Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.627
Filter
1.
Cancer Immunol Immunother ; 73(9): 176, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38954030

ABSTRACT

BACKGROUND: Tissue-resident memory CD103+CD8+ T cells (CD103+CD8+ TRMs) are important components of anti-tumor immunity. However, the significance of CD103+CD8+ TRMs in colorectal cancer (CRC) and their advantages remain unclear. METHODS: Clinical data and specimens were used to evaluate the significance of CD103+CD8+ TRMs in CRC. A mouse subcutaneous tumorigenesis model and colony-formation assay were conducted to evaluate the anti-tumor effects of CD103+CD8+ TRMs. Finally, the infiltration density and function of CD103+CD8+ TRMs in the tumors were evaluated using flow cytometry. RESULTS: In this study, we showed that highly infiltrated CD103+CD8+ TRMs were associated with earlier clinical stage and negative VEGF expression in CRC patients and predicted a favorable prognosis for CRC/CRC liver metastases patients. Interestingly, we also found that CD103+CD8+ TRMs may have predictive potential for whether CRC develops liver metastasis in CRC. In addition, we found a positive correlation between the ratio of the number of α-SMA+ vessels to the sum of the number of α-SMA+ and CD31+ vessels in CRC, and the infiltration level of CD103+CD8+ TRMs. In addition, anti-angiogenic therapy promoted infiltration of CD103+CD8+ TRMs and enhanced their ability to secrete interferon (IFN)-γ, thus further improving the anti-tumor effect. Moreover, in vivo experiments showed that compared with peripheral blood CD8+ T cells, CD103+CD8+ TRMs infused back into the body could also further promote CD8+ T cells to infiltrate the tumor, and they had a stronger ability to secrete IFN-γ, which resulted in better anti-tumor effects. CONCLUSION: We demonstrated that CD103+CD8+ TRMs have the potential for clinical applications and provide new ideas for combined anti-tumor therapeutic strategies, such as anti-tumor angiogenesis therapy and CAR-T combined immunotherapy.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Colorectal Neoplasms , Immunologic Memory , Integrin alpha Chains , Liver Neoplasms , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Integrin alpha Chains/metabolism , Integrin alpha Chains/immunology , Animals , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Antigens, CD/metabolism , Prognosis , Female , Male , Biomarkers, Tumor/metabolism , Memory T Cells/immunology , Memory T Cells/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Middle Aged
2.
Cell Rep ; 43(6): 114258, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38781073

ABSTRACT

Transforming growth factor ß (TGF-ß) represents a well-established signal required for tissue-resident memory T cell (TRM) formation at intestinal surfaces, regulating the expression of a large collection of genes coordinately promoting intestinal TRM differentiation. The functional contribution from each TGF-ß-controlled transcription factor is not entirely known. Here, we find that TGF-ß-induced T-bet downregulation and Hic1 induction represent two critical events during intestinal TRM differentiation. Importantly, T-bet deficiency significantly rescues intestinal TRM formation in the absence of the TGF-ß receptor. Hic1 induction further strengthens TRM maturation in the absence of TGF-ß and T-bet. Our results reveal that provision of certain TGF-ß-induced molecular events can partially replace TGF-ß signaling to promote the establishment of intestinal TRMs, which allows the functional dissection of TGF-ß-induced transcriptional targets and molecular mechanisms for TRM differentiation.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Integrin alpha Chains , Kruppel-Like Transcription Factors , T-Box Domain Proteins , Transforming Growth Factor beta , Animals , Transforming Growth Factor beta/metabolism , Kruppel-Like Transcription Factors/metabolism , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Integrin alpha Chains/metabolism , Antigens, CD/metabolism , Cell Differentiation , Mice, Inbred C57BL , Intestines/immunology , Signal Transduction , Memory T Cells/metabolism , Memory T Cells/immunology , Immunologic Memory , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology
3.
Commun Biol ; 7(1): 583, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755265

ABSTRACT

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.


Subject(s)
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
4.
J Immunol Res ; 2024: 5582151, 2024.
Article in English | MEDLINE | ID: mdl-38690552

ABSTRACT

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.


Subject(s)
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
5.
Nat Commun ; 15(1): 4326, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773113

ABSTRACT

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.


Subject(s)
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
6.
Transplantation ; 108(7): 1551-1557, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38557650

ABSTRACT

BACKGROUND: Machine perfusion is the preferred preservation method for deceased donor kidneys. Perfusate fluid, which contains a complex mixture of components, offers potential insight into the organ's viability and function. This study explored immune cell release, particularly tissue-resident lymphocytes (TRLs), during donor kidney machine perfusion and its correlation with injury markers. METHODS: Perfusate samples from hypothermic machine perfusion (HMP; n = 26) and normothermic machine perfusion (NMP; n = 16) of human donor kidneys were analyzed for TRLs using flow cytometry. Residency was defined by expressions of CD69, CD103, and CD49as. TRL release was quantified exclusively in NMP. Additionally, levels of cell-free DNA, neutrophil gelatinase-associated lipocalin, and soluble E-cadherin (sE-cadherin) were measured in NMP supernatants with quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS: Both HMP and NMP samples contained a heterogeneous population of TRLs, including CD4 + tissue-resident memory T cells, CD8 + tissue-resident memory T cells, tissue-resident natural killer cells, tissue-resident natural killer T cells, and helper-like innate lymphoid cells. Median TRL proportions among total CD45 + lymphocytes were 0.89% (NMP) and 0.84% (HMP). TRL quantities in NMP did not correlate with donor characteristics, perfusion parameters, posttransplant outcomes, or cell-free DNA and neutrophil gelatinase-associated lipocalin concentrations. However, CD103 + TRL release positively correlated with the release of sE-cadherin, the ligand for the CD103 integrin. CONCLUSIONS: Human donor kidneys release TRLs during both HMP and NMP. The release of CD103 + TRLs was associated with the loss of their ligand sE-cadherin but not with general transplant injury biomarkers.


Subject(s)
Antigens, CD , Kidney Transplantation , Kidney , Lipocalin-2 , Organ Preservation , Perfusion , Humans , Kidney Transplantation/methods , Perfusion/methods , Male , Middle Aged , Female , Lipocalin-2/metabolism , Lipocalin-2/analysis , Adult , Organ Preservation/methods , Antigens, CD/metabolism , Kidney/immunology , Kidney/blood supply , Tissue Donors , Lymphocytes/immunology , Lymphocytes/metabolism , Biomarkers/metabolism , Cadherins/metabolism , Aged , Integrin alpha Chains/metabolism , Flow Cytometry , Lipocalins/metabolism , Hypothermia, Induced
7.
J Immunol ; 212(12): 1904-1912, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38668728

ABSTRACT

NK cells have been shown to exhibit inflammatory and immunoregulatory functions in a variety of healthy and diseased settings. In the context of chronic viral infection and cancer, distinct NK cell populations that inhibit adaptive immune responses have been observed. To understand how these cells arise and further characterize their immunosuppressive role, we examined in vitro conditions that could polarize human NK cells into an inhibitory subset. TGF-ß1 has been shown to induce regulatory T cells in vitro and in vivo; we therefore investigated if TGF-ß1 could also induce immunosuppressive NK-like cells. First, we found that TGF-ß1/IL-15, but not IL-15 alone, induced CD103+CD49a+ NK-like cells from peripheral blood NK cells, which expressed markers previously associated with inhibitory CD56+ innate lymphoid cells, including high expression of GITR and CD101. Moreover, supernatant from ascites collected from patients with ovarian carcinoma also induced CD103+CD49a+ NK-like cells in vitro in a TGF-ß-dependent manner. Interestingly, TGF-ß1/IL-15-induced CD103+CD56+ NK-like cells suppressed autologous CD4+ T cells in vitro by reducing absolute number, proliferation, and expression of activation marker CD25. Collectively, these findings provide new insight into how NK cells may acquire an inhibitory phenotype in TGF-ß1-rich environments.


Subject(s)
Interleukin-15 , Killer Cells, Natural , Transforming Growth Factor beta1 , Humans , Killer Cells, Natural/immunology , Interleukin-15/immunology , Interleukin-15/metabolism , Transforming Growth Factor beta1/metabolism , Female , Antigens, CD/metabolism , Antigens, CD/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Integrin alpha Chains/metabolism , Integrin alpha Chains/immunology , CD56 Antigen/metabolism , Cells, Cultured , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocyte Activation/immunology
8.
Int J Biol Macromol ; 267(Pt 1): 131334, 2024 May.
Article in English | MEDLINE | ID: mdl-38582475

ABSTRACT

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.


Subject(s)
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
9.
Nature ; 628(8009): 854-862, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570678

ABSTRACT

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.


Subject(s)
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
10.
Nat Immunol ; 25(5): 834-846, 2024 May.
Article in English | MEDLINE | ID: mdl-38561495

ABSTRACT

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.


Subject(s)
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
12.
Pflugers Arch ; 476(6): 963-974, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38563997

ABSTRACT

Complex interactions of the branching ureteric bud (UB) and surrounding mesenchymal cells during metanephric kidney development determine the final number of nephrons. Impaired nephron endowment predisposes to arterial hypertension and chronic kidney disease. In the kidney, extracellular matrix (ECM) proteins are usually regarded as acellular scaffolds or as the common histological end-point of chronic kidney diseases. Since only little is known about their physiological role in kidney development, we aimed for analyzing the expression and role of fibronectin. In mouse, fibronectin was expressed during all stages of kidney development with significant changes over time. At embryonic day (E) 12.5 and E13.5, fibronectin lined the UB epithelium, which became less pronounced at E16.5 and then switched to a glomerular expression in the postnatal and adult kidneys. Similar results were obtained in human kidneys. Deletion of fibronectin at E13.5 in cultured metanephric mouse kidneys resulted in reduced kidney sizes and impaired glomerulogenesis following reduced cell proliferation and branching of the UB epithelium. Fibronectin colocalized with alpha 8 integrin and fibronectin loss caused a reduction in alpha 8 integrin expression, release of glial-derived neurotrophic factor and expression of Wnt11, both of which are promoters of UB branching. In conclusion, the ECM protein fibronectin acts as a regulator of kidney development and is a determinant of the final nephron number.


Subject(s)
Fibronectins , Kidney , Animals , Fibronectins/metabolism , Fibronectins/genetics , Mice , Humans , Kidney/metabolism , Kidney/embryology , Wnt Proteins/metabolism , Wnt Proteins/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Cell Proliferation , Integrins/metabolism , Integrins/genetics , Mice, Inbred C57BL , Extracellular Matrix/metabolism , Integrin alpha Chains
13.
Gastroenterology ; 166(6): 1069-1084, 2024 06.
Article in English | MEDLINE | ID: mdl-38445519

ABSTRACT

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.


Subject(s)
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
14.
Eur J Immunol ; 54(5): e2350839, 2024 May.
Article in English | MEDLINE | ID: mdl-38430190

ABSTRACT

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.


Subject(s)
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
15.
Eur J Immunol ; 54(5): e2350873, 2024 May.
Article in English | MEDLINE | ID: mdl-38501878

ABSTRACT

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.


Subject(s)
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
16.
Cancer Cell ; 42(4): 682-700.e12, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38428409

ABSTRACT

Cancer-associated fibroblasts (CAFs) exhibit considerable heterogeneity in advanced cancers; however, the functional annotation and mechanism of CAFs in early-stage cancers remain elusive. Utilizing single-cell RNA sequencing and spatial transcriptomic, we identify a previously unknown PDGFRα+ITGA11+ CAF subset in early-stage bladder cancer (BCa). Multicenter clinical analysis of a 910-case cohort confirms that PDGFRα+ITGA11+ CAFs are associated with lymphovascular invasion (LVI) and poor prognosis in early-stage BCa. These CAFs facilitate LVI and lymph node (LN) metastasis in early-stage BCa, as evidenced in a PDGFRα+ITGA11+ CAFs-specific deficient mouse model. Mechanistically, PDGFRα+ITGA11+ CAFs promote lymphangiogenesis via recognizing ITGA11 surface receptor SELE on lymphatic endothelial cells to activate SRC-p-VEGFR3-MAPK pathway. Further, CHI3L1 from PDGFRα+ITGA11+ CAFs aligns the surrounding matrix to assist cancer cell intravasation, fostering early-stage BCa LVI and LN metastasis. Collectively, our study reveals the crucial role of PDGFRα+ITGA11+ CAFs in shaping metastatic landscape, informing the treatment of early-stage BCa LVI.


Subject(s)
Cancer-Associated Fibroblasts , Receptor, Platelet-Derived Growth Factor alpha , Animals , Humans , Mice , Cancer-Associated Fibroblasts/pathology , Endothelial Cells , Fibroblasts/metabolism , Integrin alpha Chains , Lymphatic Metastasis/pathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism
17.
Mol Carcinog ; 63(3): 479-493, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38174862

ABSTRACT

Cancer-associated fibroblasts (CAFs) represent a major cellular component of the tumor (pre-)metastatic niche and play an essential role in omental dissemination of ovarian cancer. The omentum is rich in adipose, and adipose-derived mesenchymal stem cells (ADSCs) have been identified as a source of CAFs. However, the molecular events driving the phenotype shift of ADSCs remain largely unexplored. In this research, we focus on integrins, transmembrane receptors that have been widely involved in cellular plasticity. We found that integrin α7 (ITGA7) was the only member of the integrin family that positively correlated with both overall survival and progression-free survival in ovarian cancer through GEPIA2. The immunohistochemistry signal of ITGA7 was apparent in the tumor stroma, and a lower omental ITGA7 level was associated with metastasis. Primary ADSCs were isolated from the omentum of patients with ovarian cancer and identified by cellular morphology, biomarkers, and multilineage differentiation. The conditional medium of ovarian cancer cells induced ITGA7 expression decrease and phenotypic changes in ADSCs. Downregulation of ITGA7 in primary omental ADSCs led to decrease in stemness properties and emerge of characteristic morphology and biomarkers of CAFs. Moreover, the conditioned medium of ADSCs with ITGA7 depletion exhibited enhanced abilities to improve the migration and invasion of ovarian cancer cells in vitro. Overall, these findings indicate that loss of ITGA7 may induce the differentiation of ADSCs to CAFs that contribute to a tumor-supportive niche.


Subject(s)
Antigens, CD , Cancer-Associated Fibroblasts , Integrin alpha Chains , Integrins , Mesenchymal Stem Cells , Ovarian Neoplasms , Female , Humans , Cancer-Associated Fibroblasts/pathology , Cell Proliferation , Mesenchymal Stem Cells/metabolism , Biomarkers , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Adipose Tissue/metabolism
18.
Appl Biochem Biotechnol ; 196(1): 245-260, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37119505

ABSTRACT

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.


Subject(s)
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
19.
J Invest Dermatol ; 144(6): 1344-1352.e7, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38135026

ABSTRACT

Local immune memory develops at the site of antigen exposure and facilitates a rapid and strong local adaptive defense upon re-exposure. Resident memory T (TRM) cells play a role in local immune memory, and their cell-surface molecules CD69 and CD103 promote their tissue residency. However, the contribution of these molecules to skin immune memory remains unclear. In this study, by inducing contact hypersensitivity (CHS) in CD69KO (CD69-deficient) and CD103-deficient mice, where different degrees of TRM cell contribution are observed by repeated challenges on the right ear and a single challenge on the left ear, we found that the deficiency of CD69 but not CD103 leads to impaired CHS upon repeated antigen challenges, even although TRM cells-like CD8 T cells developed at the challenged site of CD69KO. CHS responses in both ears were diminished in CD69KO by FTY720 or CD8 neutralization, suggesting that CHS in CD69KO is ascribed to circulating CD8 T cells and that the developed TRM cell-like CD8 T cells do not behave as TRM cells. The infiltration of macrophages was reduced in the rechallenged site of CD69KO, along with the downregulation of Cxcl1 and Cxcl2. Thus, CD69 is considered essential for an effective recall response, involving the development of functional TRM cells and the recruitment of macrophages.


Subject(s)
Antigens, CD , Antigens, Differentiation, T-Lymphocyte , CD8-Positive T-Lymphocytes , Dermatitis, Contact , Immunologic Memory , Lectins, C-Type , Mice, Knockout , Animals , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, CD/metabolism , Mice , Dermatitis, Contact/immunology , Lectins, C-Type/metabolism , CD8-Positive T-Lymphocytes/immunology , Mice, Inbred C57BL , Disease Models, Animal , Integrin alpha Chains/metabolism , Skin/immunology , Skin/pathology
20.
J Pathol ; 261(2): 184-197, 2023 10.
Article in English | MEDLINE | ID: mdl-37565309

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...