Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 798
1.
J Immunol Res ; 2024: 5582151, 2024.
Article En | MEDLINE | ID: mdl-38690552

Unlike T cells in other tissues, uterine T cells must balance strong immune defense against pathogens with tolerance to semiallogeneic fetus. Our previous study fully elucidated the characteristics of γδT cells in nonpregnant uterus and the mechanism modulated by estrogen. However, comprehensive knowledge of the immunological properties of αßT (including CD4+T cells and CD8+T) cells in nonpregnancy uterus has not been acquired. In this study, we fully compared the immunological properties of αßT cells between uterus and blood using mouse and human sample. It showed that most of CD4+T cells and CD8+T cells in murine uterus and human endometrium were tissue resident memory T cells which highly expressed tissue residence markers CD69 and/or CD103. In addition, both CD4+T cells and CD8+T cells in uterus highly expressed inhibitory molecular PD-1 and cytokine IFN-γ. Uterine CD4+T cells highly expressed IL-17 and modulated by transcription factor pSTAT3. Moreover, we compared the similarities and differences between human and murine uterine T cell phenotype. Together, uterine CD4+T cells and CD8+ cells exhibited a unique mixed signature of T cell dysfunction, activation, and effector function which enabled them to balance strong immune defense against pathogens with tolerance to fetus. Our study fully elucidated the unique immunologic properties of uterine CD4+T and CD8+T cells and provided a base for further investigation of functions.


Antigens, CD , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Uterus , Female , CD8-Positive T-Lymphocytes/immunology , Animals , Humans , Mice , CD4-Positive T-Lymphocytes/immunology , Uterus/immunology , Antigens, CD/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Integrin alpha Chains/metabolism , Memory T Cells/immunology , STAT3 Transcription Factor/metabolism , Interferon-gamma/metabolism , Lectins, C-Type/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Interleukin-17/metabolism , Lymphocyte Activation/immunology , Immunologic Memory
2.
Commun Biol ; 7(1): 583, 2024 May 16.
Article En | MEDLINE | ID: mdl-38755265

Tumor-associated macrophages of the M2 phenotype promote cancer initiation and progression. Importantly, M2 macrophage-derived exosomes play key roles in the malignancy of cancer cells. Here, we report that circTMCO3 is upregulated in ovarian cancer patients, and its high expression indicates poor survival. M2-derived exosomes promote proliferation, migration, and invasion in ovarian cancer, but these effects are abolished by knockdown of circTMCO3. Furthermore, circTMCO3 functions as a competing endogenous RNA for miR-515-5p to reduce its abundance, thus upregulating ITGA8 in ovarian cancer. miR-515-5p inhibits ovarian cancer malignancy via directly downregulating ITGA8. The decreased oncogenic activity of circTMCO3-silencing exosomes is reversed by miR-515-5p knockdown or ITGA8 overexpression. Exosomal circTMCO3 promotes ovarian cancer progression in nude mice. Thus, M2 macrophage-derived exosomes promote malignancy by delivering circTMCO3 and targeting the miR-515-5p/ITGA8 axis in ovarian cancer. Our findings not only provide mechanistic insights into ovarian cancer progression, but also suggest potential therapeutic targets.


Exosomes , Mice, Nude , MicroRNAs , Ovarian Neoplasms , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Humans , Exosomes/metabolism , Exosomes/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Mice , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Cell Proliferation , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Cell Movement
3.
Nat Commun ; 15(1): 4326, 2024 May 21.
Article En | MEDLINE | ID: mdl-38773113

Resolving inflammation is thought to return the affected tissue back to homoeostasis but recent evidence supports a non-linear model of resolution involving a phase of prolonged immune activity. Here we show that within days following resolution of Streptococcus pneumoniae-triggered lung inflammation, there is an influx of antigen specific lymphocytes with a memory and tissue-resident phenotype as well as macrophages bearing alveolar or interstitial phenotype. The transcriptome of these macrophages shows enrichment of genes associated with prostaglandin biosynthesis and genes that drive T cell chemotaxis and differentiation. Therapeutic depletion of post-resolution macrophages, inhibition of prostaglandin E2 (PGE2) synthesis or treatment with an EP4 antagonist, MF498, reduce numbers of lung CD4+/CD44+/CD62L+ and CD4+/CD44+/CD62L-/CD27+ T cells as well as their expression of the α-integrin, CD103. The T cells fail to reappear and reactivate upon secondary challenge for up to six weeks following primary infection. Concomitantly, EP4 antagonism through MF498 causes accumulation of lung macrophages and marked tissue fibrosis. Our study thus shows that PGE2 signalling, predominantly via EP4, plays an important role during the second wave of immune activity following resolution of inflammation. This secondary immune activation drives local tissue-resident T cell development while limiting tissue injury.


Dinoprostone , Disease Models, Animal , Lung , Macrophages , Mice, Inbred C57BL , Pneumonia, Pneumococcal , Receptors, Prostaglandin E, EP4 Subtype , Streptococcus pneumoniae , Animals , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/pathology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/metabolism , Mice , Dinoprostone/metabolism , Streptococcus pneumoniae/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Macrophages/immunology , Macrophages/metabolism , Lung/immunology , Lung/pathology , Lung/microbiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Integrin alpha Chains/metabolism , Integrin alpha Chains/genetics , Female , Antigens, CD/metabolism , Antigens, CD/genetics , T-Lymphocytes/immunology
5.
Int J Biol Macromol ; 267(Pt 1): 131334, 2024 May.
Article En | MEDLINE | ID: mdl-38582475

Chitin and its derivative chitosan (Q) are abundant structural elements in nature. Q has modulatory and anti-inflammatory effects and also regulates the expression of adhesion molecules. The interaction between cells expressing the αEß7 integrin and E-cadherin facilitates tolerogenic signal transmission and localization of lymphocytes at the frontline for interaction with luminal antigens. In this study we evaluated the ability of orally administered Q to stimulate E-cadherin and CD103 expression in vitro and in vivo. Our findings show that Q promoted epithelial cell migration, accelerated wound healing and increased E-cadherin expression in IEC-18 cells and isolated intestinal epithelial cells (IECs) after Q feeding. The upregulation of E-cadherin was dependent on TLR4 and IFNAR signaling, triggering CD103 expression in lymphocytes. Q reinforced the E-cadherin-αEß7 axis, crucial for intestinal barrier integrity and contributed to the localization of lymphocytes on the epithelium.


Antigens, CD , Cadherins , Chitosan , Integrin alpha Chains , Intestinal Mucosa , Signal Transduction , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Chitosan/pharmacology , Chitosan/chemistry , Cadherins/metabolism , Signal Transduction/drug effects , Integrin alpha Chains/metabolism , Mice , Antigens, CD/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Cell Movement/drug effects , Cell Line , Intestines/drug effects , Rats , Male
6.
Nature ; 628(8009): 854-862, 2024 Apr.
Article En | MEDLINE | ID: mdl-38570678

The intestinal immune system is highly adapted to maintaining tolerance to the commensal microbiota and self-antigens while defending against invading pathogens1,2. Recognizing how the diverse network of local cells establish homeostasis and maintains it in the complex immune environment of the gut is critical to understanding how tolerance can be re-established following dysfunction, such as in inflammatory disorders. Although cell and molecular interactions that control T regulatory (Treg) cell development and function have been identified3,4, less is known about the cellular neighbourhoods and spatial compartmentalization that shapes microorganism-reactive Treg cell function. Here we used in vivo live imaging, photo-activation-guided single-cell RNA sequencing5-7 and spatial transcriptomics to follow the natural history of T cells that are reactive towards Helicobacter hepaticus through space and time in the settings of tolerance and inflammation. Although antigen stimulation can occur anywhere in the tissue, the lamina propria-but not embedded lymphoid aggregates-is the key microniche that supports effector Treg (eTreg) cell function. eTreg cells are stable once their niche is established; however, unleashing inflammation breaks down compartmentalization, leading to dominance of CD103+SIRPα+ dendritic cells in the lamina propria. We identify and validate the putative tolerogenic interaction between CD206+ macrophages and eTreg cells in the lamina propria and identify receptor-ligand pairs that are likely to govern the interaction. Our results reveal a spatial mechanism of tolerance in the lamina propria and demonstrate how knowledge of local interactions may contribute to the next generation of tolerance-inducing therapies.


Intestinal Mucosa , Mucous Membrane , T-Lymphocytes, Regulatory , Animals , Female , Male , Mice , Antigens, CD/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Profiling , Helicobacter hepaticus/immunology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Immune Tolerance/immunology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Integrin alpha Chains/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mucous Membrane/cytology , Mucous Membrane/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/immunology , Single-Cell Gene Expression Analysis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/cytology , Transcriptome
7.
Nat Immunol ; 25(5): 834-846, 2024 May.
Article En | MEDLINE | ID: mdl-38561495

Cancer remains one of the leading causes of mortality worldwide, leading to increased interest in utilizing immunotherapy strategies for better cancer treatments. In the past decade, CD103+ T cells have been associated with better clinical prognosis in patients with cancer. However, the specific immune mechanisms contributing toward CD103-mediated protective immunity remain unclear. Here, we show an unexpected and transient CD61 expression, which is paired with CD103 at the synaptic microclusters of T cells. CD61 colocalization with the T cell antigen receptor further modulates downstream T cell antigen receptor signaling, improving antitumor cytotoxicity and promoting physiological control of tumor growth. Clinically, the presence of CD61+ tumor-infiltrating T lymphocytes is associated with improved clinical outcomes, mediated through enhanced effector functions and phenotype with limited evidence of cellular exhaustion. In conclusion, this study identified an unconventional and transient CD61 expression and pairing with CD103 on human immune cells, which potentiates a new target for immune-based cellular therapies.


Antigens, CD , Apyrase , Integrin alpha Chains , Receptors, Antigen, T-Cell , Signal Transduction , Animals , Humans , Mice , Antigens, CD/metabolism , Antigens, CD/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Integrin alpha Chains/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology
8.
Gastroenterology ; 166(6): 1069-1084, 2024 Jun.
Article En | MEDLINE | ID: mdl-38445519

BACKGROUND & AIMS: Although the presence of tertiary lymphoid structures (TLS) correlates with positive responses to immunotherapy in many solid malignancies, the mechanism by which TLS enhances antitumor immunity is not well understood. The present study aimed to investigate the underlying cross talk circuits between B cells and tissue-resident memory T (Trm) cells within the TLS and to understand their role in the context of immunotherapy. METHODS: Immunostaining and H&E staining of TLS and chemokine (C-X-C motif) ligand 13 (CXCL13)+ cluster of differentiation (CD)103+CD8+ Trm cells were performed on tumor sections from patients with gastric cancer (GC). The mechanism of communication between B cells and CXCL13+CD103+CD8+ Trm cells was determined in vitro and in vivo. The effect of CXCL13+CD103+CD8+ Trm cells in suppressing tumor growth was evaluated through anti-programmed cell death protein (PD)-1 therapy. RESULTS: The presence of TLS and CXCL13+CD103+CD8+ Trm cells in tumor tissues favored a superior response to anti-PD-1 therapy in patients with GC. Additionally, our research identified that activated B cells enhanced CXCL13 and granzyme B secretion by CD103+CD8+ Trm cells. Mechanistically, B cells facilitated the glycolysis of CD103+CD8+ Trm cells through the lymphotoxin-α/tumor necrosis factor receptor 2 (TNFR2) axis, and the mechanistic target of rapamycin signaling pathway played a critical role in CD103+CD8+ Trm cells glycolysis during this process. Moreover, the presence of TLS and CXCL13+CD103+CD8+ Trm cells correlated with potent responsiveness to anti-PD-1 therapy in a TNFR2-dependent manner. CONCLUSIONS: This study further reveals a crucial role for cellular communication between TLS-associated B cell and CXCL13+CD103+CD8+ Trm cells in antitumor immunity, providing valuable insights into the potential use of the lymphotoxin-α/TNFR2 axis within CXCL13+CD103+CD8+ Trm cells for advancing immunotherapy strategies in GC.


Antigens, CD , B-Lymphocytes , CD8-Positive T-Lymphocytes , Chemokine CXCL13 , Immune Checkpoint Inhibitors , Integrin alpha Chains , Memory T Cells , Programmed Cell Death 1 Receptor , Stomach Neoplasms , Tertiary Lymphoid Structures , Chemokine CXCL13/metabolism , Humans , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/drug effects , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Stomach Neoplasms/drug therapy , Antigens, CD/metabolism , Integrin alpha Chains/metabolism , Integrin alpha Chains/immunology , Memory T Cells/immunology , Memory T Cells/metabolism , Animals , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Granzymes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Immunologic Memory , Signal Transduction/immunology , Tumor Microenvironment/immunology , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Mice , Immunotherapy/methods , Cell Line, Tumor
9.
Eur J Immunol ; 54(5): e2350873, 2024 May.
Article En | MEDLINE | ID: mdl-38501878

Resident memory T (TRM) cells have been recently established as an important subset of memory T cells that provide early and essential protection against reinfection in the absence of circulating memory T cells. Recent findings showing that TRM expand in vivo after repeated antigenic stimulation indicate that these memory T cells are not terminally differentiated. This suggests an opportunity for in vitro TRM expansion to apply in an immunotherapy setting. However, it has also been shown that TRM may not maintain their identity and form circulating memory T cells after in vivo restimulation. Therefore, we set out to determine how TRM respond to antigenic activation in culture. Using Listeria monocytogenes and LCMV infection models, we found that TRM from the intraepithelial compartment of the small intestine expand in vitro after antigenic stimulation and subsequent resting in homeostatic cytokines. A large fraction of the expanded TRM retained their phenotype, including the expression of key TRM markers CD69 and CD103 (ITGAE). The optimal culture of TRM required low O2 pressure to maintain the expression of these and other TRM-associated molecules. Expanded TRM retained their effector capacity to produce cytokines after restimulation, but did not acquire a highly glycolytic profile indicative of effector T cells. The proteomic analysis confirmed TRM profile retention, including expression of TRM-related transcription factors, tissue retention factors, adhesion molecules, and enzymes involved in fatty acid metabolism. Collectively, our data indicate that limiting oxygen conditions supports in vitro expansion of TRM cells that maintain their TRM phenotype, at least in part, suggesting an opportunity for therapeutic strategies that require in vitro expansion of TRM.


Immunologic Memory , Listeria monocytogenes , Memory T Cells , Animals , Memory T Cells/immunology , Immunologic Memory/immunology , Mice , Listeria monocytogenes/immunology , Antigens, CD/metabolism , Antigens, CD/immunology , Integrin alpha Chains/metabolism , Mice, Inbred C57BL , Listeriosis/immunology , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Cytokines/metabolism , Cytokines/immunology , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , Intestinal Mucosa/immunology , CD8-Positive T-Lymphocytes/immunology , Intestine, Small/immunology , Cells, Cultured
10.
Eur J Immunol ; 54(5): e2350839, 2024 May.
Article En | MEDLINE | ID: mdl-38430190

The active vitamin A metabolite, all-trans-retinoic acid (RA), primes precursor dendritic cells (DCs) into a mucosal phenotype with tolerogenic properties characterized by the expression of integrin CD103. CD103+ DCs can counteract pathogenic Th1 and Th17 in inflammatory bowel disease (IBD) or celiac disease (CD). Tolerogenic manipulation of DCs using nanoparticles carrying tolerogenic adjuvants and disease-specific antigens is a valuable treatment strategy to induce antigen-specific mucosal tolerance in vivo. Here, we investigated the effects of RA-loaded liposomes on human DC phenotype and function, including DC-driven T-cell development, both during the generation of monocyte-derived DCs (moDCs) as well as by priming immature moDCs. RA liposomes drove CD103+ DC differentiation as well as ALDH1A2 expression in DCs. Neutrophil-dependent Th17 cell development was reduced by RA-liposome-differentiated and RA-liposome-primed DCs. Moreover, RA liposome treatment shifted T-cell development toward a Th2 cell profile. Importantly, RA liposomes induced the development of IL-10-producing and FoxP3+ regulatory T cells (Tregs) of various Treg subsets, including ICOS+ Tregs, that were potent inhibitors of bystander memory T-cell proliferation. Taken together, RA-loaded liposomes could be a novel treatment avenue for IBD or CD patients.


Aldehyde Dehydrogenase 1 Family , Antigens, CD , Cell Differentiation , Dendritic Cells , Integrin alpha Chains , Liposomes , Retinal Dehydrogenase , T-Lymphocytes, Regulatory , Th17 Cells , Tretinoin , Humans , Tretinoin/pharmacology , Integrin alpha Chains/metabolism , Th17 Cells/immunology , Dendritic Cells/immunology , Dendritic Cells/drug effects , Antigens, CD/immunology , Antigens, CD/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Cell Differentiation/drug effects , Cell Differentiation/immunology , Retinal Dehydrogenase/metabolism , Immune Tolerance/drug effects , Cells, Cultured , Interleukin-10/metabolism , Interleukin-10/immunology , Forkhead Transcription Factors/metabolism , Inflammatory Bowel Diseases/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Celiac Disease/immunology
11.
Appl Biochem Biotechnol ; 196(1): 245-260, 2024 Jan.
Article En | MEDLINE | ID: mdl-37119505

DNA methylation plays a vital role during the development of tumorigenesis. The purpose of this study is to identify candidate DNA methylation drivers during progression of bladder cancer (BLCA). The methylation spectrum in bladder cancer tissues was detected by CHARM analysis, and methylated ITGA8 was selected for further study due to its low expression. Methylation levels in BLCA tissues and cells were detected with methylated-specific PCR (MSP), while mRNA expression and methylation of ITGA8 were detected by qRT-PCR and MSP. After treatment with 5-Aza-dC (DNA methylation inhibitor), the proliferation, migration, and invasion abilities of BLCA cells were determined by MTT, wound healing, and transwell assays, respectively. Flow cytometric analysis was performed to evaluate any variance in the cell cycle. In addition, the effect of demethylated ITGA8 on BLCA tumor growth was verified with an in vivo xenograft tumor model. Based on the methylation profiling of BLCA, ITGA8 was identified to be hypermethylated. ITGA8 methylation levels in BLCA tissues and cells were upregulated, and 5-Aza-dC significantly suppressed ITGA8 methylation levels and increased ITGA8 mRNA expression. Furthermore, after treatment with 5-Aza-dC, the propagation, migration, and invasiveness of the cancer cells were inhibited, and more cancer cells were arrested at the G0/G1 phase. In vivo assays further demonstrated that 5-Aza-dC could impede BLCA tumor growth by repressing methylation levels of ITGA8 and increasing ITGA8 mRNA expression. Hypermethylated ITGA8 facilitated BLCA progression, and 5-Aza-dC treatment inhibited BLCA cell propagation and metastasis by decreasing methylation levels of ITGA8 and inducing cell cycle arrest.


DNA Methylation , Urinary Bladder Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation/genetics , Azacitidine/pharmacology , Azacitidine/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , RNA, Messenger/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism
12.
J Pathol ; 261(2): 184-197, 2023 10.
Article En | MEDLINE | ID: mdl-37565309

Psoriasis is a chronic inflammatory skin condition. Repeated epicutaneous application of Aldara® (imiquimod) cream results in psoriasiform dermatitis in mice. The Aldara®-induced psoriasiform dermatitis (AIPD) mouse model has been used to examine the pathogenesis of psoriasis. Here, we used a forward genetics approach in which we compared AIPD that developed in 13 different inbred mouse strains to identify genes and pathways that modulated disease severity. Among our primary results, we found that the severity of AIPD differed substantially between different strains of inbred mice and that these variations were associated with polymorphisms in Itga11. The Itga11 gene encodes the integrin α11 subunit that heterodimerizes with the integrin ß1 subunit to form integrin α11ß1. Less information is available about the function of ITGA11 in skin inflammation; however, a role in the regulation of cutaneous wound healing, specifically the development of dermal fibrosis, has been described. Experiments performed with Itga11 gene-deleted (Itga11-/- ) mice revealed that the integrin α11 subunit contributes substantially to the clinical phenotype as well as the histopathological and molecular findings associated with skin inflammation characteristic of AIPD. Although the skin transcriptomes of Itga11-/- and WT mice do not differ from one another under physiological conditions, distinct transcriptomes emerge in these strains in response to the induction of AIPD. Most of the differentially expressed genes contributed to extracellular matrix organization, immune system, and metabolism of lipids pathways. Consistent with these findings, we detected a reduced number of fibroblasts and inflammatory cells, including macrophages, T cells, and tissue-resident memory T cells in skin samples from Itga11-/- mice in response to AIPD induction. Collectively, our results reveal that Itga11 plays a critical role in promoting skin inflammation in AIPD and thus might be targeted for the development of novel therapeutics for psoriasiform skin conditions. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Dermatitis , Integrin alpha Chains , Psoriasis , Animals , Mice , Dermatitis/genetics , Dermatitis/pathology , Disease Models, Animal , Imiquimod/adverse effects , Inflammation/pathology , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Psoriasis/chemically induced , Psoriasis/genetics , Skin/pathology
13.
J Immunol ; 211(4): 551-562, 2023 08 15.
Article En | MEDLINE | ID: mdl-37341508

Dermal regulatory T cells (Tregs) are essential for maintenance of skin homeostasis and control of skin inflammatory responses. In mice, Tregs in the skin are characterized by high expression of CD103, the αE integrin. Evidence indicates that CD103 promotes Treg retention within the skin, although the mechanism underlying this effect is unknown. The main ligand of CD103, E-cadherin, is predominantly expressed by cells in the epidermis. However, because Tregs are predominantly located within the dermis, the nature of the interactions between E-cadherin and CD103-expressing Tregs is unclear. In this study, we used multiphoton intravital microscopy to examine the contribution of CD103 to Treg behavior in resting and inflamed skin of mice undergoing oxazolone-induced contact hypersensitivity. Inhibition of CD103 in uninflamed skin did not alter Treg behavior, whereas 48 h after inducing contact hypersensitivity by oxazolone challenge, CD103 inhibition increased Treg migration. This coincided with E-cadherin upregulation on infiltrating myeloid leukocytes in the dermis. Using CD11c-enhanced yellow fluorescent protein (EYFP) × Foxp3-GFP dual-reporter mice, inhibition of CD103 was found to reduce Treg interactions with dermal dendritic cells. CD103 inhibition also resulted in increased recruitment of effector CD4+ T cells and IFN-γ expression in challenged skin and resulted in reduced glucocorticoid-induced TNFR-related protein expression on Tregs. These results demonstrate that CD103 controls intradermal Treg migration, but only at later stages in the inflammatory response, when E-cadherin expression in the dermis is increased, and provide evidence that CD103-mediated interactions between Tregs and dermal dendritic cells support regulation of skin inflammation.


Dermatitis, Contact , T-Lymphocytes, Regulatory , Animals , Mice , Cadherins/metabolism , Dermatitis, Contact/metabolism , Inflammation/metabolism , Integrin alpha Chains/metabolism , Oxazolone/metabolism , T-Lymphocytes, Regulatory/metabolism
14.
Cell Rep ; 42(6): 112550, 2023 06 27.
Article En | MEDLINE | ID: mdl-37224018

Diabetic kidney disease (DKD) is the most prevalent chronic kidney disease. Macrophage infiltration in the kidney is critical for the progression of DKD. However, the underlying mechanism is far from clear. Cullin 4B (CUL4B) is the scaffold protein in CUL4B-RING E3 ligase complexes. Previous studies have shown that depletion of CUL4B in macrophages aggravates lipopolysaccharide-induced peritonitis and septic shock. In this study, using two mouse models for DKD, we demonstrate that myeloid deficiency of CUL4B alleviates diabetes-induced renal injury and fibrosis. In vivo and in vitro analyses reveal that loss of CUL4B suppresses migration, adhesion, and renal infiltration of macrophages. Mechanistically, we show that high glucose upregulates CUL4B in macrophages. CUL4B represses expression of miR-194-5p, which leads to elevated integrin α9 (ITGA9), promoting migration and adhesion. Our study suggests the CUL4B/miR-194-5p/ITGA9 axis as an important regulator for macrophage infiltration in diabetic kidneys.


Diabetes Mellitus , Diabetic Nephropathies , MicroRNAs , Animals , Mice , Cullin Proteins/genetics , Cullin Proteins/metabolism , Diabetes Mellitus/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Integrin alpha Chains/metabolism , Macrophages/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
15.
Cell Rep ; 42(5): 112501, 2023 05 30.
Article En | MEDLINE | ID: mdl-37178117

Locoregional monotherapy with heterodimeric interleukin (IL)-15 (hetIL-15) in a triple-negative breast cancer (TNBC) orthotopic mouse model resulted in tumor eradication in 40% of treated mice, reduction of metastasis, and induction of immunological memory against breast cancer cells. hetIL-15 re-shaped the tumor microenvironment by promoting the intratumoral accumulation of cytotoxic lymphocytes, conventional type 1 dendritic cells (cDC1s), and a dendritic cell (DC) population expressing both CD103 and CD11b markers. These CD103intCD11b+DCs share phenotypic and gene expression characteristics with both cDC1s and cDC2s, have transcriptomic profiles more similar to monocyte-derived DCs (moDCs), and correlate with tumor regression. Therefore, hetIL-15, a cytokine directly affecting lymphocytes and inducing cytotoxic cells, also has an indirect rapid and significant effect on the recruitment of myeloid cells, initiating a cascade for tumor elimination through innate and adoptive immune mechanisms. The intratumoral CD103intCD11b+DC population induced by hetIL-15 may be targeted for the development of additional cancer immunotherapy approaches.


Antineoplastic Agents , Neoplasms , Mice , Animals , Integrin alpha Chains/metabolism , Neoplasms/metabolism , Cytokines/metabolism , Dendritic Cells/metabolism , Lymphocytes/metabolism , Antineoplastic Agents/metabolism , Immunologic Factors/metabolism , Mice, Inbred C57BL , Tumor Microenvironment
16.
Cell Rep ; 42(5): 112431, 2023 05 30.
Article En | MEDLINE | ID: mdl-37099426

While dysbiosis in the gut is implicated in the impaired induction of oral tolerance generated in mesenteric lymph nodes (MesLNs), how dysbiosis affects this process remains unclear. Here, we describe that antibiotic-driven gut dysbiosis causes the dysfunction of CD11c+CD103+ conventional dendritic cells (cDCs) in MesLNs, preventing the establishment of oral tolerance. Deficiency of CD11c+CD103+ cDCs abrogates the generation of regulatory T cells in MesLNs to establish oral tolerance. Antibiotic treatment triggers the intestinal dysbiosis linked to the impaired generation of colony-stimulating factor 2 (Csf2)-producing group 3 innate lymphoid cells (ILC3s) for regulating the tolerogenesis of CD11c+CD103+ cDCs and the reduced expression of tumor necrosis factor (TNF)-like ligand 1A (TL1A) on CD11c+CD103+ cDCs for generating Csf2-producing ILC3s. Thus, antibiotic-driven intestinal dysbiosis leads to the breakdown of crosstalk between CD11c+CD103+ cDCs and ILC3s for maintaining the tolerogenesis of CD11c+CD103+ cDCs in MesLNs, responsible for the failed establishment of oral tolerance.


Dysbiosis , Immunity, Innate , Humans , Dysbiosis/metabolism , Lymphocytes/metabolism , Integrin alpha Chains/metabolism , Dendritic Cells/metabolism , Anti-Bacterial Agents/metabolism , Intestinal Mucosa/metabolism
17.
J Ethnopharmacol ; 308: 116191, 2023 May 23.
Article En | MEDLINE | ID: mdl-36731809

ETHNOPHARMACOLOGICAL RELEVANCE: Dahuang Zhechong pill (DHZCP), a traditional Chinese medicine, was derived from the famous book Unk "Synopsis of Prescriptions of the Golden Chamber" during the Han dynasty. Owing to its ability to invigorate the circulation of blood in Chinese medicine, DHZCP is usually used for treating liver cirrhosis (LC) and hepatocellular carcinoma (HCC). Clinical application have shown that DHZCP exhibits satisfactory therapeutic effects in HCC adjuvant therapy; however, little is known about its underlying mechanisms. AIM OF THE STUDY: We aimed to clarify the mechanism of DHZCP against hepatic sinusoidal capillarization in rats with LC and HCC by inhibiting the MK/integrin signaling pathway of liver sinusoidal endothelial cells (LSECs). MATERIALS AND METHODS: The contents of 29 characteristic components in DHZCP were determined by ultraperformance liquid chromatography-tandem mass spectrometry. DEN (Diethylnitrosamine)-induced LC and HCC rat models were constructed, and DHZCP was administered when the disease entered the LC stage. After 4 or 12 weeks of administration, hematoxylin and eosin staining, Masson staining, Metavir score, and SSCP (Single strand conformation polymorphism) gene mutation detection were used to confirm tissue fibrosis and cancer. The levels of NO, ET-1 and TXA2, which can regulate vasomotor functions and activate the MK/Itgα6/Src signaling pathway were evaluated by using immunohistochemistry, chemiluminescence, immunofluorescence, Western blot analysis, and enzyme-linked immunosorbent assay (ELISA). Similar methods were also used to evaluate the levels of VEGF, VEGFR, Ang-2 and Tie, which can promote pathological angiogenesis and activate the MK/Itgα4/NF-κB signaling pathway. In vitro cell experiments were performed using potential pharmacodynamic molecules targeting integrins in DHZCP were selected by molecular docking, and the effects of these molecules on the function of LSECs were studied by Itgα4+ and Itgα6+ cell models. RESULTS: At the stage of LC, the animal experiments demonstrated that DHZCP mainly inhibited the MK/Itgα6 signaling pathway to increase the number and size of hepatic sinus fenestration, reversed the ET-1/NO and TXA2/NO ratios, regulated hepatic sinus relaxation and contraction balance, reduced the portal vein pressure, and inhibited cirrhotic carcinogenesis. At the HCC stage, DHZCP could also significantly inhibit the MK/Itgα4 signaling pathway, reduce pathological angiogenesis, and alleviate disease progression. The results of the cell experiments showed that Rhein, Naringenin, Liquiritin and Emodin-8-O-ß-D-glucoside (PMEG) were involved in vascular regulation by affecting the MK/integrin signaling pathway. Liquiritin and PMEG mainly blocked the MK/α6 signal, which is important in regulating the vasomotor function of the liver sinus. Naringenin and Rhein mainly acted by blocked the signaling of MK/α4 action signal, which are potent molecules that inhibit pathological angiogenesis. CONCLUSIONS: DHZCP could improve the hepatic sinusoidal capillarization of LC and HCC by inhibiting the MK/Itgα signaling pathway and inhibited disease progression. Rhein, Naringenin, Liquiritin and PMEG were the main active molecules that affected the MK/Itgα signaling pathway.


Carcinoma, Hepatocellular , Drugs, Chinese Herbal , Integrin alpha Chains , Liver Cirrhosis , Liver Neoplasms , Neovascularization, Pathologic , Animals , Rats , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Disease Progression , Endothelial Cells/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Neoplasms/blood supply , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Molecular Docking Simulation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Signal Transduction , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Capillaries/drug effects , Liver/blood supply , Liver/drug effects , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism
18.
Mol Med Rep ; 26(5)2022 Nov.
Article En | MEDLINE | ID: mdl-36069233

Extracellular matrix tenascin­X (TNX) is the largest member of the tenascin family. Our previous study demonstrated that TNX was involved in hepatic dysfunction, including fibrosis, in mice that were administered a high­fat and high­cholesterol diet with high levels of phosphorus and calcium. The present study investigated whether overexpression of both the fibrinogen domain of TNX (TNX­FG) and integrin α11, one of the TNX cell surface receptors, induces in vitro fibrosis in LX­2 human hepatic stellate cells. Overexpression of both a 15­amino acid peptide (hTNX­FGFFFF) derived from the TNX­FG domain and integrin α11 induced the expression of type I collagen α1 chain (COL1A1). Treatment with verteporfin [YAP (Yes­associated protein) inhibitor] attenuated the elevated COL1A1 expression elicited by overexpression of both hTNX­FGFFFF and integrin α11. In addition, small interfering RNA­mediated knockdown of YAP1 resulted in a decrease in COL1A1 expression induced by overexpression of both hTNX­FGFFFF and integrin α11. These results indicated that overexpression of both hTNX­FGFFFF and integrin α11 induced COL1A1 expression via the YAP signaling pathway.


Integrins , Tenascin , Amino Acids , Animals , Extracellular Matrix/metabolism , Fibrinogen , Fibrosis , Humans , Integrin alpha Chains/metabolism , Integrins/metabolism , Mice , Peptides , Tenascin/genetics
19.
Placenta ; 128: 73-82, 2022 10.
Article En | MEDLINE | ID: mdl-36088840

INTRODUCTION: Maternal glucocorticoid exposure increases the risk of preterm delivery; however, the association between glucocorticoids and preterm premature rupture of membranes (pPROM)-a direct cause of preterm delivery-has rarely been investigated. METHODS: To examine this association, we evaluated the clinical data of patients with systemic lupus erythematosus (SLE). Mechanism analysis was performed in both human amnion-derived mesenchymal cells (as a model for fetal membranes) and the amnion from SLE patients. We characterized the effects of glucocorticoids on the amnion in both models through comprehensive gene expression profiling and by electric cell-substrate impedance sensing in the mesenchymal cells. RESULTS: The average glucocorticoid dose in cases with pPROM (13.3 mg/day, n = 10) was significantly higher than in those without pPROM (8.5 mg/day, n = 65; P < 0.01) among pregnant patients with well-controlled SLE (SLEDAI <4, n = 75); however, we did not observe a statistically significant difference in it between cases with or without chorioamnionitis. Glucocorticoid-treated human amnion mesenchymal cells showed decreased electric resistance between cells, indicating increased permeability. Differentially expressed genes upon glucocorticoid treatment were significantly enriched with cell adhesion-related genes. Among them, ITGA8 was strikingly induced in both the amnion mesenchymal cells and in amnion derived from patients with SLE. DISCUSSION: We observed an association between glucocorticoids and pPROM with non-infectious etiology. Our findings indicate that glucocorticoids increase amnion permeability and modulate cell-adhesion related genes. ITGA8 represents a primary molecule that triggers pPROM through fibrotic remodeling and preventing resealing of the rupture site in fetal amnion.


Fetal Membranes, Premature Rupture , Glucocorticoids , Integrin alpha Chains , Lupus Erythematosus, Systemic , Premature Birth , Amnion/metabolism , Female , Fetal Membranes, Premature Rupture/metabolism , Gene Expression , Glucocorticoids/adverse effects , Humans , Infant, Newborn , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Lupus Erythematosus, Systemic/metabolism , Pregnancy , Premature Birth/metabolism
20.
Am J Physiol Heart Circ Physiol ; 322(6): H1057-H1071, 2022 06 01.
Article En | MEDLINE | ID: mdl-35522553

Pathological myocardial hypertrophy in response to an increase in left ventricular (LV) afterload may ultimately lead to heart failure. Cell surface receptors bridge the interface between the cell and the extracellular matrix (ECM) in cardiac myocytes and cardiac fibroblasts and have been suggested to be important mediators of pathological myocardial hypertrophy. We identify for the first time that integrin α11 (α11) is preferentially upregulated among integrin ß1 heterodimer-forming α-subunits in response to increased afterload induced by aortic banding (AB) in wild-type (WT) mice. Mice were anesthetized in a chamber with 4% isoflurane and 95% oxygen before being intubated and ventilated with 2.5% isoflurane and 97% oxygen. For pre- and postoperative analgesia, animals were administered 0.02-mL buprenorphine (0.3 mg/mL) subcutaneously. Surprisingly, mice lacking α11 develop myocardial hypertrophy following AB comparable to WT. In the mice lacking α11, we further show a compensatory increase in the expression of another mechanoreceptor, syndecan-4, following AB compared with WT AB mice, indicating that syndecan-4 compensated for lack of α11. Intriguingly, mice lacking mechanoreceptors α11 and syndecan-4 show ablated myocardial hypertrophy following AB compared with WT mice. Expression of the main cardiac collagen isoforms col1a2 and col3a1 was significantly reduced in AB mice lacking mechanoreceptors α11 and syndecan-4 compared with WT AB.NEW & NOTEWORTHY Despite their putative importance in stress sensing, the specific integrin α-subunit(s) involved in cardiac hypertrophy has not been identified. Here, we show that α11 and syndecan-4 are critical and interdependent mediators of the hypertrophic response to increased LV afterload. We demonstrate in cells lacking both receptors an interdependent reduction in cell attachment to the major cardiac extracellular matrix components, suggesting that their interplay represents an important mechanism for stress sensing in cardiac cells.


Isoflurane , Syndecan-4 , Animals , Cardiomegaly/metabolism , Integrin alpha Chains/metabolism , Integrins/metabolism , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Receptors, Collagen , Syndecan-4/genetics , Syndecan-4/metabolism
...