of "non-melanoma skin cancer". International delegates also voted in favour of the revised terminology. The more precise and, by means of etiopathogenesis, correct term "keratinocyte cancer" should be consistently used for malignancies originated from keratinocytes. This is expected to have a positive impact on patient-physician communication and gives better justice to this important group of keratinocyte-derived cancers.


Asunto(s)
Carcinoma Basocelular , Carcinoma de Células Escamosas , Consenso , Queratinocitos , Queratosis Actínica , Neoplasias Cutáneas , Terminología como Asunto , Humanos , Neoplasias Cutáneas/patología , Queratinocitos/patología , Carcinoma Basocelular/patología , Carcinoma de Células Escamosas/patología , Queratosis Actínica/patología , Queratosis Actínica/diagnóstico , Enfermedad de Bowen/patología , Europa (Continente)
6.
Cell Death Dis ; 15(7): 535, 2024 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-39069531

RESUMEN

Atopic dermatitis (AD), a prevalent chronic inflammatory disease with multifactorial etiology, features epidermal barrier defects and immune overactivation. Synthetic glucocorticoids (GCs) are widely prescribed for treating AD due to their anti-inflammatory actions; however, mechanisms are incompletely understood. Defective local GC signaling due to decreased production of endogenous ligand and/or GC receptor (GR) levels was reported in prevalent inflammatory skin disorders; whether this is a consequence or contributing factor to AD pathology is unclear. To identify the chromatin-bound cell-type-specific GR protein interactome in keratinocytes, we used rapid immunoprecipitation of endogenous proteins and mass spectrometry identifying 145 interactors that increased upon dexamethasone treatment. GR-interacting proteins were enriched in p53/p63 signaling, including epidermal transcription factors with critical roles in AD pathology. Previous analyses indicating mirrored AD-like phenotypes between P63 overexpression and GR loss in epidermis, and our data show an intricate relationship between these transcription factors in human keratinocytes, identifying TP63 as a direct GR target. Dexamethasone treatment counteracted transcriptional up-regulation of inflammatory markers by IL4/IL13, known to mimic AD, causing opposite shifts in GR and P63 genomic binding. Indeed, IL4/IL13 decreased GR and increased P63 levels in cultured keratinocytes and human epidermal equivalents (HEE), consistent with GR down-regulation and increased P63 expression in AD lesions vs normal skin. Moreover, GR knockdown (GRKD) resulted in constitutive increases in P63, phospho-P38 and S100A9, IL6, and IL33. Also, GRKD culture supernatants showed increased autocrine production of TH2-/TH1-/TH17-TH22-associated factors including IL4, CXCL10, CXCL11, and CXCL8. GRKD HEEs showed AD-like features including hyperplasia and abnormal differentiation, resembling phenotypes observed with GR antagonist or IL4/IL13 treatment. The simultaneous GR/P63 knockdown partially reversed constitutive up-regulation of inflammatory genes in GRKD. In summary, our data support a causative role for GR loss in AD pathogenesis via functional interactions with P63 and autocrine signaling in epidermal keratinocytes.


Asunto(s)
Comunicación Autocrina , Dermatitis Atópica , Dexametasona , Queratinocitos , Receptores de Glucocorticoides , Queratinocitos/metabolismo , Queratinocitos/patología , Humanos , Dermatitis Atópica/patología , Dermatitis Atópica/metabolismo , Dermatitis Atópica/genética , Receptores de Glucocorticoides/metabolismo , Dexametasona/farmacología , Epidermis/metabolismo , Epidermis/patología , Inflamación/patología , Inflamación/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética
8.
Sci Adv ; 10(27): eado2365, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38959302

RESUMEN

Pityriasis rubra pilaris (PRP) is a rare inflammatory skin disease with a poorly understood pathogenesis. Through a molecularly driven precision medicine approach and an extensive mechanistic pathway analysis in PRP skin samples, compared to psoriasis, atopic dermatitis, healed PRP, and healthy controls, we identified IL-1ß as a key mediator, orchestrating an NF-κB-mediated IL-1ß-CCL20 axis, including activation of CARD14 and NOD2. Treatment of three patients with the IL-1 antagonists anakinra and canakinumab resulted in rapid clinical improvement and reversal of the PRP-associated molecular signature with a 50% improvement in skin lesions after 2 to 3 weeks. This transcriptional signature was consistent with in vitro stimulation of keratinocytes with IL-1ß. With the central role of IL-1ß underscoring its potential as a therapeutic target, our findings propose a redefinition of PRP as an autoinflammatory keratinization disorder. Further clinical trials are needed to validate the efficacy of IL-1ß antagonists in PRP.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1beta , Queratinocitos , Pitiriasis Rubra Pilaris , Humanos , Pitiriasis Rubra Pilaris/tratamiento farmacológico , Pitiriasis Rubra Pilaris/patología , Pitiriasis Rubra Pilaris/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Queratinocitos/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Masculino , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/antagonistas & inhibidores , Femenino , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Piel/patología , Piel/metabolismo , Piel/efectos de los fármacos , Interleucina-1/antagonistas & inhibidores , Interleucina-1/metabolismo , Interleucina-1/genética , Persona de Mediana Edad , Guanilato Ciclasa/metabolismo , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/genética , Adulto , Transducción de Señal/efectos de los fármacos , Proteínas de la Membrana
10.
Cell Death Dis ; 15(6): 449, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926337

RESUMEN

Psoriasis is an IL-23/Th17-mediated skin disorder with a strong genetic predisposition. The impact of its susceptibility gene nitric oxide synthase 2 (NOS2) remains unknown. Here, we demonstrate strong NOS2 mRNA expression in psoriatic epidermis, an effect that is IL-17 dependent. However, its complete translation to protein is prevented by the IL-17-induced miR-31 implying marginally upregulated NO levels in psoriatic skin. We demonstrate that lower levels of NO, as opposed to higher levels, increase keratinocyte proliferation and mediate IL-17 downstream effects. We hypothesized that the psoriatic phenotype may be alleviated by either eliminating or increasing cellular NO levels. In fact, using the imiquimod psoriasis mouse model, we found a profound impact on the psoriatic inflammation in both IMQ-treated NOS2 KO mice and wild-type mice treated with IMQ and the NO-releasing berdazimer gel. In conclusion, we demonstrate that IL-17 induces NOS2 and fine-tunes its translation towards a window of proinflammatory and hyperproliferative effects and identify NO donor therapy as a new treatment modality for psoriasis.


Asunto(s)
Interleucina-17 , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico , Psoriasis , Psoriasis/genética , Psoriasis/patología , Animales , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Ratones , Humanos , Óxido Nítrico/metabolismo , Interleucina-17/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Queratinocitos/efectos de los fármacos , Imiquimod , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Proliferación Celular/efectos de los fármacos
11.
Br J Cancer ; 131(3): 589-600, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38886556

RESUMEN

BACKGROUND: Occurrence of squamous cell carcinoma (SCC) even in early-stage, untreated chronic lymphocytic leukemia (CLL) patients can be a significant morbidity issue with occasional transformation into metastatic skin lesions. METHODS: CLL cells and extracellular vesicles (EVs) from CLL patients' blood/plasma were purified and used. Expression/activation of AXL and its functions in normal keratinocytes (HEKa) were assessed in vitro co-culture system and in SCC tissues. RESULTS: We detected aberrant activation of AXL, AKT and ERK-1/2 in SCC cell lines compared to HEKa. We also detected increased expression of AXL in primary SCC tissues obtained from CLL patients. Increased activation of AXL, AKT, ERK-1/2 and Src was discernible in HEKa upon co-culturing with CLL cells. Further analysis suggests that Gas6, a ligand of AXL, regulates AXL activation in co-cultured HEKa. Interestingly, exposure of HEKa cells to CLL plasma-derived EVs induced expression of AXL, P-AKT, and EMT-associated markers leading to migration of the cells. Finally, pharmacologic inhibition of AXL induced cell death in SCC lines in a dose dependent manner. CONCLUSIONS: Our findings that CLL cells likely are involved in driving SCC progression, at least in part, via activation of the AXL signaling axis, indicating that AXL inhibition may be beneficial for our CLL patients with SCC.


Asunto(s)
Tirosina Quinasa del Receptor Axl , Carcinoma de Células Escamosas , Progresión de la Enfermedad , Vesículas Extracelulares , Leucemia Linfocítica Crónica de Células B , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Humanos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Proteínas Proto-Oncogénicas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Vesículas Extracelulares/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/genética
12.
JCI Insight ; 9(15)2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38916963

RESUMEN

Despite epidermal turnover, the skin is host to a complex array of microbes, including viruses, such as HPV, which must infect and manipulate skin keratinocyte stem cells (KSCs) to survive. This crosstalk between the virome and KSC populations remains largely unknown. Here, we investigated the effect of HPV8 on KSCs using various mouse models. We observed that the HPV8 early region gene E6 specifically caused Lrig1+ hair follicle junctional zone KSC proliferation and expansion, which would facilitate viral transmission. Within Lrig1+ KSCs specifically, HPV8 E6 bound intracellular p300 to phosphorylate the STAT3 transcriptional regulatory node. This induced ΔNp63 expression, resulting in KSC expansion into the overlying epidermis. HPV8 was associated with 70% of human actinic keratoses. Together, these results define the "hit-and-run" mechanism for HPV8 in human actinic keratosis as an expansion of KSCs, which lack melanosome protection and are thus susceptible to sun light-induced malignant transformation.


Asunto(s)
Proliferación Celular , Queratinocitos , Queratosis Actínica , Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Factor de Transcripción STAT3 , Células Madre , Factor de Transcripción STAT3/metabolismo , Queratinocitos/virología , Queratinocitos/metabolismo , Queratinocitos/patología , Humanos , Queratosis Actínica/patología , Queratosis Actínica/metabolismo , Queratosis Actínica/virología , Animales , Ratones , Células Madre/metabolismo , Células Madre/virología , Proteínas Oncogénicas Virales/metabolismo , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/virología , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/complicaciones , Modelos Animales de Enfermedad , Femenino
13.
Mol Cell Probes ; 76: 101967, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38942130

RESUMEN

Psoriasis is a chronic inflammatory disease characterized by increased keratinocyte proliferation and local inflammation. Long noncoding RNAs (lncRNAs) play important regulatory roles in many immune-mediated diseases, including psoriasis. In this study, we aimed to investigate the role and mechanism of lnc-SPRR2G-2 (SPRR2G) in M5-treated psoriatic keratinocytes. Fluorescence in situ hybridization and quantitative real-time polymerase chain reaction (qRT-PCR) showed that lnc-SPRR2G-2 was significantly upregulated in psoriasis tissues and psoriatic keratinocytes. In psoriatic keratinocytes, functional and molecular experiment analyses demonstrated that SPRR2G regulated proliferation, cell cycle and apoptosis, and induced the expression of S100 calcium binding protein A7 (S100A7), interleukin (IL)-1ß, IL-8 and C-X-C motif chemokine ligand 10 (CXCL10). The function of SPRR2G in psoriasis is related to the STAT3 signaling pathway and can be inhibited by a STAT3 inhibitor. Moreover, KH-type splicing regulatory protein (KHSRP) was proved to be regulated by lnc-SPRR2G-2 and to control the mRNA decay of psoriasis-related cytokines (p < 0.05). In summary, we reported the functions of lnc-SPRR2G-2 and KHSRP in psoriasis. Our findings provide new insights for the further exploration of the pathogenesis and treatment of psoriasis.


Asunto(s)
Proliferación Celular , Inflamación , Queratinocitos , Psoriasis , ARN Largo no Codificante , Factor de Transcripción STAT3 , Transducción de Señal , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Humanos , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Psoriasis/genética , Psoriasis/patología , Psoriasis/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Proliferación Celular/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Regulación hacia Abajo/genética , Proteína A7 de Unión a Calcio de la Familia S100/genética , Proteína A7 de Unión a Calcio de la Familia S100/metabolismo , Apoptosis/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Masculino , Femenino , Adulto
15.
Sci Transl Med ; 16(752): eabq7074, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38896602

RESUMEN

Epidermal growth factor receptor inhibitors (EGFRis) are used to treat many cancers, but their use is complicated by the development of a skin rash that may be severe, limiting their use and adversely affecting patient quality of life. Most studies of EGFRi-induced rash have focused on the fully developed stage of this skin disorder, and early pathological changes remain unclear. We analyzed high-throughput transcriptome sequencing of skin samples from rats exposed to the EGFRi afatinib and identified that keratinocyte activation is an early pathological alteration in EGFRi-induced rash. Mechanistically, the induction of S100 calcium-binding protein A9 (S100A9) occurred before skin barrier disruption and led to keratinocyte activation, resulting in expression of specific cytokines, chemokines, and surface molecules such as interleukin 6 (Il6) and C-C motif chemokine ligand 2 (CCL2) to recruit and activate monocytes through activation of the Janus kinase (JAK)-signal transducers and activators of transcription (STAT) pathway, further recruiting more immune cells. Topical JAK inhibition suppressed the recruitment of immune cells and ameliorated the severity of skin rash in afatinib-treated rats and mice with epidermal deletion of EGFR, while having no effect on EGFRi efficacy in tumor-bearing mice. In a pilot clinical trial (NCT05120362), 11 patients with EGFRi-induced rash were treated with delgocitinib ointment, resulting in improvement in rash severity by at least one grade in 10 of them according to the MASCC EGFR inhibitor skin toxicity tool (MESTT) criteria. These findings provide a better understanding of the early pathophysiology of EGFRi-induced rash and suggest a strategy to manage this condition.


Asunto(s)
Receptores ErbB , Exantema , Inhibidores de las Cinasas Janus , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Ratas , Administración Tópica , Afatinib/farmacología , Afatinib/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Exantema/inducido químicamente , Exantema/patología , Inhibidores de las Cinasas Janus/farmacología , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus/metabolismo , Quinasas Janus/antagonistas & inhibidores , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Estudios Prospectivos
16.
Front Immunol ; 15: 1398120, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38903528

RESUMEN

Psoriasis is an inflammatory disease with systemic manifestations that most commonly presents as itchy, erythematous, scaly plaques on extensor surfaces. Activation of the IL-23/IL-17 pro-inflammatory signaling pathway is a hallmark of psoriasis and its inhibition is key to clinical management. Granzyme K (GzmK) is an immune cell-secreted serine protease elevated in inflammatory and proliferative skin conditions. In the present study, human psoriasis lesions exhibited elevated GzmK levels compared to non-lesional psoriasis and healthy control skin. In an established murine model of imiquimod (IMQ)-induced psoriasis, genetic loss of GzmK significantly reduced disease severity, as determined by delayed plaque formation, decreased erythema and desquamation, reduced epidermal thickness, and inflammatory infiltrate. Molecular characterization in vitro revealed that GzmK contributed to macrophage secretion of IL-23 as well as PAR-1-dependent keratinocyte proliferation. These findings demonstrate that GzmK enhances IL-23-driven inflammation as well as keratinocyte proliferation to exacerbate psoriasis severity.


Asunto(s)
Proliferación Celular , Granzimas , Inflamación , Interleucina-23 , Queratinocitos , Psoriasis , Psoriasis/inmunología , Psoriasis/patología , Animales , Queratinocitos/metabolismo , Queratinocitos/inmunología , Queratinocitos/patología , Humanos , Ratones , Granzimas/metabolismo , Granzimas/genética , Interleucina-23/metabolismo , Inflamación/inmunología , Inflamación/patología , Imiquimod , Modelos Animales de Enfermedad , Ratones Noqueados , Femenino , Masculino , Ratones Endogámicos C57BL
17.
Int J Mol Sci ; 25(11)2024 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-38891963

RESUMEN

Cutaneous field cancerization (CFC) refers to a skin region containing mutated cells' clones, predominantly arising from chronic exposure to ultraviolet radiation (UVR), which exhibits an elevated risk of developing precancerous and neoplastic lesions. Despite extensive research, many molecular aspects of CFC still need to be better understood. In this study, we conducted ex vivo assessment of cell differentiation, oxidative stress, inflammation, and DNA damage in CFC samples. We collected perilesional skin from 41 patients with skin cancer and non-photoexposed skin from 25 healthy control individuals. These biopsies were either paraffin-embedded for indirect immunofluorescence and immunohistochemistry stain or processed for proteins and mRNA extraction from the epidermidis. Our findings indicate a downregulation of p53 expression and an upregulation of Ki67 and p16 in CFC tissues. Additionally, there were alterations in keratinocyte differentiation markers, disrupted cell differentiation, increased expression of iNOS and proinflammatory cytokines IL-6 and IL-8, along with evidence of oxidative DNA damage. Collectively, our results suggest that despite its outwardly normal appearance, CFC tissue shows early signs of DNA damage, an active inflammatory state, oxidative stress, abnormal cell proliferation and differentiation.


Asunto(s)
Diferenciación Celular , Daño del ADN , Inflamación , Estrés Oxidativo , Neoplasias Cutáneas , Rayos Ultravioleta , Humanos , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/genética , Inflamación/metabolismo , Inflamación/genética , Inflamación/patología , Masculino , Femenino , Persona de Mediana Edad , Rayos Ultravioleta/efectos adversos , Anciano , Queratinocitos/metabolismo , Queratinocitos/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Adulto , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Piel/metabolismo , Piel/patología , Piel/efectos de la radiación , Antígeno Ki-67/metabolismo , Antígeno Ki-67/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Interleucina-6/metabolismo , Interleucina-6/genética
18.
Cell Death Dis ; 15(6): 392, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834617

RESUMEN

Keratinocyte proliferation and differentiation in epidermis are well-controlled and essential for reacting to stimuli such as ultraviolet light. Imbalance between proliferation and differentiation is a characteristic feature of major human skin diseases such as psoriasis and squamous cell carcinoma. However, the effect of keratinocyte metabolism on proliferation and differentiation remains largely elusive. We show here that the gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1) promotes differentiation while inhibits proliferation of keratinocyte and suppresses psoriasis development. FBP1 is identified among the most upregulated genes induced by UVB using transcriptome sequencing and is elevated especially in upper epidermis. Fbp1 heterozygous mice exhibit aberrant epidermis phenotypes with local hyperplasia and dedifferentiation. Loss of FBP1 promotes proliferation and inhibits differentiation of keratinocytes in vitro. Mechanistically, FBP1 loss facilitates glycolysis-mediated acetyl-CoA production, which increases histone H3 acetylation at lysine 9, resulting in enhanced transcription of proliferation genes. We further find that the expression of FBP1 is dramatically reduced in human psoriatic lesions and in skin of mouse imiquimod psoriasis model. Fbp1 deficiency in mice facilitates psoriasis-like skin lesions development through glycolysis and acetyl-CoA production. Collectively, our findings reveal a previously unrecognized role of FBP1 in epidermal homeostasis and provide evidence for FBP1 as a metabolic psoriasis suppressor.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Fructosa-Bifosfatasa , Histonas , Queratinocitos , Psoriasis , Animales , Humanos , Ratones , Acetilcoenzima A/metabolismo , Acetilación , Modelos Animales de Enfermedad , Fructosa-Bifosfatasa/metabolismo , Fructosa-Bifosfatasa/genética , Glucólisis , Histonas/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones Endogámicos C57BL , Psoriasis/patología , Psoriasis/metabolismo , Psoriasis/genética
20.
Cell Death Differ ; 31(8): 970-982, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38714880

RESUMEN

Obesity is endemic to many developed countries. Overweight or obesity is associated with an increased risk of developing cancer. Dysfunctional adipose tissue alters cancer cell proliferation and migration; however, whether and how neoplastic epithelial cells communicate with adipose tissue and the underlying mechanism are less clear. BTG3 is a member of the anti-proliferative BTG/Tob family and functions as a tumor suppressor. Here, we demonstrated that BTG3 levels are downregulated in basal cell carcinoma and squamous cell carcinoma compared to normal skin tissue, and Btg3 knockout in mice augmented the development of papilloma in a mouse model of DMBA/TPA-induced skin carcinogenesis. Mechanistically, BTG3-knockout keratinocytes promoted adipocyte differentiation mainly through the release of IL1α, IL10, and CCL4, as a result of elevated NF-κB activity. These adipocytes produced CCL20 and FGF7 in a feedback loop to promote keratinocyte migration. Thus, our findings showcased the role of BTG3 in guarding the interplay between keratinocytes and adjacent adipocytes, and identified the underlying neoplastic molecular mediators that may serve as possible targets in the treatment of skin cancer.


Asunto(s)
Adipocitos , Queratinocitos , Ratones Noqueados , Transducción de Señal , Neoplasias Cutáneas , Animales , Queratinocitos/metabolismo , Queratinocitos/patología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/genética , Adipocitos/metabolismo , Adipocitos/patología , Ratones , Humanos , Carcinogénesis/patología , Carcinogénesis/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma Basocelular/metabolismo , Carcinoma Basocelular/patología , Diferenciación Celular , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Movimiento Celular , Factor 7 de Crecimiento de Fibroblastos/metabolismo , FN-kappa B/metabolismo , Quimiocina CCL20/metabolismo , Quimiocina CCL20/genética , Proteínas de Ciclo Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...