Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.608
Filter
1.
Adv Exp Med Biol ; 1447: 21-35, 2024.
Article in English | MEDLINE | ID: mdl-38724781

ABSTRACT

The pathophysiology of atopic dermatitis is complex and multifactorial, involving elements of barrier dysfunction, alterations in cell-mediated immune responses, IgE-mediated hypersensitivity, and environmental factors. Loss-of-function mutations in filaggrin have been implicated in severe atopic dermatitis due to a potential increase in trans-epidermal water loss, pH alterations, and dehydration. Other genetic changes have also been identified, which may alter the skin's barrier function, resulting in an atopic dermatitis phenotype. The imbalance of Th2 to Th1 cytokines observed in atopic dermatitis can create alterations in the cell-mediated immune responses and can promote IgE-mediated hypersensitivity, both of which appear to play a role in the development of atopic dermatitis. One must additionally take into consideration the role of the environment on the causation of atopic dermatitis and the impact of chemicals such as airborne formaldehyde, harsh detergents, fragrances, and preservatives. Use of harsh alkaline detergents in skin care products may also unfavorably alter the skin's pH causing downstream changes in enzyme activity and triggering inflammation. Environmental pollutants can trigger responses from both the innate and adaptive immune pathways. This chapter will discuss the multifaceted etiology of atopic dermatitis, which will help us to elucidate potential therapeutic targets. We will also review existing treatment options and their interaction with the complex inflammatory and molecular triggers of atopic dermatitis.


Subject(s)
Dermatitis, Atopic , Filaggrin Proteins , Dermatitis, Atopic/immunology , Dermatitis, Atopic/genetics , Dermatitis, Atopic/physiopathology , Humans , Skin/pathology , Skin/immunology , Animals , Cytokines/metabolism , Immunoglobulin E/immunology , Environmental Exposure/adverse effects
2.
Front Immunol ; 15: 1352469, 2024.
Article in English | MEDLINE | ID: mdl-38711504

ABSTRACT

Vibriosis, caused by Vibrio, seriously affects the health of fish, shellfish, and shrimps, causing large economic losses. Teleosts are represent the first bony vertebrates with both innate and adaptive immune responses against pathogens. Aquatic animals encounter hydraulic pressure and more pathogens, compared to terrestrial animals. The skin is the first line of defense in fish, constituting the skin-associated lymphoid tissue (SALT), which belongs to the main mucosa-associated lymphoid tissues (MALT). However, little is known about the function of immunity related proteins in fish. Therefore, this study used iTRAQ (isobaric tags for relative and absolute quantitation) to compare the skin proteome between the resistant and susceptible families of Cynoglossus semilaevis. The protein integrin beta-2, the alpha-enolase isoform X1, subunit B of V-type proton ATPase, eukaryotic translation initiation factor 6, and ubiquitin-like protein ISG15, were highly expressed in the resistant family. The 16S sequencing of the skin tissues of the resistant and susceptible families showed significant differences in the microbial communities of the two families. The protein-microbial interaction identified ten proteins associated with skin microbes, including immunoglobulin heavy chain gene (IGH), B-cell lymphoma/leukemia 10 (BCL10) and pre-B-cell leukemia transcription factor 1 isoform X2 (PBX2). This study highlights the interaction between skin proteins and the microbial compositions of C. semilaevis and provides new insights into understanding aquaculture breeding research.


Subject(s)
Disease Resistance , Fish Diseases , Fish Proteins , Flatfishes , Microbiota , Skin , Vibrio Infections , Vibrio , Animals , Skin/immunology , Skin/microbiology , Skin/metabolism , Fish Diseases/immunology , Fish Diseases/microbiology , Disease Resistance/immunology , Vibrio Infections/immunology , Vibrio Infections/veterinary , Flatfishes/immunology , Flatfishes/microbiology , Microbiota/immunology , Vibrio/immunology , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/immunology , Proteome , Proteomics/methods
3.
JCI Insight ; 9(9)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38716729

ABSTRACT

Atopic dermatitis (AD) is an inflammatory skin condition with a childhood prevalence of up to 25%. Microbial dysbiosis is characteristic of AD, with Staphylococcus aureus the most frequent pathogen associated with disease flares and increasingly implicated in disease pathogenesis. Therapeutics to mitigate the effects of S. aureus have had limited efficacy and S. aureus-associated temporal disease flares are synonymous with AD. An alternative approach is an anti-S. aureus vaccine, tailored to AD. Experimental vaccines have highlighted the importance of T cells in conferring protective anti-S. aureus responses; however, correlates of T cell immunity against S. aureus in AD have not been identified. We identify a systemic and cutaneous immunological signature associated with S. aureus skin infection (ADS.aureus) in a pediatric AD cohort, using a combined Bayesian multinomial analysis. ADS.aureus was most highly associated with elevated cutaneous chemokines IP10 and TARC, which preferentially direct Th1 and Th2 cells to skin. Systemic CD4+ and CD8+ T cells, except for Th2 cells, were suppressed in ADS.aureus, particularly circulating Th1, memory IL-10+ T cells, and skin-homing memory Th17 cells. Systemic γδ T cell expansion in ADS.aureus was also observed. This study suggests that augmentation of protective T cell subsets is a potential therapeutic strategy in the management of S. aureus in AD.


Subject(s)
Dermatitis, Atopic , Staphylococcal Skin Infections , Staphylococcus aureus , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Humans , Staphylococcus aureus/immunology , Child , Female , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Male , Child, Preschool , Skin/microbiology , Skin/immunology , Skin/pathology , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Th17 Cells/immunology , Bayes Theorem , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/metabolism , Interleukin-10/immunology , Intraepithelial Lymphocytes/immunology , Antigens, Differentiation, T-Lymphocyte , Membrane Glycoproteins
4.
Front Immunol ; 15: 1388690, 2024.
Article in English | MEDLINE | ID: mdl-38803495

ABSTRACT

Introduction: Psoriasis is a chronic skin disease characterized by unique scaling plaques. However, during the acute phase, psoriatic lesions exhibit eczematous changes, making them difficult to distinguish from atopic dermatitis, which poses challenges for the selection of biological agents. This study aimed to identify potential diagnostic genes in psoriatic lesions and investigate their clinical significance. Methods: GSE182740 datasets from the GEO database were analyzed for differential analysis; machine learning algorithms (SVM-RFE and LASSO regression models) are used to screen for diagnostic markers; CIBERSORTx is used to determine the dynamic changes of 22 different immune cell components in normal skin lesions, psoriatic non-lesional skin, and psoriatic lesional skin, as well as the expression of the diagnostic genes in 10 major immune cells, and real-time quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry are used to validate results. Results: We obtained 580 differentially expressed genes (DEGs) in the skin lesion and non-lesion of psoriasis patients, 813 DEGs in mixed patients between non-lesions and lesions, and 96 DEGs in the skin lesion and non-lesion of atopic dermatitis, respectively. Then 144 specific DEGs in psoriasis via a Veen diagram were identified. Ultimately, UGGT1, CCNE1, MMP9 and ARHGEF28 are identified for potential diagnostic genes from these 144 specific DEGs. The value of the selected diagnostic genes was verified by receiver operating characteristic (ROC) curves with expanded samples. The the area under the ROC curve (AUC) exceeded 0.7 for the four diagnosis genes. RT-qPCR results showed that compared to normal human epidermis, the expression of UGGT1, CCNE1, and MMP9 was significantly increased in patients with psoriasis, while ARHGEF28 expression was significantly decreased. Notably, the results of CIBERSORTx showed that CCNE1 was highly expressed in CD4+ T cells and neutrophils, ARHGEF28 was also expressed in mast cells. Additionally, CCNE1 was strongly correlated with IL-17/CXCL8/9/10 and CCL20. Immunohistochemical results showed increased nuclear expression of CCNE1 in psoriatic epidermal cells relative to normal. Conclusion: Based on the performance of the four genes in ROC curves and their expression in immune cells from patients with psoriasis, we suggest that CCNE1 possess higher diagnostic value.


Subject(s)
Biomarkers , Machine Learning , Psoriasis , Skin , Psoriasis/immunology , Psoriasis/diagnosis , Psoriasis/genetics , Humans , Skin/immunology , Skin/pathology , Skin/metabolism , Gene Expression Profiling , Dermatitis, Atopic/immunology , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/genetics , Transcriptome , Databases, Genetic , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Oncogene Proteins , Cyclin E
6.
Front Immunol ; 15: 1378359, 2024.
Article in English | MEDLINE | ID: mdl-38779662

ABSTRACT

Skin tissue-resident memory T (Trm) cells are produced by antigenic stimulation and remain in the skin for a long time without entering the peripheral circulation. In the healthy state Trm cells can play a patrolling and surveillance role, but in the disease state Trm cells differentiate into various phenotypes associated with different diseases, exhibit different localizations, and consequently have local protective or pathogenic roles, such as disease recurrence in vitiligo and maintenance of immune homeostasis in melanoma. The most common surface marker of Trm cells is CD69/CD103. However, the plasticity of tissue-resident memory T cells after colonization remains somewhat uncertain. This ambiguity is largely due to the variation in the functionality and ultimate destination of Trm cells produced from memory cells differentiated from diverse precursors. Notably, the presence of Trm cells is not stationary across numerous non-lymphoid tissues, most notably in the skin. These cells may reenter the blood and distant tissue sites during the recall response, revealing the recycling and migration potential of the Trm cell progeny. This review focuses on the origin and function of skin Trm cells, and provides new insights into the role of skin Trm cells in the treatment of autoimmune skin diseases, infectious skin diseases, and tumors.


Subject(s)
Cell Plasticity , Homeostasis , Immunologic Memory , Memory T Cells , Skin Diseases , Skin , Humans , Homeostasis/immunology , Memory T Cells/immunology , Memory T Cells/metabolism , Skin/immunology , Skin/pathology , Cell Plasticity/immunology , Animals , Skin Diseases/immunology , Antigens, CD/metabolism , Antigens, CD/immunology
7.
Front Immunol ; 15: 1367602, 2024.
Article in English | MEDLINE | ID: mdl-38774875

ABSTRACT

Background: There is a significant imbalance of mitochondrial activity and oxidative stress (OS) status in patients with atopic dermatitis (AD). This study aims to screen skin and peripheral mitochondria-related biomarkers, providing insights into the underlying mechanisms of mitochondrial dysfunction in AD. Methods: Public data were obtained from MitoCarta 3.0 and GEO database. We screened mitochondria-related differentially expressed genes (MitoDEGs) using R language and then performed GO and KEGG pathway analysis on MitoDEGs. PPI and machine learning algorithms were also used to select hub MitoDEGs. Meanwhile, the expression of hub MitoDEGs in clinical samples were verified. Using ROC curve analysis, the diagnostic performance of risk model constructed from these hub MitoDEGs was evaluated in the training and validation sets. Further computer-aided algorithm analyses included gene set enrichment analysis (GSEA), immune infiltration and mitochondrial metabolism, centered on these hub MitoDEGs. We also used real-time PCR and Spearman method to evaluate the relationship between plasma circulating cell-free mitochondrial DNA (ccf-mtDNA) levels and disease severity in AD patients. Results: MitoDEGs in AD were significantly enriched in pathways involved in mitochondrial respiration, mitochondrial metabolism, and mitochondrial membrane transport. Four hub genes (BAX, IDH3A, MRPS6, and GPT2) were selected to take part in the creation of a novel mitochondrial-based risk model for AD prediction. The risk score demonstrated excellent diagnostic performance in both the training cohort (AUC = 1.000) and the validation cohort (AUC = 0.810). Four hub MitoDEGs were also clearly associated with the innate immune cells' infiltration and the molecular modifications of mitochondrial hypermetabolism in AD. We further discovered that AD patients had considerably greater plasma ccf-mtDNA levels than controls (U = 92.0, p< 0.001). Besides, there was a significant relationship between the up-regulation of plasma mtDNA and the severity of AD symptoms. Conclusions: The study highlights BAX, IDH3A, MRPS6 and GPT2 as crucial MitoDEGs and demonstrates their efficiency in identifying AD. Moderate to severe AD is associated with increased markers of mitochondrial damage and cellular stress (ccf=mtDNA). Our study provides data support for the variation in mitochondria-related functional characteristics of AD patients.


Subject(s)
Biomarkers , Computational Biology , Dermatitis, Atopic , Machine Learning , Mitochondria , Skin , Humans , Dermatitis, Atopic/genetics , Dermatitis, Atopic/blood , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/immunology , Biomarkers/blood , Mitochondria/metabolism , Mitochondria/genetics , Computational Biology/methods , Skin/metabolism , Skin/immunology , Male , DNA, Mitochondrial/genetics , Female , Gene Expression Profiling
10.
Front Immunol ; 15: 1343987, 2024.
Article in English | MEDLINE | ID: mdl-38690268

ABSTRACT

Autophagy is a cellular process that functions to maintain intracellular homeostasis via the degradation and recycling of defective organelles or damaged proteins. This dynamic mechanism participates in various biological processes, such as the regulation of cellular differentiation, proliferation, survival, and the modulation of inflammation and immune responses. Recent evidence has demonstrated the involvement of polymorphisms in autophagy-related genes in various skin autoimmune diseases. In addition, autophagy, along with autophagy-related proteins, also contributes to homeostasis maintenance and immune regulation in the skin, which is associated with skin autoimmune disorders. This review aims to provide an overview of the multifaceted role of autophagy in skin autoimmune diseases and shed light on the potential of autophagy-targeting therapeutic strategies in dermatology.


Subject(s)
Autoimmune Diseases , Autophagy , Skin Diseases , Humans , Autophagy/immunology , Autoimmune Diseases/immunology , Skin Diseases/immunology , Animals , Skin/immunology , Skin/pathology , Skin/metabolism , Homeostasis/immunology
11.
Arch Dermatol Res ; 316(5): 176, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758283

ABSTRACT

Psoriasis is a chronic immune mediated inflammatory skin disease with systemic manifestations. It has been reported that caloric restriction could improve severity of psoriasis patients. However, the mechanism of intermittent fasting effects on psoriasis has not been investigated. Caloric restriction is known to reduce the number of circulating inflammatory monocytes in a CCL2-dependent manner. However, it is still unknown whether caloric restriction can improve psoriasis by regulating monocytes through CCL2. In this study, we used imiquimod (IMQ)-induced psoriasis-like mouse model to explore the effects and the mechanisms of intermittent fasting on psoriasis-like dermatitis. We found that intermittent fasting could significantly improve IMQ-induced psoriasis-like dermatitis, and reduce the number of γδT17 cells and IL-17 production in draining lymph nodes and psoriatic lesion via inhibiting proliferation and increasing death of γδT17 cells. Furthermore, intermittent fasting could significantly decrease monocytes in blood, and this was associated with decreased monocytes, macrophages and DC in psoriasis-like skin inflammation. Reduced monocytes in circulation and increased monocytes in BM of fasting IMQ-induced psoriasis-like mice is through reducing the production of CCL2 from BM to inhibit monocyte egress to the periphery. Our above data shads light on the mechanisms of intermittent fasting on psoriasis.


Subject(s)
Chemokine CCL2 , Disease Models, Animal , Fasting , Imiquimod , Monocytes , Psoriasis , Animals , Psoriasis/immunology , Psoriasis/chemically induced , Psoriasis/pathology , Monocytes/immunology , Monocytes/metabolism , Mice , Fasting/blood , Chemokine CCL2/metabolism , Th17 Cells/immunology , Interleukin-17/metabolism , Skin/pathology , Skin/immunology , Humans , Mice, Inbred C57BL , Male , Cell Proliferation , Caloric Restriction , Intermittent Fasting
14.
BMC Immunol ; 25(1): 30, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734636

ABSTRACT

BACKGROUND: Immune-mediated necrotizing myopathy (IMNM) is an idiopathic inflammatory myopathy (IIM). Though patients with IMNM were not considered to show skin rash, several reports have showed atypical skin conditions in patients with anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) antibody-positive IMNM (HMGCR-IMNM). The incidence and phenotype of skin conditions in patients with HMGCR-IMNM are not fully known. RESULTS: Among the 100 IIM patients diagnosed from April 2015 through August 2022, 34 (34%) presented some form of skin condition, with 27 having typical skin rashes; this included 13 patients with dermatomyositis (DM), 8 with anti-synthetase syndrome (ASS), and 6 with IMNM. Meanwhile, 8 of 19 patients with HMGCR-IMNM (42%) presented atypical skin lesions, but no patients with other IIMs did (p < 0.001). Skin eruption with ash-like scales was observed in four HMGCR-IMNM patients, and non-scaly red patches and lumps in the other four patients; accordingly, their skin manifestations were considered as other dermal diseases except for IIM. However, skin and muscle biopsies revealed the atypical skin conditions of patients with HMGCR-IMNM to have the same pathological background, formed by Bcl-2-positive lymphocyte infiltrations. CONCLUSIONS: HMGCR-IMNM patients frequently have atypical skin conditions of the neck and back. Skin biopsy specimens from these lesions showed the same Bcl-2-positive lymphocytic infiltrations as muscle biopsy specimens regardless of the different gross dermal findings. Thus, such atypical skin conditions may be suggestive for HMGCR-IMNM.


Subject(s)
Autoantibodies , Hydroxymethylglutaryl CoA Reductases , Myositis , Skin , Humans , Hydroxymethylglutaryl CoA Reductases/immunology , Female , Male , Middle Aged , Autoantibodies/immunology , Autoantibodies/blood , Adult , Skin/pathology , Skin/immunology , Myositis/immunology , Myositis/diagnosis , Aged , Skin Diseases/immunology , Skin Diseases/etiology , Muscular Diseases/immunology , Muscular Diseases/diagnosis , Biopsy
15.
Arch Dermatol Res ; 316(5): 156, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734816

ABSTRACT

Atopic dermatitis (AD) is an inflammatory skin disease with intense pruritus, and chronic skin colonization by Staphylococcus aureus. To understand the inflammatory status in AD, we investigated the inflammasome complex, that activates ASC (Apoptosis-associated speck-like protein containing a CARD), caspase-1 and GSDMD (gasdermin-D), and production of IL-1ß and IL-18. We aimed to evaluate the expression of the inflammasome pathway in the skin of adults with AD. Thirty patients with moderate to severe AD and 20 healthy controls were enrolled in the study. We performed the analysis of the inflammasome components NLRP1, NLRP3, AIM-2, IL-1ß, IL-18, Caspase-1, ASC, GSDMD, and CD68 expression (macrophage marker) by immunohistochemistry and immunofluorescence. The main findings included increased expression of NLRP3, NLRP1 and AIM-2 at dermal level of severe AD; augmented IL-18 and IL-1ß expression at epidermis of moderate and severe patients, and in the dermis of severe AD; augmented expression of ASC, caspase-1 and GSDMD in both epidermis and dermis of moderate and severe AD. We detected positive correlation between caspase-1, GSDMD and IL-1ß (epidermis) and caspase-1 (dermis) and AD severity; NLRP3, AIM-2 and IL-1ß, and NLRP3 with IL-18 in the epidermis; ASC, GSDMD and IL-1ß, and NLRP3, AIM-2, caspase-1, and IL-18 in the dermis. We also evidenced the presence of CD68+ macrophages secreting GSDMD, ASC and IL-1ß in moderate and severe AD. Cutaneous macrophages, early detected in moderate AD, have its role in the disease inflammatory mechanisms. Our study indicates a canonical activation pathway of inflammasomes, reinforced by the chronic status of inflammation in AD. The analysis of the inflammasome complex evidenced an imbalance in its regulation, with increased expression of the evaluated components, which is remarkably in severe AD, emphasizing its relevance as potential disease biomarkers and targets for immunomodulatory interventions.


Subject(s)
CARD Signaling Adaptor Proteins , Caspase 1 , Dermatitis, Atopic , Inflammasomes , Interleukin-18 , Interleukin-1beta , Intracellular Signaling Peptides and Proteins , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphate-Binding Proteins , Humans , Inflammasomes/metabolism , Inflammasomes/immunology , CARD Signaling Adaptor Proteins/metabolism , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Macrophages/metabolism , Macrophages/immunology , Interleukin-1beta/metabolism , Male , Female , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Adult , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-18/metabolism , Caspase 1/metabolism , Skin/pathology , Skin/immunology , Skin/metabolism , Severity of Illness Index , Middle Aged , Antigens, Differentiation, Myelomonocytic/metabolism , Young Adult , Apoptosis Regulatory Proteins/metabolism , Antigens, CD/metabolism , NLR Proteins/metabolism , Case-Control Studies , Epidermis/immunology , Epidermis/metabolism , Epidermis/pathology , Gasdermins , CD68 Molecule , DNA-Binding Proteins
16.
Front Immunol ; 15: 1383263, 2024.
Article in English | MEDLINE | ID: mdl-38736879

ABSTRACT

Acne vulgaris is one of the most common skin diseases. The current understanding of acne primarily revolves around inflammatory responses, sebum metabolism disorders, aberrant hormone and receptor expression, colonization by Cutibacterium acnes, and abnormal keratinization of follicular sebaceous glands. Although the precise mechanism of action remains incompletely understood, it is plausible that macrophages exert an influence on these pathological features. Macrophages, as a constituent of the human innate immune system, typically manifest distinct phenotypes across various diseases. It has been observed that the polarization of macrophages toward the M1 phenotype plays a pivotal role in the pathogenesis of acne. In recent years, extensive research on acne has revealed an increasing number of natural remedies exhibiting therapeutic efficacy through the modulation of macrophage polarization. This review investigates the role of cutaneous macrophages, elucidates their potential significance in the pathogenesis of acne, a prevalent chronic inflammatory skin disorder, and explores the therapeutic mechanisms of natural plant products targeting macrophages. Despite these insights, the precise role of macrophages in the pathogenesis of acne remains poorly elucidated. Subsequent investigations in this domain will further illuminate the pathogenesis of acne and potentially offer guidance for identifying novel therapeutic targets for this condition.


Subject(s)
Acne Vulgaris , Macrophages , Acne Vulgaris/immunology , Acne Vulgaris/drug therapy , Humans , Macrophages/immunology , Macrophages/metabolism , Biological Products/therapeutic use , Biological Products/pharmacology , Animals , Skin/immunology , Skin/pathology , Skin/metabolism
17.
Pediatr Allergy Immunol ; 35(5): e14130, 2024 May.
Article in English | MEDLINE | ID: mdl-38693814

ABSTRACT

While the early introduction of food allergens in the infant diet has been shown to be effective at preventing the development of food allergy (FA), its implementation in real life has been associated with various challenges. Interventions aimed at correcting skin barrier dysfunction have been explored in recent decades as a distinct or complementary mean to prevent allergic sensitization through the skin and subsequent development of FA. Studies assessing the application of emollient from birth have yielded conflicting results, and meta-analyses have demonstrated either no effect or only a slight positive effect on FA prevention. However, a careful review of the clinical trials reveals that different emollients were used, which may have explained some of the discrepancies between study results. Emollient application protocols also varied widely between studies. While firm conclusions cannot be drawn with regard to their overall efficacy at preventing FA, the available data provide valuable insight into the characteristics that could be associated with a more effective intervention. Namely, successful trials tended to use emollients with an acidic pH of 5.5, applied over the entire body, and combined with topical corticosteroids in affected areas. Consensus on the optimal strategy to restore skin barrier function could help improve the homogeneity and clinical relevance of future trials on this topic. In the meantime, clinicians should avoid products associated with worse outcomes.


Subject(s)
Emollients , Food Hypersensitivity , Skin , Humans , Food Hypersensitivity/prevention & control , Emollients/administration & dosage , Skin/drug effects , Skin/immunology , Infant , Allergens/immunology , Allergens/administration & dosage , Clinical Trials as Topic , Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/therapeutic use , Infant, Newborn
18.
Arthritis Res Ther ; 26(1): 94, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702742

ABSTRACT

BACKGROUND: Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular injury and inflammation, followed by excessive fibrosis of the skin and other internal organs, including the lungs. CX3CL1 (fractalkine), a chemokine expressed on endothelial cells, supports the migration of macrophages and T cells that express its specific receptor CX3CR1 into targeted tissues. We previously reported that anti-CX3CL1 monoclonal antibody (mAb) treatment significantly inhibited transforming growth factor (TGF)-ß1-induced expression of type I collagen and fibronectin 1 in human dermal fibroblasts. Additionally, anti-mouse CX3CL1 mAb efficiently suppressed skin inflammation and fibrosis in bleomycin- and growth factor-induced SSc mouse models. However, further studies using different mouse models of the complex immunopathology of SSc are required before the initiation of a clinical trial of CX3CL1 inhibitors for human SSc. METHODS: To assess the preclinical utility and functional mechanism of anti-CX3CL1 mAb therapy in skin and lung fibrosis, a sclerodermatous chronic graft-versus-host disease (Scl-cGVHD) mouse model was analyzed with immunohistochemical staining for characteristic infiltrating cells and RNA sequencing assays. RESULTS: On day 42 after bone marrow transplantation, Scl-cGVHD mice showed increased serum CX3CL1 level. Intraperitoneal administration of anti-CX3CL1 mAb inhibited the development of fibrosis in the skin and lungs of Scl-cGVHD model, and did not result in any apparent adverse events. The therapeutic effects were correlated with the number of tissue-infiltrating inflammatory cells and α-smooth muscle actin (α-SMA)-positive myofibroblasts. RNA sequencing analysis of the fibrotic skin demonstrated that cGVHD-dependent induction of gene sets associated with macrophage-related inflammation and fibrosis was significantly downregulated by mAb treatment. In the process of fibrosis, mAb treatment reduced cGVHD-induced infiltration of macrophages and T cells in the skin and lungs, especially those expressing CX3CR1. CONCLUSIONS: Together with our previous findings in other SSc mouse models, the current results indicated that anti-CX3CL1 mAb therapy could be a rational therapeutic approach for fibrotic disorders, such as human SSc and Scl-cGVHD.


Subject(s)
Antibodies, Monoclonal , Chemokine CX3CL1 , Disease Models, Animal , Graft vs Host Disease , Pulmonary Fibrosis , Scleroderma, Systemic , Skin , Animals , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/pathology , Scleroderma, Systemic/immunology , Mice , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , Skin/pathology , Skin/drug effects , Skin/metabolism , Skin/immunology , Fibrosis , Female , Mice, Inbred C57BL , Humans , Lung/pathology , Lung/drug effects , Lung/metabolism , Lung/immunology
19.
PLoS Pathog ; 20(5): e1012020, 2024 May.
Article in English | MEDLINE | ID: mdl-38743761

ABSTRACT

Scrub typhus is an acute febrile disease due to Orientia tsutsugamushi (Ot) infection and can be life-threatening with organ failure, hemorrhage, and fatality. Yet, little is known as to how the host reacts to Ot bacteria at early stages of infection; no reports have addressed the functional roles of type I versus type II interferon (IFN) responses in scrub typhus. In this study, we used comprehensive intradermal (i.d.) inoculation models and two clinically predominant Ot strains (Karp and Gilliam) to uncover early immune events. Karp infection induced sequential expression of Ifnb and Ifng in inflamed skin and draining lymph nodes at days 1 and 3 post-infection. Using double Ifnar1-/-Ifngr1-/- and Stat1-/- mice, we found that deficiency in IFN/STAT1 signaling resulted in lethal infection with profound pathology and skin eschar lesions, which resembled to human scrub typhus. Further analyses demonstrated that deficiency in IFN-γ, but not IFN-I, resulted in impaired NK cell and macrophage activation and uncontrolled bacterial growth and dissemination, leading to metabolic dysregulation, excessive inflammatory cell infiltration, and exacerbated tissue damage. NK cells were found to be the major cellular source of innate IFN-γ, contributing to the initial Ot control in the draining lymph nodes. In vitro studies with dendritic cell cultures revealed a superior antibacterial effect offered by IFN-γ than IFN-ß. Comparative in vivo studies with Karp- and Gilliam-infection revealed a crucial role of IFN-γ signaling in protection against progression of eschar lesions and Ot infection lethality. Additionally, our i.d. mouse models of lethal infection with eschar lesions are promising tools for immunological study and vaccine development for scrub typhus.


Subject(s)
Interferon-gamma , Mice, Knockout , Orientia tsutsugamushi , Scrub Typhus , Signal Transduction , Animals , Scrub Typhus/immunology , Scrub Typhus/microbiology , Orientia tsutsugamushi/immunology , Mice , Interferon-gamma/metabolism , Interferon-gamma/immunology , Mice, Inbred C57BL , Disease Models, Animal , Skin/microbiology , Skin/pathology , Skin/immunology , STAT1 Transcription Factor/metabolism
20.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791342

ABSTRACT

Psoriasis is a systemic autoimmune/autoinflammatory disease that can be well studied in established mouse models. Skin-resident macrophages are classified into epidermal Langerhans cells and dermal macrophages and are involved in innate immunity, orchestration of adaptive immunity, and maintenance of tissue homeostasis due to their ability to constantly shift their phenotype and adapt to the current microenvironment. Consequently, both macrophage populations play dual roles in psoriasis. In some circumstances, pro-inflammatory activated macrophages and Langerhans cells trigger psoriatic inflammation, while in other cases their anti-inflammatory stimulation results in amelioration of the disease. These features make macrophages interesting candidates for modern therapeutic strategies. Owing to the significant progress in knowledge, our review article summarizes current achievements and indicates future research directions to better understand the function of macrophages in psoriasis.


Subject(s)
Disease Models, Animal , Macrophages , Psoriasis , Psoriasis/immunology , Psoriasis/pathology , Animals , Macrophages/immunology , Macrophages/metabolism , Mice , Humans , Langerhans Cells/immunology , Langerhans Cells/pathology , Immunity, Innate , Skin/pathology , Skin/immunology , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...