Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(7): 114377, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38889005

ABSTRACT

Bone tissue represents the most frequent site of cancer metastasis. We developed a hemichannel-activating antibody, Cx43-M2. Cx43-M2, directly targeting osteocytes in situ, activates osteocytic hemichannels and elevates extracellular ATP, thereby inhibiting the growth and migration of cultured breast and osteosarcoma cancer cells. Cx43-M2 significantly decreases breast cancer metastasis, osteosarcoma growth, and osteolytic activity, while improving survival rates in mice. The antibody's inhibition of breast cancer and osteosarcoma is dose dependent in both mouse and human cancer metastatic models. Furthermore, Cx43-M2 enhances anti-tumor immunity by increasing the population and activation of tumor-infiltrating immune-promoting effector T lymphocytes, while reducing immune-suppressive regulatory T cells. Our results suggest that the Cx43-M2 antibody, by activating Cx43 hemichannels and facilitating ATP release and purinergic signaling, transforms the cancer microenvironment from a supportive to a suppressive state. Collectively, our study underscores the potential of Cx43-M2 as a therapeutic for treating breast cancer bone metastasis and osteosarcoma.

2.
Cell Rep ; 43(6): 114258, 2024 May 22.
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.

3.
bioRxiv ; 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38076957

ABSTRACT

Resident Memory T cells (TRM) play a vital role in regional immune defense in barrier organs. Although laboratory rodents have been extensively used to study fundamental TRM biology, poor isolation efficiency, sampling bias and low cell survival rates have limited our ability to conduct TRM-focused high-throughput assays. Here, we engineered a murine vaginal epithelial organoid (VEO)-CD8 T cell co-culture system that supports CD8 TRM differentiation in vitro. The three-dimensional VEOs established from murine adult stem cells resembled stratified squamous vaginal epithelium and induced gradual differentiation of activated CD8 T cells into epithelial TRM. These in vitro generated TRM were phenotypically and transcriptionally similar to in vivo TRM, and key tissue residency features were reinforced with a second cognate-antigen exposure during co-culture. TRM differentiation was not affected even when VEOs and CD8 T cells were separated by a semipermeable barrier, indicating soluble factors' involvement. Pharmacological and genetic approaches showed that TGF-ß signaling played a crucial role in their differentiation. We found that the VEOs in our model remained susceptible to viral infections and the CD8 T cells were amenable to genetic manipulation; both of which will allow detailed interrogation of antiviral CD8 T cell biology in a reductionist setting. In summary, we established a robust model which captures bonafide TRM differentiation that is scalable, open to iterative sampling, and can be subjected to high throughput assays that will rapidly add to our understanding of TRM.

4.
J Anim Sci ; 1012023 Jan 03.
Article in English | MEDLINE | ID: mdl-37422771

ABSTRACT

This study aimed to investigate the effects of ellagic acid (EA) supplementation on body weight (BW), nutrient digestibility, fecal microbiota, blood biochemical indices, and urolithin A metabolism in 1-yr-old Thoroughbred horses. A group of 18 1-yr-old Thoroughbred horses, with an average weight of 339.00 ±â€…30.11 kg, were randomly allocated into three groups of six horses each (three males and three females). The control group (n = 6) received only the basal diet, whereas test groups I (n = 6) and II (n = 6) were fed the basal diet supplemented with 15 mg/kg BW/d and 30 mg/kg BW/d of EA, respectively, for 40-d. The results showed that test group I and II horses had a significant increase in total weight gain by 49.47% and 62.74%, respectively, compared to the control group. The digestibility of various components in the diets of the test group horses was improved, including dry matter, organic matter, gross energy, neutral detergent fiber, acid detergent fiber, and calcium. Additionally, the digestibility of crude protein and phosphorus (P) in test group II horses increased significantly by 10.96% and 33.56% (P < 0.05), respectively. Moreover, EA supplementation significantly increased the fecal abundance of Firmicutes, Bacteroidetes (P < 0.05), Fibrobacterota, p-251-o5, Desemzia incerta (P < 0.05), and Fibrobacter sp. (P < 0.05), while reducing the abundance of Proteobacteria, Pseudomonadaceae, Pseudomonas, and Cupriavidus pauculus (P < 0.05 or P < 0.01). Fecal samples from test group II showed 89.47%, 100%, and 86.15% increases in the concentrations of acetic acid, valeric acid, and total volatile fatty acids, respectively. In addition, the plasma levels of total protein, and globulin increased significantly in test groups I (7.88% and 11.35%, respectively) and II (13.44% and 16.07%, respectively) compared to those in the control group (P < 0.05). The concentration of urolithin A in fecal and urine samples was positively correlated with increasing doses of EA. These findings suggest that supplemental feeding of EA improved nutrient digestibility, blood biochemical indices, and fecal microbiota in 1-yr-old Thoroughbred horses, promoting growth and development.


Ellagic acid (EA), a plant-derived feed additive, has beneficial physiological effects, including antioxidant and anti-inflammatory properties as well as intestinal microbiota regulation. Young Thoroughbred horses exhibit rapid growth and require ample nourishment. However, the underdeveloped functional anatomy of their gastrointestinal tract restricts the rate of feed utilization. Therefore, improving digestive tract function in horses at this stage promotes intestinal homeostasis, improves antioxidant and anti-inflammatory capabilities, and supports rapid growth and health. This study revealed that supplemental feeding of 1-yr-old Thoroughbred horses with EA improved nutrient digestibility and fecal floral diversity, leading to enhanced growth performance. The optimal dose was 30 mg/kg body weight.


Subject(s)
Ellagic Acid , Microbiota , Animals , Female , Male , Animal Feed/analysis , Diet/veterinary , Dietary Fiber/metabolism , Digestion , Ellagic Acid/pharmacology , Feces/microbiology , Horses , Nutrients/metabolism , Weight Gain
5.
Front Immunol ; 13: 1074698, 2022.
Article in English | MEDLINE | ID: mdl-36569850

ABSTRACT

During chronic antigen exposure, a subset of exhausted CD8+ T cells differentiate into stem cell-like or progenitor-like T cells expressing both transcription factor Tcf-1 (T cell factor-1) and co-inhibitory receptor PD-1. These Tcf-1+ stem-like or progenitor exhausted T cells represent the key target for immunotherapies. Deeper understanding of the biology of Tcf-1+PD-1+ CD8+ T cells will lead to rational design of future immunotherapies. Here, we summarize recent findings about the migratory and resident behavior of Tcf-1+ T cells. Specifically, we will focus on TGF-ß-dependent lymphoid tissue residency program of Tcf-1+ T cells, which may represent a key to understanding the differentiation and maintenance of Tcf-1+ stem-like CD8+ T cells during persistent antigen stimulation.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Differentiation , Lymphoid Tissue , Programmed Cell Death 1 Receptor
6.
Nat Commun ; 13(1): 6043, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36229613

ABSTRACT

TGF-ß signaling is necessary for CD8+ T cell differentiation into tissue resident memory T cells (TRM). Although higher frequency of CD8+ TRM cells in the tumor microenvironment is associated with better prognosis, TGF-ß-blockade typically improves rather than worsens outcomes. Here we show that in a mouse melanoma model, in the tumor-draining lymph nodes (TDLN) rather than in the tumors themselves, stem-like CD8+ T cells differentiate into TRMs in a TGF-ß and tumor antigen dependent manner. Following vaccination against a melanoma-specific epitope, most tumour-specific CD8+ T cells are maintained in a stem-like state, but a proportion of cells lost TRM status and differentiate into CX3CR1+ effector CD8+ T cells in the TDLN, which are subsequently migrating into the tumours. Disruption of TGF-ß signaling changes the dynamics of these developmental processes, with the net result of improving effector CD8+ T cell migration into the tumours. In summary, TDLN stem-like T cells transiently switch from a TGF-ß-dependent TRM differentiation program to an anti-tumor migratory effector development upon vaccination, which transition can be facilitated by targeted TGF-ß blockade.


Subject(s)
AIDS Vaccines , Cancer Vaccines , Influenza Vaccines , Melanoma , Papillomavirus Vaccines , Respiratory Syncytial Virus Vaccines , SAIDS Vaccines , Animals , Mice , Antigens, Neoplasm , BCG Vaccine , CD8-Positive T-Lymphocytes , Diphtheria-Tetanus-Pertussis Vaccine , Epitopes , Immunologic Memory , Lymphoid Tissue , Measles-Mumps-Rubella Vaccine , Melanoma/pathology , Transforming Growth Factor beta
7.
J Exp Med ; 219(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-35980385

ABSTRACT

Stem-like CD8+ T cells sustain the antigen-specific CD8+ T cell response during chronic antigen exposure. However, the signals that control the maintenance and differentiation of these cells are largely unknown. Here, we demonstrated that TGF-ß was essential for the optimal maintenance of these cells and inhibited their differentiation into migratory effectors during chronic viral infection. Mechanistically, stem-like CD8+ T cells carried a unique expression pattern of α4 integrins (i.e., α4ß1hi and α4ß7lo) controlled by TGF-ß. In the absence of TGF-ß signaling, greatly enhanced expression of migration-related markers, including altered expression of α4 integrins, led to enhanced egress of stem-like CD8+ T cells into circulation accompanied by further differentiation into transitional states. Blocking α4 integrin significantly promoted their lymphoid tissue retention and therefore partially rescued the defective maintenance of Tcf-1+ subset in the absence of TGF-ß signaling. Thus, TGF-ß promotes the maintenance and inhibits the further differentiation of stem-like T cells at least partially via enforcing their lymphoid tissue residency.


Subject(s)
CD8-Positive T-Lymphocytes , Transforming Growth Factor beta , Integrins/metabolism , Lymphoid Tissue/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism
8.
J Exp Med ; 219(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-35980386

ABSTRACT

Recent studies have defined a novel population of PD-1+ TCF-1+ stem-like CD8 T cells in chronic infections and cancer. These quiescent cells reside in lymphoid tissues, are critical for maintaining the CD8 T cell response under conditions of persistent antigen, and provide the proliferative burst after PD-1 blockade. Here we examined the role of TGF-ß in regulating the differentiation of virus-specific CD8 T cells during chronic LCMV infection of mice. We found that TGF-ß signaling was not essential for the generation of the stem-like CD8 T cells but was critical for maintaining the stem-like state and quiescence of these cells. TGF-ß regulated the unique transcriptional program of the stem-like subset, including upregulation of inhibitory receptors specifically expressed on these cells. TGF-ß also promoted the terminal differentiation of exhausted CD8 T cells by suppressing the effector-associated program. Together, the absence of TGF-ß signaling resulted in significantly increased accumulation of effector-like CD8 T cells. These findings have implications for immunotherapies in general and especially for T cell therapy against chronic infections and cancer.


Subject(s)
Lymphocytic Choriomeningitis , Neoplasms , Animals , CD8-Positive T-Lymphocytes , Lymphocytic choriomeningitis virus/physiology , Mice , Persistent Infection , Programmed Cell Death 1 Receptor , Transforming Growth Factor beta
9.
Front Immunol ; 12: 708874, 2021.
Article in English | MEDLINE | ID: mdl-34484208

ABSTRACT

Regulatory T cells (Treg) are essential to maintain immune homeostasis and prevent autoimmune disorders. While the function and molecular regulation of Foxp3+CD4+ Tregs are well established, much of CD8+ Treg biology remains to be revealed. Here, we will review the heterogenous subsets of CD8+ T cells have been named "CD8+ Treg" and mainly focus on CD122hiLy49+CD8+ Tregs present in naïve mice. CD122hiLy49+CD8+ Tregs, which depends on transcription factor Helios and homeostatic cytokine IL-15, have been established as a non-redundant regulator of germinal center (GC) reaction. Recently, we have demonstrated that TGF-ß (Transforming growth factor-ß) and transcription factor Eomes (Eomesodermin) are essential for the function and homeostasis of CD8+ Tregs. In addition, we will discuss several open questions regarding the differentiation, function and true identity of CD8+ Tregs as well as a brief comparison between two regulatory T cell subsets critical to control GC reaction, namely CD4+ TFR (follicular regulatory T cells) and CD8+ Tregs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/classification , Germinal Center/immunology , Humans , Interleukin-2 Receptor beta Subunit/analysis , Mice , NK Cell Lectin-Like Receptor Subfamily A/analysis , T-Lymphocytes, Regulatory/classification , Transforming Growth Factor beta/physiology
10.
iScience ; 24(1): 101975, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33474536

ABSTRACT

Stepwise induction of CD69 and CD103 marks distinct differentiation stages of mucosal Trms. But the majority of non-mucosal Trm lacks CD103 expression. The expression of CD69 alone cannot faithfully define Trm cells in heavily vascularized non-mucosal tissues, such as the kidney. Here, we found that a subset of kidney Trms downregulated IL-18 receptor during differentiation. Via global transcriptional analysis and parabiosis experiments, we have discovered that the downregulation of interleukin-18 receptor (IL-18R) is associated with the establishment of tissue residency. Together with the expression of CD69, IL-18Rlo exclusively identify tissue-resident cells whereas IL-18Rhi population contains both tissue-resident and migratory ones. Local cytokines including transforming growth factor ß (TGF-ß) and interferon α (IFN-α)/ß as well as TGF-ß-dependent suppression of transcription factor Tcf-1 are essential for IL-18R downregulation during kidney Trm differentiation. Together, we identified a convenient surface marker to distinguish bona fide kidney-resident CD8+ T cells as well as underlying molecular mechanisms controlling this differentiation process.

11.
J Exp Med ; 218(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32991667

ABSTRACT

In addition to Foxp3+ CD4+ regulatory T cells (CD4+ T reg cells), Foxp3- CD8+ regulatory T cells (CD8+ T reg cells) are critical to maintain immune tolerance. However, the molecular programs that specifically control CD8+ but not CD4+ T reg cells are largely unknown. Here, we demonstrate that simultaneous disruption of both TGF-ß receptor and transcription factor Eomesodermin (Eomes) in T cells results in lethal autoimmunity due to a specific defect in CD8+ but not CD4+ T reg cells. Further, TGF-ß signal maintains the regulatory identity, while Eomes controls the follicular location of CD8+ T reg cells. Both TGF-ß signal and Eomes coordinate to promote the homeostasis of CD8+ T reg cells. Together, we have identified a unique molecular program designed for CD8+ T reg cells.


Subject(s)
Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Signal Transduction/immunology , T-Box Domain Proteins/immunology , Transforming Growth Factor beta/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , CD8-Positive T-Lymphocytes/pathology , Mice , Mice, Transgenic , Signal Transduction/genetics , T-Box Domain Proteins/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics
12.
J Vis Exp ; (160)2020 06 27.
Article in English | MEDLINE | ID: mdl-32658202

ABSTRACT

Tissue-resident memory T cell (TRM) is a rapidly expanding field of immunology research. Isolating T cells from various non-lymphoid tissues is one of the key steps to investigate TRMs. There are slight variations in lymphocyte isolation protocols for different organs. Kidney is an essential non-lymphoid organ with numerous immune cell infiltration especially after pathogen exposure or autoimmune activation. In recent years, multiple labs including our own have started characterizing kidney resident CD8+ T cells in various physiological and pathological settings in both mouse and human. Due to the abundance of T lymphocytes, kidney represents an attractive model organ to study TRMs in non-mucosal or non-barrier tissues. Here, we will describe a protocol commonly used in TRM-focused labs to isolate CD8+ T cells from mouse kidneys following systemic viral infection. Briefly, using an acute lymphocytic choriomeningitis virus (LCMV) infection model in C57BL/6 mice, we demonstrate intravascular CD8+ T cell labeling, enzymatic digestion, and density gradient centrifugation to isolate and enrich lymphocytes from mouse kidneys to make samples ready for the subsequent flow cytometry analysis.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Flow Cytometry , Kidney/cytology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Immunologic Memory/immunology , Kidney/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL
13.
Immunity ; 53(1): 158-171.e6, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32640257

ABSTRACT

Tissue-resident memory (Trm) CD8+ T cells mediate protective immunity in barrier tissues, but the cues promoting Trm cell generation are poorly understood. Sensing of extracellular adenosine triphosphate (eATP) by the purinergic receptor P2RX7 is needed for recirculating CD8+ T cell memory, but its role for Trm cells is unclear. Here we showed that P2RX7 supported Trm cell generation by enhancing CD8+ T cell sensing of TGF-ß, which was necessary for tissue residency. P2RX7-deficient Trm cells progressively decayed in non-lymphoid tissues and expressed dysregulated Trm-specific markers. P2RX7 was required for efficient re-expression of the receptor TGF-ßRII through calcineurin signaling. Forced Tgfbr2 expression rescued P2RX7-deficient Trm cell generation, and TGF-ß sensitivity was dictated by P2RX7 agonists and antagonists. Forced Tgfbr2 also rescued P2RX7-deficient Trm cell mitochondrial function. Sustained P2RX7 signaling was required for long-term Trm cell maintenance, indicating that P2RX7 signaling drives induction and CD8+ T cell durability in barrier sites.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Purinergic P2X7/metabolism , Transforming Growth Factor beta/immunology , Adenosine Triphosphate/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Calcineurin/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
Sci Immunol ; 4(36)2019 06 14.
Article in English | MEDLINE | ID: mdl-31201259

ABSTRACT

CD8+ tissue-resident memory T (TRM) cells provide frontline immunity in mucosal tissues. The mechanisms regulating CD8+ TRM maintenance, heterogeneity, and protective and pathological functions are largely elusive. Here, we identify a population of CD8+ TRM cells that is maintained by major histocompatibility complex class I (MHC-I) signaling, and CD80 and CD86 costimulation after acute influenza infection. These TRM cells have both exhausted-like phenotypes and memory features and provide heterologous immunity against secondary infection. PD-L1 blockade after the resolution of primary infection promotes the rejuvenation of these exhausted-like TRM cells, restoring protective immunity at the cost of promoting postinfection inflammatory and fibrotic sequelae. Thus, PD-1 serves to limit the pathogenic capacity of exhausted-like TRM cells at the memory phase. Our data indicate that TRM cell exhaustion is the result of a tissue-specific cellular adaptation that balances fibrotic sequelae with protective immunity.


Subject(s)
B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Influenza A virus , Lung/immunology , Orthomyxoviridae Infections/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Female , Fibrosis , Humans , Lung/pathology , Lung Diseases, Interstitial/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic
15.
Crit Rev Immunol ; 38(2): 79-103, 2018.
Article in English | MEDLINE | ID: mdl-29953389

ABSTRACT

Tissue-resident memory T (TRM) cells have emerged as a major component of T cell biology. Recent investigations have greatly advanced our understanding of TRMs. Common features have been discovered to distinguish memory T cells residing in various mucosal and non-mucosal tissues from their circulating counterparts. Given that most organs and tissues contain a unique microenvironment, local signal-induced tissue-specific features are tightly associated with the differentiation, homeostasis, and protective functions of TRMs. Here, we discuss recent advances in the TRM field with a special emphasis on the interaction between local signals and TRMs in the context of individual tissue environment.


Subject(s)
Immunity, Mucosal/immunology , Immunologic Memory/immunology , Organ Specificity , T-Lymphocytes/immunology , Animals , Homeostasis , Humans
16.
J Immunol ; 198(2): 749-756, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27903738

ABSTRACT

Tissue-resident memory T (TRM) cells, a population of noncirculating memory T cells, are one of the essential components of immunological memory in both mouse and human. Although CD69+CD103+ TRM cells represent a major TRM cell population in barrier tissues including the mucosal surface and the skin, CD69+CD103- TRM cells dominate most nonbarrier tissues, such as the kidney. TGF-ß is required for the differentiation of CD69+CD103+ TRM cells in barrier tissues. However, the developmental control of CD69+CD103- TRM cells in nonbarrier tissues remains largely unknown and the involvement of TGF-ß signaling is less clear. In this study we demonstrated that TGF-ß promoted the formation of kidney-resident T cells via enhancing the tissue entry of effector T cells. Mechanistically, TGF-ß enhanced E- and P-selectin and inflammatory chemokine-mediated extravasation of effector T cells. Thus TGF-ß controls the first developmental checkpoint of TRM cell differentiation in nonbarrier tissues.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Kidney/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Flow Cytometry , Mice , Mice, Transgenic
17.
Biomed Res Int ; 2016: 1208476, 2016.
Article in English | MEDLINE | ID: mdl-26989676

ABSTRACT

The pathophysiological functions of cardiac histamine level and related histamine receptors during the development of chronic heart failure (CHF) were intensively investigated previously. However, the relevance of polymorphisms in histamine-related genes, such as HRH2, HRH3, DAO, and HNMT, with CHF remains largely neglected. This study herein aims to analyze the clinical associations of polymorphisms in those genes with CHF risk. A total of 333 unrelated Chinese Han CHF patients and 354 ethnicity-matched healthy controls were recruited and 11 single nucleotide polymorphisms (SNPs) were genotyped. We found that the HRH3 rs3787429 polymorphism was associated with CHF risk (p < 0.001). The T allele of rs3787429 exhibited protective effect against CHF under the dominant (ORs = 0.455; 95% CIs = 0.322-0.642) and additive models (ORs = 0.662; 95% CIs = 0.523-0.838), while, for SNPs in HRH2, DAO, and HNMT, no significant associations were observed in the present study. These findings for the first time screen out one SNP (rs3787429) of HRH3 gene that was significantly associated with CHF in Chinese Han population, which may be a novel biomarker for personal prevention and treatment of CHF and provides novel highlights for investigating the contribution of this disease.


Subject(s)
D-Amino-Acid Oxidase/genetics , Heart Failure/genetics , Histamine N-Methyltransferase/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine H3/genetics , Receptors, Histamine/genetics , Adult , Aged , Chronic Disease , Female , Genetic Association Studies , Genotype , Haplotypes , Heart Failure/pathology , Histamine/genetics , Humans , Male , Middle Aged , Receptors, Histamine H4
18.
PLoS One ; 10(11): e0143863, 2015.
Article in English | MEDLINE | ID: mdl-26606133

ABSTRACT

Staphylococcus aureus is one of the most frequently occurring hospital- and community-associated pathogenic bacteria featuring high morbidity and mortality. The occurrence of methicillin-resistant S. aureus (MRSA) has increased persistently over the years. Therefore, developing novel anti-MRSA drugs to circumvent drug resistance of S. aureus is highly important. Roemerine, an aporphine alkaloid, has previously been reported to exhibit antibacterial activity. The present study aimed to investigate whether roemerine can maintain these activities against S.aureus in vivo and further explore the underlying mechanism. We found that roemerine is effective in vitro against four S. aureus strains as well as in vivo against MRSA insepticemic BALB/c mice. Furthermore, roemerine was found to increase cell membrane permeability in a concentration-dependent manner. These findings suggest that roemerine may be developed as a promising compound for treating S. aureus, especially methicillin-resistant strains of these bacteria.


Subject(s)
Alkaloids/pharmacology , Cell Membrane Permeability/drug effects , Sepsis/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Alkaloids/administration & dosage , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Drugs, Chinese Herbal , Mice , Mice, Inbred BALB C , Sepsis/drug therapy , Sepsis/mortality , Staphylococcal Infections/drug therapy , Staphylococcal Infections/mortality
19.
Molecules ; 20(10): 17913-28, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26426004

ABSTRACT

Roemerine (RM) is an aporphine alkaloid isolated from the fresh rattan stem of Fibraurea recisa, and it has been demonstrated to have certain antifungal activity. This study aimed to investigate the antifungal activity of RM and the underlying mechanisms in Candida albicans (C. albicans). The in vitro antifungal activity of RM was evaluated by a series of experiments, including the XTT reduction assay, confocal laser scanning microscopy assay, scanning electron microscope assay. Results showed that 1 µg/mL RM inhibited biofilm formation significantly (p < 0.01) both in Spider medium and Lee's medium. In addition, RM could inhibit yeast-to-hyphae transition of C. albicans in a dose-dependent manner. The biofilm-specific and hypha-specific genes such as YWP1, SAP5, SAP6, HWP1, ECE1 were up-regulated and EFG1 was down-regulated after 8 µg/mL RM treatment. Furthermore, the toxicity of RM was investigated using C. elegans worms, three cancer cells and one normal cell. The date showed that RM had no significant toxicity. In conclusion, RM could inhibited the formation of C. albicans biofilm in vitro, but it had no fungicidal effect on planktonic C. albicans cells, and the anti-biofilm mechanism may be related to the cAMP pathway.


Subject(s)
Alkaloids/pharmacology , Antifungal Agents/pharmacology , Candida albicans/drug effects , Alkaloids/chemistry , Animals , Antifungal Agents/chemistry , Biofilms/drug effects , Caenorhabditis elegans/drug effects , Candida albicans/genetics , Gene Expression Regulation, Fungal/drug effects , Hyphae/drug effects , Microbial Sensitivity Tests , Plant Extracts/chemistry , Plant Extracts/pharmacology
20.
Proc Natl Acad Sci U S A ; 112(35): 11013-7, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26283373

ABSTRACT

The long-term maintenance of memory T cells is essential for successful vaccines. Both the quantity and the quality of the memory T-cell population must be maintained. The signals that control the maintenance of memory T cells remain incompletely identified. Here we used two genetic models to show that continuous transforming growth factor-ß signaling to antigen-specific T cells is required for the differentiation and maintenance of memory CD8(+) T cells. In addition, both infection-induced and microbiota-induced inflammation impact the phenotypic and functional identity of memory CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Proliferation , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...